NZ790341A - Inhibitors of short-chain dehydrogenase activity for promoting neurogenesis and inhibiting nerve cell death - Google Patents

Inhibitors of short-chain dehydrogenase activity for promoting neurogenesis and inhibiting nerve cell death Download PDF

Info

Publication number
NZ790341A
NZ790341A NZ790341A NZ79034117A NZ790341A NZ 790341 A NZ790341 A NZ 790341A NZ 790341 A NZ790341 A NZ 790341A NZ 79034117 A NZ79034117 A NZ 79034117A NZ 790341 A NZ790341 A NZ 790341A
Authority
NZ
New Zealand
Prior art keywords
alkyl
aryl
mmol
methyl
esi
Prior art date
Application number
NZ790341A
Inventor
Sanford Markowitz
Andrew Pieper
Joseph Ready
Original Assignee
Case Western Reserve University
University Of Iowa Research Foundation
University Of Texas Southwestern Medical Center
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 Case Western Reserve University, University Of Iowa Research Foundation, University Of Texas Southwestern Medical Center filed Critical Case Western Reserve University
Publication of NZ790341A publication Critical patent/NZ790341A/en

Links

Abstract

method of promoting neuroprotection in a subject from axonal degeneration, neuronal cell death, and/or glia cell damage after injury, augmenting neuronal signaling underlying learning and memory, stimulating neuronal regeneration after injury, and/or treating a disease, disorder, and/or condition of the nervous system in a subject in need thereof includes administering to the subject a therapeutically effective amount of a 15-PGDH inhibitor. of the nervous system in a subject in need thereof includes administering to the subject a therapeutically effective amount of a 15-PGDH inhibitor.

Description

INHIBITORS OF SHORT-CHAIN OGENASE ACTIVITY FOR PROMOTING NEUROGENESIS AND INHIBITING NERVE CELL DEATH D APPLICATIONS This application is a divisional application from New Zealand patent application no. 749870.
The disclosures of New Zealand patent application no. 749870 and corresponding international patent application no. , are incorporated herein by reference in their entirety. This application claims priority from U.S. Provisional Application Nos. 62/363,441, filed July 18, 2016 and 62/372,203 filed August 8, 2016, the subject matter of which are incorporated herein by reference in their entirety.
GOVERNMENT FUNDING This invention was made with government support under Grant No. P50CA150964 awarded by The National Institutes of Health. The United States government has n rights in the ion.
BACKGROUND Prostaglandins, via their specific G protein coupled receptors, have a variety of logical functions in the central nervous . The major prostaglandin, prostaglandin E2 (PGE2) can te receptor types EP1, 2, 3, and 4. Activation of EP2 and EP4 receptors can regulates adenylate cyclase and the generation of 3, 5′-cyclic adenosine monophosphate (cAMP), whereas the activation of EP1 and EP3 receptors can regulates Ca2+ signaling. EP1 and EP2 receptors are expressed in cultured neurons and lia as well as neurons of the cerebral cortex, striatum, and hippocampus.
Also, activation of the EP2 receptor by PGE2 is involved in long-term synaptic plasticity and ive on, as EP2−/− mice showed impaired hippocampal synaptogenesis. (Chemtob et al. Semin Perinatol. 1994 Feb; 18(1):23-9; Yang et al., J Neurochem.2009 Jan; :295-304). ing activation, different PGE2 ors can contribute or protect against N-methyl-D-aspartate (NMDA) neurotoxicity and ischemic stroke. For example, in a mouse model of focal cerebral ischemia, pretreatment with an EP2 receptor selective agonist was able to significantly decrease neurological deficits and deletion of EP2 receptors aggravated ic brain damage. (Ahmad et al., Exp Transl Stroke Med.2010 Jul 8; 2(1):12). Activation of the EP2 receptors with butaprost protected neurons from amyloid β-peptide neurotoxicity in vitro. (Echeverria et al., Eur J Neurosci.2005 Nov; 22(9):2199-206).
Several studies suggest that the mechanism by which PGE2 s neuroprotection is through EP2 or EP4 receptors, as they both increases cAMP, followed by a protein kinase A (PKA)- dependent pathway. (Echeverria et al. Eur J Neurosci.2005 Nov; 22(9):2199-206; McCullough et al., J Neurosci.2004 Jan 7; 24(1):257-68). stration of PGE2 has not been shown to be therapeutically useful against the EP2 receptor as the half- W0 2018f017582 life of PGE2 is less than 1 min. ing intravenous injection and approximately 30 sec. in the circulatory system. (Fitzpatrick et al., Prostaglandins. 1980 Jun; 19(6):917—31; Kimball et a1. Prostaglandins. 1980 Sep; 20(3):559-69).
Embodiments described herein relate generally to compositions and methods that promote the generation or the survival of neurons in the mammalian brain as well as to compositons and methods of treating diseases, ers, and/or conditions of the nervous system. As described in the Examples below, it was found that compounds that inhibit, reduce, and/or antagonize short-chain dehydrogenase activity, such as 15-PGDH inhibitors, can be used to increase PGE2 levels in the nervous system (e. g., brain) of a mammal. PGE2 elevates cyclic AMP via binding to EP2 and EP4 receptors, which are highly sed in the cerebral cortex, hippocampus, and striatum. Stimulation of these receptors with PGE2 by administration of a compound that inhibits, reduces, and/or antagonizes 15—PGDH acivity, such as with a 15—PGDH inhibitor described herein, can promote neuroprotection in a subject from axonal degeneration, neuronal cell death, and/or glia cell damage after injury, augment neuronal signaling underlying learning and , stimulate neuronal regeneration after injury, and/or treat diseases, disorders, and/or conditions of the nervous system.
In some embodiments, the e, disorder, and/or condition of the s system, which can be treated with the 15-PGDH inhibitors, can e at least one of a neurological disorder, a neuropsychiatric disorder, a neural injury, a neural toxicity disorder, a neuropathic pain, or a neural degenerative disorder.
For example, the neurological disorder can include at least one of traumatic or toxic injuries to peripheral or l nerves, spinal cord or brain, such as traumatic brain injury, stroke, al aneurism, and spinal cord injury. The neurological disorder can also include at least one of Alzheimer's e, dementias related to Alzheimer‘s e, Parkinson‘s, Lewy e body diseases, senile dementia, Huntington‘s disease, Gilles de Ia Tourette‘s syndrome, multiple sclerosis, amyotrophic l sclerosis, hereditary motor and sensory neuropathy, ic neuropathy, progressive supranuclear palsy, epilepsy, or Jakob— Creutzfieldt disease.
In some embodiments, the neural inj ury can be caused by or associated with at least one of epilepsy, cerebrovascular diseases, autoimmune diseases, sleep disorders, W0 2018/‘017582 autonomic disorders, urinary bladder disorders, abnormal metabolic states, disorders of the muscular system, infectious and parasitic es, neoplasms, endocrine diseases, nutritional and metabolic diseases, immunological diseases, diseases of the blood and blood—forming organs, mental disorders, diseases of the nervous system, diseases of the sense organs, diseases of the circulatory , diseases of the respiratory system, diseases of the digestive system, diseases of the genitourinary system, diseases of the skin and subcutaneous tissue, diseases of the musculoskeletal system and connective , congenital anomalies, or conditions originating in the perinatal period.
In certain embodiments, the H inhibitors can be administered to a subject or neurons of the subject to promote the survival, growth, development and/or function of the s, particularly, the central nervous system (CNS), brain, cerebral, and hippocampal neurons. In certain embodiments, the lS-PGDH inhibitors can be used ate hippocampal neurogenesis, for the treatment of neuropsychiatric and egenerative diseases, including (but not limited to) schizophrenia, major depression, bipolar disorder, normal aging, epilepsy, traumatic brain injury, post-traumatic stress disorder, Parkinson's e, Alzheimer's disease, Down syndrome, spinocerebellar ataxia, amyotrophic lateral sclerosis, gton's disease, stroke, radiation therapy, chronic stress, and abuse of neuro—active drugs, such as alcohol, opiates, methamphetamine, phencyclidine, and cocaine.
In some embodiments, the lS-PGDH inhibitors can be administered to a subject at an amount effective to increase prostaglandin levels in the nervous system (e.g., brain).
The H inhibitor can include a compound having formula (I): Y1—X1 If \R1 Y2 (I) wherein n is 0—2; Y1, Y2, and R1 are the same or different and are each selected from the group ting of en, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3—C20 aryl, heteroaryl, heterocycloalkenyl containing from 5—6 ring atoms (wherein W0 2018/‘017582 from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O) (C 1-C6 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1—C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5—C20 aryloxy, acyl (including C2—C24 alkylcarbonyl alky1) and C6-C20 rbonyl (—CO—ary1)), acyloxy (-O-acyl), C2—C24 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl —O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O-alkyl), C6-C20 arylcarbonato (-O-(CO)-O-ary1), carboxy ), carboxylato (-COO'), oyl (-(CO)-NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl (-(CO)-NH-aryl), thiocarbamoyl (-(CS)—NH2), carbamido (—NH-(CO)-NH2), cyano(-CN), isocyano (-N+C'), cyanato (-O-CN), isocyanato (-O-N+=C‘), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alky1), C6-C20 arylamido (-NH-(CO)-aryl), imino H where R is hydrogen, C1-C24 alkyl, C5—C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(ary1), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo OH), ato (-SOg-O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed "alkylthio"), arylsulfanyl (—S—ary1; also termed hio"), C1-C24 alkylsulfinyl (-(SO)-a1ky1), C5—C20 arylsulfinyl (-(SO)—ary1), C1—C24 alkylsulfonyl (-S02-alkyl), C5-C20 arylsulfonyl (-SOZ—ary1), amide (-SOz-NH2, —S02NY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (O')), phospho (—P02), ino (--PH2), polyalkylethers, phosphates, phosphate esters, groups orating amino acids or other moieties expected to bear positive or negative charge at physiological pH, combinations thereof, and wherein Y1 and Y2 may be linked to form a cyclic or polycyclic ring, n the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C—Rz, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)H10R’ (wherein n1=1, 2, or 3), CF3, CH2—CH2X, O-CHz- CH2X, CH2-CH2-CH2X, CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, W0 2018/‘017582 IOR’ (wherein nl=1, 2, or 3), CF3, CHz-CHZX, 2-CH2X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; X1 and X2 are independently N or C, and wherein when X1 and/or X2 are N, Y1 and/or Y2, respectively, are absent; Z1 is O, S, CRZ‘Rb or NRa, wherein Ra and Rb are ndently H or a C1_g alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically able salts thereof.
In other embodiments, the lS-PGDH inhibitor can include a compound having the following (V): 8 NS\R1 R? (V) wherein n is 0-2 X6 is independently is N or CRC R1, R6, R7, and RC are each independently selected from the group consisting of hydrogen, tuted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 l, C3-C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O)(C1-C6 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2—C24 loxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (--CO-alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (—O—acyl), C2-C24 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2—C24 alkylcarbonato (-O-(CO)-O-alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), y (— COOH), carboxylato (—COO'), carbamoyl (-(CO)-NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1—C24 alkyl)), arylcarbamoyl (-(CO)-NH-aryl), thiocarbamoyl (—(CS)—NH2), carbamido (-NH-(CO)-NH2), cyano(-CN), no (-N+C‘), o (-O-CN), isocyanato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (-—(CS)——H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2—C24 alkylamido W0 2018f017582 (-NH-(CO)-alkyl), C6-C20 arylamido CO)-ary1), imino H where R is hydrogen, C1—C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (-CR=N(alkyl), where R=hydrogen, alkyl, aryl, l, aralkyl, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo (—SOz—OH), sulfonato (—SOg—O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1—C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOg-aryl), sulfonamide (-SOz-NH2, -S02NY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (-P(O)(O')), phospho (—P02), phosphino (——PH2), polyalkylethers, phosphates, phosphate esters, groups orating amino acids or other moieties expected to bear positive or negative charge at physiological pH, combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or tituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or tituted heterocyclyl; U1 is N, C-RZ, or 4, n R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ in n1=1, 2, or 3), CF3, CHz—CHzX, O-CHz- CHZX, z—CHZX, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz-CHZX, CHz-CHz-CHZX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; and pharmaceutically acceptable salts thereof.
In some embodiments, R1 is selected from the group consisting of branched or linear alkyl including —(CH2)n1CH3 (n1=0-7), n2 wherein n2=0-6 and X is any of the following: CFyHZ (y + z = 3), CClsz (y + z = 3), OH, OAc, OMe, R71, 0R”, CN, N(R73)2, “A)m WR74 “3 ”4 (n3=0—5, m=1-5), and (114:0—5).
In other embodiments, R6 and R7 can each independently be one of the following: Rar s s s s 0 §_ Roll/ wa >,§_R11|: Rafi I/ I/IN/l/w/I§_ R1 l— E— \ “i” “/ >7 _ o 0 NR19 NR21 ll/ R18'—/ II/ RI14 “/0/R"\N/§ 18‘I/Nil—JE— I / E‘RI / \ \ \ R22ES>§—R23 “/NR24 NR26 R25I— l'\N/ l N / 7fNR>ZE: — Nl/OLN/E / ~\N/0> _E W ”‘1‘, N/o N/NR32 N/NR34 N/NR35 N/NR38 N/NR“° “ / NLN/>§_ 33L _R35k WK RI /§ /l' / g— NW Wm;Mu/ \ \ /NR42 N/NR43 NR45’N R46 NR47 \ R39\ TI I I \ T)E-RML N / N / I WWI—(N N\N / \ / M, N\J\~'\ \ J‘R‘” \ \ N\ N ORGO OR51 \N \ R58{j\ R56:_K2353R57T: —3_R59 J AJrR62 /, 3‘53\N / )1?“ \ = R53 o 0 Jk R69 R55 fi fi a, 0262a (A R66— _s w _§_ _7 ,R68 §_ _ /.7 “\* each R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39 R40 R41 R42 R43 R44 R45 R46 R47 R48 R49 R50 R51 R52 R53 R54 R55 R56 R57 R58 R59 R60 R61 R62 R63 R64 R65 R66 R67 R68 R69 W0 2018/‘017582 R70, R71~ Rn~ R73, and R74, are the same or different and are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2—C24 alkynyl, C3—C20 aryl, heterocycloalkenyl containing from 5-6 ring atoms, (wherein from 1—3 of the ring atoms is independently selected from N, NH, 6 alkyl), NC(O)(C1- C6 alkyl), 0, and S), heteroaryl or heterocyclyl containing from 5-14 ring atoms, (wherein from 1-6 of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, silyl, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2—C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (--CO- alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (-O-acyl), C2-C24 alkoxycarbonyl (- (CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O- alkyl), C6—C20 arylcarbonato (-O-(CO)-O-aryl), carboxy ), ylato (-COO'), oyl (-(CO)--NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl (-(CO)-NH-ary1), thiocarbamoyl -NH2), carbamido (-NH-(CO)-NH2), cyano(-CN), isocyano (—N+C‘), cyanato (—O-CN), isocyanato (-O-N+=C'), isothiocyanato (—S—CN), azido (-N=N+=N'), fonnyl )--H), thioformyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alkyl), C6-C20 arylamido (-NH-(CO)-aryl), sulfanamido (—S02N(R)2 where R is independently H, alkyl, aryl or heteroaryl), imino (- CR=NH where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6—C24 aralkyl, etc.), alkylimino (—CR=N(alkyl), where R=hydrogen, alkyl, aryl, l, aralkyl, etc.), arylimino (- CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (—NO), sulfo OH), sulfonato (-SOz-O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed "alkylthio"), arylsulfanyl (—S—aryl; also termed ”arylthio”), C1-C24 ulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl -aryl), C1-C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (—SOz-aryl), amide (-SOz-NH2, -SOZNY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (-P(O)(O')), phospho (—P02), phosphino ), polyalkyl ethers (-[(CH2)nO]m), phosphates, phosphate esters [- OP(O)(OR)2 where R = H, methyl or other , groups orating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations thereof, and pharmaceutically acceptable salts thereof.
In some embodiments, the 15-PGDH inhibitor can inhibit the enzymatic activity of recombinant 15—PGDH at an IC50 of less than 1 uM, or preferably at an IC50 of less than 250 nM, or more ably at an IC50 of less than 50 nM, or more preferably at an IC50 of WO 2018f017582 less than 10 nM, or more preferably at an IC50 of less than 5 nM at a recombinant 15-PGDH concentration of about 5 nM to about 10 nM.
BRIEF DESCRIPTION OF THE GS Fig. 1 illustrates a plot showing the pharmokinetics of the l5—PGDH inhibitor (+) 91 when administered at lOmg/kg by intraperitoneal injection into female CD-1 mice and then measured at mg/ml in plasma or at mg/gm of wet tissue weight in brain.
Fig. 2 illustrates a schematic diagram and graphs showing the concentration of of prostaglandin E2 (PGE2) in 3 regions of the brain, as averaged from 3 mice, samples 3 hours after intraperitoneal injection with vehicle (VE) or with (+) SW033291 at 2.5 mg/kg.
Figs. 3(A-B) illustrate a plot and graphs showing impact of administering (+) SW033291 on mouse performance in learning and memory ing traumatic brain injury.
Fig. 4 is an image showing detection of 15-PGDH mRNA expression in neurons of mouse hippocampus.
Figs. 5(A—B) rate plots showing showing pharmacokinetics of the 15— PGDH inhibitor (+) 415 when administered at 2.5 and at 25 mg/kg by intraperitoneal injection into female C57BL/7 mice and then measured at mg/ml in plasma or at mg/gm of wet tissue weight in brain.
Figs. 6(A-C) rate graphs showing 15-PGDH activity in the cortex (A), cerebellum (B), and pons and medulla (C) of mouse brain following IP injection of 15-PGDH inhibitor (+) SW033291 at 2.5 mpk.
Fig. 7 illustrates a graph showing PGE2 levels in rat brain cortex following IP injection of (+) SW033291 at 2.5, 5.0, and 10.0 mg/kg.
Figs. 8(A-B) illustrate Western blots and graphs showing levels of 15-PGDH in brain tissue of ts with Alzheimer’s disease relative to age d control subjects t Alzheimer’s disease.
DETAILED DESCRIPTION For convenience, certain terms employed in the specification, examples, and appended claims are collected here. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly tood by one of ordinary skill in the art to which this application belongs.
W0 2018/‘017582 The articles "a" and "an" are used herein to refer to one or to more than one (i.€., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
The terms "comprise, H Hcomprising, H H'include, H H'including,” "have," and ”having" are used in the inclusive, open sense, meaning that additional elements may be included. The terms "such as”, ”e. g. ”, as used herein are non-limiting and are for illustrative purposes only. "Including” and ding but not limited to” are used hangeably.
The term ”or” as used herein should be tood to mean ”and/or", unless the context clearly indicates otherwise.
As used herein, the term ”about" or ”approximately” refers to a quantity, level, value, number, frequency, tage, dimension, size, amount, weight or length that varies by as much as 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% to a nce quantity, level, value, number, frequency, percentage, ion, size, amount, weight or length. In one embodiment, the term "about” or "approximately” refers a range of quantity, level, value, , frequency, percentage, ion, size, amount, weight or length i 15%, i 10%, i 9%, i 8%, i 7%, i 6%, i 5%, i 4%, i 3%, i 2%, or i 1% about a reference quantity, level, value, number, frequency, tage, dimension, size, amount, weight or length.
It will be noted that the structure of some of the compounds of the application include asymmetric (chiral) carbon or sulfur atoms. It is to be understood accordingly that the isomers arising from such asymmetry are included herein, unless indicated otherwise.
Such s can be obtained in substantially pure form by classical tion techniques and by stereochemically controlled synthesis. The compounds of this application may exist in isomeric form, therefore can be produced as individual stereoisomers or as mixtures.
The term ”isomerism” means compounds that have identical molecular formulae but that differ in the nature or the ce of bonding of their atoms or in the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers". Stereoisomers that are not mirror images of one another are termed ”diastereoisomers", and stereoisomers that are non-superimposable mirror images are termed "enantiomers", or sometimes optical isomers. A carbon atom bonded to four nonidentical substituents is termed a "chiral center" whereas a sulfur bound to three or four different substitutents, e. g., sulfoxides or sulfinimides, is likewise termed a “chiral center”.
W0 2018/‘017582 The term "chiral isomer" means a nd with at least one chiral center. It has two enantiomeric forms of opposite chirality and may exist either as an individual enantiomer or as a mixture of enantiomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a ic mixture". A nd that has more than one chiral center has 2n-1 enantiomeric pairs, where n is the number of chiral centers. Compounds with more than one chiral center may exist as either an individual diastereomer or as a mixture of reomers, termed a ”diastereomeric mixture".
When one chiral center is present, a stereoisomer may be characterized by the absolute uration (R or S) of that chiral center. atively, when one or more chiral centers are present, a stereoisomer may be characterized as (+) or (-). Absolute configuration refers to the arrangement in space of the substituents ed to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al, Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511; Cahn et al., Angew. Chem. 1966, 78, 413; Cahn and Ingold, J Chem. Soc. 1951 (London), 612; Cahn et al., Experientia 1956, 12, 81; Cahn, J., Chem. Educ. 1964, 41, 116).
The term der" refers to any disorder, disease, or condition that may benefit from an agent that es neuroprotection, augments neuronal signaling, and/or stimulated neuronal regeneration after injury.
The term "geometric Isomers” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold- Prelog rules. Further, the structures and other compounds discussed in this application include all atropic isomers thereof.
The term ”atropic isomers” are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. c isomers owe their nce to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques, it has been possible to separate mixtures of two atropic isomers in select cases.
The terms "crystal polymorphs" or ”polymorphs" or "crystal forms" means crystal structures in which a compound (or salt or solvate f) can crystallize in different W0 2018/‘017582 crystal packing arrangements, all of which have the same elemental composition. Different crystal forms usually have different X-ray diffraction patterns, infrared a], melting points, density ss, crystal shape, optical and electrical properties, ity and solubility. tallization solvent, rate of crystallization, storage temperature, and other factors may cause one crystal form to dominate. Crystal polymorphs of the compounds can be prepared by crystallization under different conditions.
The term "derivative” refers to compounds that have a common core structure, and are tuted with various groups as described herein.
The term "bioisostere" refers to a compound resulting from the exchange of an atom or of a group of atoms with another, broadly similar, atom or group of atoms. The objective of a bioisosteric replacement is to create a new compound with similar biological properties to the parent compound. The bioisosteric replacement may be physicochemically or topologically based. Examples of carboxylic acid bioisosteres include acyl sulfonimides, oles, sulfonates, and phosphonates. See, e. g., Patani and LaVoie, Chem. Rev. 96, 3147— 3176 (1996).
The phrases "parenteral stration" and "administered parenterally" are art- recognized terms, and e modes of administration other than enteral and topical administration, such as ions, and include, t limitation, intravenous, intramuscular, intrapleural, intravascular, intrapericardial, intraarterial, intrathecal, apsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intra-articular, sular, subarachnoid, intraspinal and intrastemal injection and infusion.
The term "treating” is art-recognized and includes inhibiting a disease, disorder or condition in a subject, e. g., impeding its progress; and ing the disease, disorder or condition, e.g., causing regression of the disease, disorder and/or condition. ng the disease or condition includes ameliorating at least one symptom of the particular disease or condition, even if the underlying pathophysiology is not affected.
The term "preventing” is art-recognized and includes stopping a disease, disorder or ion from occurring in a subject, which may be posed to the disease, disorder and/or condition but has not yet been diagnosed as having it. Preventing a ion related to a disease includes stopping the condition from occurring after the disease has been diagnosed but before the condition has been diagnosed.
W0 2018f017582 The term "pharmaceutical composition” refers to a formulation containing the disclosed compounds in a form suitable for administration to a subject. In a preferred ment, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol r, or a vial. The quantity of active ingredient (e.g., a formulation of the disclosed compound or salts f) in a unit dose of ition is an effective amount and is varied ing to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of stration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, eritoneal, intranasal, inhalational, and the like. Dosage forms for the topical or transdermal administration of a compound described herein includes powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, nebulized nds, and nts. In a preferred embodiment, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.
The term ”flash dose" refers to compound formulations that are rapidly dispersing dosage forms.
The term "immediate release” is defined as a release of compound from a dosage form in a relatively brief period of time, lly up to about 60 minutes. The term ”modified release" is defined to include delayed release, extended release, and pulsed release.
The term "pulsed release” is defined as a series of releases of drug from a dosage form. The term ”sustained release” or ded e” is defined as continuous release of a compound from a dosage form over a prolonged period.
The phrase ”pharmaceutically acceptable” is art-recognized. In certain embodiments, the term includes compositions, polymers and other materials and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or cation, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically able r" is art—recognized, and includes, for example, pharmaceutically acceptable materials, compositions or vehicles, such W0 2018/‘017582 as a liquid or solid filler, t, excipient, t or encapsulating material, involved in carrying or orting any subject composition from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be ”acceptable" in the sense of being ible with the other ingredients of a subject composition and not injurious to the patient. In certain embodiments, a pharmaceutically acceptable carrier is rogenic.
Some examples of materials which may serve as pharmaceutically acceptable carriers include: ( 1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl ose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, wer oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen—free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl l; (20) ate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical ations.
The compounds of the application are capable of further forming salts. All of these forms are also contemplated herein.
"Pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological ty of the parent compound. For example, the salt can be an acid addition salt. One embodiment of an acid addition salt is a hydrochloride salt. The pharmaceutically acceptable salts can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a iometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile being preferred. Lists of salts are found in Remington's Pharmaceutical Sciences, 18th ed. (Mack Publishing Company, 1990).
The compounds bed herein can also be prepared as esters, for example pharmaceutically acceptable esters. For example, a carboxylic acid function group in a nd can be converted to its corresponding ester, e. g., a methyl, ethyl, or other ester.
W0 2018/‘017582 Also, an alcohol group in a compound can be converted to its corresponding ester, e. g., an acetate, propionate, or other ester.
The compounds described herein can also be prepared as prodrugs, for example pharmaceutically acceptable gs. The terms ”pro-drug” and ug" are used interchangeably herein and refer to any compound, which releases an active parent drug in viva. Since prodrugs are known to e numerous ble qualities of pharmaceuticals (e. g., solubility, bioavailability, manufacturing, etc.) the compounds can be delivered in prodrug form. Thus, the compounds described herein are intended to cover prodrugs of the presently claimed compounds, methods of delivering the same and itions containing the same. "Prodrugs" are ed to include any covalently bonded carriers that e an active parent drug in vivo when such prodrug is administered to a subject. Prodrugs are prepared by modifying onal groups present in the compound in such a way that the modifications are d, either in routine manipulation or in vivo, to the parent compound.
Prodrugs include compounds wherein a hydroxy, amino, sulfhydryl, carboxy, or carbonyl group is bonded to any group that may be cleaved in vivo to form a free hydroxyl, free amino, free sulfhydryl, free carboxy or free yl group, respectively. Prodrugs can also include a precursor (forerunner) of a compound bed herein that undergoes chemical sion by metabolic processes before becoming an active or more active pharmacological agent or active compound described herein.
Examples of prodrugs include, but are not limited to, esters (e. g., acetate, dialkylaminoacetates, formates, phosphates, sulfates, and benzoate derivatives) and carbamates (e.g., N,N-dimethylaminocarbonyl) of hydroxy functional groups, ester groups (e. g., ethyl , morpholinoethanol esters) of carboxyl functional groups, N-acyl derivatives (e.g., N-acetyl) N-Mannich bases, Schiff bases and enaminones of amino onal groups, oximes, acetals, ketals and enol esters of ketone and aldehyde functional groups in compounds, and the like, as well as sulfides that are oxidized to form sulfoxides or sulfones..
The term "protecting group" refers to a grouping of atoms that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity. es of protecting groups can be found in Green and Wuts, Protective Groups in Organic Chemistry, (Wiley, 2.sup.nd ed. 1991); Harrison and Harrison et al., Compendium of Synthetic Organic W0 2018f017582 Methods, Vols. 1-8 (John Wiley and Sons, 1971-1996); and Kocienski, Protecting Groups, (Verlag, 3rd ed. 2003).
The term "amine ting group” is intended to mean a functional group that converts an amine, amide, or other nitrogen-containing moiety into a different chemical group that is substantially inert to the conditions of a particular chemical reaction. Amine protecting groups are preferably removed easily and selectively in good yield under conditions that do not affect other functional groups of the le. Examples of amine protecting groups include, but are not limited to, formyl, acetyl, benzyl, t—butyldimethylsilyl, t-butyldiphenylsilyl, t-butyloxycarbonyl (Boc), p-methoxybenzyl, methoxymethyl, tosyl, trifluoroacetyl, trimethylsilyl (TMS), fluorenyl-methyloxycarbonyl, 2-trimethylsilyl- ethyoxycarbonyl, 1-methyl(4-biphenylyl) ethoxycarbonyl, allyloxycarbonyl, oxycarbonyl (CBZ), ethylsilyl-ethanesulfonyl (SES), trityl and substituted trityl groups, renylmethyloxycarbonyl (FMOC), nitro-veratryloxycarbonyl (NVOC), and the like. Those of skill in the art can identify other suitable amine protecting groups. entative hydroxy protecting groups include those where the hydroxy group is either acylated or alkylated such as benzyl, and trityl ethers as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers and allyl ethers.
Additionally, the salts of the compounds described herein, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
Non-limiting examples of hydrates include monohydrates, dihydrates, etc. Nonlimiting examples of solvates e ethanol solvates, acetone solvates, etc.
The term ”solvates” means solvent addition forms that contain either stoichiometric or non-stoichiometric amounts of solvent. Some nds have a tendency to trap a fixed molar ratio of t molecules in the crystalline solid state, thus forming a solvate. If the t is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H20, such combination being able to form one or more hydrate.
The compounds, salts and gs described herein can exist in several tautomeric forms, including the enol and imine form, and the keto and enamine form and geometric isomers and es thereof. Tautomers exist as mixtures of a tautomeric set in on. In solid form, usually one tautomer predominates. Even though one tautomer may W0 2018/‘017582 be described, the present application includes all tautomers of the present compounds. A tautomer is one of two or more structural isomers that exist in equilibrium and are y converted from one isomeric form to another. This reaction results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. In solutions where tautomerization is possible, a al equilibrium of the tautomers will be d.
The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. The concept of ers that are interconvertable by tautomerizations is called tautomerism.
Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. erizations can be catalyzed by: Base: 1. deprotonation; 2. formation of a delocalized anion (e.g., an enolate); 3. protonation at a different position of the anion; Acid: 1. protonation; 2. formation of a delocalized cation; 3. deprotonation at a different position adjacent to the .
The term "analogue" refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a ent element or in the presence of a particular functional group, or the replacement of one functional group by r functional group). Thus, an ue is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.
A "patient,” ”subject,” or ”host” to be treated by the subject method may mean either a human or non-human animal, such as a mammal, a fish, a bird, a reptile, or an amphibian. Thus, the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or . The term does not denote a ular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. In one , the subject is a mammal.
A patient refers to a subject afflicted with a disease or disorder.
The terms "prophylactic” or “therapeutic” treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical station of the unwanted condition ((3. g., e or other ed state of the host animal) then the treatment is prophylactic, i.€., it protects the host W0 2018/‘017582 against developing the unwanted condition, whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic (126., it is ed to diminish, ameliorate, or stabilize the ng unwanted condition or side effects f).
The terms "therapeutic agent”, ”drug”, ”medicament” and ”bioactive substance" are art-recognized and include molecules and other agents that are biologically, physiologically, or pharmacologically active substances that act locally or systemically in a t or subject to treat a e or condition. The terms include without limitation pharmaceutically acceptable salts thereof and prodrugs. Such agents may be , basic, or salts; they may be neutral molecules, polar molecules, or molecular xes capable of hydrogen bonding; they may be prodrugs in the form of ethers, esters, amides and the like that are biologically activated when administered into a patient or subject.
The phrase ”therapeutically effective ” or “pharmaceutically effective amount” is an art-recognized term. In n embodiments, the term refers to an amount of a therapeutic agent that produces some desired effect at a reasonable benefitfrisk ratio applicable to any medical treatment. In n ments, the term refers to that amount necessary or sufficient to eliminate, reduce or maintain a target of a particular therapeutic regimen. The effective amount may vary depending on such factors as the disease or condition being treated, the particular ed constructs being administered, the size of the subject or the severity of the disease or condition. One of ordinary skill in the art may empirically determine the effective amount of a particular compound without necessitating undue experimentation. In certain embodiments, a therapeutically effective amount of a therapeutic agent for in viva use will likely depend on a number of factors, including: the rate of release of an agent from a polymer matrix, which will depend in part on the chemical and physical characteristics of the polymer; the identity of the agent; the mode and method of administration; and any other materials incorporated in the polymer matrix in addition to the agent.
The term ”ED50" is art-recognized. In certain ments, ED50 means the dose of a drug, which produces 50% of its maximum response or effect, or alternatively, the dose, which produces a pre-determined response in 50% of test subjects or preparations. The term "LD50" is art-recognized. In certain embodiments, LD50 means the dose of a drug, which is lethal in 50% of test subjects. The term ”therapeutic index" is an art—recognized term, which refers to the therapeutic index of a drug, defined as LD50/ED50.
W0 2018/‘017582 The terms "IC50," or “half maximal inhibitory concentration” is intended to refer to the concentration of a substance (e. g., a compound or a drug) that is required for 50% inhibition of a biological process, or component of a process, including a protein, subunit, organelle, cleoprotein, etc.
With respect to any chemical compounds, the present application is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, es of hydrogen include tritium and deuterium, and isotopes of carbon include C-13 and C-l4.
When a bond to a tuent is shown to cross a bond connecting two atoms in a ring, then such substituent can be bonded to any atom in the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent can be bonded via any atom in such substituent. ations of tuents and/or variables are permissible, but only if such combinations result in stable compounds.
When an atom or a chemical moiety is followed by a subscripted numeric range (6. g., C1_6), it is meant to ass each number within the range as well as all intermediate ranges. For example, "C1_6 alkyl" is meant to e alkyl groups with l, 2, 3, 4, 5, 6, 1-6, 1- , 1-4, 1—3, 1-2, 2-6, 2-5, 2-4, 2-3, 3-6, 3-5, 3-4, 4-6, 4-5, and 5-6 carbons.
The term "alkyl" is intended to include both branched (e. g., isopropyl, tert-butyl, isobutyl), ht—chain e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl), and cycloalkyl (e. g., alicyclic) groups (e. g., cyclopropyl, cyclopentyl, exyl, cycloheptyl, cyclooctyl), alkyl substituted lkyl , and cycloalkyl substituted alkyl groups. Such aliphatic arbon groups have a specified number of carbon atoms. For example, C1_6 alkyl is intended to include C1, C2, C3, C4, C5, and C6 alkyl groups. As used herein, "lower alkyl” refers to alkyl groups having from 1 to 6 carbon atoms in the backbone of the carbon chain. "Alkyl" further includes alkyl groups that have oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more hydrocarbon backbone carbon atoms. In certain embodiments, a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e. g., C1-C6 for straight chain, C3-C6 for branched chain), for example four or fewer. Likewise, certain cycloalkyls have from three to eight carbon atoms in their ring structure, such as five or six carbons in the ring structure.
W0 2018/‘017582 The term "substituted ” refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, rbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino ding alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, io, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic . Cycloalkyls can be further substituted, e.g., with the substituents described above. An aryl” or an "aralkyl" moiety is an alkyl substituted with an aryl (e. g., phenylmethyl (benzyl)). If not otherwise indicated, the terms "alkyl" and "lower alkyl” include linear, branched, cyclic, unsubstituted, substituted, and/or heteroatom-containing alkyl or lower alkyl, tively.
The term "alkenyl" refers to a , branched or cyclic hydrocarbon group of 2 to about 24 carbon atoms ning at least one double bond, such as ethenyl, n—propenyl, isopropenyl, n—butenyl, enyl, octenyl, decenyl, tetradecenyl, hexadecenyl, eicosenyl, tetracosenyl, cyclopentenyl, cyclohexenyl, cyclooctenyl, and the like. Generally, although again not necessarily, alkenyl groups can contain 2 to about 18 carbon atoms, and more particularly 2 to 12 carbon atoms. The term ”lower alkenyl” refers to an alkenyl group of 2 to 6 carbon atoms, and the specific term ”cycloalkenyl” intends a cyclic alkenyl group, preferably having 5 to 8 carbon atoms. The term ”substituted alkenyl” refers to l substituted with one or more substituent groups, and the terms ”heteroatom—containing alkenyl" and "heteroalkenyl" refer to alkenyl or heterocycloalkenyl (e. g., heterocylcohexenyl) in which at least one carbon atom is replaced with a atom. If not otherwise indicated, the terms "alkenyl" and "lower alkenyl” include linear, branched, cyclic, unsubstituted, substituted, and/or heteroatom-containing alkenyl and lower alkenyl, respectively.
The term "alkynyl” refers to a linear or branched hydrocarbon group of 2 to 24 carbon atoms containing at least one triple bond, such as ethynyl, n-propynyl, and the like.
Generally, gh again not necessarily, alkynyl groups can contain 2 to about 18 carbon atoms, and more particularly can contain 2 to 12 carbon atoms. The term "lower alkynyl" W0 2018/‘017582 intends an alkynyl group of 2 to 6 carbon atoms. The term "substituted alkynyl" refers to alkynyl substituted with one or more substituent groups, and the terms ”heteroatom—containing alkynyl" and "heteroalkynyl" refer to alkynyl in which at least one carbon atom is replaced with a heteroatom. If not otherwise ted, the terms "alkynyl" and ”lower alkynyl" include linear, branched, unsubstituted, substituted, and/or heteroatom- ning alkynyl and lower l, respectively.
The terms "alkyl”, ”alkenyl”, and ”alkynyl” are intended to include moieties which are diradicals, i.e., having two points of attachment. A nonlimiting example of such an alkyl moiety that is a diradical is --CH2CH2--, i.e., a C2 alkyl group that is covalently bonded via each terminal carbon atom to the remainder of the molecule.
The term ”alkoxy” refers to an alkyl group bound through a single, al ether e; that is, an "alkoxy" group may be represented as --O-alkyl where alkyl is as defined above. A "lower " group intends an alkoxy group containing 1 to 6 carbon atoms, and includes, for example, y, ethoxy, n-propoxy, isopropoxy, t—butyloxy, etc. red substituents identified as "C1-C6 alkoxy" or "lower alkoxy" herein contain 1 to 3 carbon atoms, and particularly red such substituents contain 1 or 2 carbon atoms (i.e., methoxy and ethoxy).
The term "aryl" refers to an aromatic substituent containing a single aromatic ring or multiple aromatic rings that are fused together, directly linked, or indirectly linked (such that the different aromatic rings are bound to a common group such as a methylene or ethylene moiety). Aryl groups can contain 5 to 20 carbon atoms, and particularly preferred aryl groups can contain 5 to 14 carbon atoms. Examples of aryl groups include benzene, phenyl, pyrrole, furan, thiophene, thiazole, isothiazole, imidazole, triazole, tetrazole, pyrazole, e, isooxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
Furthermore, the term ”aryl” includes multicyclic aryl , e. g., tricyclic, bicyclic, e. g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl, ine, isoquinoline, napthridine, indole, benzofuran, purine, benzofuran, deazapurine, or indolizine. Those aryl groups having heteroatoms in the ring structure may also be referred to as ”aryl heterocycles", "heterocycles," "heteroaryls" or "heteroaromatics”. The aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for e, halogen, yl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, W0 2018/‘017582 aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminocarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino ding alkylamino, dialkylamino, arylamino, diaryl amino, and a1 kylaryl amino), acylamino (including alkylcarbonylamino, rbonylamino, oyl and ureido), o, imino, dryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or aromatic . Aryl groups can also be fused or bridged with lic or heterocyclic rings, which are not aromatic so as to form a multicyclic system (e. g., tetralin, methylenedioxyphenyl). If not otherwise indicated, the term ”aryl" includes unsubstituted, substituted, and/or heteroatom—containing ic substituents.
The term ”alkaryl" refers to an aryl group with an alkyl substituent, and the term "aralkyl" refers to an alkyl group with an aryl tuent, wherein "aryl" and "alkyl" are as defined above. Exemplary aralkyl groups contain 6 to 24 carbon atoms, and particularly preferred aralkyl groups contain 6 to 16 carbon atoms. Examples of aralkyl groups include, without limitation, benzyl, 2-phenyl-ethyl, yl-propyl, yl—butyl, 5—phenyl—pentyl, 4-phenylcyclohexyl, 4-benzylcyclohexyl, 4-phenylcyclohexylmethyl, 4-benzylcyclohexylmethyl, and the like. Alkaryl groups include, for example, p-methylphenyl, 2,4-dimethylphenyl, p-cyclohexylphenyl, 2,7-dimethylnaphthy1, 7-cyclooctylnaphthyl, 3-ethyl-cyclopenta-1,4-diene, and the like.
The terms ”heterocyclyl” or ”heterocyclic group” include closed ring structures, e. g., 3- to 10-, or 4- to 7-membered rings, which include one or more heteroatoms.
”Heteroatom" includes atoms of any t other than carbon or hydrogen. Examples of heteroatoms include nitrogen, oxygen, sulfur and phosphorus.
Heterocyclyl groups can be saturated or unsaturated and include pyrrolidine, oxolane, thiolane, piperidine, piperazine, morpholine, lactones, lactams, such as azetidinones and pyrrolidinones, sultams, and sultones. Heterocyclic groups such as pyrrole and furan can have aromatic character. They include fused ring structures, such as quinoline and isoquinoline. Other examples of heterocyclic groups include ne and purine. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, W0 2018/‘017582 alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, l, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including arbonylamino, arylcarbonylamino, oyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, or an aromatic or heteroaromatic moiety. Heterocyclic groups can also be substituted at one or more tuent atoms with, for example, a lower alkyl, a lower alkenyl, a lower alkoxy, a lower hio, a lower alkylamino, a lower alkylcarboxyl, a nitro, a hydroxyl, --CF3, or --CN, or the like.
The term ”halo” or ”halogen” refers to fluoro, chloro, bromo, and iodo.
”Counterion" is used to ent a small, negatively d species such as fluoride, chloride, bromide, iodide, hydroxide, acetate, and sulfate. The term sulfoxide refers to a sulfur attached to 2 ent carbon atoms and one oxygen and the 8-0 bond can be graphically represented with a double bond (S=O), a single bond without charges (5-0) or a single bond with charges [S(+)-O(-)].
The terms ”substituted" as in ituted alkyl,” ”substituted aryl," and the like, as alluded to in some of the aforementioned tions, is meant that in the alkyl, aryl, or other moiety, at least one hydrogen atom bound to a carbon (or other) atom is replaced with one or more non—hydrogen substituents. Examples of such substituents include, without limitation: functional groups such as halo, hydroxyl, silyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (-CO-alkyl) and C6-C20 arylcarbonyl ryl)), acyloxy (-O-acyl), C2-C24 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)—O—alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato (-COO-), carbamoyl (-(CO)-NH2), mono-(Cl-C24 -substituted carbamoyl (-(CO)- NH(C1-C24 alky1)), di-(Cl-C4 alkyl)-substituted carbamoyl (-(CO)--N(C1-C24 alky1)2), mono-substituted arylcarbamoyl (-(CO)-NH—ary1), thiocarbamoyl (-(CS)-NH2), ido (-NH-(CO)-NH2), -CN), isocyano (-N+C'), cyanato (-O--CN), isocyanato (-ON+C’), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (-(CO)--H), thioformyl (-(CS)-H), amino (—NH2), mono— and di-(Cl-C24 alkyl)-substituted amino, mono— and di—(Cs—Czo aryl)— tuted amino, C2-C24 alkylamido (-NH—(CO)-alkyl), C6-C20 arylamido (—NH—(CO)-aryl), W0 2018/‘017582 imino H where R=hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (-—CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), arylimino (-CR=N(aryl), where ogen, alkyl, aryl, alkaryl, etc.), nitro (—N02), nitroso (—NO), sulfo (—S02 —OH), sulfonato (-S02-O'), C1-C24 alkylsulfanyl kyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed ”arylthio”), C1-C24 alkylsulfinyl )-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-S02 -aryl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (-P(O)(O‘)), o (-P02), and phosphino (-PH2); and the hydrocarbyl moieties C1-C24 alkyl, C2-C24 alkenyl, C2- C24 alkynyl, C5-C20 aryl, C6-C24 alkaryl, and C6-C24 aralkyl.
In on, the entioned functional groups may, if a particular group permits, be further substituted with one or more additional functional groups or with one or more hydrocarbyl moieties such as those specifically enumerated above. Analogously, the above-mentioned hydrocarbyl moieties may be further substituted with one or more functional groups or additional hydrocarbyl moieties such as those specifically enumerated.
When the term "substituted” s prior to a list of possible substituted groups, it is intended that the term apply to every member of that group. For example, the phrase ”substituted alkyl, alkenyl, and aryl" is to be interpreted as ”substituted alkyl, substituted alkenyl, and substituted aryl.” Analogously, when the term ”heteroatom— containing" appears prior to a list of possible heteroatom—containing groups, it is ed that the term apply to every member of that group. For example, the phrase "heteroatom- containing alkyl, alkenyl, and aryl” is to be interpreted as ”heteroatom-containing alkyl, substituted alkenyl, and substituted aryl. nal" or ”optionally” means that the uently described circumstance may or may not occur, so that the description includes instances where the circumstance occurs and instances where it does not. For example, the phrase ”optionally substituted” means that a non-hydrogen tuent may or may not be present on a given atom, and, thus, the description includes ures wherein a non-hydrogen substituent is present and structures wherein a non—hydrogen substituent is not present.
The terms "stable compound” and "stable ure" are meant to indicate a compound that is sufficiently robust to survive isolation, and as appropriate, purification from a reaction mixture, and formulation into an efficacious therapeutic agent.
W0 2018f017582 The terms "free compound” is used herein to describe a compound in the unbound state.
Throughout the description, where itions are described as having, ing, or comprising, specific components, it is contemplated that itions also consist essentially of, or consist of, the recited components. Similarly, where methods or processes are described as having, including, or comprising specific process steps, the ses also t essentially of, or consist of, the recited processing steps. Further, it should be understood that the order of steps or order for performing certain actions is rial so long as the compositions and methods bed herein remains operable. er, two or more steps or actions can be conducted simultaneously.
The term ”small le” is an art-recognized term. In certain embodiments, this term refers to a molecule, which has a molecular weight of less than about 2000 amu, or less than about 1000 amu, and even less than about 500 amu.
All percentages and ratios used herein, unless otherwise indicated, are by weight.
The terms " gene expression" or "protein expression" includes any information pertaining to the amount of gene transcript or protein present in a sample, as well as information about the rate at which genes or proteins are ed or are accumulating or being degraded (e. g., reporter gene data, data from nuclear runoff experiments, pulse-chase data etc.). Certain kinds of data might be Viewed as relating to both gene and protein expression. For example, protein levels in a cell are ive of the level of protein as well as the level of transcription, and such data is intended to be included by the phrase gene or protein expression information”. Such information may be given in the form of amounts per cell, s relative to a control gene or protein, in unitless measures, etc.; the term ”information" is not to be limited to any particular means of representation and is intended to mean any representation that provides relevant information. The term ”expression levels” refers to a quantity reflected in or derivable from the gene or n expression data, whether the data is directed to gene transcript accumulation or protein accumulation or protein synthesis rates, etc.
The terms "healthy" and "normal” are used interchangeably herein to refer to a subject or particular cell or tissue that is devoid (at least to the limit of detection) of a disease condition.
W0 2018/‘017582 The term "nucleic acid" refers to polynucleotides, such as deoxyribonucleic acid (DNA), and, where riate, ribonucleic acid (RNA). The term should also be understood to include analogues of either RNA or DNA made from nucleotide analogues, and, as applicable to the embodiment being described, single-stranded (such as sense or antisense) and double—stranded polynucleotides. In some embodiments, ”nucleic acid" refers to inhibitory nucleic acids. Some categories of inhibitory nucleic acid compounds include antisense nucleic acids, RNAi ucts, and catalytic c acid constructs. Such categories of nucleic acids are well-known in the art. ments described herein relate generally to compositions and methods that promote the generation or the survival of neurons in the mammalian nervouse system (e. g., brain) as well as to compositons and methods of treating diseases, disorders, and/or ions of the nervous system. As described in the Examples below, it was found that compounds, which inhibit, , and/or antagonize short-chain dehydrogenase activity, such as 15—PGDH inhibitors, can be used to increase PGE2 levels in the nervous system ((3. g., brain) of a mammal. PGE2 elevates cyclic AMP via binding to EP2 and EP4 receptors, which are highly expressed in the cerebral cortex, hippocampus, and striatum. Stimulation of these receptors with PGE2 by administration of a compound that inhibits, reduces, and/or antagonizes 15—PGDH acivity, such as with a H inhibitor described herein, can promote neuroprotection in a subject from axonal degeneration, neuronal cell death, and/or glia cell damage after injury, augment neuronal signaling underlying learning and memory, stimulate neuronal regeneration after injury, and/or treat diseases, disorders, and/or conditions of the nervous system.
In some embodiments, the disease, disorder, and/or condition of the nervous system that can be treated with the H inhibitors can include at least one of a ogical disorder, a neuropsychiatric disorder, a neural injury, a neural toxicity disorder, neuropathic pain, and a neural degenerative disorder.
In some embodiments, the 15-PGDH inhibitors described herein can be used in methods for treating (e. g., controlling, relieving, ameliorating, alleviating, or g the progression of) or s for preventing (e. g., ng the onset of or reducing the risk of developing) one or more diseases, disorders, or conditions caused by, or associated with insufficient (e.g., aberrant) neurogenesis or ed neuronal cell death in a subject in need thereof. The methods include stering to the subject an effective amount of a lS-PGDH W0 2018/‘017582 inhibitor described herein (and/or a compound of any of the other formulae described herein) or a salt (e. g., a pharmaceutically acceptable salt) thereof as defined anywhere herein to the subject. The one or more diseases, disorders, or conditions can include neuropathies, nerve trauma, and neurodegenerative diseases.
In some embodiments, the one or more diseases, disorders, or ions can be diseases, disorders, or conditions caused by or ated with insufficient neurogenesis (e. g., aberrant hippocampal enesis) as is believed to occur in neuropsychiatric diseases or aberrant neuronal cell death as is ed to occur in neurodegenerative diseases.
Examples of the one or more es, disorders, or conditions include, but are not limited to, schizophrenia, major depression, bipolar disorder, normal aging, epilepsy, traumatic brain , post-traumatic stress disorder, Parkinson's e, Alzheimer‘s disease, Down syndrome, spinocerebellar ataxia, amyotrophic lateral sclerosis, Huntington‘s disease, stroke, radiation y, chronic stress, and abuse of neuro-active drugs, such as alcohol, opiates, methamphetamine, phencyclidine, and cocaine.
In some embodiments, the t can be a t in need thereof (e. g., a subject identified as being in need of such treatment), such as a subject having, or at risk of having, one or more of the diseases or conditions described herein. Identifying a subject in need of such treatment can be in the judgment of the subject or a health care professional and can be subjective (e.g., opinion) or objective (e.g., able by a test or diagnostic method). In some embodiments, the subject can be a mammal. In n embodiments, the subject can be a human.
In other embodiments, the lS-PGDH tors can be used to treat diseases, disorders, or conditions associated with elements of the nervous system, including the central, somatic, autonomic, sympathetic, and parasympathetic components of the nervous system, neurosensory s within the eye, ear, nose, mouth or other organs, as well as glial tissues associated with neuronal cells and structures. Such neurological disorders may be caused by an injury to a neuron, such as a mechanical injury or an injury due to a toxic compound, by the abnormal growth or development of a neuron, or by the misregulation, such as gulation, of an activity of a neuron.
Neurological disorders can detrimentally affect nervous system functions such as the sensory function (the ability to sense changes within the body and the outside environment); the integrative function (the ability to interpret the changes); and the motor W0 2018/‘017582 function (the ability to d to the interpretation by initiating an action, such as a muscular contraction or glandular secretion). es of neurological disorders that can be treated by administration of the -PGDH inhibitors to a subject in need f include traumatic or toxic injuries to peripheral or cranial nerves, spinal cord, or brain, such as traumatic brain injury, stroke, cerebral aneurism, and spinal cord injury. Other ogical disorders that can be treated by administration of the 15-PGDH tors to a subject in need thereof include cognitive and neurodegenerative disorders, such as Alzheimer's disease, dementias related to Alzheimer's disease (such as Pick's disease), Parkinson's and other Lewy diffuse body diseases, senile dementia, gton's disease, Gilles de la Tourette’s syndrome, multiple sclerosis, amyotrophic lateral sclerosis, tary motor and sensory athy (Charcot—Marie- Tooth disease), ic neuropathy, progressive supranuclear palsy, epilepsy, and Jakob- fieldt disease. Autonomic function disorders include hypertension and sleep disorders.
Also to be treated with 15-PGDH inhibitors described herein are neuropsychiatric disorders, such as sion, schizophrenia, schizoaffective disorder, Korsakoff’s psychosis, mania, y disorders, or phobic disorders, learning or memory disorders (such as amnesia and age-related memory loss), attention t disorder, dysthymic er, major depressive disorder, mania, obsessive-compulsive disorder, psychoactive substance use disorders, anxiety, phobias, panic disorder, r ive disorder, psychogenic pain syndromes, and eating disorders.
Other examples of neurological disorders that can be treated by administration of the 15-PGDH inhibitors to a subject in need thereof include injuries to the nervous system due to an infectious disease (such as meningitis, high fevers of various etiologies, HIV, syphilis, or post-polio syndrome) and injuries to the nervous system due to electricity (including contact with electricity or lightning, and complications from o—convulsive psychiatric therapy). Other neurological disorders can be associated with ophthalmic conditions including retina and optic nerve damage, glaucoma and age related macular degeneration.
The developing brain is a target for neurotoxicity in the developing l nervous system through many stages of pregnancy as well as during infancy and early childhood, and the 15-PGDH inhibitors described herein may be utilized in preventing or treating neurological deficits in embryos or fetuses in utero, in premature infants, or in W0 2018f017582 children with need of such treatment, including those with ogical birth defects. Further neurological ers e, for example, those listed in HARRISON‘S PRINCIPLES OF INTERNAL MEDICINE (Braunwald et al., McGraW-Hill, 2001) and in the AMERICAN PSYCHIATRIC ATION’S DIAGNOSTIC AND STATISTICAL MANUAL OF MENTAL ERS DSM-IV (American Psychiatric Press, 2000).
] The 15-PGDH inhibitors described herein can also be used in a method of to treat a medical condition associated with a neural injury. The medical condition associated with a neural injury can refer to any movement disorders, epilepsy, cerebrovascular diseases, autoimmune diseases, sleep disorders, autonomic disorders, urinary bladder disorders, abnormal metabolic states, disorders of the muscular system, infectious and parasitic diseases neoplasms, endocrine diseases, nutritional and metabolic diseases, logical diseases, diseases of the blood and blood-forming , mental disorders, diseases of the nervous system, diseases of the sense organs, diseases of the circulatory system, diseases of the respiratory system, diseases of the digestive system, diseases of the genitourinary system, diseases of the skin and aneous tissue, diseases of the musculoskeletal system and connective , congenital anomalies, certain conditions originating in the perinatal period, and symptoms, signs, and fined conditions.
Cerebrovascular disease treatable may be caused by conditions including, but not limited to, aneurysms, strokes, arrhythmia, dial infarction, ischemia reperfusion injury, and cerebral hemorrhage.
Autoimmune diseases treatable e, but are not limited to, le sis.
Sleep disorders treatable by the 15-PGDH inhibitors may be caused by conditions including, but not d to, sleep apnea and parasomnias.
Autonomic disorders treatable by the 15-PGDH inhibitors may be caused by conditions including, but not limited to, intestinal disorders, including but not limited to gastrointestinal motility disorders, nausea, vomiting, diarrhea, chronic hiccups, gastroesphageal reflux disease, and hypersecretion of gastric acid, autonomic insufficiency; excessive epiphoresis, ive rhinorrhea; and cardiovascular disorders including, but not limited, to cardiac dysrythmias and mias, hypertension, and carotid sinus disease.
Urinary bladder disorders treatable by the 15-PGDH inhibitors may be caused by conditions including, but not limited to, spinal cord injury and spastic or flaccid bladder.
W0 2018/‘017582 Abnormal lic states treatable by the 15-PGDH inhibitors may be caused by conditions including, but not limited to, hyperthyroidism or hypothyroidism.
Disorders of the muscular system treatable by the 15-PGDH inhibitors can include, but are not limited to, muscular dystrophy, and spasms of the upper atory tract and face.
The 15-PGDH inhibitors can also be used to treat athic pain caused by ions including, but not limited to, migraine headaches, including migraine hes with aura, migraine headaches without aura, menstrual migraines, migraine ts, atypical migraines, complicated migraines, hemiplegic migraines, transformed migraines, and c daily migraines, episodic n headaches, chronic tension headaches, analgesic rebound headaches, episodic cluster headaches, chronic cluster headaches, cluster variants, chronic paroxysmal hemicranias, hemicrania continua, post-traumatic headache, post-traumatic neck pain, erpetic neuralgia involving the head or face, pain from spine fracture secondary to osteoporosis, arthritis pain in the spine, headache related to cerebrovascular disease and stroke, headache due to vascular disorder, reflex sympathetic dystrophy, cervicalgia (which may be due to various causes, including, but not limited to, muscular, discogenic, or degenerative, including arthritic, posturally related, or metastatic), glossodynia, carotidynia, cricoidynia, otalgia due to middle ear lesion, gastric pain, sciatica, maxillary neuralgia, laryngeal pain, myalgia of neck muscles, trigeminal neuralgia (sometimes also termed tic douloureux), post—lumbar puncture headache, low cerebro-spinal fluid pressure headache, temporomandibular joint disorder, atypical facial pain, ciliary neuralgia, paratrigeminal neuralgia (sometimes also termed Raeder‘s me); petrosal neuralgia, Eagle‘s syndrome, idiopathic intracranial hypertension, orofacial pain, myofascial pain syndrome involving the head, neck, and shoulder, chronic migraneous neuralgia, cervical headache, paratrigeminal paralysis, SPG neuralgia (sometimes also termed lower-half headache, lower facial neuralgia syndrome, Sluder‘s neuralgia, and Sluder‘s syndrome), dynia, vidian gia, causalgia, and/or a combination of the above.
As used herein, the term che" can refer to migraines, tension headaches, cluster headaches, trigeminal neuralgia, secondary headaches, tension-type headaches, chronic and epsisodic headaches, tion e/rebound headaches, chronic paroxysmal hemicrinia hes, hemicranias continua hes, post—traumatic headaches, post—herpetic headaches, vascular hes, reflex sympathetic dystrophy-related W0 2018/‘017582 headaches, crvicalgia headaches, caroidynia headaches, sciatica headaches, trigeminal headaches, occipital headaches, maxillary hes, diary headaches, paratrigeminal headaches, petrosal headaches, Sluder’s headache, Vidian headaches, low CSF pressure headaches, TMJ headaches, causalgia headaches, myofascial headaches, all primary headaches (e. g., primary ng headache, primary cough headache, primary exertional he, primary headache ated with sexual activity, hypnic he, and new daily persistent headache), all trigeminal autonomic cephalagias (e. g., episodic paroxysmal hemicranias, SUNCT, all probable TACs, and SUNA), chronic daily hes, occipital gia, atypical facial pain, neuropathic trigeminal pain, and miscellaneous-type headaches.
In still other ments, the lS-PGDH inhibitors can be used to promote neural stem cell or progenitor cell survival, plasticity, and/or growth. The lS-PGDH inhibitors can be administered to the stem cell or itor cells ex vivo, in vitro, or in viva.
When administered ex vivo or in vitro to the stem cells or progenitor cells, the stem cell or progenitor can then be transplanted to a subject for therapeutic applications.
For the neural stem/progenitor cell, for example, a method of transplanting a neural stem/progenitor cell(s) to a d area that is generally used in the field of regenerative medicine may be employed in conjunction with administration of the lS—PGDH tor to the cells or area. More specifically, there can be exemplified, for example, a method of transplanting a neural stem/progenitor cell(s) to an area of interest by: suspending neural stem/progenitor cells in phosphate buffered saline with the lS-PGDH tor; and adding/injecting the resultant cell suspension to the area.
In other embodiments, the lS-PGDH inhibitors described herein can be applied to a nerve graft. The graft can include any tissue intended for implantation within a human or animal. Various types of graft are encompassed within the t invention, such as autografts, syngrafts, allografts, and xenografts. The size (e. g., length and diameter) of the graft is not critical. For example, the length of the nerve graft can be from about 1 centimeter to about 10 centimeters, or over about 10 centimeters. The diameter of the nerve graft can match that of any injured nerve or part of a nerve, as needed. The nerve graft can be a structurally te segment of nerve to bridge a gap along the length of the ent's nerve or to replace the distal end, i.e., for end-to-end grafting. Alternatively, the nerve graft can be a partial nerve segment, or eccentrically-shaped (e. g., a nerve flap), and intended to W0 2018f017582 reconstruct a lacerated nerve that has some ural disruption, but retains its physical continuity.
When the l5-PGDH inhibitors are applied to a nerve graft, the entire graft can be treated. The l5—PGDH inhibitors can be applied to the entire nerve graft, en bloc. The en bloc treatment can be applied to living (fresh) or previously frozen nerve grafts. The 15- PGDH inhibitors can also be applied to a nerve graft before, , or after implantation.
The l5-PGDH inhibitors can be d to any n of the graft, such as the end or ends to be joined to the stump of a damaged nerve. If the l5-PGDH inhibitor is applied to the damaged nerve, the l5-PGDH inhibitor can be applied to any area of the damaged nerve that promotes repair of the damaged nerve, such as at the site of damage or adjacent to the site of damage.
The 15-PGDH inhibitors can be placed in a e medium for application to the nerve graft. The culture medium can be undefined medium, defined medium, or defined medium supplemented with serum for e. Embodiments described herein also include storage solutions for storage of nerve grafts prior to implantation. The storage solution contains a culture medium and at least one 15-PGDH inhibitor. The storage solution can also include other biologically active agents, such as the growth factors described below.
In some embodiments, l5-PGDH inhibitors used to treat the e, disorder or condition of the nervous system can be identified using assays in which putative inhibitor compounds are applied to cells expressing H and then the functional effects on l5-PGDH activity are determined. Samples or assays sing l5-PGDH that are treated with a potential inhibitor are compared to control samples t the inhibitor to examine the extent of effect. Control samples (untreated with modulators) are assigned a relative l5-PGDH activity value of 100%. Inhibition of l5-PGDH is achieved when the 15-PGDH activity value relative to the control is about 80%, optionally 50% or 25%, 10%, 5% or 1%.
Agents tested as inhibitors of l5-PGDH can be any small chemical molecule or compound. Typically, test compounds will be small chemical molecules, l products, or peptides. The assays are ed to screen large chemical libraries by automating the assay steps and providing compounds from any convenient source to assays, which are typically run in parallel (e. g., in microtiter formats on iter plates in c assays).
In some embodiments, the lS-PGDH inhibitor can include a compound having the following formula (I): W0 2018/‘017582 ( ll ) n Y1—X1/Z1 I‘JJ".8\R1 \\ /U1 Y2 (I) n n is 0-2; Y1, Y2, and R1 are the same or different and are each selected from the group ting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3—C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O) (C1-C6 alkyl), 0, and S), C6-C24 l, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1—C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (--CO-alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (-O-acyl), C2—C24 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (—(CO)—O—aryl), C2-C24 alkylcarbonato (-O-(CO)-O-alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato (-COO'), carbamoyl (-(CO)-NH2), C1-C24 alkyl-carbamoyl (—(CO)—NH(C1—C24 alkyl)), arylcarbamoyl (-(CO)-NH—aryl), thiocarbamoyl (—(CS)—NH2), carbamido (—NH—(CO)-NH2), cyano(-CN), isocyano (-N+C'), cyanato (—O—CN), nato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), rmyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-a1kyl), C6-C20 arylamido (-NH-(CO)-aryl), imino (-CR=NH where R is hydrogen, C1-C24 alkyl, C5—C20 aryl, C6-C24 alkaryl, C6-C24 l, etc.), alkylimino (-CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, l, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo (-S02—OH), sulfonato (—SOz-O‘), C1-C24 ulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed ”arylthio"), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1—C24 alkylsulfonyl alkyl), C5-C20 arylsulfonyl (-SOg-aryl), sulfonamide (-SOz-NH2, 2 (wherein Y is independently H, arlyl or , phosphono (-P(O)(OH)2), onato (-P(O)(O')2), phosphinato (-P(O)(O')), phospho (-P02), phosphino (--PH2), polyalkylethers, phosphates, phosphate esters, groups incorporating amino acids or other moieties expected to bear positive or ve charge at physiological W0 2018f017582 pH, combinations thereof, and wherein Y1 and Y2 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or tituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C—Rz, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz—CHZX, O-CHz-CHZX, CHz-CHz-CHZX, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz-CHZX, CHz-CHz-CHZX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; X1 and X2 are independently N or C, and wherein when X1 and/or X2 are N, Y1 and/or Y2, respectively, are ; Z1 is O, S, CRE‘Rb or NRa, wherein R2’1 and Rb are independently H or a C1_3 alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically acceptable salts thereof.
Examples of H tors having formulas (I) include the ing ; and pharmaceutically acceptable salts thereof.
W0 2018/‘017582 In other embodiments, the 15-PGDH inhibitor can e a compound having the following formula (II): ('0'), z1 IJJJ3\R1 REX/$3“ \\ u1 \Xei); (11) wherein n is 0-2 X4, X5, X6, and X7 are independently N or CR“; R1, R6, R7, and RC are independently selected from the group consisting of hydrogen, tuted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3-C20 aryl, aryl, heterocycloalkenyl containing from 5-6 ring atoms (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O)(C1—C6 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (——CO-alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (—O—acyl), C2-C24 alkoxycarbonyl (—(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2—C24 alkylcarbonato (—O—(CO)-O-alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (- COOH), carboxylato (-COO'), carbamoyl (-(CO)-NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1—C24 alkyl)), arylcarbamoyl (-(CO)-NH-aryl), thiocarbamoyl (-(CS)—NH2), carbamido (-NH—(CO)-NH2), cyano(-CN), isocyano (-N+C'), cyanato (-O-CN), nato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl )--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)—alkyl), C6-C20 arylamido (-NH-(CO)-aryl), imino (-CR=NH where R is hydrogen, C1-C24 alkyl, C5—C20 aryl, C6-C24 l, C6-C24 aralkyl, etc.), alkylimino (—CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(ary1), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo (-SOz-OH), sulfonato (—SOg—O‘), C1—C24 ulfanyl (-S-alkyl; also termed "alkylthio"), arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyl -alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOz-aryl), sulfonamide (-SOz—NH2, —SOZNY2 in Y is ndently H, arlyl or , phosphono W0 2018/‘017582 (-P(O)(OH)2), phosphonato (-P(O)(O‘)2), phosphinato (-P(O)(O')), phospho (-P02), phosphino (--PH2), polyalkylethers, phosphates, phosphate esters, groups incoporating amino acids or other moieties ed to bear positive or negative charge at physiological pH, combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a tuted or unsubstituted heterocyclyl; U1 is N, C—RZ, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ in n1=1, 2, or 3), CF3, CHz—CHzX, O-CHz-CHZX, CHz-CHg-CHZX, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (R’)2, ’, COOR’ (wherein R’ is H or a lower alkyl group), and n R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein n1=1, 2, or 3), CF3, CHz-CHzX, CHz-CHz-CHQX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R” is H or a lower alkyl group), and R3 or R4 may be absent; Z1 is O, S, CRaRb or NRa, wherein R31 and Rb are ndently H or a C1_g alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically acceptable salts thereof.
] Examples of lS-PGDH inhibitors having formulas (11) include the following compounds: W0 2018/‘017582 2017/042620 NH2 - OPO3H2 ; and pharmaceutically able salts thereof.
In yet other embodiments, the 15-PGDH tor can include a compound having the following formula (III) or (IV): (0),, (o), g S 21 Jud" \R1 U1\¢r’ \R1 R7 (111), or R’ (IV) wherein n is 0-2 X6 is independently is N or CR“; R1, R6, R7, and RC are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 l, C2-C24 alkynyl, C3—C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms (wherein from 1—3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O)(C1—C6 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, -C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2—C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (——CO-alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (—O—acyl), C2-C24 W0 017582 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-ary1), C2-C24 alkylcarbonato (-O-(CO)-O-alkyl), C6-C20 arylcarbonato (-O-(CO)-O-ary1), carboxy (- COOH), carboxylato (-COO'), oyl (-(CO)-NH2), C1-C24 alkyl—carbamoyl (-(CO)—NH(C1—C24 alkyl)), arylcarbamoyl -NH-aryl), thiocarbamoyl (—(CS)—NH2), carbamido (—NH—(CO)-NH2), cyano(-CN), no (-N+C'), cyanato (-O-CN), isocyanato (-O-N+=C‘), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (--(CS)--H), amino ), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alkyl), C6-C20 arylamido (-NH-(CO)-aryl), imino (-CR=NH where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (—CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, l, etc.), nitro (-N02), nitroso (-NO), sulfo (—SOz-OH), sulfonato (—SOg-O‘), C1-C24 alkylsulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1—C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOg—ary1), sulfonamide (-SOz-NH2, -SOgNY2 (wherein Y is independently H, arlyl or , phosphono (-P(O)(OH)2), phosphonato (-P(O)(O‘)2), phosphinato (-P(O)(O')), phospho (-P02), phosphino (——PH2), polyalkylethers, phosphates, phosphate , groups orating amino acids or other moieties expected to bear positive or negative charge at physiological pH, combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a tuted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C—RZ, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz-CHQX, O-CHg-CH2X, CHz-CHz-CHZX, O-CHz-CHZX, X, in X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl , and wherein R1 and R2 may be linked to form a cyclic or clic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)nIOR’ (wherein n1=1, 2, or 3), CF3, CH2-CH2X, CH2-CH2-CH2X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R” is H or a lower alkyl , and R3 or R4 may be absent; Z1 is O, S, CRaRb or NRa, wherein Rat and Rb are independently H or a C1_3 alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically able salts thereof.
In some embodiments, R1 is selected from the group consisting of branched or linear alkyl including —(CH2)n1CH3 (n1=0-7), n2 wherein n2=0-6 and X is any of the following: CFyHZ (y + z = 3), CClyHZ (y + z = 3), OH, OAC, OMe, R71, 0R”, CN, N(R73)2, WAh WW4 ”3 "4 , m=l-5), and (114:0—5).
In other embodiments, R6 and R7 can each independently be one of the following: airs _ ell/S (3 “/8 “/8 O R g R I | / I / 7R | N>E—R @712 III / ng '— g_ M 3 3 \ \ nil—KO 0 15E >7 _ O 0 NR” 1%I/ NRZ‘ g ”r R13'—l/ R H/ I / R I N/ R R IU— / g— NI /, I / g R I— /7 ”t“ “x” NV“ 22(53>, —R23/NR24 26 28 O R30 “/NR RNL/NR N/ O R r{II\N/ E J" / R III— / é— “\N/>§_ I / N|\N/>’§_ N/O N/NR32 N/NR34 />§RI/§ IAMH/E|| 33”— _R35 5R N/NR38 N/NR37II 31 _ N , .NW W31" \ \ R39 N/NR42 N/NR43 NR45‘N R45 \ \ || R41 || | R44 / / | R49I_/\“ N R“ “\ N\/RR'/,f'/>§_48|_ i N \ ~I- ”V ”\“n N /\ N R; \ \ \ \ R.— 1R~R mm; /:’S; /;\KN/.~5¢\ / \N / \KN/ £\ W0 2018/‘017582 N N OR60 OR61 R56|_// \ N R57L\\ R55|_( \ I ” _3_R59 J ¥ $75 R52 49:“ at“ Kai V“ / R53 5 0 0 back O=m=O R69 R66 —§—CN -§—fl—Re8 —§— / N/Fg: /R54:LL) l ’ \R70 each R8, R9, R10, R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22, R23, R24, R25, R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39 R40 R41 R42 R43 R44 R45 R46 R47 R48 R49 R50 R51 R52 R53 R54 R55 R56 R57 R58 R59 R60 R61 R62 R63 R64 R65 R66 R67 R68 R69 R70, R71, R? R73, and R74 are the same or different and are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2—C24 alkenyl, C2-C24 alkynyl, C3—C20 aryl, heterocycloalkenyl containing from 5-6 ring atoms, (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O) (C1-C6 alkyl), 0, and S), heteroaryl or heterocyclyl containing from 5-14 ring atoms, (wherein from 1-6 of the ring atoms is ndently selected from N, NH, N(C1-C3 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, silyl, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2—C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (--CO—alky1) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (-O-acyl), C2—C24 alkoxycarbonyl (-(CO)-O—a1ky1), C6-C20 aryloxycarbonyl -O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O—alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato (-COO'), carbamoyl (-(CO)--NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl -NH-aryl), thiocarbamoyl (-(CS)-NH2), carbamido (-NH-(CO)-NH2), cyano(-CN), no (-N+C'), cyanato (-O-CN), isocyanato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), rmyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alkyl), C6—C20 arylamido (—NH-(CO)-aryl), amido (R)2 where R is independently H, alkyl, aryl or heteroaryl), imino (—CR=NH where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6—C24 l, etc.), mino (-CR=N(alky1), where R=hydrogen, alkyl, aryl, alkaryl, l, etc), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo (-SOz-OH), sulfonato (-SOg-O‘), C1-C24 alkylsulfanyl W0 2018f017582 2017/042620 (-S-alkyl; also termed "alkylthio"), arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyl (-(SO)—alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 alkylsulfonyl (—SOz—alkyl), C5—C3) arylsulfonyl (-SOz-aryl), sulfonamide (-SOz-NHZ, —SOzNY2 in Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (—P(O)(O')2), phosphinato (—P(O)(O')), phospho (-POZ), phosphino (--PH2), polyalkyl ethers (—[(CH2)nO]m), phosphates, phosphate esters [-OP(O)(OR)2 where R = H, methyl or other alkyl], groups orating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations thereof, and pharmaceutically able salts thereof.
In still other embodiments, R6 and R7 can ndently be a group that improves aqueous solubility, for example, a phosphate ester (-OPO3H2), a phenyl ring linked to a phosphate ester H2), a phenyl ring substituted with one or more methoxyethoxy groups, or a morpholine, or an aryl or heteroaryl ring substituted with such a group.
Examples of lS-PGDH inhibitors having as (III) or (IV) include the following compounds: WO 17582 W0 2018f017582 ; and pharmaceutically acceptable salts f.
In other embodiments, the lS-PGDH inhibitor can include a compound having the following formula (V): ( fl)“ s ‘p‘ \R1 R7 (V) wherein n is 0-2 X6 is independently is N or CRC R1, R6, R7, and RC are each independently ed from the group consisting of en, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3-C20 aryl, heteroaryl, heterocycloalkenyl ning from 5-6 ring atoms (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O)(C1—C6 alkyl), 0, and S), C6—C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2—C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (-—CO—alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy (-O-acy1), C2—C24 alkoxycarbonyl —O—alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2—C24 alkylcarbonato O)—O—alky1), C6-C20 arylcarbonato (-O-(CO)-O-ary1), carboxy (-COOH), carboxylato (-COO'), carbamoyl (-(CO)-NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl -NH-ary1), thiocarbamoyl (-(CS)-NH2), carbamido (—NH—(CO)-NH2), cyano(-CN), isocyano (-N+C'), cyanato (—O—CN), isocyanato W0 2018f017582 (-O-N+=C'), isothiocyanato (-S-CN), azido =N'), formyl (--(CO)--H), thiofonnyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido (—NH—(CO)—alkyl), C6-C20 arylamido (-NH-(CO)-aryl), imino (-CR=NH where R is hydrogen, C1-C24 alkyl, C5—C20 aryl, C6-C24 l, C6-C24 aralkyl, etc.), alkylimino (—CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro , nitroso (-NO), sulfo (-SOz—OH), sulfonato (-SOg—O‘), C1-C24 ulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed ”arylthio”), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOz-aryl), sulfonamide (-SOz-NH2, -SOzNY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (-P(O)(O')), o (-P02), phosphino (——PH2), polyalkylethers, phosphates, ate , groups incorporating amino acids or other moieties expected to bear ve or ve charge at physiological pH, combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C—Rz, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz—CHZX, O-CHz-CHZX, CHg-CHz-CHZX, CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each ed from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CHz-CHZX, CHz-CHz-CHzX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; and pharmaceutically able salts thereof.
In some embodiments, R1 is selected from the group consisting of branched or linear alkyl including —(CH2)n1CH3 (n1=0-7), n2 wherein n2=0-6 and X is any of the following: (:13sz (y + z = 3), cc1sz (y + z = 3), OH, OAC, OMe, R71, 0R”, CN, N(R73)2, WAR WW4 ”3 "4 (n3=0—5, n1=1-5), and —5).
In other embodiments, R6 and R7 can each independently be one of the Brs rs (3 (S ° RI /5_ (3RI /f‘ I—N/fir‘“? r/ E—Rn— €- J‘RN J\r\\f"‘ 14|_l/O 0 “/ >7 _ O 0 NR19 NR“ R I R18'—/ ||/ i /R“\N/§ RNI15‘|/ /7R'!‘|—/§— I / g—RI /) “a” N?” ”t” 22£S> —R23 I/NR24 “/NR26 28 R30 RnyR o N/ O Rl'\/§ l' / Rr/ i‘ L/fl—l / L}?— N ’ N \ Ni” N/0 N/NRsz N/NR34 N/NRas N/NRaa N/NRM R37 I 31 || 33”— _R35 .l R' _ / L); J5 NJ/_ l' /§ N1” NV/ R39 N/NR42 N/NR43 NR45—N R46 NR47 \ \ ILi R41 / NI / 'L\\ Rh/ />§—R48|_ R49|—/\N i i ’ N\N " KS;\ / M \ NV“ \ ”l" ’ N /\ N Rsofi \ \ R5:ill |_//\N \N RMAN 53L \ N\ N OR“ OR61 \N \ 56'_ R57,Ell R53|_‘/ \ 3 R59 R w _ _ [ill [ii R52 /j\ /, 5‘s;\ /, 5‘54; / /‘3) L 0 fi ii /R69 R64, /R65 R66 _ —CN _§_S_R68 —§—S—N 31!, 0/ ”$7 N E , /EJI\K (1 Ci \R7o‘ W0 2018/‘017582 each each R8!R9,R10,R11,R12'R13,R14,R15,R16’R17’R18‘R19‘R20~R21~R22,R23§ R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39 R40 R41 R42 R43 R44 R45 R46 R47 R48 R49 R50 R51 R52 R53 R54 R55 R56 R57 R58 R59 R60 R61 R62 R63 R64 R65 R66 R67 R68, R69, R70, R71, R72, R73, and R74, are the same or different and are independently ed from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2—C24 alkenyl, C2-C24 alkynyl, C3-C20 aryl, heterocycloalkenyl ning from 5-6 ring atoms, (wherein from 1—3 of the ring atoms is ndently selected from N, NH, N(C1—C6 alkyl), C1-C6 , 0, and S), heteroaryl or heterocyclyl containing from 5-14 ring atoms, (wherein from 1-6 of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S), C6—C24 alkaryl, C6-C24 aralkyl, halo, silyl, hydroxyl, sulfhydryl, C1—C24 alkoxy, C2-C24 loxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (--CO-alkyl) and C6-C20 arylcarbonyl ryl)), acyloxy (-O-acyl), C2-C24 alkoxycarbonyl (-(CO)-O-alky1), C6—C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O- alkyl), C6—C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato (—COO‘), carbamoyl (-(CO)--NH2), C1-C24 carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl (-(CO)-NH-aryl), thiocarbamoyl (-(CS)-NH2), carbamido (-NH-(CO)-NH2), cyano(-CN), isocyano ), cyanato (-O-CN), isocyanato (-O-N+=C'), isothiocyanato (—S—CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (--(CS)--H), amino (--NH2), C1—C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido CO)-alkyl), C6-C20 arylamido (—NH—(CO)-aryl), sulfanamido (—S02N(R)2 where R is independently H, alkyl, aryl or aryl), imino (- CR=NH where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (-CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (-N02), nitroso (-NO), sulfo (-SOz—OH), sulfonato (-SOz-O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed ”arylthio”), C1-C24 alkylsulfinyl (—(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOz-ary1), sulfonamide (-SOz-NH2, -SOZNY2 (wherein Y is independently H, arlyl or alkyl), phosphono (—P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (O‘)), phospho (-P02), phosphino (--PH2), polyalkyl ethers (-[(CH2)nO]m), phosphates, phosphate CStCI'S W0 2018f017582 [-OP(O)(OR)2 where R = H, methyl or other , groups incorporating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and ations thereof, and pharmaceutically acceptable salts f.
In still other embodiments, R6 and R7 can independently be a group that improves aqueous solubility, for example, a phosphate ester (-OPO3H2), a phenyl ring linked to a phosphate ester (-OPO3H2), a phenyl ring substituted with one or more methoxyethoxy groups, or a line, or an aryl or heteroaryl ring substituted with such a group.
In other embodiments, the 15-PGDH inhibitor can include a compound having the following formula (VI): Red/K S (f) / S\R1 n R? (VI) wherein n = 0—2; X6 is N or CRC; R1 is selected from the group consisting of branched or linear alkyl including — (CH2)n1CH3 (n1=0—7), n2 wherein n2=0-6 and X is any of the following: CFyHZ (y + z = 3), CClsz (y + z = 3), OH, OAc, OMe, R71, OR72, CN, N(R73)2, ”3 (113:0—5, m=1-5), and (n4=0-5).
R5 is ed from the group consisting of H, Cl, F, NH2, and N(R76)2; R6 and R7 can each independently be one of the following: each R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26, R27, R28, R29, R305 R31” R32, R33, R34, R35, R36, R37, R38, R39, R40, R41, R42, R43, R44 R45, R46” R47, R48,R49,R50, R51, R52,R53,R54, R55,R56, R57, R583R593R60a R615R62, R63, R64,R65,R66, 1167,1168, R69” R70‘ R71‘ R? R73~ R74, R76, and R0 are the same or different and are ndently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, W0 2018/‘017582 C2-C24 alkynyl, C3-C20 aryl, heterocycloalkenyl containing from 5-6 ring atoms, (wherein from 1-3 of the ring atoms is independently ed from N, NH, N(C1-C6 alkyl), NC(O) (C1—C6 alkyl), 0, and S), aryl or heterocyclyl containing from 5—14 ring atoms, in from 1—6 of the ring atoms is independently selected from N, NH, N(C1—C3 alkyl), 0, and S), C6—C24 alkaryl, C6-C24 aralkyl, halo, silyl, hydroxyl, sulfhydryl, C1—C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (--CO-alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), y yl), C2-C24 carbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O—alkyl), C6-C20 arylcarbonato (-O-(CO)-O-aryl), carboxy (-COOH), carboxylato (-COO'), carbamoyl (-(CO)--NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl (—(CO)-NH-aryl), thiocarbamoyl (-(CS)-NH2), carbamido (—NH—(CO)-NH2), cyano(-CN), isocyano ), cyanato (-O-CN), isocyanato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, C5—C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alkyl), C6—C20 ido (-NH-(CO)-aryl), sulfanamido (-SOZN(R)2 where R is independently H, alkyl, aryl or heteroaryl), imino (-CR=NH where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6—C24 aralkyl, etc.), alkylimino (-CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(aryl), where R=hydrogen, alkyl, aryl, l, etc.), nitro (-N02), nitroso (-NO), sulfo (-SOz-OH), ato (-SOz-O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed "alkylthio”), arylsulfanyl (-S-aryl; also termed hio"), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 ulfonyl (-SOz-alkyl), C5—C20 arylsulfonyl (-SOz-aryl), sulfonamide NH2, -SOZNY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (—P(O)(O')), phospho (-P02), phosphino (--PH2), polyalkyl ethers (-[(CH2)nO]m), phosphates, phosphate esters )(OR)2 where R = H, methyl or other alkyl], groups incorporating amino acids or other moieties expected to bear positive or ve charge at physiological pH, and combinations thereof, and pharmaceutically acceptable salts thereof.
In other embodiments, the 15-PGDH inhibitor can include a compound having the following formula (VII): C/ l N\ S l (5/0) n X6 / / \R1 R7 (VII) wherein n = 0-2; X6 is N or CR“; R1 is selected from the group consisting of ed or linear alkyl including — 1CH3 (n1=0-7), n2 wherein n2=0-6 and X is any of the following: CFyHZ (y + z = 3), CClyHZ (y + z = 3), OH, OAc, OMe, R71, OR72, CN, N(R73)2, ”3 (n3=0—5, m=l-5), and (114:0-5).
R5 is selected from the group consisting of H, Cl, F, NH2, and N(R76)2; R7 can each independently be one of the following: Jr «J 14|_|/O O “/ > _ o 0 NR19 wil/ NRZ‘ RI/R"\N/§RNI—\2RH—/§—l/§—RI/,17ll/ Rmfi ll/ “Y” NV” NV“ 22r8> —R23 /NR24 l/NR26 28 0 R30 R27l‘I/NR N/ 0 R lil|\N/ g rLI I / R [1"— / g— “\N/>’§_ / N|\N/>~§_ W 38w N/0 N/NR32 N/NR34 N/NR36 N/NR N/NR40 31“ LN)E 33“— /,§ R35 .. R _ 37K RI / RI _ I/MI /l' /§ NY“ W/ R39 W0 2018f017582 /NR42 /NR43 45¢N R46 Tl R“‘Nl TR \ RW)~§—48| \ R49|—/\N / N R .— N / N M9 " N\N /3555'\%\ JV" \JUV‘ , \N \\ \ \ R63 O O O 0 Jk R69 ,5 II II / /R'5“:1,7 /R 31L . 0 5);, Ree—E—CN —§—S—R68—§—s—N , H I \ 0 R70 each R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 Ra1 R532 N33 N34 Ros Ros N37 Ros Raw RS40 Ra“ Ra42 12,43 R,“ R5 R,46 Rm 5 R48 R49 R50 R51 R52 R53 R54 R55 R56 R57 R58 R59 R60 R61 R62 R63 R64 R65 R66 R67 R68 R69 R70, R71, R”, R73, R74, R76, and Re are the same or different and are independently selected from the group consisting of en, substituted or unsubstituted C1-C24 alkyl, C2—C24 alkenyl, C2-C24 alkynyl, C3—C20 aryl, heterocycloalkenyl containing from 5-6 ring atoms, (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O)(C1- C6 alkyl), 0, and S), aryl or heterocyclyl containing from 5-14 ring atoms, (wherein from 1-6 of the ring atoms is independently selected from N, NH, N(C1-C3 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, silyl, yl, dryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2-C24 alkylcarbonyl (--CO- alkyl) and C6-C20 arylcarbonyl (-CO-aryl)), acyloxy yl), C2-C24 alkoxycarbonyl (- (CO)-O-alky1), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O- , C6—C20 arylcarbonato (-O-(CO)-O-aryl), carboxy ), carboxylato (-COO'), carbamoyl (-(CO)--NH2), C1-C24 alkyl-carbamoyl (-(CO)-NH(C1-C24 alkyl)), arylcarbamoyl (-(CO)-NH—ary1), thiocarbamoyl (-(CS)-NH2), carbamido (-NH—(CO)-NH2), cyano(—CN), isocyano (-N+C'), cyanato ), isocyanato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (--(CS)--H), amino (--NH2), C1-C24 alkyl amino, W0 2018f017582 2017/042620 C5-C20 aryl amino, C2-C24 alkylamido (-NH-(CO)-alky1), C6-C20 arylamido CO)-ary1), sulfanamido (-SOgN(R)2 where R is independently H, alkyl, aryl or heteroaryl), imino H where R is hydrogen, C1-C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6—C24 aralkyl, etc.), alkylimino (—CR=N(alky1), where ogen, alkyl, aryl, alkaryl, aralkyl, etc), ino (—CR=N(ary1), where R=hydrogen, alkyl, aryl, alkaryl, etc.), nitro (—N02), nitroso (-NO), sulfo (—S02-OH), sulfonato (-SOz-O'), C1-C24 alkylsulfanyl (-S-alkyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed ”arylthio”), C1-C24 alkylsulfinyl (—(SO)-alky1), C5-C20 arylsulfinyl (-(SO)-aryl), C1-C24 ulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOZ- aryl), sulfonamide (-SOz-NH2, -SOZNY2 (wherein Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (O‘)), phospho (-P02), phosphino (--PH2), polyalkyl ethers (-[(CH2)HO]m), phosphates, phosphate esters [-OP(O)(OR)2 where R = H, methyl or other alkyl], groups incorporating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations thereof, and pharmaceutically acceptable salts thereof.
Examples of compounds having formulas (V), (VI), or (VII) are selected from the group consisting of: [l)Yg/fw gym/N:N\NN / 8/ K361 CN \ U HN \ . \N— W0 017582 PCT/U82017/042620 WO 17582 WO 17582 WO 17582 WO 17582 PCT/USZOl7/042620 WO 17582 WO 17582 W0 17582 WO 17582 N S \ // N \ // I/ / SLL I / / S\_\_ NH2 CH3 NH2 CH3 CANNOH 1 ; NH2 N/ f8 IN\ S S//O N/ N\ S //O L\_ l/ / NH2 CH3 8L\_ NH2 CH3 N S \ // N S I \ // /' S | S NHz CH3 NH2 CH3 W0 017582 W0 17582 W0 2018/‘017582 pharmaceutically acceptable salts thereof.
In certain ments, the 15-PGDH inhibitor having formula (I), (II), (IV), (V), (VI), and (VII) can be selected that can ia) at 2.5 uM concentration, stimulate a Vac0503 reporter cell line expressing a lS-PGDH luciferase fusion construct to a luciferase output level of greater than 70 (using a scale on which a value of 100 tes a doubling of reporter output over baseline); iia) at 2.5 uM tration stimulate a V9m er cell line expressing a 15—PGDH luciferase fusion construct to a luciferase output level of r than 75; iiia) at 7.5 uM concentration stimulate a LSl74T reporter cell line expressing a 15-PGDH luciferase fusion construct to a luciferase output level of greater than 70; and iva) at 7.5 uM concentration, does not activate a negative control V9m cell line expressing TK—renilla luciferase reporter to a level r than 20; and va) inhibits the enzymatic ty of recombinant l5-PGDH protein at an IC50 of less than 1 uM.
In other embodiments, the l5-PGDH inhibitor can ib) at 2.5 uM concentration, stimulate a Vac0503 reporter cell line expressing a l5-PGDH luciferase fusion construct to increase luciferase output; iib) at 2.5 uM concentration stimulate a V9m reporter cell line expressing a lS-PGDH luciferase fusion construct to increase luciferase output; iiib) at 7.5 uM concentration stimulate a LSl74T er cell line expressing a lS—PGDH luciferase fusion construct to increase luciferase output; ivb) at 7.5 uM concentration, does not activate a negative control V9m cell line expressing TK-renilla rase reporter to a luciferase level W0 17582 greater than 20% above background; and vb) inhibits the enzymatic activity of recombinant H protein at an IC50 of less than 1 uM.
In other embodiments, the 15-PGDH inhibitor can t the enzymatic activity of recombinant 15—PGDH at an IC50 of less than 1 nM, or ably at an IC50 of less than 250 nM, or more preferably at an IC50 of less than 50 nM, or more preferably at an IC50 of less than 10 nM, or more preferably at an IC50 of less than 5 nM at a recombinant 15-PGDH concentration of about 5 nM to about 10 nM.
In other embodiments, the 15-PGDH inhibitor can increase the cellular levels of PGE-2 following stimulation of an A459 cell with an appropriate agent, for example ILl-beta.
In some embodiments, a15-PGDH inhibitor can include a compound having the following formula (VIII): R6 N / $0» l 3 \ / R1 R7 2 (VIII) wherein n is 0-2; R1, R6, and R7 are the same or different and are each selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2—C24 alkynyl, C3-C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms (wherein from 1-3 of the ring atoms is independently selected from N, NH, N(C1-C6 alkyl), NC(O) (C1-C6 alkyl), 0, and S), C6-C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, yl, dryl, C1-C24 , C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl (including C2—C24 alkylcarbonyl (--CO-alkyl) and C6-C20 arylcarbonyl (-CO—ary1)), acyloxy (-O-acyl), C2—C24 alkoxycarbonyl (-(CO)-O-alkyl), C6-C20 aryloxycarbonyl (-(CO)-O-aryl), C2-C24 alkylcarbonato (-O-(CO)-O-alkyl), C6-C20 arylcarbonato O)—O—aryl), carboxy (-COOH), carboxylato (-COO'), carbamoyl (-(CO)-NH2), C1-C24 carbamoyl (-(CO)-NH(C1—C24 alkyl)), arylcarbamoyl -NH—aryl), thiocarbamoyl (—(CS)—NH2), carbamido (-NH-(CO)-NH2), cyano(-CN), isocyano (-N+C'), cyanato (-O-CN), isocyanato (-O-N+=C'), isothiocyanato (-S-CN), azido (-N=N+=N'), formyl (--(CO)--H), thioformyl (——(CS)——H), amino (--NH2), C1-C24 alkyl amino, C5-C20 aryl amino, C2—C24 alkylamido W0 2018/‘017582 (-NH-(CO)-alkyl), C6-C20 arylamido (-NH—(CO)-ary1), imino (-CR=NH where R is hydrogen, C1—C24 alkyl, C5-C20 aryl, C6-C24 alkaryl, C6-C24 aralkyl, etc.), alkylimino (-CR=N(alkyl), where R=hydrogen, alkyl, aryl, alkaryl, aralkyl, etc.), arylimino (-CR=N(aryl), where ogen, alkyl, aryl, l, etc.), nitro (-N02), o (-NO), sulfo (—SOz—OH), sulfonato (—SOg—O'), C1-C24 alkylsulfanyl kyl; also termed ”alkylthio"), arylsulfanyl (-S-aryl; also termed "arylthio"), C1-C24 alkylsulfinyl (-(SO)-alkyl), C5-C20 arylsulfinyl (-(SO)-aryl), C1—C24 alkylsulfonyl (-SOz-alkyl), C5-C20 arylsulfonyl (-SOg-aryl), sulfonamide (-SOz-NH2, -S02NY2 in Y is independently H, arlyl or alkyl), phosphono (-P(O)(OH)2), phosphonato (-P(O)(O')2), phosphinato (-P(O)(O')), phospho (—P02), phosphino (——PH2), polyalkylethers, phosphates, ate esters, groups incorporating amino acids or other moieties expected to bear positive or negative charge at physiological pH, combinations thereof, and n R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a tuted or unsubstituted cyclyl; and pharmaceutically acceptable salts thereof. 15-PGDH inhibitors having formula (VIII) can be synthesized as shown: O 0 8 CN H HN R R7 )k/ l SR1 R R7 CI 8 \/ «I?» \R1 H202 R5 N 3 SR1 S —> A—COH» / \/ Et3N CN / I I \ R6 R7 R7 R6 N KOH, H o S (O)n DM2F / I so \ / \R1 Any reaction solvent can be used in the above preparation process as long as it is not ed in the reaction. For example, the reaction solvent includes ethers such as diethyl ether, tetrahydrofuran and dioxane; halogenized arbons, such as dichloromethane and chloroform; amines such as pyridine, piperidine and triethylamine; alkylketones, such as acetone, methylethylketone and methylisobutyl; alcohols, such as methanol, ethanol and W0 2018/‘017582 propanol; non-protonic polar solvent, such as N,N-dimethylformamide, N,N- dimethylacetamide, itrile, dimethylsulfoxide and hexamethyl phosphoric acid triamide.
Among non—reactive organic solvents that are ordinarily used in the organic synthesis, preferable ts are those from which water generated in the reaction can be d by a Dean-Stark trap. The examples of such solvents include, but are not limited to benzene, toluene, xylene and the like. The reaction product thus obtained may be isolated and purified by condensation, tion and the like, which is ordinarily ted in the field of the organic synthesis, if desired, by silica gel column chromatography. The individual enantiomers of PGDH inhibitors having the formula III can be separated by a preparative HPLC using chromatography columns containing chiral stationary phases.
Further, embodiments of this application include any modifications for the preparation method of the 15-PGDH inhibitors described above. In this connection, any intermediate product obtainable from any step of the preparation method can be used as a starting material in the other steps. Such starting material can be formed in situ under certain reaction conditions. Reaction reagents can also be used in the form of their salts or optical Depending on the kinds of the substituents to be used in the preparation of the -PGDH inhibitors, and the intermediate product and the ation method selected, novel -PGDH inhibitors can be in the form of any possible isomers such as substantially pure geometrical (cis or trans) isomers, l isomers (enantiomers) and racemates.
In some embodiments, a 15-PGDH inhibitor having formula (VIII) can include a compound with the following formula (IX): (IX), and pharmaceutically able salts thereof.
Advantageously, the 15-PDGH inhibitor having formula (IX) was found to: i) t recombinant 15-PGDH at 1 nM tration; ii) inhibit 15-PGDH in cell lines at 100 nM tration, iii) increase PGE2 production by cell lines; iv) is ally stable in aqueous solutions over broad pH range; v) is chemically stable when incubated with W0 2018/‘017582 hepatocyte extracts, vi) is chemically stable when incubated with hepatocyte cell lines; vii) shows 253 minutes plasma half-life when injected IP into mice; and viii) shows no immediate toxicity over 24 hours when injected IP into mice at 0.6 umole/per mouse and at 1.2 umole/per mouse and also no toxicity when injected IP into mice at 0.3 umole/per mouse twice daily for 21 days.
] In other embodiments, a 15-PGDH inhibitor having formula (IX) can include a compound with the following formula (IXa): (IXa) and phannaceutically acceptable salts thereof.
In still other ments, a 15-PGDH tor having formula (IX) can include a compound with the following formula (IXb): and pharrnaceutically acceptable salts thereof.
In other ments, the 15-PDHG inhibitor can comprise a (+) or (-) optical isomer of a H inhibitor having formula (IX). In still other embodiments, the -PDHG inhibitor can comprise a mixture at least one of a (+) or (-) optical isomer of a -PGDH tor having a (IX). For example, the 15-PGDH inhibitor can comprise a mixture of: less than about 50% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (IX) and greater than about 50% by weight of the (+) optical isomer of a W0 2018/‘017582 -PGDH inhibitor having formula (IX), less than about 25% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (IX) and greater than about 75% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (IX), less than about 10% by weight of the (—) optical isomer of a 15-PGDH inhibitor having formula (IX) and greater than about 90% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (IX), less than about 1% by weight of the (-) l isomer of a 15-PGDH inhibitor having formula (IX) and greater than about 99% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (IX), greater than about 50% by weight of the (-) optical isomer of a -PGDH inhibitor having formula (IX) and less than about 50% by weight of the (+) optical isomer of a 15-PGDH inhibitor having a (IX), r than about 75% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (IX) and less than about 25% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (IX), greater than about 90% by weight of the (-) l isomer of a 15-PGDH inhibitor having formula (IX) and less than about 10% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (IX), or greater than about 99% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (IX) and less than about 1% by weight of the (+) optical isomer of a —PGDH inhibitor having formula (IX).
In a still further embodiment, the 15-PDGH inhibitor can consist essentially of or t of the (+) optical isomer of a 15-PGDH inhibitor having a (IX). In yet another embodiment, the PDGH inhibitor can consist essentially of or consist of the (-) optical isomer of a 15-PGDH inhibitor having formula (IX).
In other ments, a 15-PGDH tor having formula (VIII) can include a compound with the following formula (X): and pharmaceutically acceptable salts thereof.
] Advantageously, the 15-PDGH tor having formula (X) was found to: i) inhibit recombinant 15-PGDH at 3 nM concentration; ii) increase PGEZ production by cell W0 2018/‘017582 lines at 20nM; iii) is chemically stable in aqueous solutions over broad pH range; iv) is chemically stable when ted with mouse, rat and human liver extracts, v) shows 33 minutes plasma ife when injected IP into mice; viii) shows no immediate toxicity over 24 hours when injected IP into mice at 50 mg/kg body weight, and ix) is soluble in water (pH=3) at 1 mg/mL.
In other embodiments, a lS-PGDH inhibitor having formula (X) can e a compound with the following formula (Xa): (X21) and pharmaceutically acceptable salts thereof.
In still other embodiments, a 15-PGDH inhibitor having formula (X) can e a compound with the following formula (Xb): (Xb), and pharmaceutically acceptable salts f.
In other embodiments, the lS-PDHG inhibitor can comprise a (+) or (-) optical isomer of a 15-PGDH inhibitor having formula (X). In still other embodiments, the lS-PDHG inhibitor can comprise a mixture at least one of a (+) or (-) optical isomer of a -PGDH inhibitor having formula (X). For example, the 15-PGDH inhibitor can comprise a mixture of: less than about 50% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (X) and greater than about 50% by weight of the (+) optical isomer of a W0 2018f017582 -PGDH inhibitor having formula (X), less than about 25% by weight of the (-) optical isomer of a 15-PGDH inhibitor having a (X) and greater than about 75% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (X), less than about 10% by weight of the (—) optical isomer of a 15-PGDH tor having formula (X) and greater than about 90% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (X), less than about 1% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (X) and r than about 99% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (X), greater than about 50% by weight of the (-) optical isomer of a -PGDH inhibitor having a (X) and less than about 50% by weight of the (+) optical isomer of a 15-PGDH tor having formula (X), greater than about 75% by weight of the (-) optical isomer of a H inhibitor having formula (X) and less than about 25% by weight of the (+) optical isomer of a 15-PGDH inhibitor having formula (X), greater than about 90% by weight of the (-) optical isomer of a 15-PGDH inhibitor having formula (X) and less than about 10% by weight of the (+) optical isomer of a H inhibitor having formula (X), or greater than about 99% by weight of the (-) optical isomer of a H inhibitor having formula (X) and less than about 1% by weight of the (+) optical isomer of a —PGDH inhibitor having formula (X).
] In a still further embodiment, the 15-PDGH inhibitor can consist essentially of or consist of the (+) optical isomer of a 15-PGDH inhibitor having formula (X). In yet another embodiment, the PDGH inhibitor can consist essentially of or consist of the (-) optical isomer of a 15-PGDH inhibitor having formula (X).
It will be appreciated that the other 15-PGDH inhibitors can be used in the methods described bed herein. These other 15-PGDH inhibitors can include known H inhibitors including, for example, tetrazole compounds of formulas (I) and (II), 2-alkylideneaminooxyacetamidecompounds of formula (I), heterocyclic compounds of fourrnulas (VI) and (VII), and pyrazole compounds of formula (III) described in US. Patent Application Publication No. 2006/0034786 and US. Patent No. 7,705,041; benzylidene—1,3—thiazolidine compounds of formula (I) described in US. Patent Application Publication No. 2007/0071699; phenylfurylmethylthiazolidine-2,4-dione and thienylmethylthiazolidine-2,4-dione compounds described in US. Patent Application Publication No. 2007/0078175; thiazolidenedione derivatives described in US. Patent Application ation No. 2011/0269954; phenylfuran, phenylthiophene, or W0 2018f017582 phenylpyrrazole compounds described in US. Patent No. 641, 5-(3,5-disubstituted phenylazo)hydroxybenzene-acetic acids and salts and lactones described in US. Patent No. 676, and azo compounds described in US. Patent No. 4,889,846.
The 15-PGDH inhibitors described herein can be provided in a ceutical composition or ic composition depending on the pathological or cosmetic condition or disorder being treated. A pharmaceutical composition containing the H inhibitors described herein as an active ingredient may be manufactured by mixing the tive with a pharmaceutically acceptable carrier(s) or an excipient(s) or diluting the 15-PGDH inhibitors with a diluent in accordance with conventional methods. The pharmaceutical composition may r n fillers, anti-cohesives, lubricants, wetting agents, flavoring agents, emulsifying agents, preservatives and the like. The pharmaceutical composition may be formulated into a suitable formulation in accordance with the methods known to those skilled in the art so that it can provide an ate, controlled or sustained release of the 15—PGDH inhibitors after being administered into a mammal.
In some embodiments, the pharmaceutical composition may be formulated into a parenteral or oral dosage form. The solid dosage form for oral administration may be manufactured by adding excipient, if necessary, together with binder, disintegrants, lubricants, ng agents, and/or flavoring , to the 15-PGDH inhibitors and shaping the resulting mixture into the form of tablets, coated pills, granules, powder or capsules. The additives that can be added in the composition may be ordinary ones in the art.
For example, examples of the excipient include lactose, sucrose, sodium chloride, glucose, starch, calcium ate, kaolin, microcrystalline ose, silicate and the like. Exemplary binders include water, ethanol, propanol, sweet syrup, sucrose solution, starch solution, gelatin solution, carboxymethylcellulose, hydroxypropyl cellulose, hydroxypropyl starch, methylcellulose, ethylcellulose, shellac, calcium phosphonate and polypyrrolidone. es of the disintegrant include dry starch, sodium arginate, agar powder, sodium bicarbonate, calcium carbonate, sodium lauryl sulfate, stearic monoglyceride and lactose.
Further, ed talc, stearates, sodium borate, and polyethylene glycol may be used as a lubricant; and sucrose, bitter orange peel, citric acid, tartaric acid, may be used as a flavoring agent. In some embodiments, the pharmaceutical composition can be made into aerosol formulations (e. g., they can be nebulized) to be administered via inhalation.
W0 2018/‘017582 The lS-PGDH inhibitors described herein may be ed with flavoring agents, buffers, stabilizing agents, and the like and incorporated into oral liquid dosage forms such as solutions, syrups or elixirs in accordance with conventional s. One example of the buffers may be sodium citrate. Examples of the stabilizing agents e tragacanth, acacia and gelatin.
In some embodiments, the lS-PGDH inhibitors described herein may be orated into an injection dosage form, for example, for a aneous, intramuscular or intravenous route by adding thereto pH adjusters, buffers, stabilizing agents, relaxants, topical anesthetics. Examples of the pH adjusters and the buffers e sodium citrate, sodium e and sodium phosphate. Examples of the stabilizing agents include sodium lfite, EDTA, thioglycolic acid and thiolactic acid. The topical anesthetics may be procaine HCl, lidocaine HCl and the like. The relaxants may be sodium chloride, glucose and the like.
In other embodiments, the 15-PGDH inhibitors described herein may be incorporated into suppositories in accordance with conventional methods by adding thereto pharmaceutically able carriers that are known in the art, for e, polyethylene glycol, lanolin, cacao butter or fatty acid triglycerides, if necessary, er with surfactants such as Tween.
The pharmaceutical composition may be formulated into various dosage forms as discussed above and then administered through various routes including an oral, inhalational, transdermal, subcutaneous, intravenous or intramuscular route. The dosage can be a pharmaceutically or therapeutically effective amount.
Therapeutically effective dosage amounts of the lS-PGDH inhibitor may be present in varying amounts in various embodiments. For example, in some embodiments, a eutically effective amount of the 15-PGDH inhibitor may be an amount ranging from about 10—1000 mg (e.g., about 20 mg-l,000 mg, 30 mg-l,000 mg, 40 mg-1,000 mg, 50 mg- l,000 mg, 60 mg—l,000 mg, 70 mg-l,000 mg, 80 mg-l,000 mg, 90 mg-1,000 mg, about -900 mg, 10—800 mg, 10-700 mg, 10-600 mg, 10-500 mg, 100-1000 mg, 100—900 mg, 100-800 mg, 0 mg, 100-600 mg, 100-500 mg, 100-400 mg, 100-300 mg, 200-1000 mg, 200-900 mg, 200-800 mg, 200-700 mg, 200-600 mg, 200-500 mg, 200-400 mg, 300- 1000 mg, 300—900 mg, 300-800 mg, 0 mg, 300-600 mg, 300—500 mg, 400 mg—l,000 mg, 500 mg—l,000 mg, 100 mg-900 mg, 200 mg-SOO mg, 300 mg-700 mg, 400 mg—700 mg, W0 2018f017582 2017/042620 and 500 mg-6OO mg). In some embodiments, the 15-PGDH inhibitor is present in an amount of or greater than about 10 mg, 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg, 550 mg, 600 mg, 650 mg, 700 mg, 750 mg, 800 mg. In some embodiments, the 15-PGDH inhibitor is present in an amount of or less than about 1000 mg, 950 mg, 900 mg, 850 mg, 800 mg, 750 mg, 700 mg, 650 mg, 600 mg, 550 mg, 500 mg, 450 mg, 400 mg, 350 mg, 300 mg, 250 mg, 200 mg, 150 mg, or 100 mg.
In other embodiments, a therapeutically effective dosage amount may be, for example, about 0.001 mg/kg weight to 500 mg/kg , e. g., from about 0.001 mg/kg weight to 400 mg/kg , from about 0.001 mg/kg weight to 300 mg/kg weight, from about 0.001 mg/kg weight to 200 mg/kg weight, from about 0.001 mg/kg weight to 100 mg/kg weight, from about 0.001 mg/kg weight to 90 mg/kg weight, from about 0.001 mg/kg weight to 80 mg/kg weight, from about 0.001 mg/kg weight to 70 mg/kg weight, from about 0.001 mglkg weight to 60 mg/kg weight, from about 0.001 mg/kg weight to 50 mg/kg weight, from about 0.001 mg/kg weight to 40 mg/kg weight, from about 0.001 mgfkg weight to mg/kg weight, from about 0.001 mg/kg weight to 25 mg/kg weight, from about 0.001 mg/kg weight to 20 mg/kg weight, from about 0.001 mg/kg weight to 15 mg/kg weight, from about 0.001 mg/kg weight to 10 mg/kg weight.
In still other embodiments, a therapeutically effective dosage amount may be, for example, about 0.0001 mg/kg weight to 0.1 mg/kg weight, e.g. from about 0.0001 mg/kg weight to 0.09 mglkg , from about 0.0001 mg/kg weight to 0.08 mgikg weight, from about 0.0001 mgfkg weight to 0.07 mg/kg weight, from about 0.0001 mg/kg weight to 0.06 mg/kg weight, from about 0.0001 mg/kg weight to 0.05 mg/kg weight, from about 0.0001 mg/kg weight to about 0.04 mg/kg weight, from about 0.0001 mg/kg weight to 0.03 mg/kg weight, from about 0.0001 mg/kg weight to 0.02 mg/kg weight, from about 0.0001 mgikg weight to 0.019 mg/kg weight, from about 0.0001 mg/kg weight to 0.018 mglkg weight, from about 0.0001 mg/kg weight to 0.017 mg/kg weight, from about 0.0001 mgikg weight to 0.016 mg/kg weight, from about 0.0001 mg/kg weight to 0.015 mglkg weight, from about 0.0001 mg/kg weight to 0.014 mg/kg weight, from about 00001 mg/kg weight to 0.013 mg/kg weight, from about 00001 mg/kg weight to 0.012 mg/kg weight, from about 0.0001 mg/kg weight to 0.011 mg/kg weight, from about 0.0001 mg/kg weight to 0.01 mg/kg weight, from about 0.0001 mg/kg weight to 0.009 mg/kg , from about 0.0001 mg/kg weight to 0.008 mg/kg weight, from about 0.0001 mg/kg W0 2018/‘017582 weight to 0.007 mg/kg weight, from about 0.0001 mg/kg weight to 0.006 mg/kg weight, from about 0.0001 mg/kg weight to 0.005 mg/kg weight, from about 0.0001 mg/kg weight to 0.004 mg/kg weight, from about 0.0001 mg/kg weight to 0.003 mg/kg weight, from about 0.0001 mg/kg weight to 0.002 mg/kg . In some embodiments, the eutically effective dose may be 0.0001 mg/kg weight, 0.0002 mg/kg weight, 0.0003 mg/kg weight, 0.0004 mg/kg weight, 0.0005 mg/kg weight, 0.0006 mg/kg weight, 0.0007 mg/kg weight, 0.0008 mg/kg weight, 0.0009 mg/kg weight, 0.001 mg/kg weight, 0.002 mg/kg weight, 0.003 mg/kg weight, 0.004 mg/kg weight, 0.005 mg/kg weight, 0.006 mg/kg weight, 0.007 mg/kg , 0.008 mg/kg weight, 0.009 mg/kg weight, 0.01 mg/kg weight, 0.02 mg/kg weight, 0.03 mg/kg weight, 0.04 mg/kg weight, 0.05 mg/kg weight, 0.06 mg/kg weight, 0.07 mg/kg weight, 0.08 mg/kg weight, 0.09 mg/kg weight, or 0.1 mg/kg weight.
The effective dose for a particular individual can be varied (e. g., sed or decreased) over time, depending on the needs of the individual.
In some embodiments, a therapeutically effective dosage may be a dosage of ug/kg/day, 50 ug/kg/day, 100 ug/kg/day, 250 ug/kg/day, 500 ug/kg/day, 1000 ug/kg/day or more. In various embodiments, the amount of the 15-PGDH inhibitor or pharmaceutical salt thereof is sufficient to provide a dosage to a patient of between 0.01 ug/kg and 10 ug/kg; 0.1 ug/kg and 5 ugfkg; 0.1 ug/kg and 1000 ug/kg; 0.1 ug/kg and 900 ug/kg; 0.1 ugfkg and 900 ug/kg; 0.1 ug/kg and 800 ug/kg; 0.1 ug/kg and 700 ug/kg; 0.1 ug/kg and 600 ugfkg; 0.1 ug/kg and 500 ug/kg; or 0.1 ug/kg and 400 ug/kg.
] Particular doses or amounts to be stered in accordance with the present invention may vary, for example, depending on the nature and/or extent of the desired outcome, on particulars of route and/or timing of administration, and/or on one or more characteristics (e. g., weight, age, personal history, genetic characteristic, lifestyle parameter, severity of cardiac defect and/or level of risk of c defect, etc., or combinations thereof).
Such doses or amounts can be ined by those of ordinary skill. In some embodiments, an appropriate dose or amount is determined in accordance with standard clinical techniques.
For example, in some embodiments, an appropriate dose or amount is a dose or amount sufficient to reduce a e ty index score by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, , 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100% or more.
For example, in some embodiments, an appropriate dose or amount is a dose or amount sufficient to reduce a disease severity index score by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, W0 2018f017582 , 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100%.
Alternatively or additionally, in some embodiments, an appropriate dose or amount is determined h use of one or more in vitro or in viva assays to help identify desirable or optimal dosage ranges or amounts to be administered.
Various embodiments may include differing dosing n. In some embodiments, the 15-PGDH inhibitor can be administered via continuous infusion. In some ments, the continuous infusion is intravenous. In other embodiments, the continuous infusion is subcutaneous. Alternatively or additionally, in some embodiments, the l5-PGDH inhibitor can be administered bimonthly, monthly, twice monthly, triweekly, biweekly, weekly, twice weekly, thrice weekly, daily, twice daily, or on another clinically desirable dosing schedule. The dosing regimen for a single t need not be at a fixed interval, but can be varied over time, depending on the needs of the subject.
For topical application, the ition can be administered in the form of aqueous, alcoholic, aqueous—alcoholic or oily solutions or suspensions, or of a dispersion of the lotion or serum type, of ons that have a liquid or semi-liquid consistency or are pasty, obtained by dispersion of a fatty phase in an aqueous phase (O/W) or vice versa (W/O) or multiple emulsions, of a free or compacted powder to be used as it is or to be orated into a physiologically acceptable medium, or else of microcapsules or microparticles, or of vesicular dispersions of ionic and/or nonionic type. It may thus be in the form of a salve, a tincture, milks, a cream, an ointment, a powder, a patch, an impregnated pad, a solution, an emulsion or a lar dispersion, a lotion, aqueous or anhydrous gels, a spray, a sion, a shampoo, an aerosol or a foam. It may be anhydrous or s. It may also comprise solid ations constituting soaps or cleansing cakes.
Pharmaceutical and/or cosmetic compositions including the 15—PGDH tor described herein can additionally contain, for example, at least one compound chosen from prostaglandins, in particular prostaglandin PGEl, PGEZ, their salts, their esters, their analogues and their derivatives, in particular those bed in WO 95/11003, JP 97—100091, JP 96-134242, in particular agonists of the prostaglandin receptors. It may in particular contain at least one compound such as the agonists (in acid form or in the form of a precursor, in particular in ester form) of the prostaglandin F20t receptor, such as for example latanoprost, fluprostenol, cloprostenol, bimatoprost, unoprostone, the ts (and their precursors, in particular the esters such as travoprost) of W0 2018/‘017582 the prostaglandin E2 receptors such as 17-phenyl PGEZ, viprostol, butaprost, misoprostol, stone, 16,16-dimethyl PGEZ, 11-deoxy PGE1, 1-deoxy PGEl, the agonists and their precursors, in particular esters, of the prostacycline (IP) receptor such as cicaprost, iloprost, isocarbacycline, beraprost, eprostenol, treprostinil, the agonists and their precursors, in particular the esters, of the prostaglandin D2 receptor such as BW245C ((4S)—(3—[(3R,S) cyclohexyl-3—isopropyl]-2,5-dioxo)imidazolidinehept- anoic acid), BW246C ((4R)-(3- [(3R,S)cyclohexy1—3-isopropyl]-2,5-dioxo)imidazolidinehept- anoic acid), the agonists and their sors, in particular the esters, of the or for the thromboxanes A2 (TP) such as I-BOP la,2a(Z), 3b(1E,3S),4a]]—7-[3-[3-hydroxy[4-(iodophenoxy)-l- l]—7—oxabicyclo- [2.2.1]heptyl]—5-heptenoic acid).
] Advantageously, the composition can e at least one 15-PGDH inhibitor as defined above and at least one prostaglandin or one prostaglandin derivative such as for example the prostaglandins of series 2 ing in particular PGFZU and PGE2 in saline form or in the form of sors, in particular of the esters (example isopropyl esters), their derivatives such as 16,16-dimethyl PGEZ, 17-pheny1 PGE2 and 16,16-dimethyl PGFga 17-phenyl PGan, prostaglandins of series 1 such as 11-deoxyprostaglandin E1, 1—deoxyprostaglandin E1 in saline or ester form, is their analogues, in particular latanoprost, travoprost, fluprostenol, unoprostone, bimatoprost, cloprostenol, viprostol, butaprost, misoprostol, their salts or their esters.
The invention is further illustrated by the following examples, which is not intended to limit the scope of the claims.
Example 1 The following Example describes the synthesis of SW033291 and analogues thereof as well as provides mass spectrometry and NMR confirmation of the structures.
W0 2018/‘017582 O O HJkHQ + FHJVPPh3 i’ s H Cl 5‘ 1.0 e . 0 JK/CN v q R N S F13 FI1 N\ SVS‘R M HzN i E13N 1.5 equiv | 3 F11 R2—’ / dabco or pIperdIne / CN CN CH3CN reflux 45 min EtOH, reflux ' T KOH/EtOH Fi R2 0 0 H202 1.5 equiv.
A + JL R1 CH3 H R2 AC0HICH30| 32 |3C 0 KOH or KOtBu, R1 N S / g S DMF, 37 °C ~ 0” 2 IOSI I H o / 2 15 equiv \ 5 R3 / equiv.0.6 SVSR ACOH/CH3CI 32 °C R NH2 3-phenyl(thiophenyl)propenone was prepared from benzaldehyde and 1-(thiophenyl)ethanone via aldol sation using procedure described by Azam (Parveen, H.; Iqbal, P. F.; Azam, A. Synth. C0mmu., 2008, 38, 3973). 1H NMR (400 MHZ, CDC13) 5 7.88 — 7.80 (rn, 2H), 7.67 (dd, J = 4.9, 1.1 Hz, 1H), 7.66 — 7.59 (m, 2H), 7.47 — 7.34 (m, 4H), 7.18 (dd, J: 5.0, 3.8 Hz, 1H). ESI—MS (m/z): 215 [M+H]+. 4—phenyl(thiophenyl)thioxo-1,2-dihydropyridinecarbonitrile. To a solution of 3-phenyl-l-(thiophenyl)propenone (2.34 mmol, 500 mg) and cyanothioacetamjde (7.0 mmol, 717 mg, 3.0 equiv.) in ethanol (7 mL), a few drops of piperidine were added. The on was refluxed for 3 h. The solid that formed was ted and recrystallized from acetic acid to give designed product in 46 % isolated yield. 1H NMR (400 MHz, DMSO-ds) 8 8.17 (d, J = 3.8 Hz, 1H), 7.96 (d, J: 5.0 Hz, 1H), 7.74 — 7.62 (m, 2H), 7.54 (dd, J = 5.1, 2.0 Hz, 3H), 7.31 — 7.19 (m, 1H), 7.01 (s, 1H). ESI-MS (m/z): 295 [M+H]+.
W0 2018f017582 utylthio)methyl)sulfinyl)phenyl(thiophenyl)nicotinonitrile.
Acetic Acid (900 uL) and hydrogen peroxide (0.57 mmol, 1.5 equiV., 30 % solution in water) were added to the on of 2-(((butylthio)methyl)sulfinyl)phenyl(thiophen—2- yl)nicotinonitrile (0.38 mmol, 150 mg) in chloroform (900 uL). The reaction mixture was stirring at 32 0C for 45 min. The reaction was then diluted with EtOAc and washed with saturated NaHC03 solution, dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 153 mg of designed product (98 %). 1H NMR (400 MHz, CDCl3) 5 7.75 (dd, J = 3.8, 1.1Hz, 1H), 7.66 — 7.57 (m, 2H), 7.58 — 7.51 (m, 4H), 7.47 (s, 1H), 7.16 (dd, J = 5.0, 3.8 Hz, 1H), 4.74 (d, J: 13.0 Hz, 1H), 4.41 (d, J = 13.0 Hz, 1H), 2.97 (dt, J = 13.0, 8.2 Hz, 1H), 2.81 (dt, J: 12.9, 7.3 Hz, 1H), 1.94 — 1.76 (m, 2H), 1.53 — 1.38 (m, 2H), 0.94 (t, J = 7.4 Hz, 3H). ESI—MS (m/z): 413 [M+H]+.
S /NI S 8/0 SW033291 2-(butylsulfinyl)phenyl(thiophenyl)thieno[2,3—b]pyridin amine was prepared using procedure describe by n in V.E. Russian. Chem.
Bull, Int. Ed., 2006, 55, 529). To the solution of 4-(((butylthio)methyl)sulfinyl)—2,6- diphenylpyrimidinecarbonitrile (0.53 mmol, 220 mg) in DMF (0.25 M)/ EtOH (0.5 M) was added KOH (0.32 mmol, 18 mg, 0.6 equiV., 0.1 M in water). The reaction e was stirred at 35 0C for 40 min. Once complete, the reaction was diluted with EtOAc and washed with 10 % aq. solution of acidic acid, the organic phase was separated and aqueous layer was extracted twice with EtOAc, dried over magnesium sulfate, ed and concentrated under reduced pressure to give 211 mg of SW033291 2-(butylsulfinyl)phenyl—6—(thiophen—2— yl)thieno[2,3-b]pyridinamine (96 %). 1H NMR (400 MHz, CDCl3) 5 7.67 — 7.60 (m, 1H), 7.57 — 7.35 (m, 7H), 7.10 (dd, J = 5.0, 3.7 Hz, 1H), 4.54 (s, 2H), 3.26 (ddd, J = 12.8, 9.1, 6.0 Hz, 1H), 3.09 (ddd, J = 12.8, 9.1, 6.6 Hz, 1H), 1.83 — 1.61 (m, 2H), 1.53 —1.38(m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 413 [M+H]+.
W0 2018/‘017582 ] SW208437 4-phenyl(propylsulfinyl)(thiophenyl)thieno[2,3—b]pyridin- 3-amine was prepared in 56 % ed yield using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 5 7.65 (dd, J = 3.8, 1.1 Hz, 1H), 7.61 - 7.49 (m, 4H), 7.49 - 7.41 (m, 3H), 7.12 (dd, J: 5.0, 3.7 Hz, 1H), 3.28 (ddd, J: 12.7, 8.4, 6.3 Hz, 1H), 3.07 (ddd, J: 12.7, 8.6, 7.0 Hz, 1H), 1.91 - 1.65 (m, 2H), 1.08 (t, J: 7.4 Hz, 3H). APCI—MS (m/z): 399 [M+H]+.
SW208438 propylsulfinyl)pheny1(thiopheny1)thieno[2,3- b]pyridinamine was prepared in 48 % isolated yield using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHZ, CDC13) 5 7.64 (dd, J = 3.7, 1.1 Hz, 1H), 7.58 - 7.47 (m, 5H), 7.47 -7.39 (m, 2H), 7.10 (dd, J: 5.0, 3.7 Hz, 1H), 4.59 (s, 2H), 3.38 (p, J: 6.8 Hz, 1H), 1.43 (d, J: 6.9 Hz, 3H), 1.25 (d, J: 6.8 Hz, 3H). ESI-MS (m/z): 399 [M+H]+.
SW208488 2-(butylsulfinyl)methyl(thiophenyl)thieno[2,3-b]pyridin amine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 5 7.55 (dd, J = 3.7, 1.2 Hz, 1H), 7.39 (dd, J = 5.0, 1.1 Hz, 1H), 7.25 — 7.23 (m, 1H), 7.06 (dd, J: 5.0, 3.7 Hz, 1H), 5.02 (s, 2H), 3.25 (ddd, J: 12.7, 9.1, 6.0 Hz, 1H), 3.08 (ddd, J: 12.8, 9.2, 6.4 Hz, 1H), 2.74 (s, 3H), 1.82 — 1.58 (m, 2H), 1.56 - 1.38 (m, 2H), 0.93 (t, J: 7.3 Hz, 3H). ESI—MS (m/z): 351 [M+H]+.
W0 17582 <9N /N s ,0 \'/ S NH,L\_ SW208496 2-(buty1su1finyl)(oxazoly1)pheny1thieno[2,3—b]pyridin amine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 5 7.99 (s, 1H), 7.84 (d, J = 0.8 Hz, 1H), 7.58 - 7.41 (m, 5H), 7.33 (d, J: 0.8 Hz, 1H), 4.65 (s, 2H), 3.30 (ddd, J: 12.9, 8.8, 6.2 Hz, 1H), 3.10 (ddd, J = 12.8, 8.9, 6.9 Hz, 1H), 1.86 - 1.64 (m, 2H), 1.42 — 1.54 (m, 2H), 0.93 (t, J: 7.4 Hz, 3H). ESI—MS (mlz): 398.1 [M+H]+. 36 6-(butylsulfinyl)pheny1(thiazoly1)thien0[2,3-dipyrimidin amine was prepared by synthetic procedures bed for the preparation of analog SW208065. 1H NMR (400 MHz, CDC13) 5 8.06 (dd, J = 3.1, 1H), 7.75 - 7.66 (m, 2H), 7.75 - 7.66 (m, 3H), 7.55 (dd, J: 3.1, 1H), 4.87 (s, 2H), 3.30 (ddd, J: 12.8, 8.4, 6.3 Hz, 1H), 3.12 (ddd,J=12.8, 8.6, 6.9 Hz, 1H), 1.85 - 1.65 (m, 2H), 1.55 — 1.40 (m, 2H), 0.95 (t, J: 7.3 Hz, 3H). ESI-MS (mil): 415.1 [M+H]+. n-BuLi 2.0 equiv. PdClzdppf 10 mol% BuS-SBu 4.0 equiv. PhB(OH)2 2.0 equiv.
Cl /N s s | N / Br THF _78 ac OlmsiCsco32.0equiv./ / s \ Cu.C|10equiv. i S DMF 100 0c \—\_ SW208430 HZSO4IHN03 PdCIdepf 10 mol% S PhB(OH)2 2.0 equiv. CHacIIAcOH CHSCIIAcOH \l S <—C' S / / H202,32°C H202,80°C \—\_ | s 05603 2.0 equiv. \ / CuCl 1.0 equiv.' \—\_ DMF 100 DC swzoa4a4 /N s i S NH,‘—\_ swzoams W0 2018f017582 N S / P \ / S\_\_ SW208432 2-(butylsulfinyl)phenylthieno[2,3-b]pyridine. Acetic Acid (90 uL) and hydrogen peroxide (0.06 mmol, 1.5 equiv., 30 % solution in water) were added to the solution of 2-(butylthio)phenylthieno[2,3-b]pyridine (0.04 mmol, 12 mg) in form (90 ML). The reaction mixture was stirring at 32°C for 1 h. The reaction was then diluted with EtOAc and washed with saturated NaHC03 solution, dried over magnesium sulfate, filtered and concentrated under reduced pressure to give designed product in 76 % isolated yield. 1H NMR (400 MHz, CDCl3) 5 8.14 (d, J = 8.4, 1H), 8.08 (d, J = 8.2, 2H), 7.82 (d, J: 8.4, 1H), 7.58 —7.36 (m, 4H), 3.30 - 2.73 (m, 2H), 1.90 - 1.62 (m, 2H), 1.55 - 1.41 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 316.1 [M+H]+.
] SW208434. Acetic Acid (200 uL) and hydrogen peroxide (0.15 mmol, 30 % solution in water) were added to the solution of 2-(butylthio)phenylthieno[2,3—b]pyridine (0.09 mmol, 27 mg) in chloroform (200 uL). The reaction mixture was stirring at 100°C for min. The reaction was then diluted with EtOAc and washed with saturated NaHC03 solution, dried over magnesium sulfate, filtered and concentrated under d pressure to give designed product in 81 % ed yield. 1H NMR (400 MHz, CDCl3) 5 8.23 (d, J = 8.5 Hz, 1H), 8.09 (dd, J: 8.2, 1.6 Hz, 2H), 7.89 (d, J = 2.1 Hz, 1H), 7.59 - 7.39 (m, 4H), 3.39 - 3.10 (m, 2H), 1.92 - 1.68 (m, 2H), 1.54 - 1.27 (m, 2H), 0.91 (t, J: 7.3 Hz, 3H). ESI—MS (m/z): 332.1 [M+H]+. \ i 8w SW208430 ylthio)phenylthieno[2,3-b]pyridine. Phenylboronic acid (0.39 mmol, 2.0 , 2-(butylthio)chlorothieno[2,3-b]pyridine (50 mg, 0.195 mmol, 1.0 equiv), Cesium Carbonate (0.39 mmol, 2.0 equiv.), PdClzdppf (10 mol%), Copper Chloride (0.195 mmol, 1.0 equiv.) were heated in DMF at 100°C for 12 h. After cooling to r.t. the W0 2018/‘017582 reaction mixture was d with EtOAc and washed with water and next brine. The organic layer was dried over magnesium sulfate and the solvent was removed under reduced pressure.
The crude t was purified by flash chromatography (Hexanes/EtOAc: 8/2) to afford designed product in 32 % yield. 1H NMR (400 MHZ, CDC13) 5 8.09 - 8.00 (m, 2H), 7.93 (d, J: 8.3 Hz, 1H), 7.69 (d, J: 8.3 Hz, 1H), 7.52 - 7.34 (m, 4H), 2.99 (t, J: 7.9 Hz, 2H), 1.77 - 1.63 (m, 2H), 1.53 - 1.38 (m, 2H), 0.92 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 300.1 [M+H]+.
CI N wsis/ 2—(buty1thio)chlorothieno[2,3-b]pyridine. To the solution of 2—bromo chlorothieno[2,3-b]pyridine (40 mg, 0.16 mmol) in THF (2 mL) at -78°C was added n-BuLi (0.32 mmol, 2.0 equiv.; 1.6 M solution in hexanes). The traction mixture was stirred for 5 min. and 1,2—dibutyldisulfane (0.48 mmol, 85.4 mg) was then added. The on mixture was stirred at —78°C for additional 1h, quenched with water and diluted with EtOAc. The organic layer was separated, dried over MgSO4, filtered and concentrated to give crude t, which was purified by flash column tography (95/5 Hexane/EtOAc) to give designed product in 91 % isolated yield. 1H NMR (400 MHz, CDC13) 5 7.81 (d, J: 8.3 Hz, 1H), 7.24 (d, J = 8.3 Hz, 1H), 7.13 (s, 1H), 3.01 - 2.89 (m, 2H), 1.74 - 1.59 (m, 2H), 1.52 - 1.36 (m, 2H), 0.91 (t, J: 7.4 Hz, 3H). ESI-MS (m/z): 258.0 [M+H]+. cu mi/N| s 8 2—(butylthio)chloronitrothieno[2,3-b]pyridine was prepared in 53 % yield according procedure described by Nardine Cohn, 0.; Narine, B. Telmhedon Lett.. 1978, 23, 2045.). 1H NMR (400 MHz, CDC13) 5 8.68 (d, J = 8.6 Hz, 1H), 7.45 (d, J: 8.6 Hz, 1H), 3.15 (t, J: 7.4Hz, 2H), 1.95 - 1.73 (m, 2H), 1.68 - 1.41 (m, 2H), 0.99 (t, J: 7.4 Hz, 3H). ESI—MS (m/z): 303.0 [M+H]+.
W0 2018/‘017582 N S \\ / S\_fi\__ 35 2-(butylthio)phenylthieno[2,3-b]pyridinamine. Phenylboronic acid (37 mg, 0.30 mmol, 2.0 equiv), 2-(butylthio)chloronitrothieno[2,3-b]pyridine (46 mg, 0.15 mmol, 1.0 equiv), Cesium Carbonate (0.30 mmol, 2.0 equiv.), PdClgdppf (10 mol%), Copper Chloride (0.15 mmol, 15 mg, 1.0 equiv.) were heated in DMF at 100°C for 12 h. After cooling to r.t. the reaction mixture was diluted with EtOAc and washed with water and next brine. The c layer was dried over magnesium sulfate and the t was removed under reduced pressure. The crude product was ed by preparative TLC (AcOEt/Hexanes: 2/8) to afford 2-(butylthio)nitrophenylthieno[2,3-b]pyridine. ESI-MS (m/z): 345.1 [M+H]+. 2-(butylthio)nitrophenylthieno[2,3-b]pyridine {0.017 mmol, 6 mg) was dissolved in a mixed t of acetic acid (0.12 ml.) and cone. hydrochloric: acid (one drop). Zinc (13 mg) was added at 0°C. After the mixture was stirred for 30 minutes. the reaction mixture was ed, and the filtrate was neutralized with an aqueous solution of NaHC03, and extracted with DCM. The organic layer was washed with water and then with a saturated aqueous solution of sodium chloride, and dried over sodium sulfate.
Subsequently, the solvent was evaporated to obtain designed product. 1H NMR (400 MHZ, CDC13) 5 7.65 —7.54 (m, 3H), 7.50 - 7.40 (m, 2H), 7.35 -7.28 (m, 1H), 7.14 (d, J: 8.4 Hz, 1H), 3.35 - 3.18 (m, 2H), 1.80 - 1.65 (m, 2H), 1.54 -1.38 (m, 2H), 0.95 (t, J: 7.3 Hz, 3H).
ESI-MS (m/z): 315.1 [M+H]+. 2-bromochlorothieno[2,3-b]pyridine was prepared according procedure dmammedeme1IHNMRQMOMHLCDCQS787fiLJ=84HL1HL728@JHL 7.27 (d, J = 8.4 Hz, 1H). ESI-MS (m/z): 249 [M+H]+.
W0 2018/‘017582 / \ / Jk / \ l H N NH N S 2 N \ N\ NH? HZN ”Hz 8 \ I Zn 10 equiv. S I I )=S / —> / N02 / 5-0 9‘1“”, N NH4C| 15 equiv. NH2 H PdClzdppf 10 mol% K20031.1 equiv.
ThB(OH)2 2.0 equiv.
M1'0 equw.. Br 05003 2.0 equiv.
CuCI 1.0 equiv.
DMF 100°C / l /_/_ / H H2021.5 equlv l 3 N\ s N\ “ /_/_ I />—s I />—8 / ‘ / NH2 N o N SW2084-95 94 N NH S 2 ] 3-nitro—6-(thiophenyl)pyridinamine. Thiophene boronic acid (742 mg, .8 mmol, 2.0 equiv), ronitropyridinamine (500 mg, 2.9 mmol, 1.0 equiv), Cesium Carbonate (5.8 mmol, 2.0 equiv), PdClgdppf (10 mol%), Copper Chloride (2.9 mmol, 1.0 equiv.) in DMF were heated at 100°C for 12 h. After cooling to r.t. the reaction mixture was diluted with EtOAc and washed with water and next brine. The organic layer was dried over magnesium sulfate and the solvent was removed under reduced re.
The crude product was purified by column tography (hexanes/ EtOAc: 8/2) to afford 3-nitro-6—(thiophen—2-yl)pyridinamine in 63 % yield. 1H NMR (400 MHZCDC13) 6 8.42 (d, J: 8.7 Hz, 1H), 7.70 (dd, J: 3.8, 1.1Hz, 1H), 7.54 (dd, J: 5.0, 1.1Hz, 1H), 7.15 (dd, J = 5.0, 3.8 Hz, 1H), 7.09 (d, J: 8.7 Hz, 1H). ESI-MS (m/z): 222 [M+H]+. s N\ NH2 6-(thiophenyl)pyridine-2,3-diamine. The starting material, 3-nitro (thiophen—2—yl)pyridin—2—amine (1.20 mmol, 265.4 mg), was dissolved in a 5:1 acetone/water mixture. Zinc (12.0 mmol, 784 mg, 10 eq) and ammonium chloride (18 mmol, 962.5 mg, 15 eq) were added to the solution, which was stirred at room temperature for 1 hour. The solution was then filtered through a celite pad and washed with ethyl acetate. The filtrate was W0 2018/‘017582 extracted twice with brine then the aqueous layer was back ted with EtOAc. The combined organic layers were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. Further cation by column chromatography gave 118.2 mg of 6- (thiophen—2—y1)pyridine-2,3-diamine (52 %). 1H NMR (400 MHz, CD3OD) 5 7.34 (dd, J = 3.6, 1.1Hz, 1H), 7.25 (dd, J: 5.1, 1.1Hz, 1H), 7.00 (dd, J: 5.1, 3.6 Hz, 1H), 6.96 — 6.86 (m, 2H), 4.85 (s, 4H). ESI-MS (m/z): 192 [M+H]+.
/ \ H 3 IN\ N>=s 5-(thiophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridine-2—thione. Thiourea (16.97 mmol, 223.0 mg, 5 eq) was added to 6-(thiophenyl)pyridine-2,3—diamine. The solution was heated at 170°C for 2 hours. The addition of ethanol room temperature produced solid which was filtered to give 112.5 mg of 5-(thiophenyl)-1,3-dihydro-2H— imidazo[4,5—b]pyridine—2—thione (82 %). 1H NMR (400 MHZ, (CD3)2SO) 5 7.69 (dd, J = 3.7, 1.2 Hz, 1H), 7.66 (d, J: 8.3 Hz, 1H), 7.56 (dd, J: 5.1, 1.1Hz, 1H), 7.47 (d, J: 8.2 Hz, 1H), 7.15 — 7.09 (m, 1H). ESI-MS (m/z): 235 [M+2H]+. 94 ylthio)(thiophenyl)-3H-imidazo[4,5-b]pyridine. A mixture of 5-(thiophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridinethione (0.39 mmol, 92 mg), potassium carbonate (0.45 mmol, 61.9 mg, 1.1 eq), 1-bromobutane (0.39 mmol, 42.8 uL, 1 eq), 18-Crown-6 (0.039 mmol, 10.5 mg, 0.1 eq), and DMF (2.67 mL) was heated at 80 0C for 3 hours. This solution was then diluted with EtOAc and washed with water. The organic layer was dried over magnesium sulfate, filtered, and concentrated under high re to give 74.4 mg of SW208494 2-(butylthio)(thiophenyl)-3H-imidazo[4,5- b]pyridine (65 %). 1H NMR (400 MHZ, CDC13) 5 7.95 — 7.83 (m, 1H), 7.61 — 7.53 (m, 2H), 7.36 (d, J: 5.1, 1H), 7.16 — 7.06 (m, 1H), 3.28 (t, J: 7.3 Hz, 2H), 1.76 — 1.62 (m, 2H), 1.48 — 1.32 (m, 2H), 0.90 (t, J = 7.4 Hz, 3H). ESI—MS (m/z): 290 [M+H]+.
W0 2018/‘017582 /S\ N z: ,0 I; %z\ S\_\— SW208495. ylsulfinyl)(thiophenyl)-3H—imidazo[4,5—b]pyridine.
Chloroform (450 uL), acetic acid (450 uL), and hydrogen peroxide (0.376 mmol, 2.0 eq, 40 uL) were added to SW208494 2-(butylthio)(thiophenyl)-3H-imidazo[4,5-b]pyridine and heated at 45°C for 2.5 hours. The solution was then diluted with EtOAc and washed with 10 % acetic acid. The organic layer was separated, dried with magnesium e, filtered, concentrated, and purified to give 16.8 mg of SW208495. 1H NMR (400 MHZ, CDC13) 8 8.06 (d, J = 8.5 Hz, 1H), 7.83 — 7.67 (m, 1H), 7.68 — 7.60 (m, 1H), 7.41 (d, J = 5.3Hz, 1H), 720—715(nL1HL344—3J7(nL2HL189—158(nL2HL159—140(nL2HL093(LJ = 7.3 Hz, 3H). ESI—MS (m/z): 306 [M+H]+.
...... Luz/9‘ S‘fk-‘l-itz-SE’Es‘i Si'u'K’LEUSYEIHZ @N/| S 800 N / \ \/\/ SW208662. 6-(butylsulfinyl)phenyl(piperidinyl)thieno[2,3— d]pyrimidine. Acetic acid (50 pl) and hydrogen peroxide (5.0 pl, 30 % solution in water) were added to the solution of 2-(butylthio)phenyl(piperidinyl)thieno[2,3— b]pyrimidine (10 mg, 0.026 mmol) in chloroform (50 pl). The on mixture was stirred at 32 0C for 45 min. Once complete, the reaction was d with EtOAc and was washed with saturated NaHC03 on, dried over magnesium sulfate, filtered and concentrated under reduce pressure to give designed product. 1H NMR (400 MHZ, CDCl3) 5 8.54 — 8.34 (m, 2H), 7.73 (s, 1H), 7.55 — 7.36 (m, 3H), 4.09 — 3.86 (m, 4H), 3.24 — 2.94 (m, 2H), 1.97 — 1.36 (m, 10H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 400.1 .
Gus/kw 2-(butylthio)phenyl(piperidinyl)thieno[2,3 -b]pyrimidine. 2—(butylthio)- 6-chloro-4—(piperidinyl)thieno[2,3-b]pyrimidine (52 mg, 0.15 mmol), boronic acid (27 mg, 0.22 mmol, 1.5 equiv), Potassium Carbonate (0.3 mmol, 2.0 equiv.), PdCl2dtbpf (10 mol mol%), in CH3CN:H2O (2: 1) were heated at 100°C overnight. After cooling to r.t. the reaction mixture was diluted with EtOAc and washed with water. The organic layer was dried over magnesium sulfate and the solvent was removed under reduced pressure. The crude product was purified by flash chromatography to afford designed product. 1H NMR (400 MHZ, CHC13) 8 8.49 — 8.36 (m, 2H), 7.51 — 7.36 (m, 3H), 7.29 (s, 1H), 3.95-3.85 (m, 4H), 2.90 (t, J = 7.4 Hz, 2H), 1.76 — 1.73 (m, 6H), 1.70 — 1.59 (m, 2H), 1.48 — 1.39 (m, 2H), 0.91 (t, J = 7.4 Hz, 3H). ESI-MS (m/z): 384.0 [M+H]+.
W0 2018f017582 2—(butylthio)chloro(piperidinyl)thieno[2,3-b]pyrimidine. To the solution of ro(piperidinyl)thieno[2,3-b]pyrimidine (52 mg, 0.20 mmol) in THF was added n-BuLi (0.4 mmol, 2.0 equiv., 1.6 M solution in s) at -78°C. The reaction mixture was stirred for 5 min and 1,2-dibutyldisulfane (0.80 mmol, 4.0 equiv.) in THF was added. The reaction mixture was stirred for additional 1h at -78 0C and then quenched. The crude product was purified by flash tography to afford designed product in 74 % yield. 1H NMR (400 MHz, CHC13) 5 7.24 (s, 1 H), 3.93 — 3.74 (m, 4H), 2.83 (t, J: 7.3 Hz, 2H), 1.82 — 1.66 (m, 6H), 1.66 — 1.53 (m, 2H), 1.49 — 1.33 (m, 2H), 0.89 (t, J: 7.3 Hz, 3H).
ESI—MS (mfz): 342.1 [M+H]+.
CIYN s N \ / 6—chloro(piperidinyl)thieno[2,3-b]pyrimidine. 4,6-dichlorothieno[2,3- b]pyrimidine (50 mg, 0.24 mmol) and piperidine ( 0.36 mmol, 1.5 equiv.) in EtOH were stirred at room temperature overnight. The solvent was evaporated and crude compound purified by flash chromatography to give designed product in quantitative yield. 1H NMR (400 MHZ, CDC13) 5 7.28 (d, J = 6.1 Hz, 1H), 7.18 (d, J = 6.2 Hz, 1H), 4.01 — 3.67 (m, 4H), 1.92 — 1.63 (m, 6H). ESI-MS (m/z): 254.0 [M+H]+.
SW208776. 6—(butylsulfinyl)-2,4-diphenylthieno[2,3-d]pyrimidine. Acetic acid (250 pl) and hydrogen de (20 pl, 30 % solution in water) were added to the solution of 6-(butylthio)-2,4-diphenylthieno[2,3-d]pyrimidine (35 mg, 0.1 mmol) in chloroform (250 pl).
The on mixture was stirred at 32 °C for 45 min. Once complete, the reaction was W0 2018/‘017582 diluted with EtOAc and was washed with saturated NaHCO3 solution, dried over magnesium sulfate, filtered and concentrated under reduce pressure to give designed product. 1H NMR (400 MHZ, CDC13) 8 8.69 — 8.59 (m, 2H), 8.09 — 7.99 (m, 2H), 7.95 (s, 1H), 7.65 — 7.56 (m, 3H), 7.56 — 7.45 (m, 3H), 3.18 — 3.02 (m, 2H), 1.87 — 1.64 (m, 2H), 1.54 — 1.42 (m, 2H), 0.94 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 393.1 . 6-(butylthio)-2,4-diphenylthieno[2,3-d]pyrimidine. To the solution of 2,4- diphenylthieno[2,3-d]pyrimidine (53 mg, 0.28 mmol) in THF was added n—BuLi (0.56 mmol, 2.0 equiv., 225 ”L, 2.5 M solution in hexanes) at -78°C. The reaction mixture was stirred for min and 1,2-dibutyldisulfane (1.14 mmol, 4.0 equiv.) in THF was added. The reaction e was stirred for additional 1h at -78°C and then quenched. The crude product was purified by flash chromatography to afford designed product. 1H NMR (400 MHz, CDC13) 8 8.62 — 8.56 (m, 2H), 8.06 — 7.98 (m, 2H), 7.61 — 7.41 (m, 7H), 3.01 (t, J: 7.3, 2H), 1.76 — 1.62 (m, 2H), 1.55 — 1.38 (m, 2H), 0.92 (t, J = 7.4 Hz, 3H). ESI-MS (m/z): 377.1 [M+H]+. 2,4-diphenylthieno[2,3-d]pyrimidine. 2,4-dichlorothieno[2,3-d]pyrimidine (100 mg, 0.50 mmol), phenylboronic acid (242 mg, 2.0 mmol, 4.0 equiv), Potassium ate (1.5 mmol, 3.0 ), Pd(OAc)2 (5 mol mol%), SPhos (10 mol%) in CH3CN2H20 (1.521) were heated at 100 0C overnight. After cooling to r.t. the on mixture was diluted with EtOAc and washed with water. The organic layer was dried over magnesium sulfate and the t was removed under reduced pressure. The crude product was purified by flash chromatography to afford designed product. 1H NMR (400 MHZ, CDC13) 5 8.70 — 8.59 (m, 2H), 8.14 — 8.02 (m, 2H), 7.65 — 7.44 (m, 8H). ESI-MS (m/z): 289.0 [M+H]+.
W0 2018/‘017582 SW208777. 2-(butyl(}t1-oxidanyl)-}t3-sulfanyl)(pyridinyl)—6—(thiazol yl)thieno[2,3—b]pyridinan1ine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDCl3) 5 8.80 (s, 1 H), 8.78 (dd, J = 4.9, 1.7 Hz, 1H), 8.04 (s, 1H), 7.91 (d, J: 3.2 Hz, 1H), 7.86 (d, J: 6.4 Hz, 1H), 7.51 (d, J: 3.1 Hz, 1H), 7.47 (dd, J: 7.8, 4.8 Hz, 1H), 4.53 (s, 2H), 3.28 (ddd, J: 12.8, 8.8, 6.3 Hz, 1H), 3.11 (ddd, J: 12.8, 8.9, 6.9 Hz, 1H), 1.86 — 1.70 (m, 2H), 1.57 — 1.38 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 415.0 [M+H]+.
SW208780. 2-(isopropletl-oxidanyl)-}t3-sulfanyl)(pyridiny1)(thiazol- 2—yl)thieno[2,3—b]pyridinamine was ed using tic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 5 8.87 — 8.70 (m, 2H), 8.05 (s, 1H), 7.92 (d, J = 3.1 Hz, 1H), 7.85 (dd, J = 7.8, 2.4 Hz, 1H), 7.51 (d, J = 3.2 Hz, 1H), 7.47 (dd, J = 7.9, 4.9 Hz, 1H), 4.57 (s, 2H), 3.38 (p, J: 6.8 Hz, 1H), 1.43 (d, J: 6.8 Hz, 3H), 1.29 (d, J = 6.8 Hz, 3H). ESI-MS (m/z): 400.1 [M+H]+.
[If] N\Sl,0 SW209123. Ethyl 3-amino(4-bromophenyl)(butylsulfinyl)—6—(thiazol eno[2,3—b]pyridine—5—carboxylate was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 8 7.86 (d, J = 3.2 Hz, 1H), 7.69 — 7.60 (m, 2H), 7.48 (d, J = 3.2 Hz, 1H), 7.36 — 7.27 (m, 2H), 4.12 (q, J: 7.2 Hz, 2H), 3.26 (ddd, J: 12.9, 8.8, 6.3 Hz, 1H), 3.08 (ddd, J: 12.9, 8.8, 6.3 Hz, 1H), 1.80 — 1.63 W0 2018f017582 (m, 2H), 1.58 — 1.3? (m, 2H), 1.06 (t, J = 7.2 Hz, 3H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 564.0 [M+H]+. //\—\_ SW209124. 2-(butylsulfinyl)-4,6-di(thiazolyl)thieno[2,3-b]pyridinamine was ed using synthetic procedures described for the preparation of analog SW03 3291. 1H NMR (400 MHz, CDCl3) 5 8.48 (s, 1H), 8.01 (d, J = 3.1 Hz, 1H), 7.96 (d, J = 3.2 Hz, 1H), 7.61 (d, J = 3.3 Hz, 1H), 7.52 (d, J: 3.1 Hz, 1H), 6.69 (s, 2H), 3.30 (ddd, J = 12.8, 9.2, 6.0 Hz, 1H), 3.14 (ddd, J: 12.8, 9.2, 6.4 Hz, 1H), 1.83 — 1.60 (m, 2H), 1.43 — 1.53 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 421.0 [M+H]+. {iN N S \ [/0 l/ / S\_\_ \N \ N SW209125. 2-(butylsulfinyl)(1-methyl-1H-imidazol—2—y1)—6-(thiazol yl)thieno[2,3—b]pyridinamine was ed using synthetic ures described for the preparation of analog SW033291. 1H NMR (400 MHZ, CDCl3) 5 8.13 (s, 1H), 7.91 (d, J = 3.2 Hz, 1H), 7.50 (d, J: 3.1 Hz, 1H), 7.24 (d, J: 1.2 Hz, 1H), 7.13 (d, J: 1.2 Hz, 1H), 5.78 (s, 2H), 3.80 (s, 3H), 3.26 (ddd, J: 12.8, 9.1, 6.0 Hz, 1H), 3.10 (ddd, J: 12.8, 9.2, 6.5 Hz, 1H), 1.82 — 1.57 (m, 2H), 1.56 — 1.35 (m, 2H), 0.92 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 418.1 [M+H]+. 0N N l |\SS//O// SW209126. 2-(butylsulfinyl)(1-methy1-1H-imidazoly1) phenylthieno[2,3-b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHZ, CDC13) 5 8.08 (s, 1H), 7.58 — W0 2018/‘017582 7.32 (m, 5H), 7.11 (d, J = 1.1 Hz, 1H), 7.00 (d, J: 1.1 Hz, 1H), 4.58 (s, 2H), 4.19 (s, 3H), 3.27 (ddd, J = 12.7, 9.0, 6.0 Hz, 1H), 3.08 (ddd, J = 12.8, 9.1, 6.6 Hz, 1H), 1.79 — 1.60 (m, 2H), 1.56 — 1.37011, 2H), 0.92 (I, J = 7.3 Hz, 3H). ESI—MS (m/z): 411.1 [M+H]+.
{/N‘J‘VYN s [p I l / 8 N\ W 77. 6-(butylsulfinyl)(1-methyl-1H-in1idazolyl)-4— phenylthieno[2,3-af|pyrimidinan1ine was prepared using synthetic procedures described for the preparation of analog SW208065. 1H NMR (400 MHz, CDC13) 5 7.71 (dd, J = 6.9, 2.8 Hz, 2H), 7.63 — 7.49 (m, 3H), 7.29 (s, 1H), 7.07 (s, 1H), 4.85 (s, 2H), 4.18 (s, 3H), 3.29 (ddd, J: 12.8, 8.6, 6.3 Hz, 1H), 3.11 (ddd, J: 12.8, 8.7, 6.9 Hz, 1H), 1.83 — 1.65 (m, 2H), 1.59 — 1.39 (m, 2H), 0.94 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 412.1 .
OWN S ,9 ”\I / Si SW209278. 6-(butylsulfinyl)(oxazolyl)phenylthieno[2,3—d]pyrirnidin- e was prepared using synthetic procedures described for the preparation of analog SW208065. 1H NMR (400 MHz, CDC13) 5 8.51 (d, J: 1.1 Hz, 1H), 8.01 (d, J: 1.1 Hz, 1H), 7.75 — 7.61 (m, 2H), 7.62 — 7.48 (m, 3H), 4.56 (s, 2H), 3.29 (ddd, J: 12.9, 8.8, 6.3 Hz, 1H), 3.09 (ddd, J: 12.9, 8.9, 6.9 Hz, 1H), 1.81 — 1.64 (m, 2H), 1.56 —1.39(m,2H), 0.93 (t, J: 7.3 Hz, 3H). ESI—MS (m/z): 399.1 [M+H]+.
S /ISS/9 SW209279. 2-(isopropylsulfinyl)(1-rnethy1-1H-imidazoly1)(thiazol yl)thieno[2,3-b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHZ, CDC13) 5 8.14 (s, 1H), 7.92 (d, J = W0 2018/‘017582 3.2 Hz, 1H), 7.51 (d, J = 3.2 Hz, 1H), 7.24 (s, 1H), 7.13 (d, J: 1.2 Hz, 1H), 5.92 (s, 2H), 3.80 (s, 3H), 3.38 (p, J = 6.8 Hz, 1H), 1.44 (d, J = 6.8 Hz, 3H), 1.25 (d, J: 6.8 Hz, 3H). ESI- MS (m/z): 404.1 [M+H]+.
\N\_/_\N SW209280. 4-(1-methy1—1H-imidazoly1)(propylsulfiny1)—6—(thiazol yl)thieno[2,3—b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW033291. 1H NMR (400 MHz, CDC13) 5 8.14 (s, 1H), 7.92 (d, J = 3.2 Hz, 1H), 7.51(d,J= 3.1, 1H), 7.25 (d, J: 1.3 Hz, 1H), 7.14 (d, J: 1.2 Hz, 1H), 5.97 (s, 2H), 3.80 (s, 3H), 3.27 (ddd, J: 12.7, 8.3, 6.5 Hz, 1H), 3.07 (ddd, J: 12.8, 8.4, 7.1 Hz, 1H), 1.85 — 1.69 (m, 2H), 1.07 (t, J = 7.4 Hz, 3H). ESI—MS (m/z): 404.1 [M+H]+. 0 <8“ N\ SVSR3 ‘ ' / [N3% PIKE?|/>_R2 £30 / R1)2-c-yanoethanethioamide 3 0 equiv I N idine, EtOH. 80 °C CN l ’>_R2 —PPh 3 . 3 N—R1 N 2.0 equN.
=Me, H 2) CIVSR N=< R2: Me isopropyl. cyclopropyl Eth 3.0 equiv. CH3CN, 80 °C R2 R3: Bu, 03H50CH3, C2H4OCH3 H2021.5 equiv.
CHCIg/ACOH . fl KOH 0.6 equiv. N S SVSR3 DMF/MeOH \ SW209415. 2-(butylsulfinyl)(1,2-dimethy1—1H-imidazolyl)(thiazol eno[2,3-b]pyridinamine. To the solution of 2-(((butylsulfinyl)methy1)thio)( 1,2- dimethyl—1H—imidazolyl)(thiazoly1)nicotinonitrile (0.14 mmol, 60 mg) in DMF W0 2018f017582 (600 ul)/MeOH (300 pl) was added KOH (0.084 mmol, 4.70 mg, 0.6 , 2.0 M in water).
The reaction mixture was stirred at 32°C for 20 min. Once complete, the reaction was diluted with EtOAc and acidified to pH 7 with 5 % aq. solution of AcOH, the organic phase was separated and aqueous layer was extracted twice with EtOAc, dried over ium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash chromatography to afford designed product in 97 % isolated yield. 1H NMR (400 MHz, 6 8.03 (s, 1H), 7.90 (d, J: 3.1 Hz, 1H), 7.50 (d, J = 3.2 Hz, 1H), 7.11 (s, 1H), 4.76 (s, 2H), 3.39 (s, 3H), 3.27 (ddd, J: 12.9, 8.7, 6.4 Hz, 1H), 3.09 (ddd, J: 12.8, 8.8, 6.9 Hz, 1H), 2.47 (s, 3H), 1.83 — 1.62 (m, 2H), 1.57 — 1.38 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 432.1 [M+H]+. Two enantiomers of 15 can be separated by chiral HPLC: Chiralpak AD—H, 10 X 250 mm, 5 uM, 100% MeOH. fl 9 IN\ SVSW 2-(((butylsulfinyl)methyl)thio)(1,2-dimethyl-1H-imidazoly1)(thiazol yl)nicotinonitrile. To the solution of 2-(((butylthio)methyl)thio)—4—(l,2—dimethyl—1H— imidazol—5—yl)—6—(thiazolyl)nicotinonitrile (85 mg, 0.205 mmol) in CHClg/AcOH (1:1, 0.15 M) was added H202 (0.31 mmol, 1.5 equiv. 30% solution in water). The reaction mixture was stirred at 32 0C for 40 min. Once complete, the reaction was diluted with EtOAc and was washed with saturated NaHC03 solution, dried over magnesium e, filtered and concentrated under reduce pressure to give designed product in 92 % yield. 1H NMR (400 MHz, CDC13) 6 7.98 (d, J: 3.1 Hz, 1H), 7.94 (s, 1H), 7.60 (d, J: 3.1 Hz, 1H), 7.43 (s, 1H), 4.72 (d, J: 13.1 Hz, 1H), 4.41 (d, J: , 1H), 3.63 (s, 3H), 2.96 (dt, J: 12.9, 8.2 Hz, 1H), 2.84 (dt, J: 12.9, 7.5 Hz, 1H), 2.51 (s, 3H), 1.94 — 1.74 (m, 2H), 1.63 — 1.38 (m, 2H), 0.95 (t, J = 7.4 Hz, 3H). ESI—MS (m/z): 432.1 [M+H]+.
W0 2018/‘017582 / i” S |N\ SVSW utylthio)methy1)thio)( 1 ,2-dimethyl- 1 azolyl)-6—(thiazol yl)nicotinonitri1e. To a suspension of 3-(1,2-dimethyl-1H-imidazolyl)(thiazol yl)propenone (0.31 mmol, 72 mg) and 2-cyanothioacetamide (0.93 mmol, 93 mg, 3.0 equiv.) in EtOH (1.5 mL), a few drops of piperidine were added. After being stirred at 80°C for 2 h, EtOH was evaporated and crude product was olved in CH3CN.
Butyl(chloromethyl)sulfane (0.62 mmol, 85.5 mg) and Et3N (0.93 mmol, 94.1 mg, 130 uL) were then added and the reaction mixture was stirred at 80°C for 20 min. Once complete, the reaction was diluted with EtOAc and water. The organic phase was separated and aqueous layer was extracted twice with EtOAc. The combined extractions were washed with saturated NaCl solution, dried over ium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography to give 99 mg of designed product (77%). 1H NMR (400 MHz, CDC13) 5 7.96 (d, J = 3.1 Hz, 1H), 7.85 (s, 1H), 7.56 (d, J: 3.1 Hz, 1H), 7.37 (s, 1H), 4.49 (s, 2H), 3.60 (s, 3H), 2.72 (t, J: 7.4 Hz, 2H), 2.48 (s, 3H), 1.62 (p, J = 7.3 Hz, 2H), 1.40 (h, J = 7.3 Hz, 2H), 0.90 (t, J = 7.3 Hz, 3H). ESI— MS (m/z): 416.6 [M+H]+. 83%.?”/N\ / N (E)—3-(1,2-dimethyl-1H-imidazolyl)(thiazolyl)propen—1-one. To a solution of 1,5-dimethyl-1H-imidazolecarbaldehyde (2.0 mmol, 250 mg) in 6 ml of CH3CN was added 1-(thiazolyl)(triphenylphosphanylidene)ethan—1-one (4.0 mmol, 1.55 g, 2.0 equiv.) The reaction mixture was stirred at 90°C for 48 h. Once complete, solvent was evaporated and residue was purified by flash chromatography to give 331 mg of designed product (71%). 1H NMR (400 MHz, ol-d4) 6 8.08 (d, J = 3.0 Hz, 1H), 7.97 (d, J: 3.0 Hz, 1H), 7.90 (d, J: 15.9 Hz, 1H), 7.76 (d, J: 15.9 Hz, 1H), 7.60 (s, 1H), 3.72 (s, 3H), 2.43 (s, 3H).ESI-MS (m/z): 234.3 [M+H]+.
W0 2018f017582 28. 2-(butylsulfinyl)(2-methyl-1H—imidazolyl)—6-(thiazol yl)thieno[2,3—b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW209415. 1H NMR (400 MHz, CDCl3) 5 10.51 (s, 1H), 8.10 (s, 1H), 7.89 (d, J = 3.2 Hz, 1H), 7.46 (d, J: 3.2 Hz, 1H), 7.40 (s, 1H), 3.31 (ddd, J: 12.8, 9.3, 5.8 Hz, 1H), 3.15 (ddd, J: 12.8, 9.3, 6.2 Hz, 1H), 2.42 (s, 3H), 1.79 — 1.58 (m, 2H), 1.57 — 1.38 (m, 2H), 0.93 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 418.1 [M+H]+.
] SW211688. 4-(1,2-dimethyl-1H—imidazolyl)((3-methoxypropyl) sulfinyl)- 6—(thiazol—2—yl)thieno[2,3-b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW209415. 1H NMR (400 MHz, Acetone—d6) 5 8.03 (s, 1H), 7.99 (d, J: 3.2 Hz, 1H), 7.82 (d, J = 3.2 Hz, 1H), 7.09 (s, 1H), 5.06 (s, 2H), 3.51 (s, 3H), 3.48 (t, J: 6.1 Hz, 2H), 3.26 (s, 3H), 3.26 — 3.18 (m, 1H), 3.18 — 3.12 (m, 1H), 2.43 (s, 3H), 2.00 — 1.89 (m, 2H). ESI-MS (n1/z): 448.1 .
SW211689. 4-(1,2-dimethyl-1H—imidazolyl)((2-methoxyethyl) sulfiny1)- 6-(thiazoly1)thieno[2,3-b]pyridinan1ine was prepared using synthetic procedures described for the preparation of analog SW209415. 1H NMR (400 MHZ, CDC13) 5 8.05 (s, 1H), 7.92 (d, J: 3.2 Hz, 1H), 7.51 (d, J = 3.2 Hz, 1H), 7.11 (s, 1H), 4.73 (s, 2H), 3.88 — 3.82 (m, 1H), 3.75 — 3.62 (m, 1H), 3.57 (ddd, J: 13.1, 6.0, 3.9 Hz, 1H), 3.40 (s, 3H), 3.37 (s, 3H), 3.25 (ddd, J: 12.8, 8.0, 4.4 Hz, 1H), 2.48 (s, 3H). ESI-MS (m/z): 434.1 [M+H]+.
W0 2018f017582 SW212344. 2-(butylsulfinyl)(2-isopropylmethyl-1H-imidazolyl) (thiazolyl)thieno[2,3-b]pyridinamine was prepared using synthetic procedures described for the preparation of analog SW209415. 1H NMR (400 MHz, CDC13) 5 8.06 (s, 1H), 7.92 (d, J: 3.1 Hz, 1H), 7.51 (d, J: 3.2 Hz, 1H), 7.15 (s, 1H), 4.71 (s, 2H), 3.41 (s, 3H), 3.27 (ddd, J: 13.0, 8.5, 6.5 Hz, 1H), 3.19 — 2.98 (m, 2H), 1.83 — 1.59 (m, 2H), 1.58 — 1.41 (m, 2H), 1.39 (d, J: 6.7 Hz, 6H), 0.94 (t, J: 7.3 Hz, 3H). ESI—MS (m/z): 460.1 [M+H]+. 45. 2-(butylsulfinyl)(2-cyclopropy1methyl—1H—imidazol—5-yl) (thiazol—2—y1)thieno[2,3-b]pyridinamine was prepared using synthetic procedures bed for the preparation of analog SW209415. 1H NMR (400 MHz, CDC13) 5 8.04 (s, 1H), 7.91 (d, J: 3.1 Hz, 1H), 7.50 (d, J: 3.1 Hz, 1H), 7.07 (s, 1H), 4.77 (s, 2H), 3.51 (s, 3H), 3.27 (ddd, J: 12.9, 8.7, 6.4 Hz, 1H), 3.10 (ddd, J: 12.9, 8.8, 6.9 Hz, 1H), 1.95 — 1.78 (m, 1H), 1.81 — 1.62 (m, 2H), 1.58 — 1.37 (m, 2H), 1.17 — 0.98 (m, 4H), 0.93 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 458.1 [M+H]+.
[\N3)}? 2—bromo—l-(thiazolyl)ethanone. n-Butyllithium (24.7 mL, 0.061? mol, 2.5M in ) was added dropwise to a solution of zole (5.0 g, 0.059 mol) in anhydrous diethyl ether (48.8 mL) at -78°C. After 15 minutes, ethylbromoacetate (6.84 mL, 0.0617 mol) was added, the cold bath was removed and the solution was allowed to warm to room temperature. The reaction mixture was diluted with ether and water. The organic layer W0 2018/‘017582 was ted, dried over NaZSO4, ed and concetrated under reduced pressure. The crude product was suspended in hexanes and heated to reflux for 15 minutes then the product was decanted off leaving the impure oil. This was repeated 5 times to give a white solid with 88 % yield. 1H NMR (400 MHz, CDC13) 5 8.05 (d, J = 3.0 Hz, 1H), 7.77 (d, J = 3.0 Hz, 1H), 4.71 (s, 2H). ESI—MS (m/z): 207.8 [M+H]+.
S \PPh3 1-(thiazolyl)(triphenyl-l5-phosphanylidene)ethanone. To a on of 2-bromo—1-(thiazolyl)ethanone (10.7 g, 0.0517 mol) in toluene (337.7 mL), triphenylphosphine (14.1 g, 0.0539 mol) was added portion wise. The mixture was stirred at room temperature for 3 hours. The yellowish precipitate was removed by filtration, and was washed several times with toluene and then petroleum ether. Water was added to the precipitate and was treated se with 1N NaOH to pH 10 (at pH 7 there was a color change from yellow to orange). The mixture was stirred for 30 minutes at room temperature.
The precipitate was removed by filtration and washed several times with water. The resulting orange solid, was heated at 50°C under vacuum to remove any water, giving a 96 % yield. 1H NMR (400 MHz, CDC13) 5 7.82 (d, J = 3.1 Hz, 1H), 7.72 (ddd, J = 12.8, 8.3, 1.4 Hz, 6H), 7.61 — 7.54 (m, 3H), 7.51 — 7.45 (m, 6H), 7.38 (dd, J = 3.1, 1.3 Hz, 1H), 5.00 (d, J = 23.3 Hz, 1H). ESI-MS (m/z): 387.9 [M+H]+. cmN / Methyl (E)(3-oxo(thiazolyl)propenyl)benzoate. In a dried flask, 1-(thiazoly1)-2—(triphenylphosphanylidene)ethanone (1.5 g, 3.9 mmol) and methyl 4-formy1 benzoate (634 mg, 3.86 mmol) were dissolved in anhydrous chloroform (19.3 mL) and the solution d at 71°C ght. The solvent was evaporated under reduced pressure and the solid precipitate was purified using automated flash chromatography (100 % DCM) to give a white solid in 76 % yield. 1H NMR (400 MHZ, CDC3) 5 8.10 — 8.05 (m, W0 2018/‘017582 -100— 3H), 8.01 (d, J = 1.3 Hz, 2H), 7.76 (d, J = 8.4 Hz, 2H), 7.72 (d, J = 3.0 Hz, 1H), 3.93 (s, 3H).
ESI-MS (m/z): 234.0 [M+H]+. s N\ S\/s\/\/ Methyl 4-(2-(((butylthio)methyl)thio)cyano-6—(thiazol-2—y1)pyridin-4— yl)benzoate. 2-cyanothioacetamide (274.8 mg, 2.744 mmol) and methyl (E)—4—(3-oxo (thiazol-2—y1)prop-l-en-l-yl)benzoate (250.0 mg, 0.9147 mmol) were combined in a Vial that was evacuated and backfilled with 02 then ethanol (2.75 mL) and piperdine (2 drops) were added. The solution was d for a few s then d at 80°C for 4 hours. Once cooled, the solution was filtered, and the precipitate was rinsed with ethanol, and then washed in minimal amounts of acetic acid by heating at 80°C for 45 minutes. When , the washed solution was filtered leaving the crude brown/red solid product, which was carried forward to the next step. Standard alkylation procedure: Butyl(chloromethyl)sulfane (111.2 mg, 0.8059 mmol) in acetonitrile (1.32 mL), was added to the product from the first step, and Et3N (168.6 uL, 1.209 mmol) was added last. The solution was stirred at 80°C for 20 minutes. The reaction mixture was diluted with EtOAc and washed with H20, dried over NaZSO4, filtered, and concentrated under reduced re. The crude solid was purified using automated flash chromatography (80 % hexane, 20% . This produced a solid in 24 % yield. 1H NMR (400 MHz, CDCl3) 5 8.18 (d, J = 8.4 Hz, 2H), 8.02 (s, 1H), 7.98 (d, J = 3.1 Hz, 1H), 7.71 (d, J = 8.4 Hz, 2H), 7.58 (d, J = 3.2 Hz, 1H), 4.52 (s, 2H), 3.95 (s, 3H), 2.76 (t, J = 7.3 Hz, 2H), 1.64 (tt, J = 7.7, 6.3 Hz, 2H), 1.42 (h, J = 7.3 Hz, 2H), 0.91 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 456.1 [M+H]+.
W0 2018f017582 2-(((butylthio)methyl)thio)(4-(hydroxymethyl)phenyl)(thiazol-2— yl)nicotinonitri1e. To the solution of methyl 4-(2-(((butylthio)methyl)thio)cyano (thiazolyl)pyridinyl)benzoate (336 mg, 0.737 mmol) in THF (8.41 mL) LiBH4 (96.3 mg, 4.42 mmol) was added at 0°C. The reaction was stirred at room temperature for 36 hours, and the reaction was monitored by LC/MS. The reaction mixture was diluted with EtOAc and H20. The organic layer was dried over NaZSO4, filtered, and concentrated under reduced pressure, to give product in 96 % yield. 1H NMR (400 MHz, CDC13) 8 8.02 (s, 1H), 7.98 (d, J = 3.1 Hz, 1H), 7.69 — 7.62 (m, 2H), 7.56 (d, J = 3.1Hz, 1H), 7.56 — 7.49 (m, 2H), 4.79 (d, J = 4.3 Hz, 2H), 4.52 (s, 2H), 2.82 — 2.60 (m, 2H), 1.71 — 1.58 (m, 2H), 1.49 — 1.33 (m, 2H), 0.91 (t, J = 7.4 Hz, 3H). ESI-MS (m/z): 428.1 [M+H]+.
] Standard oxidation procedure: 2-(((butyl(ll-oxidanyl)-l3-sulfanyl)methyl)thio)- 4-(4-(hydroxymethyl)phenyl)(thiazolyl)nicotinonitrile. Chloroform (2.53 mL), acetic acid (1.39 mL), and hydrogen peroxide (108.0 uL, 1.057 mmol, 30 % solution in water) were added to 2—(((butylthio)methyl)thio)(4-(hydroxymethyl)phenyl)(thiazol—2- yl)nicotinonitrile. The solution was stirred at 32°C for 45 minutes. The reaction mixture was then d with EtOAc and washed with ted NaHC03, and the organic layer was dried over Na2804, filtered, and concentrated under reduced re to give the desired product in 94 % yield. 1H NMR (400 MHZ, CDC13) 5 8.03 (s, 1H), 7.93 (d, J = 3.1 Hz, 1H), 7.59 (d, J = 8.2 Hz, 2H), 7.55 (d, J = 3.1 Hz, 1H), 7.48 (d, J = 7.9 Hz, 2H), 4.73 (s, 2H), 4.66 (d, J = 13.1 Hz, 1H), 4.38 (d, J = 13.1 Hz, 1H), 2.93 (dt, J = 13.0, 8.1 Hz, 1H), 2.79 (dt, J = 13.0, 7.2 Hz, 1H), 1.84 — 1.72 (m, 2H), 1.55 — 1.33 (m, 2H), 0.91 (t, J = 7.3 Hz, 3H).). ESI—MS (m/z): 444.1 [M+H]+.
W0 2018/‘017582 SW209510 (4-(3-amino(butyl(11-oxidanyl)-l3 -sulfanyl)(thiazol—2— yl)thieno[2,3—b]pyridinyl)phenyl)methanol. t—BuOK (22.78 mg, 0.2028 mmol) was added to utyl(l1—oxidanyl)-l3-sulfanyl)methyl)thio)(4-(hydroxymethyl)pheny1)—6-(thiazol- 2-yl)nicotinonitri1e (150 mg, 0.338 mmol) and the vial was evacuated backfilled with N2 three times before adding DMF (1.3 mL). The solution was sparged with N2 for a few minutes before heating at 32°C. The reaction mixture was monitored every five minutes by TLC (80 % EtOAc. 20 % hexanes) and upon completion was diluted with EtOAc and washed with 10 % AcOH. The organic layer was then dried over Na2SO4, ed, and concentrated under reduced pressure. The product was purified using automated flash chromatography to give an isolated green solid/oil in 16 % yield. 1H NMR (400 MHz, CDC13) 5 8.02 (s, 1H), 7.90 (d, J = 3.2 Hz, 1H), 7.59 — 7.40 (m, 5H), 4.80 (s, 2H), 4.63 (s, 2H), 3.27 (ddd, J = 12.8, 9.0, 6.1 Hz, 1H), 3.10 (ddd, J: 12.8, 9.1, 6.6 Hz, 1H), 1.78 — 1.61 (m, 2H), 1.55 — 1.40 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 444.1 [M+H]+.
SW20951 1 4-(3-amino(butyl(11-oxidanyl)-l3-sulfanyl)(thiazol-2— yl)thieno[2,3-b]pyridinyl)benzyl e. This compound was formed during the workup of 10 in EtOAc (47 % yield). 1H NMR (400 MHz, CDCl3) 6 7.99 (s, 1H), 7.87 (d, J = 3.2 Hz, 1H), 7.56 — 7.40 (m, 5H), 5.18 (s, 2H), 4.62 (s, 2H), 3.26 (ddd, J = 12.8, 9.0, 6.1 Hz, 1H), 3.08 (ddd, J = 12.8, 9.1, 6.6 Hz, 1H), 2.14 (s, 3H), 1.77 — 1.59 (m, 2H), 1.53 — 1.37 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 486.1 [M+H]+.
W0 2018/‘017582 2017/042620 4-(3—amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazolyl)thieno[2,3- b]pyridiny1)benzaldehyde. MnOz (111.3 mg, 1.28 mmol) was added to a solution of SW209510 (56.8 mg, 0.128 mmol) in DCM (2.3 mL) and stirred at room temperature overnight. LC/MS indicated that the reaction was incomplete. The on was filtered over celite, washed with DCM and the filtrate was trated under reduced pressure. The crude e was redissolved in DCM (2.3 mL) and MnOz (5 eq) was added. The solution was left to stir 24 hours at room temperature, was filtered over celite and washed with DCM.
The filtrate was concentrated under reduced pressure and the resulting crude product was ed using automated flash chromatography (55 % EtOAc, 45 % hexanes) resulting in 24 % isolated yield. 1H NMR (400 MHz, CDClg) 5 10.13 (s, 1H), 8.11 — 7.99 (m, 3H), 7.92 (d, J = 3.1 Hz, 1H), 7.75 — 7.62 (m, 2H), 7.51 (d, J = 3.2 Hz, 1H), 4.56 (s, 2H), 3.29 (ddd, J = 12.8, 8.8, 6.3 Hz, 1H), 3.11 (ddd, J: 12.8, 8.9, 6.9 Hz, 1H), 1.82 — 1.66 (m, 2H), 1.54 — 1.41 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 442.1 [M+H]+.
S} 0 S \ I / / NH?“— SW209513. 2-(butyl(11-oxidanyl)-l3 -sulfanyl)(4- ((dimethylamino)methyl)phenyl)(thiazolyl)thieno[2,3-b]pyridinamine. To a solution of 4-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazolyl)thieno[2,3—b]pyridin yl)benzaldehyde (13.3 mg, 0.0301) in ol (802.7 uL), dimethylamine (174 ”L. 0.301 mmol, 2.0M in THF) and acetic acid (1.72 uL, 0.0301 mmol) were added and the reaction was stirred at room temperature for 90 minutes. The reaction was then cooled down to 0°C and sodium cyanoborohydride (3.7 mg, 0.060 mmol) was added and the reaction stirred for 2 hours at this temperature before allowing to warm up to room temperature. After 24 hours, more sodium cyanoborohydride (2 eq) was added at 0°C and left to stir at room temperature W0 2018/‘017582 —104_ another 24 hours. Nitrogen was used to evaporate the solvent, giving a solid that was diluted with EtOAc and washed with saturated NaHC03. The organic layer was dried over NaZSO4, filtered, and concentrated under reduced re. The crude product was purified using flash chromatography (7 % MeOH, 93 % DCM) isolating the product in 13 % yield. 1H NMR (400 MHz,CDC13) 6 8.07 (s, 1H), 7.93 (d, J = 3.2 Hz, 1H), 7.51 (d, J: 3.2 Hz, 1H), 7.49 — 7.41 (m, 4H), 4.67 (s, 2H), 3.55 (s, 2H), 3.36 — 3.25 (m, 1H), 3.13 (ddd, J: 12.8, 9.0, 6.7 Hz, 1H), 2.30 (s, 6H), 1.78 — 1.68 (m, 2H), 1.55 — 1.44 (m, 2H), 0.95 (t, J = 7.3 Hz, 3H).
ESI-MS (m/z): 471.2 [M+H]+. tWON\ / Methyl (E)(3 -oxo-3 -(thiazolyl)propenyl)benzoate. Followed procedure for methyl (E)(3-oxo(thiazolyl)propenyl)benzoate using methyl 3- formyl benzoate as the starting material. Purified the crude product using automated flash chromatography (50 % EtOAc, 50 % hexanes) ing the product in 51 % yield. 1H NMR (400 MHz, CDC13) 8 8.41 — 8.35 (m, 1H), 8.11 — 8.05 (m, 2H), 8.02 (d, J= 1.3 Hz, 2H), 7.89 — 7.83 (m, 1H), 7.72 (d, J: 3.0 Hz, 1H), 7.50 (t, J: 7.8 Hz, 1H), 3.95 (s, 3H). ESI-MS (m/z): 274.1 Methyl 3-(2-(((butylthio)methyl)thio)cyano(thiazolyl)pyridin yl)benzoate. Followed the procedure for methyl ((butylthio)methyl)thio)-3—cyano (thiazol—2—y1)pyridinyl)benzoate using methyl (3-oxo(thiazol-2—yl)prop-l-en-l- zoate as the starting material. Isolated product in 87 % yield. 1H NMR (400 MHZ, CDC13) 5 8.32 — 8.26 (m, 1H), 8.20 (dt, J = 7.9, 1.3 Hz, 1H), 8.04 (s, 1H), 7.99 (d, J = 3.1 Hz, 1H), 7.85 (ddd, J: 7.7, 2.0, 1.1 Hz, 1H), 7.62 (td, J: 7.8, 0.6 Hz, 1H), 7.58 (d, J = 3.1 Hz, 1H), 4.53 (s, 2H), 3.95 (s, 3H), 2.76 (t, J: 7.3 Hz, 2H), 1.71 — 1.59 (m, 2H), 1.49 — 1.36 (m, 2H), 0.91 (t, J: 7.3 Hz, 3H). ESI—MS (m/z): 456.1 [M+Z]+. 2-(((butylthio)n1ethyl)thio)(3-(hydroxymethyl)phenyl)(thiazol-2— yl)nicotinonitri1e. Followed procedure for 2-(((butylthio)methyl)thio)(4- (hydroxymethyl)phenyl)(thiazolyl)nicotinonitrile using methyl 3-(2- (((butylthio)methyl)thio)cyano(thiazolyl)pyridinyl)benzoate as the starting material. Isolated product in 84 % yield. 1H NMR (400 MHZ, CDC13) 5 8.00 (s, 1H), 7.95 (d, J = 3.1 Hz, 1H), 7.64 — 7.61 (m, 1H), 7.58 — 7.52 (m, 2H), 7.52 — 7.46 (m, 2H), 4.76 (s, 2H), 4.50 (s, 2H), 2.74 (t, J = 7.3 Hz, 2H), 1.69 — 1.54 (m, 2H), 1.46-1.37 (m, 2H), 0.90 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 428.1 [M+H]+ 2—(((butyl(l1-oxidanyl)-l3-sulfanyl)methyl)thio)(3-(hydroxymethy1)phenyl)- 6-(thiazol—2—y1)nicotinonitrile. Followed standard oxidation ure using 2— (((butylthio)methyl)thio)(3-(hydroxymethyl)phenyl)(thiazolyl)nicotinonitrile as the starting material. Isolated product in 88 % yield. 1H NMR (400 MHZ, CDCl3) 5 8.08 (s, 1H), 7.96 (d, J = 3.1 Hz, 1H), 7.68 — 7.64 (m, 1H), 7.58 — 7.53 (m, 2H), 7.53 — 7.48 (m, 2H), 4.77 (s, 2H), 4.71 (d, J: 13.1 Hz, 1H), 4.36 (d, J: 13.1 Hz, 1H), 2.96 (dt, J: 13.0, 8.2 Hz, 1H), 2.81 (dt, J = 13.0, 7.3 Hz, 1H), 1.82 (p, J = 7.7 Hz, 2H), 1.58 — 1.40 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 444.1 .
S \8/0 SW209418 amino(butyl(11-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3—b]pyridinyl)phenyl)methanol. Followed procedure for SW209510 using 2- W0 2018/‘017582 yl(11-oxidanyl)-l3-sulfanyl)methyl)thio)(3-(hydroxymethyl)phenyl)(thiazol yl)nicotinonitrile as the starting material to give an isolated product in 68 % yield. 1H NMR (400 MHz, CDC13) 8 8.01 (s, 1H), 7.88 (d, J = 3.1 Hz, 1H), 7.55 — 7.30 (m, 5H), 4.75 (s, 2H), 4.62 (s, 2H), 3.26 (ddd, J: 12.8, 9.1, 6.0 Hz, 1H), 3.09 (ddd, J: 12.8, 9.2, 6.5 Hz, 1H), 1.76 — 1.61 (m, 2H), 1.51 — 1.38 (m, 2H), 0.92 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 444.1 [M+H]+.
Methyl 3-(2-(((butyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)-3—cyano—6—(thiazol- 2-yl)pyridinyl)benzoate. Followed standard oxidation procedure, using methyl 3-(2- ylthio)methyl)thio)cyano(thiazolyl)pyridinyl)benzoate as the starting material. This gave an isolated product in 86 % yield. 1H NMR (400 MHZ, CDC13) 8 8.27 (t, J: 1.6 Hz, 1H), 8.17 (dt, J: 7.9, 1.4 Hz, 1H), 8.09 (s, 1H), 7.97 (d, J: 3.1 Hz, 1H), 7.82 (ddd, J: 7.7, 1.9, 1.1 Hz, 1H), 7.61 (m, 1H), 7.58 (d, J: 3.1Hz, 1H), 4.72 (d, J=13.1 Hz, 1H), 4.42 (d, J: 13.1 Hz, 1H), 3.92 (s, 3H), 2.95 (dt, J: 13.0, 8.1 Hz, 1H), 2.83 (dt, J = 13.0, 7.3 Hz, 1H), 1.81 (p, J: 7.7 Hz, 2H), 1.57 — 1.36 (m, 2H), 0.92 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 472.1 [M+H]+.
|\SS’N O //\—\_ SW209416. Methyl 3-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3-b]pyridinyl)benzoate. Followed ure for SW209510 using methyl 3- (2-(((butyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)cyano(thiazolyl)pyridin-4— yl)benzoate as the starting material to give an isolated product in 68 % yield. 1H NMR (400 MHZ, CDC13) 8 8.26 — 8.11 (m, 2H), 8.02 (s, 1H), 7.89 (d, J: 3.2 Hz, 1H), 7.76 — 7.56 (m, 2H), 7.49 (d, J: 3.1 Hz, 1H), 4.54 (s, 2H), 3.93 (s, 3H), 3.27 (ddd, J: 12.8, 9.0, 6.2 Hz, 1H), 3.09 (ddd, J = 12.8, 9.0, 6.7 Hz, 1H), 1.79 — 1.61 (m, 2H), 1.55 — 1.39 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 472.1 [M+H]+.
W0 2018f017582 2017/042620 <3:/ i“ |N\ SVSW Standard Hydrolysis Procedure of Ester to Carboxylic Acid: 3-(2— ylthio)methyl)thio)cyano(thiazolyl)pyridinyl)benzoic acid. THF (214.3 uL), MeOH (214.3 uL), and H20 (71.4 uL) were added to methyl 3-(2- (((butylthio)methyl)thio)cyano(thiazolyl)pyridinyl)benzoate (50 mg, 0.110 mmol), and last LiOH (7.9 mg, 0.329 mmol) was added. The solution was stirred at room ature for 3 hours. The reaction mixture was diluted with EtOAc and washed with 1M HCl. The organic layer was dried over , filtered, and concentrated under reduced pressure. The resulting product gave a 94 % yield. 1H NMR (400 MHZ, CDC13) 8 8.41 (t, J = 1.7 Hz, 1H), 8.26 (dt, J: 8.0, 1.3 Hz, 1H), 8.13 (s, 1H), 8.02 (d, J: 3.1 HZ, 1H), 7.94 — 7.89 (m, 1H), 7.65 (t, J = 7.8 Hz, 1H), 7.59 (d, J: 3.1 Hz, 1H), 4.53 (s, 2H), 2.75 (t, J = 7.3 Hz, 2H), 1.64 (p, J: 7.5 Hz, 2H), 1.43 (m, 2H), 0.91 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 442.1 [M+Z]+.
Standard amide bond coupling procedure: 3-(2-(((butylthio)methyl)thio) cyano(thiazolyl)pyridinyl)-N,N-dimethylbenzamide. Dimethylamine hydrochloride (9.25 mg, 0.114 mmol) was added to a solution of 3-(2-(((butylthio)methyl)thio)—3-cyano (thiazol-2—y1)pyridinyl)benzoic acid (45.6 mg, 0.103 mmol), HATU (43.2 mg, 0.114 mmol), and DMF (266 uL) followed by DIPEA (36 uL, 0.21 mmol). The solution was stirred at room temperature for 3 hours, then diluted with EtOAc and washed with water. The organic layer was dried over Na2S04, filtered, and concentrated under reduced pressure. The isolated solid gave an 86 % yield. 1H NMR (400 MHZ, CDC13) 5 7.99 (s, 1H), 7.97 (d, J = 3.1 Hz, 1H), 7.69 —7.61 (m, 2H), 7.58 — 7.51 (m, 3H), 4.50 (s, 2H), 3.11 (s, 3H), 3.03 (s, 3H), 2.73 (t, J = 7.3 Hz, 2H), 1.62 (p, J: 7.4 Hz, 2H), 1.40 (h, J = 7.3 Hz, 2H), 0.89 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 469.1 [M+H]+.
W0 2018f017582 o 9 IN\ SVSW 3—(2—(((butyl(l1-oxidanyl)-l3-sulfanyl)methyl)thio)cyano(thiazol—2- yl)pyridinyl)—N,N-dimethylbenzamide. Followed standard oxidation procedure, using 3- (2-(((butylthio)methyl)thio)cyano(thiazolyl)pyridinyl)-N,N-dimethy1benzamide as the starting material to give the isolated product in 96 % yield. 1H NMR (400 MHz, Chloroform—d) 8 8.08 (s, 1H), 7.98 (d, J = 3.1 Hz, 1H), 7.70 — 7.64 (m, 2H), 7.61 — 7.54 (m, 3H), 4.70 (d, J: 13.1 Hz, 1H), 4.42 (d, J: 13.1 Hz, 1H), 3.11 (s, 3H), 3.03 (s, 3H), 2.95 (dt, J: 12.9, 8.2 Hz, 1H), 2.81 (dt, J: 12.9, 7.2 Hz, 1H), 1.82 (p, J: 7.7 Hz, 2H), 1.56 — 1.36 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 485.1 [M+H]+.
SW209417. mino(buty1(11-oxidanyl)-l3-sulfanyl)—6—(thiazol—2— yl)thieno[2,3—b]pyridinyl)-N,N-dimethylbenzamide. Followed procedure for SW209510 using ((butyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)cyano(thiazol-2—yl)pyridin yl)-N,N-dimethylbenzamide as the starting material to give the isolated product in 63 % yield. 1H NMR (400 MHz, Chloroform-d) 5 8.02 (s, 1H), 7.90 (d, J = 3.1 Hz, 1H), 7.62 — 7.51 (m, 4H), 7.49 (d, J: 3.1 Hz, 1H), 4.59 (s, 2H), 3.27 (ddd, J: 12.8, 9.0, 6.1 Hz, 1H), 3.15 — 2.97 (m, 7H), 1.78 — 1.64 (m, 2H), 1.55 — 1.39 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). 0N N S \S/O |//S\_\¥ ] SW209419. 3-(3-amino(butyl(11-oxidany1)sulfany1)(thiazol yl)thieno[2,3—b]pyridinyl)benzoic acid. Using SW209416 as the starting material, follow W0 2018/‘017582 2017/042620 the standard hydrolysis procedure of ester to carboxylic acid. This gave an isolated yield of 98 %. 1H NMR (400 MHz, (CD3)2CO)) 5 8.28 — 8.18 (m, 2H), 8.07 (s, 1H), 7.98 (d, J = 3.2 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.82 (d, J = 3.2 Hz, 1H), 7.76 (t, J = 7.6 Hz, 1H), 4.82 (s, 2H), 3.20 (ddd, J: 12.8, 8.8, 6.3 Hz, 1H), 3.09 (ddd, J: 12.9, 8.8, 6.8 Hz, 1H), 1.76 — 1.66 (m, 2H), 1.54 — 1.43 (m, 2H), 0.92 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 458.1 [M+H]+.
Q” NV.) fr |//S.
] SW209420. (3-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)-6—(thiazol yl)thieno[2,3-b]pyridinyl)phenyl)(4-methylpiperazinyl)methanone. Followed the standard amide bond coupling procedure, using SW209419 as the starting material and 1- methylpiperazine as the substrate. The product was purified using automated flash chromatography, recovering 38 % ed yield. 1H NMR (400 MHz, CDC13) 8 8.04 (s, 1H), 7.91 (d, J = 3.2 Hz, 1H), 7.65 — 7.42 (m, 5H), 4.56 (s, 2H), 3.79 (m, 2H), 3.46 (m, 2H), 3.28 (ddd, J: 12.9, 8.9, 6.1 Hz, 1H), 3.10 (ddd, J: 12.9, 9.2, 7.0 Hz, 1H), 2.48 (m, 2H), 2.35 (m, 2H), 2.31 (s, 3H), 1.77 — 1.58 (m, 2H), 1.54 — 1.38 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 540.2 [M+H]+.
S \S,0 |//S\_\— SW209508. N-allyl(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3-b]pyridinyl)benzamide. Followed the standard amide bond coupling procedure using SW209419 as the starting material and mine as the substrate. The isolated product gave a 92 % yield. 1H NMR (400 MHz, CDCl3) 5 8.05 — 7.91 (m, 3H), 7.88 (d, J: 3.2 Hz, 1H), 7.68 — 7.53 (m, 2H), 7.48 (d, J: 3.1 Hz, 1H), 6.01 — 5.82 (m, 1H), 5.25 (d, J: 17.2 Hz, 1H), 5.16 (dd, J: 10.2, 1.4 Hz, 1H), 4.52 (s, 2H), 4.19 — 3.98 (m, 2H), 3.24 (ddd, J: 12.8, 9.0, 5.9 Hz, 1H), 3.16 — 2.98 (m, 1H), 1.78 - 1.56 (m, 2H), 1.57 — 1.38 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 497.1 [M+H]+. f?! N O SW209509. 3-(3-amino(butyl(ll-oxidanyl)-l3-sulfanyl)(thiazol—2- eno[2,3—b]pyridinyl)-N-(2-(dimethylamino)ethyl)benzamide. ed the standard amide bond coupling procedure, using 19 as the starting material and N,n—dimethyl- ethane-1,2—diamine as the substrate. The reaction mixture was diluted with EtOAc and washed with water and NaOH was added to neutralize the pH. The organic layer was then dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude product was purified using automated flash chromatography (93% DCM, 2% Et3N, 5% MeOH) to give product in 70 % isolated yield. 1H NMR (400 MHz, CDC13) 5 8.02 (s, 1H), 8.00 — 7.95 (m, 2H), 7.88 (d, J = 3.2 Hz, 1H), 7.63 — 7.54 (m, 2H), 7.48 (d, J = 3.2 Hz, 1H), 4.55 (s, 2H), 3.59 — 3.50 (m, 2H), 3.25 (ddd, J: 12.8, 9.0, 6.0 Hz, 1H), 3.08 (ddd, J: 12.8, 9.1, 6.6 Hz, 1H), 2.58 (t, J: 5.9 Hz, 2H), 2.28 (s, 6H), 1.77 — 1.61 (m, 2H), 1.53 — 1.43 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 528.2 .
CI N Cl 2,6—dichloropyridinamine. The acetone/water mixture (297 mL, 5:1) was added to 2,6-dichloronitropyridine (3.0 g, 0.016 mol) followed by Zn (10.17 g, 0.1550 mol) and NH4C1 (12.44 g, 0.2325 mol). The solution stirred at room temperature overnight.
The reaction mixture was then filtered through celite and the filtrate was ted with EtOAc. With the help of brine, the organic layer was separated, dried over MgSO4 and concentrated under reduced pressure. 1H NMR (400 MHz, CDCl3) 5 7.07 (d, J = 8.2 Hz, 1H), 7.02 (d, J: 8.3 Hz, 1H), 4.11 (s, 2H). ESI—MS (m/z): 163.0.
W0 17582 K+-s o/\ Potassium Ethyl Xanthate. A potassium ethoxide solution was prepared by dissolving KOH (6.5 g, 0.12 mol) in EtOH (63.4 mL). Carbon disulfide (7.14 mL, 0.118 mol) was added to the solution slowly with continuous stirring. The reaction mixture was cooled down to 5°C, ed, and the precipitate was recrystallized twice from warm ethanol.
CI UNN\ 3 5—Chlorothiazolo[5,4-b]pyridinethiol. Potassium ethyl te (1.9 g, 0.012 mol) and ous N-methylpyrrolidone (14.1 mL) were added to 2,6—dichloropyridin amine (1.0 g, 0.0061 mol) under N2. The solution was refluxed (170°C) for 3.5 hours. The reaction mixture was cooled down to room temperature, acidified to pH 5 using AcOH, diluted in EtOAc, and washed several times with H20. The organic layer was separated, dried over MgSO4, and concentrated under reduced pressure. This gave a red solid in 18 % yield. 1H NMR (400 MHz, Chloroform-d) 6 7.41 (d, J = 8.4 Hz, 1H), 7.30 (d, J = 8.4 Hz, 1H). ESI—MS (mfz): 202.9.
CI UN»—8\—\_N\ s 2—(Butylthio)chlorothiazolo[5,4-b]pyridine. K2C03 (75 mg, 0.54 mmol), 1- bromobutane (53.3 m, 0,493 mmol), 18-Crown-6 (13.2 mg, 0.0493 mmol), and DMF (3.4 mL) were added to 5-chlorothiazolo[5,4-b]pyridinethiol and the solution was heated at 80°C for 3 hours. The solution was diluted with EtOAc, washed with H20, and the c layer was separated, dried over MgSO4, and concentrated under reduced re. The crude product was purified using flash chromatography to give 76 % isolated yield. 1H NMR (400 MHz, CDC13) 8 7.93 (d, J: 8.5 Hz, 1H), 7.30 (d, J: 8.5 Hz, 1H), 3.31 (t, J: 7.3 Hz, 2H), 1.76 (p, J = 7.5 Hz, 2H), 1.52 — 1.39 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 259.0 [M+H]+. 2—(Butylthio)(thiophenyl)thiazolo[5’4-b]pyridine. 2—Thieny1boronic acid (49.4 mg, 0.386 mmol), CsCO3 (126 mg, 0.386 mmol), Pd(dppf)C12 (15.8 mg, 0.0193 mmol), CuCl (19.1 mg, 0.193 mmol) and DMF (1 mL) were added to 2-(butylthio)-5— chlorothiazolo[5,4-b]pyridine (50 mg, 0.19 mmol) under N2. The reaction mixture was heated to 1000C for 30 s. Then the N2 was disconnected and the vial was capped and sealed with teflon tape and allowed to stir overnight. The reaction mixture was diluted in EtOAc, washed with H20, and the organic layer was ted, dried over MgSO4, filtered, and concentrated under reduced pressure to give the product in 31 % isolated yield. 1H NMR (400 MHz, CDC13) 8 7.98 (d, J: 8.6 Hz, 1H), 7.63 (dd, J: 3.8, 1.1 Hz, 1H), 7.51 (dd, J: .1,1.1Hz,1H), 7.23 (d, J: 8.6 Hz, 1H), 7.14 (dd, J: 5.0, 3.8 Hz, 1H), 3.24 (t, J: 7.3 Hz, 2H), 1.78 — 1.66 (m, 2H), 1.57 — 1.41 (m, 2H), 0.96 (t, J: 7.4 Hz, 3H). ESI—MS (m/z): 307.0 [M+H]+.
N O S \ S l/ N/>—S\_\_ ] SW208599. 2-(buty1(11-oxidanyl)-l3-sulfanyl)(thiophenyl)thiazolo[5,4- b]pyridine. CHC13 (142 uL), AcOH (142 uL), and H202 (12.0 uL, 0.118 mmol, 30 % solution in H20) were added to 2-(butylthio)(thiophenyl)thiazolo[5,4—b]pyridine (18 mg. 0.059 mmol) and heated at 35°C for 2.5 hours. The solution was diluted with EtOAc and washed with saturated NaHCO3. The organic layer was separated, dried over MgSO4, filtered, and concentrated under reduced pressure to give product in 60 % ed yield. 1H NMR (400 MHZ, CDC13) 5 8.39 (d, J = 8.4 Hz, 1H), 8.06 (d, J: 8.4 Hz, 1H), 7.74 (dd, J: 3.8,1.1Hz,1H), 7.61 (dd, J: 5.0, 1.1Hz, 1H), 7.19 (dd, J: 5.0, 3.8 Hz, 1H), 3.15 (ddd, J: 13.3, 9.8, 6.0 Hz, 1H), 2.96 (ddd, J: 13.3, 9.9, 4.9 Hz, 1H), 1.98 — 1.80 (m, 1H), 1.60 — 1.52 (m, 1H), 1.52 — 1.37 (m, 2H), 0.93 (t, J: 7.2 Hz, 3H). ESI-MS (m/z): 323.0 [M+H]+.
W0 2018f017582 ] 2—(((Isopropylthio)methyl)thio)(oxazolyl)phenylnicotinonitrile. Follow the standard alkylation procedure, using (chloromethyl)(isopropyl)sulfane as the alkylating substrate, and 6—(oxazolyl)phenylthioxo-1,2-dihydropyridinecarbonitrile as the starting material. The crude product was purified using flash chromatography to give a solid in 62 % isolated yield. 1H NMR (400 MHz, CDC13) 5 7.97 (s, 1H), 7.88 (d, J = 0.7 Hz, 1H), 7.67 — 7.61 (m, 2H), 7.56 — 7.50 (m, 3H), 7.37 (d, J = 0.8 Hz, 1H), 4.63 (s, 2H), 3.24 (hept, J = 6.7 Hz, 1H), 1.35 (d, J: 6.7 Hz, 6H). ESI-MS (m/z): 368.0 [M+H]+. 2—(((Isopropyl(ll-oxidanyl)-l3-sulfanyl)methyl)thio)-6—(oxazol—2—yl)—4— phenylnicotinonitrile. Follow the standard oxidation procedure using 2— (((isopropy1thio)methyl)thio)(oxazolyl)phenylnicotinonitrile as the starting material.
Recovered quatitative isolated yield. 1H NMR (400 MHZ, CDC13) 5 8.03 (s, 1H), 7.88 (d, J = 0.7 Hz, 1H), 7.67 — 7.61 (m, 2H), 7.58 — 7.50 (m, 3H), 7.39 (d, J: 0.7 Hz, 1H), 4.79 (d, J = 13.3 Hz, 1H), 4.55 (d, J: 13.3 Hz, 1H), 3.18 (hept, J: 6.9 Hz, 1H), 1.42 (d, J: 1.5 Hz, 3H), 1.40 (d, J: 1.3 Hz, 3H). ESI-MS (m/z): 384.1 [M+H]+. ] rd Final Cyclization Procedure: SW208660. 2-(Isopropyl(11-oxidany1)- l3-sulfanyl)(oxazolyl)phenylthieno[2,3-b]pyridinamine. DMF (485 uL) and MeOH (244 uL) were added to 2-(((isopropyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)—6- W0 2018/‘017582 2017/042620 —114_ (oxazolyl)phenylnicotinonitrile (47.2 mg, 0.123 mmol) dissolving it completely before KOH (4.1 mg in 100 uL of H20) was added to the solution. The reaction mixture was stirred at 35°C for 40 minutes. The on mixture was diluted with EtOAc, washed with 10 % AcOH and then washed with H20 multiple times. The organic layer was ted, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was purified using flash chromatography to give product in 40 % isolated yieldIH NMR (400 MHz, CDC13) 8 7.99 (s, 1H), 7.85 (d, J = 0.7 Hz, 1H), 7.56 — 7.44 (m, 5H), 7.34 (d, J = 0.8 Hz, 1H), 4.69 (s, 2H), 3.41 (hept, J = 6.8 Hz, 1H), 1.44 (d, J = 6.8 Hz, 3H), 1.28 (d, J = 6.9 Hz, 3H). ESI—MS (m/z): 384.1 [M+H]+.
O N CI 2-Chloromethy1—6-morpholinonicotinonitrile. Anhydrous MeOH (3.97 mL) was added to 2,6-dichloromethylnicotinonitrile (500 mg, 2.67 mmol) under N2 and the mixture was cooled down to 0 0C. line (473.7 uL, 5.493 mmol) was added dropwise to the solution and the solution stirred at room temperature ght. The reaction mixture was filtered, washing the precipitate with MeOH (500 uL) and H20 (3-4 mL). DCM was added to the precipitate, followed by MgSO4, and the solution was filtered, then concentrated under reduced pressure. The crude producte was ed using automated flash chromatography to give product in 85 % isolated yield. 1H NMR (400 MHZ, CDClg) 8 7.26 (s, 1H), 3.81 — 3.73 (m, 4H), 3.68 — 3.58 (m, 4H), 2.42 (d, J: 0.8 Hz, 3H). ESI—MS (m/z): 238.1 [M+H]+.
N S 4-Methylmorpholinothioxo-1,2-dihydropyridine-3 -carbonitrile. NaOME (73.6 mg, 1.36 mmol) and methyl 3-mercapiopropionate (151 uL, 1.363 mmol) were added to a solution of 2—chloromethyl-6morpholinonicotinonitrile (324 mg, 1.36 mmol) in DMF W0 2018/‘017582 2017/042620 (4.10 mL) and the reaction e was stirred at 80°C for 1 hour. Once cooled down, the reaction mixture was d with EtOAc and washed with H20. The organic layer was separated, dried over MgSO4, filtered, and concentrated under reduced pressure to give a crude mixture of 1:1 ng material to product, which was carried forward to the next step.
ESI (m/Z): 322.1 [M+H]+. NaH (150.8 mg, 3.769 mmol, 60 % in mineral oil) and THF (10 mL) were added to a flame dried flask under N2, followed by the crude product from the previous step dissolved in THF (10 mL). The reaction mixture was refluxed for 6 hours and addition NaH (2 eq) was and left refluxing overnight. EtOH (1.5 mL) was added then the reaction mixture was concentrated down under reduced pressure. H20 (8 mL) was added and the solution was adjusted to pH 6 with concentrated HCl before filtering to leave a crude solid that was carried forward. ESI (m/z): 236.1 [M+H]+.
K/N |N\ S\/S\/\ TYICN 4-Methylmorpholino(((propylthio)methyl)thio)nicotinonitrile. Followed the standard alkylating procedure, using 4-methylmorpholinothioxo-1,2- opyridine—3—carbonitrile as the starting material and (chloromethyl)(propyl)sulfane as the alkylating substrate. The crude product was carried forward. ESI (m/Z): 324.1 [M+H]+. 0 9 K/N N\ 3\/S\/\ 11-oxidanyl)(propyl)-l3-sulfanyl)methyl)thio)methyl morpholinonicotinonitrile. Followed the standard oxidation procedure using 4—methyl morpholino(((propylthio)methyl)thio)nicotinonitrile as the starting material. The crude product was carried forward. ESI (m/z): 340.1 [M+H]+. m/S / f” CH3 NH2 ] SW208663. 2-((11-oxidanyl)(propyl)-l3-sulfanyl)methyl morpholinothieno[2,3-b]pyridinamine. Followed the standard final cyclization procedure W0 2018f017582 2017/042620 using 2-((((11-oxidanyl)(propyl)-l3-sulfanyl)methy1)thio)methy1 morpholinonicotinonitrile as the starting material. The crude t was purified by flash chromatography, and PTLC to give isolated product in 10 % yield. 1H NMR (400 MHz, CDC13) 5 6.36 (s, 1H), 4.91 (s, 2H), 3.85 — 3.76 (m, 4H), 3.63 — 3.58 (m, 4H), 3.32 — 3.18 (m, 1H), 3.09 — 2.99 (m, 1H), 2.65 (s, 3H), 1.81 — 1.66 (m, 2H), 1.07 (t, J: 7.4 Hz, 3H). ESI (m/z): 340.1 [M+H]+. 4—Methyl(thiazolyl)thioxo-1,2-dihydropyridinecarbonitrile.
Followed same procedure as 4-Methylmorpholinothioxo-1,2-dihydropyridine carbonitrile, using methyl 3-((3-cyanomethyl(thiazolyl)pyridin-2—yl)thio)propanoate as the starting material. The crude product was carried forward. ESI-MS (mil): 234.0 [M+H]+. 4—Methy1—2-(((propylthio)methyl)thio)(thiazolyl)nicotinonitrile. Followed the rd alkylation procedure using 4-Methyl(thiazolyl)thioxo-1,2— dihydropyridine—3-carbonitrile as the starting material, and (chloromethyl)(propy1)su1fane as the alkylating ate. The crude product was purified using automated flash chromatography to give product in 23 % isolated yield. 1H NMR (400 MHZ, Chloroform-d) 7.96 (d, J = 3.1 Hz, 1H), 7.83 (s, 1H), 7.54 (d, J: 3.1 Hz, 1H), 4.47 (s, 2H), 2.70 (t, J = 7.2 Hz, 2H), 2.55 (s, 3H), 1.67 (h, J = 7.3 Hz, 2H), 0.99 (t, J = 7.3 Hz, 3H). ESI-MS (mjz): 322.0 {M+H]+.
W0 2018/‘017582 2017/042620 / ‘N 9 s N\ SVS\/\ 2—((((ll-oxidanyl)(propyl)-l3-sulfanyl)methyl)thio)methyl-6—(thiazol—2- yl)nicotinonitri1e. Followed the standard oxidation procedure using 4-methyl—2— (((propylthio)methyl)thio)(thiazolyl)nicotinonitrile as the starting material to give white solid in 91 % isolated yield. 1H NMR (400 MHz, CDC13) 5 7.97 (d, J = 3.1 Hz, 1H), 7.92 (s, 1H), 7.56 (d, J: 3.2 Hz, 1H), 4.74 (d, J: 13.2 Hz, 1H), 4.44 (d, J: 13.1 Hz, 1H), 2.89 (m, 2H), 2.57 (s, 3H), 1.93 — 1.79 (m, 2H), 1.06 (t, J = 7.4 Hz, 3H). ESI-MS (m/z): 338.0 [M+H]+. f?! N S O l , SW208661. 2-((ll-oxidanyl)(propyl)-l3-sulfanyl)methy1(thiazol yl)thieno[2,3-b]pyridinamine. Followed standard final cyclization procedure using 2- ((((l1—oxidany1)(propyl)-l3-sulfanyl)methyl)thio)methyl(thiazol—2—yl)nicotinonitrile as the starting material. ed the crude product using flash tography to give 61 % bright green isolated product. 1H NMR (400 MHZ, CDCl3) 5 7.95 (s, 1H), 7.93 (d, J = 3.2 Hz, 1H), 7.48 (d, J: 3.2 Hz, 1H), 5.16 (s, 2H), 3.37 — 3.23 (m, 1H), 3.16 — 3.05 (m, 1H), 2.85 (s, 3H), 1.83 (h, J: 7.5 Hz, 2H), 1.11 (t, J: 7.4 Hz, 3H). ESI—MS (m/z): 338.0 [M+H]+.
S\\/mom 2-(((isopropylthio)methyl)thio)methyl(thiazolyl)nicotinonitrile.
Followed standard ting procedure using (chloromethyl)(isopropyl)sulfane as the ting substrate and 4-methy1(thiazolyl)thioxo-1,2-dihydropyridine carbonitrile as the starting material. Purified using automated flash chromatography to give product in 32 % isolated yield. 1H NMR (400 MHZ, CDCl3) 5 7.94 (d, J = 3.2 Hz, 1H), 7.82 W0 2018f017582 (s, 1H), 7.52 (d, J = 3.2 Hz, 1H), 4.48 (s, 2H), 3.18 (hept, J = 6.6 Hz, 1H), 2.54 (s, 3H), 1.31 (d, J = 6.7 Hz, 6H). ESI-MS (m/z): 322.0 [M+H]+. {1“Si; v \rCNN s 3 2—(((isopropyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)methyl(thiazol yl)nicotinonitri1e. Followed standard oxidation procedure using 2- (((isopropylthio)methyl)thio)methyl(thiazolyl)nicotinonitrile as the starting material to give a solid product in 84 % yield. 1H NMR (400 MHZ, CDCl3) 5 7.97 (d, J = 3.1 Hz, 1H), 7.91 (s, 1H), 7.56 (d, J = 3.1 Hz, 1H), 4.57 (d, J = 13.3 Hz, 1H), 4.46 (d, J = 13.3 Hz, 1H), 3.05 (hept, J = 6.9 Hz, 1H), 2.57 (s, 3H), 1.38 (d, J = 7.1 Hz, 3H), 1.36 (d, J = 6.7 Hz, 3H). ESI-MS (m/z): 338.0 [M+H]+.
[P N s O I / / )— SW208664. 2-(Isopropyl(l1-oxidanyl)-l3-sulfanyl)—4—methy1—6—(thiazol—2- yl)thieno[2,3—b]pyridinamine. Followed rd final cyclization procedure using 2- propy1(11—oxidanyl)-l3-sulfanyl)methyl)thio)methyl(thiazol-2—yl)nicotinonitrile as the starting material. The crude product was purified using flash chromatography to give bright green oil/solid in 48 % isolated yield. 1H NMR (400 MHZ, Chloroform—d) 8 7.93 (s, 1H), 7.92 (d, J = 3.2 Hz, 1H), 7.46 (d, J = 3.2 Hz, 1H), 3.38 (hept, J = 6.9 Hz, 1H), 2.84 (s, 3H), 1.46 (d, J = 6.8 Hz, 3H), 1.29 (d, J = 6.8 Hz, 3H). ESI—MS (m/z): 338.0 [M+H]+.
O O Ethyl 2,4-dioxo(thiophenyl)butanoate. 2-Acetylthiophene (1.71 mL, 0.0159 mol) was added to a solution of NaOEt (730 mg Na cubes in 50 mL of EtOH) and the solution was cooled to 0°C for 1-2 hours then diethyl oxylate (3.2 mL) was added to the solution. This was left to stir at room ature overnight. The reaction mixture was diluted with EtOAc and H20 with a little brine to assist the separation. The organic layer was collected, dried with MgSO4, filtered, and trated under d pressure. The crude W0 2018/‘017582 2017/042620 product was purified using automated flash chromatography giving an oil product with 23 % yield. ESI-MS (m/z): 227.0 [M+H]+.
Ethyl 3-cyano(((propylthio)methyl)thio)(thiophen-2—yl)isonicotinate. 2- Cyanothioacetamide (250.6 mg, 2.503 mmol) and ethyl 2,4-dioxo(thiophen-2—yl)butanoate (565.7 mg, 2.503 mmol) were dissolved in EtOH (7.46 mL) under gentle heating (40°C), then Et3N (174.5 uL, 1.251 mmol) was added drop wise to the stirring solution. The reaction mixture was heated at 60°C and after 3 hours was concentrated down under reduced pressure and the crude t was carried forward to the next step. Followed standard alkylating procedure using ethyl o(thiophenyl)thioxo-1,2-dihydropyridine-4—carboxylate as the starting material and (chloromethyl)(propy1)su1fane as the alkylating reagent. The crude t was purified twice using automated flash chromatography (20 % EtOAc, 80 % hexanes) to give 34 % isolated product. 1H NMR (400 MHz, CDCl3) 5 7.70 (s, 1H), 7.60 (dd, J: 3.8, 1.1Hz, 1H), 7.44 (dd, J: 5.1, 1.1Hz, 1H), 7.04 (dd, J: 5.0, 3.8 Hz, 1H), 4.38 (q, J = 7.2 Hz, 2H), 4.32 (s, 2H), 2.59 (t, J = 7.2 Hz, 2H), 1.57 (h, J = 7.4 Hz, 2H), 1.36 (t, J = 7.1 Hz, 3H), 0.89 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 378.9 . /\o 0 Ethyl 2-((((11-oxidanyl)(propyl)-l3-sulfany1)methyl)thio)cyano(thiophen- 2-yl)isonicotinate. Followed standard oxidation procedure using ethyl 3-cyano (((propylthio)methyl)thio)(thiopheny1)isonicotinate as the starting material to give a solid with quantitative yield. 1H NMR (400 MHz, CDC13) 5 7.86 (s, 1H), 7.72 (dd, J = 3.8, 1.1Hz, 1H), 7.53 (dd, J: 5.0, 1.1Hz, 1H), 7.11 (dd, J: 5.0, 3.8 Hz, 1H), 4.68 (d, J: 13.2 Hz, 1H), 4.49 (d, J = 13.2 Hz, 1H), 4.43 (q, J = 7.1 Hz, 2H), 2.96 — 2.82 (m, 2H), 1.87 —1.75 (m, 2H), 1.40 (t, J = 7.2 Hz, 3H), 1.02 (t, J = 7.4 Hz, 3H). ESI-MS (m/z): 394.9 [M+H]+.
SW208781. Ethyl 2-((l1-oxidanyl)(propyl)-l3-sulfanyl)-3—amino—6—(thiophen yl)thieno[2,3—b]pyridinecarboxylate. t-BuOK (74.1 mg, 0.661 mmol) was added to a solution of ethyl 2-((((l1-oxidanyl)(propyl)-l3-sulfanyl)methyl)thio)cyano-6—(thiophen yl)isonicotinate (433.7 mg, 1.101 mmol) in DMF (4.3 mL) and the solution stirred for 40 minutes at 35°C. More t-BuOK (74.1 mg, 0.661 mmol) was added and allowed to stir at 35°C for an hour. The reaction mixture was diluted with EtOAc and washed with 10 % AcOH, and then multiple times with H20. The organic layer was separated, dried over MgSO4, filtered, and concentrated under reduced pressure. The crude product was ed using flash tography to give product in 30 % isolated yield. 1H NMR (400 MHZ, CDC13) 8 8.01 (s, 1H), 7.69 (dd, J: 3.8, 1.1Hz, 1H), 7.46 (dd, J: 5.0, 1.1Hz, 1H), 7.11 (dd, J: 5.0, 3.8 Hz, 1H), 6.09 (s, 2H), 4.49 (q, J = 7.1 Hz, 2H), 3.36 — 3.22 (m, 1H), 3.15 — 3.00 (m, 1H), 1.87 — 1.68 (m, 2H), 1.47 (t, J: 7.1 Hz, 3H), 1.07 (t, J: 7.4 Hz, 3H).ESI-MS (m/z): 394.9 [M+H]+.
N O S |\88I// HO 0 SW208782. -oxidanyl)(propyl)-l3-sulfanyl)amino(thiophen yl)thieno[2,3-b]pyridinecarboxylic acid. Followed the rd hydrolysis procedure using SW208781 as the starting material which gave product in 40 % isolated yield. 1H NMR (400 MHz, C3D7NO) 5 8.56 (s, 1H), 8.29 (d, J = 3.7 Hz, 1H), 8.19 (s, 1H), 7.99 (d, J = .0 Hz, 1H), 7.47 — 7.41 (m, 1H), 3.36 (ddd, J = 12.8, 8.4, 6.1 Hz, 1H), 3.24 (ddd, J: 12.8, 8.6, 6.8 Hz, 1H), 1.98 — 1.85 (m, 2H), 1.23 (t, J = 7.4 Hz, 3H). ESI—MS (m/Z): 366.8.
W0 2018f017582 2—(((isopropylthio)methyl)thio)phenyl(thiazolyl)nicotinonitrile.
Followed the standard alkylation procedure using 4-phenyl(thiazolyl)thioxo-1,2- opyridine—3-carbonitrile as the starting material and omethyl)(isopropyl)sulfane as the alkylating reagent. This was purified using automated flash chromatography to give product in 72 % isolated yield. 1H NMR (400 MHz, CDC13) 5 8.03 (s, 1H), 7.98 (d, J = 3.1 Hz, 1H), 7.70 — 7.62 (m, 2H), 7.57 (d, J = 3.1 Hz, 1H), 7.56 — 7.48 (m, 3H), 4.56 (s, 2H), 3.24 (hept, J = 6.7 Hz, 1H), 1.36 (d, J = 6.7 Hz, 6H). ESI—MS (m/z): 383.9. 2—(((isopropyl(ll-oxidanyl)-l3-sulfanyl)methyl)thio)-4—phenyl—6—(thiazol—2- yl)nicotinonitrile. Followed the standard oxidation procedure using 2- (((isopropylthio)methyl)thio)phenyl(thiazolyl)nicotinonitrile as the starting material.
This gave white solid product in 91 % yield. 1H NMR (400 MHZ, CDCl3) 5 8.12 (s, 1H), 8.00 (d, J = 3.1 Hz, 1H), 7.70 — 7.63 (m, 2H), 7.60 (d, J = 3.1 Hz, 1H), 7.58 — 7.51 (m, 3H), 4.63 (d, J = 13.2 Hz, 1H), 4.48 (d, J = 13.2 Hz, 1H), 3.09 (hept, J = 6.9 Hz, 1H), 1.42 (d, J: 10.5 Hz, 3H), 1.39 (d, J: 10.5 Hz, 3H). ESI-MS (m/z): 399.9.
SW208780. 2-(isopropyl(11-oxidanyl)-l3-sulfanyl)pheny1(thiazol eno[2,3-b]pyridinamine. t-BuOK (2.5 mg, 0.023 mmol) was added to a solution of 2—(((isopropyl(l 1—oxidanyl)-l3-sulfanyl)methyl)thio)phenyl-6—(thiazo1—2—yl)nicotinonitrile W0 2018/‘017582 2017/042620 (15 mg, 0.038 mmol) in DMF (148 11L), and stirred at 35°C for 40 minutes. The reaction mixture was diluted with EtOAc and washed with 10 % AcOH, then several times with H20.
The organic layer was dried over NaSO4, filtered, and concentrated under reduced pressure.
The crude product was purified using flash tography to give product‘ in 75 % yield. 1H NMR (400 MHz, CDC13) 6 8.05 (s, 1H), 7.91 (d, J = 3.2 Hz, 1H), 7.57 — 7.42 (m, 6H), 4.68 (s, 2H), 3.47 — 3.33 (m, 1H), 1.44 (d, J = 6.8 Hz, 3H), 1.27 (d, J = 6.8 Hz, 3H). ESI—MS (m/z): 399.9.
C” .N svi’w ] Methyl 4—(2-(((butyl(11-oxidanyl)-l3-sulfanyl)methyl)thio)cyano(thiazol- 2-yl)pyridin—4—yl)benzoate. Followed standard oxidation procedure using methyl 4—(2— (((butylthio)methyl)thio)cyano(thiazolyl)pyridinyl)benzoate as the starting material to give white solid in 98 % isolated yield. 1H NMR (400 MHz, CDC13) 8 8.15 (d, J = 8.3 Hz, 2H), 8.05 (s, 1H), 7.95 (d, J = 3.1 Hz, 1H), 7.68 (d, J = 8.3 Hz, 2H), 7.57 (d, J = 3.1 Hz, 1H), 4.68 (d, J = 13.1 Hz, 1H), 4.42 (d, J = 13.1 Hz, 1H), 3.91 (s, 3H), 3.01 — 2.86 (m, 1H), 2.87 — 2.74 (m, 1H), 1.88 — 1.72 (m, 2H), 1.55 — 1.35 (m, 2H), 0.91 (t, J = 7.3 Hz, 3H). ESI- MS (m/Z): 472.1 [M+H]+.
SW209127. Methyl 4-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3-b]pyridinyl)benzoate. t-BuOK (21.8 mg, 0.194 mmol) was added to methyl 4-(2-(((butyl(l1—oxidanyl)sulfany1)methyl)thio)cyano(thiazol-2—yl)pyridin—4— yl)benzoate (152.8 mg, 0.3239 mmol) in DMF (1.30 mL) and the solution stirred at 35°C for 40 minutes. The reaction mixture was d with EtOAc and washed with 10 % AcOH, and several times with H20. The organic layer was separated, dried over NaZSO4, ed, and concentrated under reduced pressure. The crude product was purified using automated flash W0 2018/‘017582 tography tO give the bright green t in 66 % isolated yield. 1H NMR (400 MHz, CDC13) 5 8.19 (d, J = 7.5 Hz, 2H), 8.04 (s, 1H), 7.91 (d, J = 3.2 Hz, 1H), 7.67 — 7.54 (m, 2H), 7.50 (d, 7 = 3.2 Hz, 1H), 3.97 (s, 3H), 3.27 (ddd, J = 12.8, 8.9, 6.2 Hz, 1H), 3.10 (ddd, J = 12.8, 9.0, 6.8 Hz, 1H), 1.81 — 1.63 (m, 2H), 1.54 — 1.39 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 472.1 [M+H]+.
S S / I0 \I / Si H0 0 SW209281. 4-(3 -amino(butyl(l l -oxidanyl)-l3-sulfanyl)-6—(thiazol yl)thieno[2,3-b]pyridinyl)benzoic acid. Followed standard hydrolysis procedure using SW209127 as the starting material to give bright green solid in 84 % isolated yield. 1H NMR (400 MHZ, CDC13) 8 8.16 (d, J = 8.4 Hz, 2H), 8.05 (s, 1H), 7.95 (d, J = 3.2 Hz, 1H), 7.68 — 7.55 (m, 2H), 7.52 (d, J = 3.2 Hz, 1H), 3.40 — 3.24 (m, 1H), 3.24 — 3.04 (m, 1H), 1.83 — 1.65 (m, 2H), 1.55 — 1.37 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 458.1 [M+H]+. \/\—\_ SW209282. 4-(3-amino(butyl(ll-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3—b]pyridinyl)-N,N-dimethylbenzamide. Followed rd amide bond coupling procedure using SW209281 as the starting material and dimethylamine hydrochloride as the coupling reagent. The product was purified using automated flash chromatography (20 % hexane, 80 % EtOAc) to give bright green solid in 59 % isolated yield 1H NMR (400 MHz, CDC13) 5 8.03 (s, 1H), 7.91 (d, J = 3.2 Hz, 1H), 7.66 — 7.44 (m, 5H), 3.36 — 3.21 (m, 1H), 3.14 (s, 3H), 3.13 — 3.06 (m, 1H), 3.02 (s, 3H), 1.81 — 1.64 (m, 2H), 1.55 — 1.41 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 485.1 [M+H]+.
W0 2018/‘017582 —124— HO 0 utylthio)methyl)thio)cyano(thiophenyl)isonicotinic acid.
Followed standard hydrolysis procedure using ethyl utylthio)methyl)thio)—3—cyano (thiopheny1)isonicotinate as the starting material to give isolated product in 94 % yield. 1H NMR (400 MHz, CDC13) 5 10.65 (s, 1H), 7.95 (s, 1H), 7.76 (dd, J = 3.8, 1.1 Hz, 1H), 7.57 (dd, J = 5.0, 1.0 Hz, 1H), 7.17 (dd, J = 5.0, 3.7 Hz, 1H), 4.46 (s, 2H), 2.72 (t, J = 7.2 Hz, 2H), 1.67 — 1.55 (m, 2H), 1.40 (h, J = 7.4 Hz, 2H), 0.90 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 365.0 [M+H]+. 2-(((butylthio)methyl)thio)cyano-N,N—dimethyl(thiophen y1)isonicotinamide. Followed standard amide bond coupling procedure using 2- (((butylthio)methyl)thio)cyano(thiophenyl)isonicotinic acid as the starting material and dimethylamine as the coupling reagent. The crude material was purified using automated flash tography (20 % EtOAc, 80 % hexanes) to give product in 53 % isolated yield. 1H NMR (400 MHz, Chloroform-d) 5 7.67 (d, J = 3.8 Hz, 1H), 7.54 (d, J = 5.0 Hz, 1H), 7.34 (s, 1H), 7.15 (t, J = 4.8, 3.9, 0.7 Hz, 1H), 4.49 (s, 2H), 3.16 (s, 3H), 2.98 (s, 3H), 2.72 (t, 2H), 1.62 (p, J = 7.7 Hz, 2H), 1.41 (h, J = 7.3 Hz, 2H), 0.90 (t, J = 7.7, 7.0 Hz, 3H). ESI—MS (m/z): 392.1 [M+H]+. 2-(((butyl(ll-oxidanyl)-l3-sulfanyl)methyl)thio)cyano-N,N-dimethyl-6— (thiophen—2—yl)isonicotinamide. Followed standard ion procedure using 2- (((butylthio)methyl)thio)cyano-N,N—dimethyl(thiophenyl)isonicotinamide as the starting material to give solid t in 85 % isolated yield. 1H NMR (400 MHz, CDC13) 8 7.67 (d, J = 3.8 Hz, 1H), 7.54 (d, J: 5.0 Hz, 1H), 7.34 (s, 1H), 7.20-7.06 (m, 1H), 4.49 (s, 2H), W0 2018/‘017582 3.16 (s, 3H), 2.98 (s, 3H), 2.72 (t, 2H), 1.62 (p, J = 7.7 Hz, 2H), 1.41 (h, J = 7.3 Hz, 2H), 0.90 (t, J = 7.7, 7.0 Hz, 3H). ESI-MS (m/z): 408.1 [M+H]+.
SW209283. 3-amino(butyl(11-oxidanyl)-l3-sulfanyl)-N,N-dimethyl (thiopheny1)thieno[2,3-b]pyridinecarboxamide. t-BuOK (6.5 mg, 0.058 mmol) was added to a solution of 2-(((butyl(l1-oxidanyl)-l3-sulfanyl)methyl)thio)cyano-N,N- dimethyl-6—(thiophenyl)isonicotinamide (39.2 mg, 0.962 mmol) in DMF (380 uL) and the on was heated at 35°C for 40 minutes. The reaction mixture was diluted with EtOAc and washed with 10 % AcOH, then several times with H20. The organic layer was separated and dried over NaZSO4, ed, and concentrated under reduced pressure. The crude material was ed using automated flash chromatography (20 % hexanes, 80 % EtOAc) to give the final product in 20 % isolated yield. 1H NMR (400 MHz, Chloroform-d) 8 7.66 (dd, J = 3.8, 1.1 Hz, 1H), 7.52 — 7.42 (m, 2H), 7.13 (dd, J = 5.0, 3.7 Hz, 1H), 3.34 — 3.23 (m, 1H), 3.21 (s, 3H), 3.15 — 3.02 (m, 1H), 2.96 (s, 3H), 1.79 — 1.62 (m, 2H), 1.55 — 1.36 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 408.1 . (3‘1le 0 \ I/ NH2L\_ 0 I OJ'VM ] SW212366. 4-(3-amino(butylsulfinyl)(thiazolyl)thieno[2,3-b]pyridin- 4-yl)benzyl ylglycinate. N,N-Dimethylglycine (3.5 mg, 0.034 mmol), 1—ethyl(3- dimethylaminopropyl)carbodiimide (6.5 mg, 0.034 mmol), and DMAP (4.1 mg, 0.0334 mmol) were added to SW209510 (10 mg, 0.023 mmol) and dissolved in DMF (270 uL). The reaction mixture stirred at room temperature overnight, then neutralized with 1M NaOH, washed with H20 and extracted with EtOAc. The organic layer was dried over Na2804, filtered and concentrated under reduced pressure. The crude product was purified using flash chromatography (7 % MeOH, 93 % DCM) to give a quantitative yield of green solid product 1H NMR (400 MHz, CDC13) 5 8.02 (s, 1H), 7.90 (d, J = 3.2 Hz, 1H), 7.56 — 7.43 (m, 5H), 5.25 W0 2018f017582 (s, 2H), 4.61 (s, 2H), 3.35 — 3.27 (m, 1H), 3.26 (s, 2H), 3.16 — 3.04 (m, 1H), 2.37 (s, 6H), 1.78 — 1.63 (m, 2H), 1.55 — 1.39 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 529.1 [M+H]+.
OVQ/O\/U\O/ Methyl 2—(4-formylphenoxy)acetate. To a solution of oxybenzaldehyde (3.0 g, 25 mmol) in acetone (61.4 mL), K2C03 (5.43 g, 39.3 mmol) was added and the mixture was stirred vigorously. Methylbromoacetate (2.8 mL, 29 mmol) was added and the mixture was stirred for 3.5 hrs at room temperature. The reaction mixture was concentrated down under d pressure then washed with H20 and extracted with EtOAc. The organic layer was separated, dried over NaZSO4, filtered, and trated to give a colorless oil that solidified under vacuum in 82 % isolated yield. 1H NMR (400 MHz, CDC13) 8 9.87 (s, 1H), 7.82 (d, J = 8.8 Hz, 2H), 6.98 (d, J: 8.7 Hz, 2H), 4.70 (s, 2H), 3.79 (s, 3H). ESI-MS (m/z): 195.1 [M+H]+.
[N‘ \ Methyl (E)-2—(4-(3-oxo(thiazol-2—yl)propenyl) phenoxy)acetate. 2- acetylthiazole (534 1.1L, 5.15 mmol) was added to a solution of methyl 2-(4- formylphenoxy)acetate (1.0 g, 5.2 mmol) in MeOH (11mL) under N2. NaOMe (279 mg, 5.15 mmol) was added last and the reaction mixture stirred at room temperature overnight. The reaction e was filtered, and the precipitate was washed with small amount of MeOH then diluted with DCM and washed with H20. The organic layer was separated, dried over Na2804, filtered, and concentrated under reduced pressure. This gave solid product in 21 % isolated yield. 1H NMR (400 MHz, Chloroform—d) 5 8.03 (d, J = 3.0 Hz, 1H), 7.95 (d, J = .9 Hz, 1H), 7.82 (d, J = 16.0 Hz, 1H), 7.70 — 7.61 (m, 3H), 6.92 (d, J = 8.8 Hz, 2H), 4.67 (s, 2H), 3.80 (s, 3H). ESI—MS (m/z): 304.1 [M+H]+.
W0 017582 Methyl 2-(4-(2-(((butylthio)methyl)thio)cyano(thiazolyl)pyridin noxy)acetate. EtOH (495 uL) was added to 2-cyanothioacetamide (49.5 mg, 0.494 mmol) and (E)(4-(3-oxo(thiazolyl)prop-l-en-l-yl)phenoxy)acetate (50 mg, 0.16 mmol), followed by 1 drop of piperidine. The reaction mixture stirred at 80°C for 4 hours then was concentrated under reduced pressure and the crude was carried forward to the next step. Followed the standard alkylation procedure, using methyl 2-(4-(3-cyano (thiazoly1)-2—thioxo-l,2-dihydropyridinyl)phenoxy)acetate as the starting material and buty1(chloromethy1)sulfane as the alkylating reagent. The crude product was purified using automated flash chromatography (20 % EtOAc, 80 % hexanes) to give solid t in 70 % isolated yield. 1H NMR (400 MHz, CDC13) 5 7.96 (s, 1H), 7.95 (d, J = 3.1 Hz, 1H), 7.62 (d, J = 8.8 Hz, 2H), 7.54 (d, J = 3.1 Hz, 1H), 7.02 (d, J = 8.8 Hz, 2H), 4.69 (s, 2H), 4.49 (s, 2H), 3.81 (s, 3H), 2.73 (t, J = 7.3 Hz, 2H), 1.68 — 1.56 (m, 2H), 1.46 — 1.34 (m, 2H), 0.89 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 486.1 [M+H]+. fl <2 3 N\ Svs\/\/ 09°09 Methyl 2-(4-(2-(((buty1(ll-oxidanyl)-l3-sulfanyl)methyl)thio)—3—cyano (thiazol-2—yl)pyridinyl)phenoxy)acetate. Followed the standard oxidation procedure using methyl 2—(((butylthio)methyl)thio)cyano(thiazolyl)pyridin yl)phenoxy)acetate as the starting material. The crude product was purified using automated flash chromatography (50 % EtOAc, 50 % hexanes). 1H NMR (400 MHZ, CDC13) 8 7.97 (s, 1H), 7.90 (d, J = 3.1 Hz, 1H), 7.57 (d, J = 8.8 Hz, 2H), 7.52 (d, J = 3.1 Hz, 1H), 6.98 (d, J = 8.8 Hz, 2H), 4.65 (s, 2H), 4.62 (d, J = 13.1 Hz, 1H), 4.37 (d, J = 13.1 Hz, 1H), 3.75 (s, 3H), 2.96 — 2.84 (m, 1H), 2.81 — 2.71 (m, 1H), 1.76 (p, J = 7.6 Hz, 2H), 1.51 — 1.33 (m, 2H), 0.88 (t, J: 7.3 Hz, 3H). ESI-MS (m/z): 502.1 [M+H]+.
W0 2018/‘017582 SW212365. Methyl 2-(4-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)—6- (thiazolyl)thieno[2,3-b]pyridinyl)phenoxy)acetate. Methyl 2-(4-(2-(((buty1(11- oxidanyl)-l3-su1fanyl)methyl)thio)cyano(thiazolyl)pyridinyl)phenoxy)acetate (80 mg, 0.16 mmol) and t-BuOK (10.7 mg, 0.0954 mmol) were combined in a vial that was evacuated and lled with N2 three times, then DMF (627 uL) was added and N2 was bubbled through the solution. The reaction mixture was stirred at room temperature for about minutes and then was diluted with EtOAc and washed with 10 % AcOH. The c layer was washed several times with water, dried over Na2804, filtered, and concentrated.
The crude product was purified using automated flash tography (30 % EtOAc, 70 % hexanes) to give green solid with 56 % isolated yield. 1H NMR (400 MHz, CDC13) 8 7.97 (s, 1H), 7.89 — 7.86 (m, 1H), 7.47 (d, J = 3.1 Hz, 1H), 7.46 — 7.37 (m, 2H), 7.02 (d, J = 8.5 Hz, 2H), 4.70 (s, 2H), 4.66 (s, 2H), 3.82 (s, 3H), 3.34 — 3.18 (m, 1H), 3.16 — 3.01 (m, 1H), 1.77 — 1.64 (m, 2H), 1.51 — 1.38 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). ESI-MS (m/z): 502.1 [M+H]+ SW212364. 2-(4-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)—6—(thiazol yl)thieno[2,3-b]pyridinyl)phenoxy)ethanol. Followed the same ure as for 2- (((butylthio)methyl)thio)(4-(hydroxymethyl)phenyl)(thiazolyl)nicotinonitrile using SW212365 as the starting material to give a quantitative yield of desired productlH NMR (400 MHZ, CDC13) 8 8.01 (s, 1H), 7.90 (d, J = 3.2 Hz, 1H), 7.48 (d, J: 3.1 Hz, 1H), 7.46 — 7.35 (m, 2H), 7.07 — 6.99 (m, 2H), 4.69 (s, 2H), 4.18 — 4.11 (m, 2H), 4.04 — 3.96 (m, 2H), 3.34 — 3.23 (m, 1H), 3.17 — 3.04 (m, 1H), 1.80 — 1.61 (m, 2H), 1.53 — 1.40 (m, 2H), 0.93 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 474.1 [M+H]+.
W0 2018/‘017582 PCT/U82017/042620 sw212363. 2-(4-(3-amino(butyl(l1-oxidanyl)-l3-sulfanyl)(thiazol yl)thieno[2,3-b]pyridinyl)phenoxy)acetic acid. Followed the standard hydrolysis procedure using SW212365 as the starting material to give a quantitative yield. 1H NMR (400 MHz, MeOD) 8 7.98 (s, 1H), 7.94 (d, J = 3.2 Hz, 1H), 7.74 (d, J = 3.2 Hz, 1H), 7.46 (d, J = 8.4 Hz, 2H), 7.14 (d, J = 8.9 Hz, 2H), 4.67 (s, 2H), 3.35 — 3.24 (m, 1H), 3.16 — 3.04 (m, 1H), 1.78 — 1.57 (m, 2H), 1.55 — 1.43 (m, 2H), 0.95 (t, J = 7.3 Hz, 3H). ESI—MS (m/z): 488.1 [M+H]+.
S nthesis of chlorometh 1 thio ethers 4-mercaptobutyl acetate pomnellpase HCI(g)I CH 0 HSNOH —_HSN —2- Ago/\Nsvc' ElOAo.30°C 0“ s S H 5 s .
I / N s /\/\/5v°‘ I/ N “0 S I N O / N S 9 . I; vs 202 5‘ ‘au —’ I é —> | / § / E03N,CH30N,IefluX / CN CN H / 35°C CHC|3.32°C 0N NH \2 cm OAC DA: SVVZUQ1 29 K2603IMEOH. H20 / N\ s K2003, MeOH. H20 |// S NHZW‘c SVII209271 I N I I / IN / IN 5 I: o I 6 Svs / Ac0H, H220, CHCI, 32 °c I‘N/S\’sr H20KOH, DMF Me0H. IN s \ \ I/ MsCI. E N. DCM. 0°C L-C—z—d—UDMFary. VSH \ §l “ 1L 1LNHZ SVIIZOQGZQ HS\/\/\0Ac 4-mercaptobutyl e. e lipase (2.35 g) was added to a solution of 4- mercapto—l—butanol (2.45 g, 23.10 mmol) in ethyl acetate (42.0 ml). The reaction was heated at 30°C for 6 days. Despite incomplete sion the mixture was filtered and condensed.
Purification was carried out on an automated flash chromatography system in 100% DCM to give oil in 84% yield. 1H NMR (400 MHz, CHC13) 5 4.08 (t, J = 6.2 Hz, 2H), 2.57 (q, J = 7.1 Hz, 2H), 2.05 (s, 3H), 1.85 — 1.59 (m, 4H), 1.36 (t, J = 7.9 Hz, 1H).
W0 2018f017582 AGO/\/\/S\/CI 4—((chloromethyl)thio)butyl acetate. Hydrogen chloride gas was bubbled for 40 s into 4—mercaptobutyl acetate (2.84 g, 19.2 mmol) which had been cooled in a dry ice/acetone bath and until the internal temperature stabilized before paraformaldehyde (0.815 g, 27.17 mmol) was slowly added using a solid addition funnel. The reaction was stirred cold for 3 hours during which hydrogen chloride ng was continued and then ceased as the reaction was warmed gently to ambient temperature and stirred overnight. The crude mixture was d with minimal DCM. The aqueous phase was removed and the organic layer was washed with brine and dried over NaZSO4, filtered and condensed to give an Oil in 62% yield. 1H NMR (400 MHz, CHC13) 4.75 (s, 2H), 4.10 (t, J = 6.0 Hz, 2H), 2.95 — 2.66 (m, 2H), 2.06 (s, 3H), 1.85 — 1.67 (m, 4H).
I / N |\ S\/s ““ 1L 4-((((3-cyanophenyl(thiophenyl)pyridinyl)thio)methyl)thio)butyl acetate. A mixture of loromethyl)thio)butyl acetate (602.3 mg, 3.1 mmol), 4—phenyl (thiophen—2—y1)—2—thioxo-1,2-dihydropyn'dinecarbonitrile (352.2 mg, 1.2 mmol) and triethylamine (250 ml, 1.8 mmol) in itrile (1.2 ml) was d for three hours. The crude mixture was then condensed and purified on an automated flash chromatography system in 0-40% EtOAc/hexanes. Fractions containing the desired product were further purified on an automated flash chromatography in 0-30% EtOAc/hexanes to give a clear oil in 41 % yield. 1H NMR (400 MHz, CHCl3) 5 7.72 (dd, J: 3.7, 1.1 Hz, 1H), 7.64 — 7.59 (m, 2H), 7.55 (dt, J = 5.6, 2.3 Hz, 4H), 7.44 (s, 1H), 7.17 (dd, J = 5.0, 3.8 Hz, 1H), 4.55 (s, 2H), 4.11 — 4.02 (m, 2H), 2.86 — 2.63 (m, 2H), 2.05 (s, 3H), 1.77 (t, J = 3.4 Hz, 4H). ESI-MS (m/z): 455.1 [M+H]+.
I / N I 3&3 2-((((4-hydroxybutyl)thio)methyl)thio)phenyl(thiophen yl)nicotinonitrile. K2C03 (157.7 mg, 1.14 mmol) was added to a solution of 4—((((3—cyano W0 017582 phenyl(thiophenyl)pyridinyl)thio)methyl)thio)butyl acetate. (245.4 mg, 0.54 mmol) in methanol (8.0 ml) and water (2.0 ml) and the reaction was stirred for 2 hours. The e was dried then diluted with EtOAc and washed twice with water and then brine. The organic layer was dried over Na2S04, ed and concentrated under reduce pressure to give desired product in 71% yield. 1H NMR (400 MHz, CDC13) 5 7.72 (dd, J = 3.7, 1.1 Hz, 1H), 7.61 (dd, J = 6.6, 3.0 Hz, 2H), 7.54 (dd, J = 5.1, 2.2 Hz, 4H), 7.43 (s, 1H), 7.16 (dd, J: 5.0, 3.8 Hz, 1H), 4.55 (s, 2H), 3.67 (t, J = 6.2 Hz, 2H), 2.80 (t, J = 7.1 Hz, 2H), 1.84 — 1.63 (m, 4H). ESI—MS (m/z): 413.1 [M+H]+. 4-((((3-cyanophenyl(thiophenyl)pyridinyl)thio)methyl)sulfinyl)butyl acetate. Acetic acid (370 pl) and hydrogen peroxide (29 pl, 30 % solution in water) were added to the solution of 4-((((3-cyanophenyl(thiophenyl)pyridin o)methyl)thio)butyl acetate (85.2 mg, 0.19 mmol) in chloroform (370 pl). The reaction mixture was stirred at 32 0C for 90 min. Once complete, the reaction was diluted with chloroform and washed with saturated NaHCO3 on, and extracted three times with chloroform. The combined organic layers was dried over NaZSO4, filtered and concentrated under reduce pressure to give designed product in 94% yield. 1H NMR (400 MHz, CDClg) 5 7.77 (d, J = 3.8 Hz, 1H), 7.62 (dd, J = 4.1, 2.3 Hz, 2H), 7.60 — 7.54 (m, 4H), 7.51 (s, 1H), 7.19 (dd, J = 5.0, 3.8 Hz, 1H), 4.78 (d, J = 13.0 Hz, 1H), 4.44 (d, J = 13.0 Hz, 1H), 4.11 (t, J = 6.4 Hz, 2H), 3.03 (dt, J = 12.9, 8.0 Hz, 1H), 2.87 (dt, J = 12.8, 7.3 Hz, 1H), 2.05 (s, 3H), 2.03 — 1.77 (m, 4H). ESI-MS (m/z): 471.1 [M+H]+.
SW209129. 4-((3-aminopheny1(thiophenyl)thieno[2,3-b]pyridin yl)su1finy1)butyl acetate. Potassium tert-butoxide (9.7 mg, 0.086 mmol) was added to a solution of 4—((((3—cyanophenyl(thiophenyl)pyridinyl)thio)methy1)sulfinyl)butyl W0 2018/‘017582 acetate (58.2 mg, 0.12 mmol) in DMF (490 pl). The reaction mixture was stirred at 35°C for 45 minutes, then diluted with EtOAc and washed several times with water. The aqueous layer was also back—extracted. The combined organic layer was washed with brine, dried over Na2S04, ed and concentrated under reduce pressure. Purification was carried out using automated flash chromatography in 0-90% EtOAc/hexanes to give the d product in 44% yield. 1H NMR (400 MHz, CDC13) 5 7.63 — 7.49 (m, 5H), 7.45 (dd, J = 4.9, 1.1 Hz, 2H), 7.41 (s, 1H), 7.10 (dd, J = 5.0, 3.7 Hz, 1H), 4.59 (bs, 2H), 4.08 (t, J = 5.8 Hz, 2H), 3.39 — 3.23 (m, 1H), 3.10 (ddd, J = 12.8, 8.4, 6.2 Hz, 1H), 2.03 (s, 3H), 1.96 — 1.72 (m, 4H). ESI—MS (m/z): 471.1 [M+H]+.
I/ IN\ 3%,,0 / / LL SW209128. 4-((3-aminophenyl(thiophenyl)thieno[2,3-b]pyridin-2— y1)su1fony1)buty1 acetate. Isolated as the over oxidation product from 4-((3-aminophenyl- 6—(thiophen—2—yl)thieno[2,3-b]pyridinyl)sulfinyl)butyl acetate in 13.5 % yield. 1H NMR (400 MHz, CDC13) 5 7.71 (dd, J = 3.7, 1.1 Hz, 1H), 7.61 — 7.55 (m, 3H), 7.53 — 7.44 (m, 4H), 7.15 (dd, J = 5.0, 3.7 Hz, 1H), 5.10 (s, 2H), 4.06 (t, J = 6.3 Hz, 2H), 3.32 — 3.18 (m, 2H), 2.02 (s, 3H), 1.98 — 1.87 (m, 2H), 1.77 (dt, J = 8.6, 6.4 Hz, 2H). ESI-MS (m/z): 487.1 .
SW209271. 4-((3-aminophenyl(thiophenyl)thieno[2,3-b]pyridin yl)sulfinyl)butanol. K2C03 (12.5 mg, 0.09 mmol) was added to a solution of SW209129 (18.7 mg, 0.04 mmol) in methanol (470 pl) and water (100 pl) and the reaction was d for 2.5 hours. The mixture was dried then diluted with EtOAc and washed twice with water and then brine. The organic layer was dried over Na2S04, filtered and concentrated under reduce pressure to give desired product in 80% yield. 1H NMR (400 MHz, CDC13) 8 7.62 (d, J = 1.1 Hz, 1H), 7.60 — 7.50 (m, 4H), 7.49 — 7.41 (m, 3H), 7.11 (dd, J = 5.0, 3.7 Hz, 1H), 4.68 W0 2018f017582 — 4.44 (s, 2H), 3.67 (t, J = 6.1 Hz, 2H), 3.42 — 3.27 (m, 1H), 3.13 (ddd, J = 12.9, 8.4, 6.9 Hz, 1H), 1.93 — 1.65 (m, 4H). ESI-MS (m/z): 429.0 [M+H]+.
'/ “\S | \,s C“ 1L 4—((((3-cyanophenyl(thiophenyl)pyridinyl)thio)methyl)thio)butyl methanesulfonate. A solution of triethylamine (38 pl, 028 mmol) in anhydrous DCM (1.0 ml) is cooled in an ice bath before the addition of 2-((((4-hydroxybutyl)thio)methy1)thio) phenyl(thiophenyl)nicotinonitrile ( 40.6 mg, 0.098 mmol) followed by dropwise on of methanesulfonyl chloride (17.5 pl, 023 mmol). After 30 minutes the crude mixture was washed with brine and dried over Na2S04, filtered and condensed to give desired product in 98% yield. 1H NMR (400 MHz, CDCl3) 5 7.74 (dd, J = 3.9, 1.1 Hz, 1H), 7.64 — 7.50 (m, 7H), 7.17 (dd, J = 5.1, 3.8 Hz, 1H), 5.46 (s, 2H), 4.01 — 3.78 (m, 2H), 2.65 (ddd, J = .1, 5.3, 1.9 Hz, 2H), 2.52 — 2.37 (m, 4H). ESI-MS (m/z): 491.1 [M+H]+. 2—((((4-chlorobutyl)thio)methyl)thio)phenyl(thiophen-2—y1)nicotinonitrile.
Lithium chloride (32.0 mg, 0.75 mmol) was added to a solution of 4-((((3-cyano—4—phenyl heny1)pyridinyl)thio)methyl)thio)butyl esulfonate (21.8 mg, 0.044 mmol) in DMF (0.4 ml). The reaction went to completion within two days. The mixture was d with EtOAc and washed several times with water and then brine. The organic layer was dried over Na2SO4, filtered and condensed. Purification was performed on an automated chromatography system in 0-40% EtOAc/hexanes and gave the d product in 76% yield. 1H NMR (400 MHz, CDC13) 5 7.72 (dd, J = 3.7, 1.1 Hz, 1H), 7.62 (dd, J = 6.5, 3.0 Hz, 2H), 7.59 — 7.52 (m, 4H), 7.44 (s, 1H), 7.17 (dd, J = 5.0, 3.8 Hz, 1H), 4.56 (s, 2H), 3.56 (t, J = 6.3 Hz, 2H), 2.80 (t, J = 7.0 Hz, 2H), 1.95 — 1.79 (m, 4H). ESI-MS (m/z): 431.0 [M+H]+. —134— 2—((((4-chlorobutyl)sulfinyl)methyl)thio)phenyl(thiophen—2— yl)nicotinonitrile. Acetic acid (70 pl) and hydrogen peroxide (5.2 pl, 30 % on in water) were added to the solution of 2-((((4-chlorobutyl)thio)methyl)thio)phenyl-6—(thiophen yl)nicotinonitrile (14.6 mg, 0.034 mmol) in chloroform (70 pl). The reaction mixture was stirred at 32°C for 40 min and then diluted with chloroform and was washed with saturated NaHCO3 on and extracted three times with chloroform. The combined organic layers was dried over Na2SO4, filtered and concentrated under reduce pressure to give ed product. 1H NMR (400 MHz, CDC13) 5 7.77 (d, J = 3.8 Hz, 1H), 7.62 (dd, J = 6.6, 2.9 Hz, 2H), 7.57 (q, J = 4.5, 3.1 Hz, 4H), 7.51 (s, 1H), 7.19 (t, J = 4.4 Hz, 1H), 4.77 (d, J = 13.0 Hz, 1H), 4.47 (d, J = 13.0 Hz, 1H), 3.58 (t, J = 6.2 Hz, 2H), 3.03 (dt, J = 13.2, 7.7 Hz, 1H), 2.87 (dt, J: 13.4, 7.0 Hz, 1H), 2.16 — 1.87 (m, 4H). ESI-MS (m/z): 447.1 [M+H]+.
S .
’/ ”\S O |// +074 NH H2 SW209329. 2-((4-chlorobutyl)sulfinyl)phenyl(thiophen—2—yl)thieno[2,3- b]pyridin-3—amine. A basic methanolic solution (1.0 mg, 0.018 mmol of potassium hydroxide in 12.0 pl water and 57.5 pl methanol) was erred to a Vial containing a solution of 2-((((4-chlorobutyl)sulfinyl)methyl)thio)phenyl(thiophen yl)nicotinonitrile (12.4 mg, 0.028 mmol) in dimethylformamide (91.5 pl). The reaction was heated at 38 0C for 30 s, before being cooled, diluted with EtOAc and washed several times with water, then brine. The organic layer was dried over Na2SO4, filtered and condensed. The crude mixture was ed using an automated chromatography system in 0- 60% EtOAc/hexanes. Isolated yield = 65 %. 1H NMR (400 MHz, CDCl3) 5 7.64 (d, J = 3.7 Hz, 1H), 7.61 — 7.50 (m, 4H), 7.50 — 7.43 (m, 3H), 7.12 (t, J = 4.4 Hz, 1H), 4.59 (s, 2H), 3.66 — 3.47 (m, 2H), 3.40 — 3.24 (m, 1H), 3.20 — 3.06 (m, 1H), 1.95 (q, J = 5.6 Hz, 4H). ESI-MS (m/z): 447.0 [M+H]+.
W0 2018/‘017582 wrcinelipase Hcl . H5 5 cl HSMOH HSMDAC (9) Was . W V EIOAC. 30 °C 3 - l H o svs N‘ :§\#—’MeOHH20NH; I N N‘ / N\ s / I AUDMSAC! l ALOH, H202 KOH MeOH I<co | / § / EHN. CchN CN \ CHcla, 32 “e \DMF35C NH2 DAD \OAC BAG DH /2C03 swzuszn swzuszu MeO‘H H20 1:]s 5 i o— s N N i 5V5 \ 5 / ‘ 5 ' / NaH CH3| DMF I vs ACQH H202 .—_ l V? KoH.MeoH, (M swzuszve / / cN CN eHeIsz: CN DMFJSDC OMe OMe OMe swzuszu s 5 - ©\/N\f p] i N O s r :5»; / 5 ‘ \,§' I § 5W209330 AwH,H202 | KoH. MeoH, / DMF 33 °c EWIDCMIUIC I CN \\\\Cl CHCIS‘ 32 “c CN \ Ms—CI LiCl DMF II”VI NH2 / 5.
Cl | S be _ . ~ . . \ s» N\ v I S I O / S swzogsst / WW Kryplcfixzzz / N\ 5&5 AcOH,H202 N\ , ‘ 5&5! KOH‘MeOH / / CN —- | fl , —D- / 32“C / DMFJE c K2C03,KF,DMAC,BU c CN CN \ NH2 0M5 / F KCN 6 s DMF 85 C N I / f H—202AoOH N\ 5&5 KoH.MeoH. (WM 5 / \\\\CN chI3 32 “c / DMF“DC CN ‘\ / / §\ smogssz CN \j EN HSMOAC 3-mercaptopropyl acetate. Porcine lipase (5.52 g) was added to a solution of 3- mercapto-l-propanol (5.03 g, 54.6 mmol) in ethyl acetate (70 ml). The reaction was heated at 28 0C for 12 days. Despite incomplete conversion the e was filtered and condensed. cation was carried out on an automated flash chromatography system in 100% DCM to give oil in 66% yield. 1H NMR (400 MHz, CHC13) 5 4.17 (t, J = 6.2 Hz, 2H), 2.60 (q, J = 7.4 Hz, 2H), 2.05 (s, 3H), 1.93 (p, J = 6.6 Hz, 2H), 1.39 (t, J = 8.1 Hz, 2H).
HSWSVCI 3-((chloromethyl)thio)propanethiol. en chloride gas was bubbled for 60 minutes into 3-((chloromethyl)thio)propanethiol (4.80 g, 35.7 mmol) which had been cooled in a dry ice/acetone bath and until the al temperature stabilized before paraformaldehyde (1.59 g, 53.3 mmol) was slowly added using a solid addition funnel. The reaction was stirred cold for 1.5 hours during which hydrogen chloride bubbling was continued and then ceased as the reaction was warmed gently to t temperature and stirred ght. The crude mixture was diluted with minimal DCM. The aqueous phase was removed and the organic layer was washed with brine and dried over Na2504, filtered and condensed to give an oil in 80% yield of a mixture of 2.4:1 desired monomer chloride to W0 2018/‘017582 diacetate dimer. 1H NMR (400 MHz, CHC13) 5 4.74 (s, 2H), 4.17 (t, J = 6.4 Hz, 2H), 2.91 — 2.77 (m, 2H), 2.06 (d, J = 1.0 Hz, 3H), 2.03 — 1.94 (m, 2H).
I / N\ | S\/S .1 B... ] 3—((((3-cyanophenyl(thiophenyl)pyridinyl)thio)methyl)thio)propyl acetate. Prepared analogously to 4-((((3-cyanophenyl(thiophen-2—yl)pyridin—2— yl)thio)methyl)thio)butyl acetate in 26% isolated). 1H NMR (400 MHz, CHC13) 8 7.72 (dd, J = 3.8, 1.1 Hz, 1H), 7.66 — 7.58 (m, 2H), 7.54 (dd, J = 4.2, 2.9 Hz, 4H), 7.44 (d, J = 1.3 Hz, 1H), 7.17 (dd, J = 5.0, 3.8 Hz, 1H), 4.55 (s, 2H), 4.18 (t, J = 6.3 Hz, 2H), 2.84 (t, J = 7.3 Hz, 2H), 2.05 (s, 3H), 2.05 — 1.97 (m, 2H). ESI—MS (m/z): 441.0 [M+H]+.
I / N\ is 3—((((3—cyanophenyl(thiophen-2—yl)pyridin—2— yl)thio)methyl)sulfinyl)propyl acetate. Prepared analogously to 4-((((3—cyano—4—phenyl (thiophen—2—y1)pyridinyl)thio)methyl)sulfinyl)butyl acetate in 90% yield. 1H NMR (400 MHz, CHC13) 5 7.77 (dd, J = 3.7, 1.1 Hz, 1H), 7.68 — 7.60 (m, 2H), 7.57 (ddd, J: 6.9, 4.5, 2.0 Hz, 4H), 7.51 (s, 1H), 7.19 (dd, J: 5.0, 3.8 Hz, 1H), 4.81 (d, J: 13.1 Hz, 1H), 4.44 (d, J = 13.1 Hz, 1H), 4.22 (td, J = 6.3, 1.3 Hz, 2H), 3.09 (dt, J: 13.0, 8.1 Hz, 1H), 2.89 (dt, J = 13.1, 7.1 Hz, 1H), 2.28 — 2.17 (m, 2H), 2.04 (s, 3H). ESI-MS (m/z): 457.1 [M+H]+.
SW209273. 3-((3-aminophenyl(thiophen-2—yl)thieno[2,3—b]pyridin—2— yl)sulfiny1)propyl acetate. Prepared ously to 4-((3-aminopheny1—6—(thiophen—2— y1)thieno[2,3-b]pyridin-2—yl)sulfinyl)butyl acetate in 37% yield. 1H NMR (400 MHz, CHC13) 7.62 — 7.52 (m, 5H), 7.45 (dd, J = 5.0, 1.1 Hz, 2H), 7.41 (s, 1H), 7.10 (dd, J = 5.0, 3.7 Hz, W0 2018/‘017582 1H), 4.65 — 4.56 (s, 2H), 4.27 — 4.14 (m, 2H), 3.42 — 3.25 (m, 1H), 3.15 (dt, J = 12.9, 7.7 Hz, 1H), 2.09 (ddd, J = 7.5, 6.2, 1.3 Hz, 2H), 2.05 (s, 3H). ESI-MS (m/z): 457.1 [M+H]+.
/ N\ s Q ’90 / / SK SW209272. 3-((3-aminophenyl(thiophenyl)thieno[2,3-b]pyridin yl)sulfonyl)propyl acetate. Isolated as the over oxidation product from 3-((3-amino (thiophen-2—yl)thieno[2,3-b]pyridinyl)sulfinyl)propyl acetate in 7 % yield. 1H NMR (400 MHz, CHC13) 5 7.72 (d, J = 3.8 Hz, 1H), 7.62 — 7.53 (m, 3H), 7.54 — 7.45 (m, 4H), 7.15 (dd, J = 5.0, 3.8 Hz, 1H), 5.12 (s, 2H), 4.15 (t, J = 6.2 Hz, 2H), 3.39 — 3.19 (m, 2H), 2.25 — 2.12 (m, 2H), 2.03 (s, 3H). ESI—MS (m/z): 457.1 . s , i / N S ,0 I S / / *\ SW209274. 3-((3-aminophenyl(thiophenyl)thieno[2,3—b]pyridin yl)sulfiny1)propan—1-ol. Was prepared analogously to SW209271. 4-((3—amino—4—phenyl (thiophen—2—y1)thieno[2,3-b]pyridinyl)sulfinyl)butanol in 84% yield. 1H NMR (400 MHz, CHC13) 5 7.63 (dd, J = 3.7, 1.1 Hz, 1H), 7.55 (p, J = 4.6, 3.2 Hz, 4H), 7.46 (dd, J: 5.0, 1.1 Hz, 2H), 7.43 (s, 1H), 7.11 (dd, J = 5.0, 3.7 Hz, 1H), 4.60 (s, 2H), 3.77 (t, J = 5.8 Hz, 2H), 3.49 — 3.33 (m, 1H), 3.21 (dt, J = 13.5, 6.9 Hz, 1H), 2.13 — 1.98 (m, 2H). ESI-MS (m/z): 415.1 [M+H]+.
/|\N3\/s «.103 2—((((3—hydroxypropyl)thio)methyl)thio)phenyl(thiophen—Z— otinonitrile. Prepared analogously to 2-((((4-hydroxybutyl)thio)methyl)thio)phenyl- 6-(thiophen-2—yl)nicotinonitrile in 98% yield. 1H NMR (400 MHz, CHC13) 7.71 (dd, J = 3.8, 1.1 Hz, 1H), 7.63 — 7.57 (m, 2H), 7.55 — 7.50 (m, 4H), 7.41 (s, 1H), 7.15 (dd, J = 5.0, 3.8 Hz, W0 2018f017582 1H), 4.54 (s, 2H), 3.76 (t, J = 6.1 Hz, 2H), 2.88 (t, J = 7.1 Hz, 2H), 1.93 (ddd, J = 13.2, 7.1, 6.1 Hz, 2H), 1.88 — 1.80 (m, 1H). ESI—MS (m/z): 399.1 [M+H]+. l/ N\ | Svs 2—((((3-methoxypropyl)thio)methyl)thio)phenyl(thiophen yl)nicotinonitrile. Sodium hydride (micro spatula ) was added to an ice-cooled solution of 2-((((3 -hydroxypropyl)thio)methyl)thio)phenyl(thiophen yl)nicotinonitrile(4l.6 mg, 0.10 mmol) in DMF( 1.0 ml). The mixture was stirred cold for 15 minutes before the on of methyl iodide (34 ml, 0.55 mmol). The mixture was stirred cold in the melting th for 2 hours, then diluted with EtOAc and washed several times with water and then brine. The organic layer was dried over Na2804, filtered and condensed.
Purification was carried out on an automated flash chromatography system in 0—40% EtOAc/hexanes with an isolated yield of 56%. 1H NMR (400 MHz, CHC13) 5 7.72 (dd, J = 3.8, 1.1Hz, 1H), 7.61 (dd, J = 6.6, 3.1 Hz, 2H), 7.57 — 7.51 (m, 4H), 7.43 (s, 1H), 7.17 (dd, J = .0, 3.8 Hz, 1H), 4.55 (s, 2H), 3.49 (t, J = 6.1 Hz, 2H), 3.34 (s, 3H), 2.85 (t, J = 7.3 Hz, 2H), 1.95 (ddd, J: 13.4, 7.3, 6.1 Hz, 2H). ESI-MS (m/z): 413.1 [M+H]+. 2-((((3-methoxypropyl)sulfinyl)methyl)thio)phenyl(thiophen-2— yl)nicotinonitrile. Prepared analogously to 3-cyanophenyl(thiophenyl)pyridin- 2-yl)thio)methy1)sulfinyl)butyl acetate in 71% yield. 1H NMR (400 MHz, CHC13) 8 7.76 (dd, J = 3.8, 1.1 Hz, 1H), 7.65 — 7.58 (m, 2H), 7.56 (td, J = 4.6, 2.0 Hz, 4H), 7.49 (s, 1H), 7.18 (dd, J = 5.0, 3.8 Hz, 1H), 4.73 (d, J = 13.1 Hz, 1H), 4.47 (d, J = 13.0 Hz, 1H), 3.54 (qt, J = 9.5, 5.8 Hz, 2H), 3.34 (s, 3H), 3.14 (dt, J = 13.1, 7.9 Hz, 1H), 2.89 (ddd, J = 13.1, 8.0, 6.4 Hz, 1H), 2.20 — 2.08 (m, 2H). ESI-MS (m/z): 429.1 [M+H]+.
W0 2018/‘017582 SW209276. 2-((3-methoxypropyl)sulfinyl)phenyl(thiophen—2— yl)thieno[2,3—b]pyridinamine. Was prepared analogously to SW209329. 2—((4— chlorobutyl)sulfinyl)phenyl(thiophenyl)thieno[2,3-b]pyridinamine. Isolated yield 2 48%. 1H NMR (400 MHz, CHC13) 5 7.64 (ddd, J = 7.2, 3.8, 1.7 Hz, 2H), 7.58 — 7.52 (m, 4H), 7.48 — 7.42 (m, 2H), 7.12 (qd, J = 3.7, 1.8 Hz, 1H), 4.57 (s, 2H), 3.50 (td, J = 6.1, 1.6 Hz, 2H), 3.40 — 3.29 (m, 4H), 3.26 — 3.13 (m, 1H), 2.09 — 1.95 (m, 2H). ESI—MS (m/z): 429.1 [M+H]+.
. N; Se. 3-((((3-cyanophenyl(thiopheny1)pyridiny1)thio)methyl)thio)propyl methanesulfonate. Prepared analogously to 4-((((3-cyanopheny1—6-(thiophen—2—yl)pyridin- hio)methyl)thio)butyl methanesulfonate in quantitative yield. 1H NMR (400 MHZ, CHC13) 5 7.69 (t, J = 3.3 Hz, 1H), 7.63 — 7.55 (m, 2H), 7.52 (p, J = 3.6, 3.0 Hz, 4H), 7.41 (q, J = 2.6, 2.2 Hz, 1H), 7.14 (p, J = 3.4, 2.5 Hz, 1H), 4.52 (q, J = 2.2 Hz, 2H), 4.40 — 4.22 (m, 2H), 2.99 (s, 3H), 2.86 (td, J = 7.2, 4.8 Hz, 2H), 2.10 (qt, J = 6.4, 2.3 Hz, 2H). ESI-MS (m/z): 477.0 [M+H]+.
I / N S | \/s CN \\\\ 2-((((3—chloropropyl)thio)methyl)thio)phenyl(thiophen-2— yl)nicotinonitrile. Prepared ously to 2-((((4-chlorobutyl)thio)methyl)thio)—4—phenyl—6— (thiophenyl)nicotinonitrile. Purification was performed using an ted flash chromatography system in 0-50% EtOAc/hexanes with an isolated yield of 69%. 1H NMR (400 MHz, CHC13) 5 7.72 (dd, J = 3.7, 1.1 Hz, 1H), 7.64 — 7.59 (m, 2H), 7.57 — 7.53 (m, 4H), W0 2018f017582 —140— 7.44 (s, 1H), 7.17 (dd, J = 5.0, 3.8 Hz, 1H), 4.56 (s, 2H), 3.68 (t, J = 6.3 Hz, 2H), 2.93 (t, J = 7.0 Hz, 2H), 2.15 (p, J = 6.7 Hz, 2H). ESI-MS (m/z): 417.0 [M+H]+.
/ / N O | \/§’ / CN \ 2-((((3-chloropropyl)sulfinyl)methyl)thio)phenyl(thiophen-2— yl)nicotinonitri1e. Prepared analogously to 2-((((4-chlorobutyl)sulfinyl)methyl)thio) phenyl(thiophenyl)nicotinonitrile in 90% yield. 1H NMR (400 MHZ, CHC13) 8 7.76 (dd, J = 3.8, 1.1 Hz, 1H), 7.62 (dq, J = 7.1, 2.6, 2.2 Hz, 2H), 7.59 — 7.53 (m, 4H), 7.51 (s, 1H), 7.18 (dd, J = 5.0, 3.8 Hz, 1H), 4.74 (d, J = 13.1 Hz, 1H), 4.51 (d, J = 13.1 Hz, 1H), 3.79 — 3.63 (m, 2H), 3.28 — 3.16 (m, 1H), 3.04 — 2.88 (m, 1H), 2.43 — 2.31 (m, 2H). ESI-MS (m/z): 433.0 [M+H]+.
S _ i / N\ S P | / § SW209330. 2-((3-chloropropyl)sulfinyl)phenyl(thiophen—2—y1)thieno[2,3- din—3—amine.Prepared analogously to SW209329. 2-((4-chlorobutyl)sulfiny1)—4- phenyl(thiophenyl)thieno[2,3-b]pyridinamine. Purification on an automated chromatography system in 0-60% EtOAc/hexanes gave the desired in 88% yield. 1H NMR (400 MHz, CHC13) 5 7.57 (h, J = 5.7, 5.3 Hz, 1H), 7.45 (t, J = 6.0 Hz, 0H), 7.40 (s, 0H), 7.09 (t, J = 4.4 Hz, 0H), 4.61 (s, 0H), 3.67 (td, J = 6.4, 3.2 Hz, 0H), 3.40 (dt, J = 14.1, 7.3 Hz, 0H), 3.24 (dt, J = 13.1, 7.6 Hz, 0H), 2.25 (p, J = 7.0 Hz, 0H).
ESI-MS (m/z): 433.0 [M+H]+. ’/ “\s | \,s 2-((((3-fluoropropyl)thio)methyl)thio)pheny1(thiophenyl)nicotinonitrile.
Kryptofix 222 (44.4 mg, 0.012 mmol), KF (6.1 mg, 0.10 mmol) and K2CO3 (3.0 mg, 0.022 W0 2018/‘017582 —141— mmol) were charged to a vial containing 3-((((3-cyanophenyl(thiophenyl)pyridin yl)thio)methyl)thio)propyl methanesulfonate (54.2 mg, 0.11 mmol). DMF (1.1 ml) was added and the reaction was heated at 85°C for 65 minutes. The cooled mixture was diluted with EtOAc and washed several times with water and then brine. The organic layer was dried over Na2S04, ed and condensed. Yield = 96%. Crude t was carried forward. 1H NMR (400 MHz, CHC13) 5 7.72 (dd, J = 3.7, 1.1 Hz, 1H), 7.64 — 7.59 (m, 2H), 7.54 (dd, J = 4.9, 2.2 Hz, 4H), 7.43 (s, 1H), 7.17 (dd, J = 5.1, 3.7 Hz, 1H), 4.63 (t, J = 5.7 Hz, 1H), 4.55 (s, 2H), 4.51 (t, J = 5.8 Hz, 1H), 3.67 (t, J = 6.3 Hz, 2H), 2.91 (dt, J = 11.2, 7.1 Hz, 1H), 2.14 (p, J = 6.7 Hz, 1H). ESI—MS (m/z): 401.1 . :8 N 6 /I\ S, /CN\\\\\/§ 2—((((3—fluoropropy1)sulfiny1)methyl)thio)phenyl(thiophen—2— yl)nicotinonitrile. Acetic acid (215 111) and hydrogen peroxide (16.75 111, 30 % solution in water) were added to the solution of 2-((((3-fluoropropyl)thio)methyl)thio)phenyl hen—2—yl)nicotinonitrile (43.3 mg, 0.034 mmol) in chloroform (215 pl). The reaction mixture was stirred at 32°C for 50 min and then diluted with chloroform and was washed with saturated NaHC03 solution and extracted three times with chloroform. The combined organic layers was dried over NaZSO4, filtered and concentrated under reduce pressure in 94 % yield. ESI-MS (m/z): 417.1 [M+H]+.
S , l / N\ S ,0 | § SW20933 1. 2-((3-fluoropropyl)sulfinyl)phenyl(thiophen-2—yl)thieno[2,3- b]pyridinamine. Prepared analogously to SW209329. 2-((4-chlorobutyl)sulfinyl)—4- phenyl-6—(thiophen—2—yl)thieno[2,3-b]pyridinamine. The crude mixture was purified atively in 4% CM. Isolated yield = 32 %. 1H NMR (400 MHz, CHC13) 8 7.68 (dd, J = 3.8, 1.1 Hz, 1H), 7.56 (q, J = 2.7 Hz, 4H), 7.53 — 7.44 (m, 3H), 7.14 (dd, J = 5.1, 3.7 Hz, 1H), 4.65 (td, J = 5.8, 3.2 Hz, 1H), 4.60 (s, 2H), 4.53 (td, J = 5.8, 3.1 Hz, 1H), 3.40 (dt, —142— J = 13.0, 7.3 Hz, 1H), 3.24 (dt, J = 13.1, 7.6 Hz, 1H), 2.19 (dtt, J = 26.4, 7.5, 5.7 Hz, 2H).
ESI-MS (m/z): 417.1 .
I/|N\S\/s 2—((((3-cyanopropyl)thio)methyl)thio)phenyl(thiophenyl)nicotinonitrile.
A on of 3—((((3-cyanophenyl(thiophenyl)pyridinyl)thio)methy1)thio)propyl methanesulfonate (54.6 mg, 0.11 mmol) and KCN (76.9 mg, 1.18 mmol) in DMF (1.14 ml) was heated at 85 0C for 4 hours. The cooled mixture was diluted with EtOAc and washed several times with water and then brine. The organic phase was dried over NaZSO4, filtered and condensed. Yield = 89 %. 1H NMR (400 MHz, CHC13) 5 7.71 (d, J = 3.5 Hz, 1H), 7.60 (dt, J = 6.4, 2.0 Hz, 3H), 7.54 (qt, J = 5.6, 2.5 Hz, 4H), 7.44 (d, J = 1.4 Hz, 1H), 7.16 (ddd, J: .2, 3.8, 1.5 Hz, 1H), 4.53 (d, J: 1.7 Hz, 2H), 2.93 — 2.82 (m, 2H), 2.52 (td, J = 7.1, 1.4 Hz, 2H), 2.09 — 1.92 (m, 2H). ESI-MS (m/z): 408.1 .
I N 0- I‘ S»? 2—((((3-cyanopropyl)sulfinyl)methyl)thio)phenyl(thiophen-2— yl)nicotinonitri1e. Acetic acid (205 pl) and hydrogen peroxide (15.6 pl, 30 % on in water) were added to the solution of 2-((((3-cyanopropyl)thio)methyl)thio)phenyl (thiophen-2—y1)nicotinonitrile (41.1 mg, 0.10 mmol) in chloroform (205 111). The reaction mixture was stirred at 32 0C for 70 min and then diluted with chloroform and was washed with saturated NaHC03 solution and extracted three times with chloroform. The combined organic layers was dried over NazSO4, filtered and concentrated under reduce pressure in 91 % yield. ESI—MS (m/z): 424.1 [M+H]+.
W0 2018f017582 —143— SW209332. 4-((3-aminophenyl(thiophenyl)thieno[2,3—b]pyridin finyl)butanenitrile. Prepared analogously to SW209329. chlorobuty1)sulfinyl) phenyl(thiophenyl)thieno[2,3-b]pyridinamine. The crude mixture was purified preparatively in 4% MeOH/DCM. Isolated yield = 45 %. 1H NMR (400 MHZ, CHC13) 8 7.65 (d, J = 3.7 Hz, 1H), 7.60 — 7.51 (m, 4H), 7.51 — 7.44 (m, 3H), 7.13 (t, J = 4.4 Hz, 1H), 4.64 (s, 2H), 3.41 (dt, J = 14.0, 7.3 Hz, 1H), 3.19 (dt, J = 13.3, 7.5 Hz, 1H), 2.59 (t, J: 7.1 Hz, 2H), 2.20 (p, J = 7.3 Hz, 2H). ESI—MS (m/z): 424.0 [M+H]+.
Example 2 Fig. 1 shows pharmacokinetics of the lS-PGDH inhibitor (+) SW033291 when administered at 10mg/kg by intraperitoneal injection into female CD-1 mice and then measured at mg/ml in plasma or at mg/gm of wet tissue weight in brain. As shown, (+) SW033291 appears to concentrate in the brain, which shows a 26-fold higher total drug re (as measured by area under the curve).
Fig. 2 shows measurement of prostaglandin E2 (PGE2) in 3 regions of the brain, as averaged from 3 mice, samples 3 hours after intraperitoneal ion with vehicle (VE) or with (+) SW033291 at 2.5mg/kg. Brain s sampled are #1, the cerebrum, # 2, the cerebellum, and # 3, medulla/pons. Basal PGE2 is highest in the um and medulla/pons, and lowest in the cerebellum. Brain PGE2 levels roughly double in all 3 regions of the brain at 3 hours after injection of (+) SW033291.
] Fig. 3 shows impact of administering (+) SW033291 on mouse performance in learning and memory following traumatic brain injury. In this study, mice were on day 0 subjected to traumatic brain injury from exposure to an adjacent blast injury in an overpressure chamber. 24 hours later, on study day 1, mice treatment with (+) SW033291 at mg/kg was initiated by daily intraperitoneal injection. A parallel cohort of control mice were initiated on injection with vehicle only. On study day 7, mice commenced 4 days of daily training to learn the location of a cup on a table with 20 holes equally spaced around the ter, in the standard Barnes maze task. Performance on training days 1—4 (study days W0 2018/‘017582 —144— 7- 10) is graphed on slide 3, Panel A, which on each day shows the average of 4 trials of the time taken for mice to locate the cup. Cohorts that are compared are sham-injured mice, mice subjected to blast injury and treated with vehicle, mice ted to blast injury and treated with the neuroprotective agent P7C3-A20, and mice subjected to blast injury and treated with (+) SW033291. Quicker time in finding the cup on day 4 is reflective of learning. Mice exposed to blast injury receiving vehicle injections showed the least learning. Mice d to blast injury and receiving ions with nd 20 or with (+) SW033291 appear similar to control mice that received only sham injury.
On study day 11, mice were returned to the Barnes maze with the escape cup removed, and memory was assessed by measuring the time mice spent within 5 cm of the cup’s prior location. As shown in Slide 3, Panel B, mice exposed to blast injury receiving e injections showed the least memory for the cup’s location. Mice exposed to blast injury and receiving injections with compound P7C3-A20 or (+) SW033291 behaved similarly to control mice that ed only sham injury, with respect to having improved memory for the cup’s location versus blast-injured mice receiving only e control.
On study day 12-14, mice were further trained to traverse a 1/2 inch cylindrical rod to reach and enter a black box. On day 14, the mice performance was videotaped and counts were made of the number of times a mouse foot d from the beam. Results are displayed graphically in slide 3, Panel C. In this assay, the worst performance is recorded for mice exposed to blast-injury receiving vehicle injections. Mice exposed to blast—inj ury and ing injections with compound P7C3-A20 behaved similarly to control mice that received only sham injury. Mice exposed to blast injury and receiving ions with (+) SW033291 showed intermediate performance between mice exposed to blast—inj ury and receiving vehicle control and mice exposed to blast-inj ury and receiving P7C3—A20.
Fig. 4 shows in situ hybridization detection of 15-PGDH mRNA sion in the neurons of the mouse hippocampus, a region of the brain involved in learning and memory, and that is an early site of damage in Alzheimer’s disease.
Fig. 5 shows pharmacokinetics of the 15-PGDH inhibitor (+) SW0209415 when administered at 2.5 and at 25 mg/kg by intraperitoneal injection into female C57BL/7 mice and then measured at mg/ml in plasma or at mg/gm of wet tissue weight in brain. As shown, at 25 mg/kg dose (+) SW209415 appears to concentrate in the brain, which shows a 1.56-fold higher total drug exposure (as measured by area under the curve).
W0 2018f017582 —145— Figs. 6(A-C) illustrate graphs showing H activity in the cortex (A), cerebellum (B), and pons and medulla (C) of mouse brain following IP injection of 15-PGDH inhibitor (+) SW033291 at 2.5 mpk. 15-PGDH activity was measured from the 3 regions of mouse brain using a 15-tritiated PGE2 substrate. A coupled enzymatic assay uses 15—PGDH and glutamate ogenase to transfer m from PGE2 onto glutamate. Brain tissues were harvested for assay at the times shown following IP injection of 15-PGDH inhibitor (+)- SW033291 at 2.5 mpk. The results show that 15-PGDH enzyme activity can be readily inhibited in the brain following IP injection of a 15-PGDH inhibitor.
Fig. 7 illustrates a graph showing PGE2 levels in rat brain cortex following IP injection of (+) 91 at 2.5, 5.0, and 10.0 mg/kg. PGE2 levels are elevated in rat brain cortex 30, 120, and 180 minutes following a single IP injection of 033291 at the noted doses. PGE2 levels were found to double at 180 minutes following dosing at 5 mpk, and double at 120 minutes ing dosing at 10 mpk.
Figs. 8(A—B) illustrate Western blots and graphs showing levels of 15—PGDH in brain tissue of subjects with Alzheimer’s disease relative to age matched control subjects without Alzheimer’s disease. Western blot with antiPGDH antibody shows markedly elevated levels of 15- PGDH enzyme in brain tissue (occipital and frontal cortex) of patients with Alzheimer’s disease (average age 85), ve to age matched (average age 85) control subjects t Alzheimer’s disease. Densitometry analysis normalized against GAPDH was compared statistically with Student’s t test. *p<0.05, and >k**p<.001. Each lane represents a separate subject.
While this ion has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention assed by the appended . All patents, publications and references cited in the ing specification are herein incorporated by reference in their entirety.
W0 2018/‘017582 The following is d: 1. A method of promoting neuroprotection in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a H inhibitor. 2. The method of claim 1, wherein the t has or is at risk of axonal degeneration, neuronal cell death, and/or glia cell damage after injury. 3. The method of claim 1, wherein the 15-PGDH inhibitor can be administered at an amount effective to stimulate hippocampal neurogenesis, for the treatment of neuropsychiatric and neurodegenerative diseases. 4. The method of claim 3, wherein the neuropsychiatric and neurodegenerative es are schizophrenia, major depression, bipolar disorder, normal aging, epilepsy, traumatic brain , post-traumatic stress disorder, Parkinson‘s disease, Alzheimer's disease, Down syndrome, spinocerebellar ataxia, amyotrophic l sclerosis, Huntington's disease, stroke, radiation therapy, chronic stress, and abuse of active drugs.
. A method of treating a disease, disorder, and/or condition of the nervous system in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a H inhibitor. 6. The method of claim 5, wherein the disease, disorder, and/or condition of the nervous system includes at least one of a neurological disorder, neuropsychiatric disorder, neural injury, neural toxicity er, a neuropathic pain, and neural degenerative disorders. 7. The method of claim 6, wherein the neurological disorder includes at least one of traumatic or toxic injuries to peripheral or l nerves, spinal cord or to the brain, cranial nerves, traumatic brain injury, stroke, al aneurism, and spinal cord injury.
W0 2018/‘017582 —147_ 8. The method of claim 6, wherein the neurological disorder includes at least one of Alzheimer's disease, dementias related to Alzheimer‘s e, Parkinson‘s, Lewy diffuse body diseases, senile dementia, Huntington's disease, Gilles de Ia Tourette‘s syndrome, multiple sclerosis, amyotrophic lateral sis, hereditary motor and sensory neuropathy, diabetic neuropathy, progressive supranuclear palsy, epilepsy, or Jakob-Creutzfieldt disease. 9. The method of claim 6, wherein the neural injury can be caused by or associated with at least one of epilepsy, cerebrovascular diseases, autoimmune diseases, sleep disorders, autonomic ers, urinary bladder disorders, abnormal metabolic states, ers of the muscular system, infectious and parasitic diseases neoplasms, endocrine es, nutritional and metabolic diseases, immunological diseases, diseases of the blood and blood-forming organs, mental disorders, diseases of the nervous system, diseases of the sense organs, diseases of the circulatory system, diseases of the respiratory system, diseases of the ive system, diseases of the genitourinary system, diseases of the skin and subcutaneous tissue, diseases of the musculoskeletal system and connective tissue, congenital anomalies, or conditions originating in the perinatal period.
. A method of augmenting neuronal ing underlying learning and memory in a subject in need thereof, the method comprising: administering to the subject a eutically effective amount of a H 11. A method of stimulating neuronal regeneration after injury in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a H 12. A method of treating Alzheimer’s disease in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a 15-PGDH inhibitor.
W0 2018/‘017582 13. A method of ng traumatic brain injury in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a 15—PGDH inhibitor. 14. The method of any of claims 1 to 13, wherein the 15-PGDH tor has the following formula (I): (11)“ 1 f I” \F“ Y—X\\ /u Y2 (I) wherein n is 0-2; Y1, Y2, and R1 are the same or different and are each selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3—C20 aryl, heteroaryl, heterocycloalkenyl containing from 5—6 ring atoms, C6-C24 alkaryl, C6—C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1—C24 alkoxy, C2-C24 alkenyloxy, C2—C24 loxy, C5-C20 aryloxy, acyl, acyloxy, C2-C24 alkoxycarbonyl, C6-C20 aryloxycarbonyl, C2-C24 alkylcarbonato, C6-C20 arylcarbonato, carboxy, ylato, carbamoyl, C1—C24 alkyl-carbamoyl, arylcarbamoyl, thiocarbamoyl, carbamido, cyano, isocyano, cyanato, isocyanato, ocyanato, azido, formyl, thioformyl, amino, C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido, C6-C20 arylamido, imino, alkylimino, arylimino, nitro, nitroso, sulfo, sulfonato, C1-C24 alkylsulfanyl, arylsulfanyl, C1-C24 alkylsulfinyl, C5-C20 arylsulfinyl, C1-C24 alkylsulfonyl, C5-C20 arylsulfonyl, sulfonamide, phosphono, phosphonato, inato, phospho, phosphino, polyalkylethers, phosphates, and phosphate esters, groups incoporating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations f, and n Y1 and Y2 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted aryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; W0 2018/‘017582 2017/042620 —149— U1 is N, C-Rz, or 4, wherein R2 is ed from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein n1=1, 2, or 3), CF3, CH2-CH2X, O-CHz- CHZX, CH2-CH2-CH2X, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or I), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein n1=1, 2, or 3), CF3, CHz-CHZX, CHz-CHz-CHZX, in X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; X1 and X2 are independently N or C, and wherein when X1 and/or X2 are N, Y1 and/or Y2, respectively, are absent; 21 is O, S, CRaRb or NRa, wherein Ra and Rb are independently H or a CH; alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically acceptable salts thereof.
. The method of any of claims 1 to 13, n the 15-PGDH inhibitor has the following formula (II): (ol) n 21 If \R1 126+! \ \X6¥x7 R7 (11) wherein n is 0-2 X4, X5 , X6, and X7 are independently N or CR“; R1, R6, R7, and RC are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 l, C2-C24 alkynyl, C3-C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms, C6-C24 alkaryl, C6—C24 aralkyl, halo, —Si(C1—C3 3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2— C24 alkynyloxy, C5-C20 aryloxy, acyl, acyloxy, C2-C24 alkoxycarbonyl, C6-C20 aryloxycarbonyl, C2-C24 alkylcarbonato, C6-C20 arylcarbonato, carboxy, carboxylato, carbamoyl, C1—C24 alkyl-carbamoyl, arylcarbamoyl, thiocarbamoyl, carbamido, cyano, W0 2018/‘017582 isocyano, cyanato, isocyanato, isothiocyanato, azido, formyl, thioformyl, amino, C1-C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido, C6-C20 arylamido, imino, alkylimino, arylimino, nitro, nitroso, sulfo, ato, C1-C24 alkylsulfanyl, arylsulfanyl, C1—C24 alkylsulfinyl, C5—C20 arylsulfinyl, C1-C24 alkylsulfonyl, C5-C20 arylsulfonyl, sulfonamide, phosphono, phosphonato, phosphinato, phospho, phosphino, polyalkylethers, phosphates, and phosphate esters, groups rating amino acids or other moieties ed to bear positive or negative charge at physiological pH, and combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted lkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C—RZ, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CH2-CH2X, O-CHz- CH2X, CH2-CH2—CH2X, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ (wherein n1=l, 2, or 3), CF3, ZX, CHz-CHz-CHZX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ in R’ is H or a lower alkyl group), and R3 or R4 may be ; Z1 is O, S, CRZ‘Rb or NRa, wherein Ra and Rb are independently H or a C1_g alkyl, which is linear, branched, or cyclic, and which is unsubstituted or substituted; and pharmaceutically acceptable salts thereof. 16. The method of any of claim 1 to 13, wherein the the lS-PGDH inhibitor has the ing formula (III) or (IV): 21 MS\1 N \ U‘ R7 (111), or W0 017582 2017/042620 U1§‘¢r~“s\R1 N \ Z1 KXA’l R7 (IV) wherein n is 0-2 X6 is independently is N or CR“; R1, R6, R7, and RC are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2-C24 alkynyl, C3-C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms, C6-C24 l, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, yl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2- C24 loxy, C5—C20 aryloxy, acyl, acyloxy, C2-C24 alkoxycarbonyl, C6—C20 aryloxycarbonyl, C2—C24 alkylcarbonato, C6-C20 arylcarbonato, carboxy, carboxylato, carbamoyl, C1-C24 alkyl-carbamoyl, arylcarbamoyl, thiocarbamoyl, carbamido, cyano, isocyano, cyanato, isocyanato, isothiocyanato, azido, formyl, thioformyl, amino, C1-C24 alkyl amino, C5—C20 aryl amino, C2-C24 alkylamido, C6-C20 arylamido, imino, alkylimino, arylimino, nitro, nitroso, sulfo, sulfonato, C1-C24 alkylsulfanyl, arylsulfanyl, C1—C24 alkylsulfinyl, C5—C20 arylsulfinyl, C1-C24 alkylsulfonyl, C5-C20 arylsulfonyl, amide, phosphono, phosphonato, phosphinato, phospho, phosphino, polyalkylethers, phosphates, and phosphate esters, groups incoporating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl, a substituted or unsubstituted lkyl, and a substituted or tituted cyclyl; U1 is N, C—Rz, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein nl=l, 2, or 3), CF3, CH2-CH2X, O-CHz- CH2X, CH2—CH2—CH2X, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or I), CN, (C=O)—R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)HIOR’ (wherein nl=l, 2, or 3), CF3, ZX, CHz-CHz-CHZX, (wherein X=H, F, Cl, W0 2018/‘017582 Br, or 1), CN, R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; Z1 is O, S, CRaRb or NRa, n R31 and Rb are independently H or a C1_8 alkyl, which is linear, branched, or , and which is unsubstituted or tuted; and pharmaceutically acceptable salts thereof. 17. The method of any of claims 1 to 13, wherein the the lS-PGDH inhibitor has the following formula (V): 8 ws\1 \” R 7 \ U‘ R7 (V) wherein n is 0-2 X6 is independently is N or CRC R1, R6, R7, and Rc are each independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2—C24 alkynyl, C3-C20 aryl, heteroaryl, heterocycloalkenyl containing from 5-6 ring atoms, C6-C24 alkaryl, C6-C24 aralkyl, halo, —Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 alkynyloxy, C5-C20 aryloxy, acyl, acyloxy, C2-C24 alkoxycarbonyl, C6-C20 aryloxycarbonyl, C2-C24 arbonato, C6-C20 arylcarbonato, carboxy, carboxylato, carbamoyl, C1-C24 alkyl-carbamoyl, arylcarbamoyl, thiocarbamoyl, carbamido, cyano, isocyano, cyanato, isocyanato, isothiocyanato, azido, formyl, thioformyl, amino, C1-C24 alkyl amino, C5—C20 aryl amino, C2-C24 alkylamido, C6-C20 arylamido, imino, alkylimino, arylimino, nitro, nitroso, sulfo, sulfonato, C1-C24 alkylsulfanyl, arylsulfanyl, C1—C24 alkylsulfinyl, C5—C20 arylsulfinyl, C1-C24 alkylsulfonyl, C5-C20 arylsulfonyl, sulfonamide, ono, phosphonato, phosphinato, phospho, phosphino, polyalkylethers, phosphates, and phosphate esters, groups incoporating amino acids or other moieties ed to bear ve or negative charge at logical pH, and combinations thereof, and wherein R6 and R7 may be linked to form a cyclic or polycyclic ring, wherein the ring is a substituted or W0 2018f017582 unsubstituted aryl, a substituted or tituted heteroaryl, a substituted or unsubstituted cycloalkyl, and a substituted or unsubstituted heterocyclyl; U1 is N, C-Rz, or C—NR3R4, wherein R2 is selected from the group consisting of a H, a lower alkyl group, O, (CH2)n10R’ (wherein n1=1, 2, or 3), CF3, CHz—CHZX, O-CHz- CH2X, CHz—CHz—CHZX, O-CHz-CHZX, X, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, O(CO)R’, COOR’ (wherein R’ is H or a lower alkyl group), and wherein R1 and R2 may be linked to form a cyclic or polycyclic ring, wherein R3 and R4 are the same or different and are each selected from the group consisting of H, a lower alkyl group, O, (CH2)n10R’ in n1=1, 2, or 3), CF3, CHz-CHZX, CHz-CHz-CHZX, (wherein X=H, F, Cl, Br, or 1), CN, (C=O)-R’, (C=O)N(R’)2, COOR’ (wherein R’ is H or a lower alkyl group), and R3 or R4 may be absent; and pharmaceutically acceptable salts thereof. 18. The method of any of claims 1 to 13, wherein the the 15-PGDH tor has the following formula (VI): R6 N Y\ S (8/0 >n X6 / / \R1 R7 (VI) wherein n = 0-2; X6 is N or CR“; R1 is selected from the group consisting of branched or linear alkyl including — (CH2)n1CH3 7), n2 wherein n2=0-6 and X is any of the ing: CFyHZ (y + z = 3), CClyHZ (y + z = 3), OH, OAc, OMe, R71, OR72, CN, N(R73)2, ”3 (n3=0—5, m=l-5), and -5).
R5 is selected from the group consisting of H, C1, F, NH2, and N(R76)2; R6 and R7 can each independently be one of the following: —154— 91“” Jw\1" ||/0 O ||/ >7 O 0 NR19 NR2‘ §N'/,Ri'/R15 E‘I/ R14_ II/ E R18'—/ _ R1_ 17— II/ ”\N ”R” ”i” 24 28 R30 NR26 R27fNR o N/ O R22 23—/NRIll|\N/ [LI I / R WE— “\N/>'§ / N|\N/>7§— R29 NW \ \ N/O N/NR32 N/NR34 N/NRSSR N/NR38 N/NR40 31 || 33”— _R35 ..
RI iL/>§ RI /3 TA T/i' /§_ / _ N , .N‘\N NV)??— R39 NR42 /NR43 45'“ R46 T|// NR47 N I' / IRS W)é—WL\ 99aI " " N\N " /,§f\ / \ ”\w JV" M " \ \ N /\ N Rsofi \ \ '15sz \ N R54II/\NR55IC \ | R551: /;\KN)9\\ /R53l11:_ (5”; \N / \KN/ 5‘: N%N \ I rill {N\ JQRGO R56|_ R51} R53“— 3—R59 / / 011° / /H/PRGZ \ /5‘5$\ 55‘: 7 R63 :0483: 9“ :02 43'" :08 “WI0z ”W: g I Zm/,.\:0 \ Z\\ o 0 each R8 R9 R10 R11 R12 R13 R14 R15 R16 R17 R18 R19 R20 R21 R22 R23 R24 R25 R26 R27 R28 R29 R30 R31 R32 R33 R34 R35 R36 R37 R38 R39 R40 R41 R42 R43 R44 R45 R46 R47 R48,R49,R50, R51, R52,R53,R54, R55,R56, R57, R583R593R60a R615R62, R63, R64,R65,R66, 1167,1168, R69” R70‘ R71‘ R? R73~ R74, R76, and R0 are the same or different and are independently selected from the group consisting of hydrogen, substituted or unsubstituted C1-C24 alkyl, C2-C24 alkenyl, C2—C24 alkynyl, C3—C20 aryl, heteroaryl, heterocycloalkenyl containing from 5—6 ring atoms, W0 017582 C6-C24 alkaryl, C6-C24 aralkyl, halo, -Si(C1-C3 alkyl)3, hydroxyl, sulfhydryl, C1-C24 alkoxy, C2-C24 alkenyloxy, C2-C24 loxy, C5-C20 aryloxy, acyl, acyloxy, C2-C24 alkoxycarbonyl, C6—C20 aryloxycarbonyl, C2-C24 arbonato, C6-C20 rbonato, carboxy, carboxylato, carbamoyl, C1—C24 alkyl-carbamoyl, arylcarbamoyl, thiocarbamoyl, carbamido, cyano, isocyano, cyanato, isocyanato, isothiocyanato, azido, formyl, thioformyl, amino, C1—C24 alkyl amino, C5-C20 aryl amino, C2-C24 alkylamido, C6-C20 arylamido, imino, alkylimino, arylimino, nitro, nitroso, sulfo, sulfonato, C1-C24 alkylsulfanyl, arylsulfanyl, C1-C24 alkylsulfinyl, C5—C20 arylsulfinyl, C1-C24 alkylsulfonyl, C5-C20 arylsulfonyl, sulfonamide, phosphono, phosphonato, inato, phospho, phosphino, polyalkylethers, phosphates, and phosphate esters, groups rating amino acids or other moieties expected to bear positive or negative charge at physiological pH, and combinations thereof, and pharmaceutically acceptable salts thereof. 19. The method of any of claims 1 to 13, wherein the 15-PGDH inhibitor has the following formula following formula: (IX), (X), or pharmaceutically acceptable salts thereof.
. The method of any of claims 1 to 19, wherein the 15-PGDH inhibitor i) at 2.5uM concentration, stimulates a Vac0503 reporter cell line expressing a H luciferase fusion construct to a luciferase output level of greater than 70 (using a scale on which a value of 100 indicates a doubling of reporter output over baseline); ii) at 2.5uM concentration ates a V9m er cell line expressing a 15-PGDH luciferase fusion uct to a luciferase output level of greater than 75; iii) at 7.5uM concentration stimulates a LS 174T reporter cell line expressing a 15-PGDH rase fusion construct to a luciferase output level of greater than 70; iv) 7.5uM concentration, does not activate a negative control W0 2018/‘017582 2017/042620 V9m cell line expressing TK-renilla rase reporter to a level greater than 20; and V) inhibits the enzymatic activity of recombinant 15-PGDH protein at an IC50 of less than luM. 21. The method of any of claims 1 to 19, wherein the l5-PGDH inhibitor i) at 2.5 uM tration stimulates a Vaco503 reporter cell line expressing a 15—PGDH luciferase fusion construct to increase luciferase output; ii) at 2.5 uM concentration stimulates a V9m reporter cell line expressing a l5-PGDH luciferase fusion construct to se luciferase output; iii) at 7.5 uM concentration stimulates a LSl74T reporter cell line expressing a l5-PGDH luciferase fusion construct to increase luciferase output; iv) at 7.5 uM concentration, does not activate a negative control V9m cell line expressing TK—renilla luciferase reporter to a luciferase level greater than 20% above background; and v) inhibits the enzymatic ty of recombinant l5-PGDH protein at an IC50 of less than 1 nM. 22. The method of any of claims 1 to 19, wherein the 15-PGDH inhibitor inhibits the enzymatic activity of recombinant l5-PGDH at an IC50 of less than luM, or preferably at an IC50 of less than 250 nM, or more preferably at an IC50 of less than 50 nM, or more preferably at an IC50 of less than 10 nM, or more preferably at an IC50 of less than 5 nM at a inant 15—PGDH concentration of about 5 nM to about 10 nM.
WO 17582 (+) SW033291 PK (10 mg/kg IP, Female CD-1 Mice) (ng/ml Concentration 100 me p|asm a M§\\\w b ra i n 100 150 200 250 Time (minutes) erminal T‘/z (min): Cmax (ng/ml org): 1133 max (min): 10 A UClast (min*ng/m| or g): 112,139 CL/F (ml or g/min): 2.00 z/F (ml or g): 190 BBB: 2.61 Fig. 1 SW033291 induces PGE-2 in mouse brain 3 hr after single dose of 2.5 mpk SW033291(+), N=3 mice : 10.011 \ \ 121 211.1111 VVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVV\ ~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~\ ~~~~~~~~ t 15.11:: § :3 .. .1 VVVVVVVV \\vvvvvvvvvvvvvvvv\ (1V \ \ N inji.’ \ V................\ :‘E.. § 1.131:- § 1 11-31“.- 77777777 rrrrrrrrrrrrrrrrrrrrrrrrrrrrrrrrrrrr ~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~~1vvvvvvvvvvvvvv .339. J 311 Q? @591 a. “$2 $551 *3 Q31 ‘1 9‘ £31 .61 m: qu'" «$11. «335‘ 111* 11“ Fig. 2 Barnes Maze-Probe Trial Barnes Maze-Training ** A Sham-vehicle d.) \\\\\\\\ Blast-vehicle U) area a.) we" Blast-A20 (U ~~~~~ SW0033921 U) target a.) in H %Time 2 3 Trial (day) Balance Test Daily intraperitoneal administration of 10 mg/kg.d SW033291, an Inhibitor of H, initiated 24 hours after blast-induced traumatic brain injury (TBI) and continued for 11 days, preserved normal learning (A) and memory (B) in the Barnes maze to levels indistinguishable from njured animals that received vehicle.
Memory was ed by the most stringent parameter in this Slip assay: percent oftime spent in the target area as defined by a 5 cm radius around the escape hole. The protective effect seen was Foot similar in magnitude to that noticed with the previously established therapeutic neuroprotective agent P7C3-A20, given at the same dose. In the balance test, a trend towards protection was noted in animals treated with SW033291, but did not achieve statistic significance as was seen in P7C3-A20-treated animals.
Every group consisted of25 male C57/Bl6 mice, aged 12-14 weeks, and data were ted and scored in an ted manner blind to treatment group. Data are represented as mean +/- SEM.
Significance was determined by two-way ANOVA with roni post hoc analysis. * , * * p<0.01, ** *p<0. 001 compared to blast-injured animals treated with vehicle.
Figs. 3A-C WO 17582 Fig. 4 SW209415 (+) 6 Mouse PK- BBB Calculation SW209415 (+) C57B6 mouse PK-ip-plasma 2.5mg[kg 25mg[kg TerminalT‘z: NA 46.51 min f Cmax: 198 2840 ng/ml Tmax: 10 10 min R1 N E AUCIast: 4536 91705 min*ng/ml § VZ: 322 ml | er‘ y, NA / 8/ CL: NA 4.8 ml/min R3 E 5 1,5, 1000 f5 \_~l ~~~..\\~~~sw209415(+)ip-2.5mg/kg U i g 500 «\\\\\\‘SW209415(+)ip—25mg/kg ‘ \\\ \\x 0 4%»»it»»}L\&\\.\.\.\.\\.xxK\\\\\\\\QN\\\\\\\\\\\\\\\\\\_\\\\\\\\\\§\\\\\\.\\\\\\\xxxxxxxxxxxxxxx\\\\\\\\\\\\\\\\\y\\\\\\\\\\\\\\\\\\\}\\§"\\‘»»»»»»»»»»»»»\55 (:3 50 100 150 200 250 300 350 400 Time(min) SW209415 (+) C5736 mouse PK-ip-Brain BBB 2.5 mpk: 0.41 2.5mg/kg 25mg/kg TerminalT‘z: NA NA min BBB 25 mpk: 1.56 3000 ‘3 Cmax: 90 6021 ng/g Tmax: 10 10 7000 min 3 I AUCIast: 1852 142614 /g The SW209415(+) average in brain is 6000 VZ: NA NA ml a \\ calculated by first subtracting the amount of 50 1555 CL: NA NA ml/min = i compound in the al blood/plasma V S 5 ; x \ within that tissue. The reference forthis E 4000 5 5 B 5 volume of residual blood for brain is shown z 3000 >~:§ 5 below. Assumes equal partitioning of 35 ‘x nd between plasma and RBCs. g 2000 § -~$\~w2.5mg/kg \\\x\\w25mg/kg U \l‘ 1000 5 ‘\ nce: § 5 . \\\\\\\\ Kwon, Y. (2001). The Handbook of Essential 0 <§§§“‘“R"""."""‘“‘\§§\\\\\\\\\m\\\\\\\\\\\\\\\xxxxx\\\\X\\‘\\\\\\.\\\\\\\\\\\\\\\\\\\x}\\\\\\\\\\\\\\\\\\\}\\\\\“\\“\“\“\“‘“\‘\I\‘...........___: Pharmacokinetics, Pharmacodynamics, and 0 50 100 150 200 250 300 350 400 _1000 Drug Metabolism for Industrial Scientists.
Time(min) Kluwer Academic /Plenum Publishers, 231— blood in brain: 30 ul/gtissue Fig. 5 . 3:. 33.333. 3333333333 33m 3333.333 W.-%kkk§.<\m¢~ $3.33. 3 333 “3 33333333333333. 3.3333w 3.3333“3‘ 33WH\\\: 3333333 3.333\3 33333333 “MW3 3 3 - 3&33333333333 B : ~ \ 3: 33$N§£N§3§233®§ 33333333333333.3333 333333333 333.33.33.33 33.333333333333333 xfifimmfi 333333 Figs. 6A-B v flaw gggfimwtmg §£§3$e§§m§gxgmgwk§$§mxmg§figwfifi mfiwfimfim wmgmmfiw Mugwfimwfia M ”mama ..........
H \\\\ mfimfifimfim mfiwwmw m”.h.h.h.h.h.mmnaaannmmn.§\\\\\\\\\\\\\\\\\\\\\\\\\\ Nwmwa N?3K mm\\\\\\\\\\\\\\\\\\\\\\\\\\\Q Fig. 7 hula a.
Humm (“WWI-II] ;aH.
Fm $.41. fimigflalm: Emfiui AH harm a mama“ [ain gr rm [:15me .2.
Fan QII; Elm! AH Fig. 8A-B
NZ790341A 2016-07-18 2017-07-18 Inhibitors of short-chain dehydrogenase activity for promoting neurogenesis and inhibiting nerve cell death NZ790341A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/363,441 2016-07-18
US62/372,203 2016-08-08

Publications (1)

Publication Number Publication Date
NZ790341A true NZ790341A (en) 2022-07-29

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017300377B2 (en) Inhibitors of short-chain dehydrogenase activity for promoting neurogenesis and inhibiting nerve cell death
AU2021200610B2 (en) Inhibitors of short-chain dehydrogenase activity for treating fibrosis
RU2678305C1 (en) Imidazopyridine derivatives as modulators of tnf activity
JP6457525B2 (en) Pyrazolopyridine derivatives as modulators of TNF activity
JP6484632B2 (en) Tetrahydroimidazopyridine derivatives as modulators of TNF activity
EP3080111B1 (en) Imidazopyridine derivatives as modulators of tnf activity
JP5680146B2 (en) Heterocyclic compounds as CRTH2 receptor antagonists
RU2684644C1 (en) Purine derivatives as tnf activity modulators
JP2018511581A5 (en)
AU2013249434A1 (en) Compositions and methods of modulating 15-PGDH activity
JP6469693B2 (en) Imidazopyridine derivatives as modulators of TNF activity
JP6483700B2 (en) Imidazothiazole Derivatives as Modulators of TNF Activity
RU2697090C1 (en) Tetrahydroimidazopyridine derivatives as modulators of tnf activity
NZ790341A (en) Inhibitors of short-chain dehydrogenase activity for promoting neurogenesis and inhibiting nerve cell death
WO2010059555A1 (en) Prolyl hydroxylase inhibitors