AU2013270464A1 - Pyridyl derivatives as CFTR modulators - Google Patents

Pyridyl derivatives as CFTR modulators Download PDF

Info

Publication number
AU2013270464A1
AU2013270464A1 AU2013270464A AU2013270464A AU2013270464A1 AU 2013270464 A1 AU2013270464 A1 AU 2013270464A1 AU 2013270464 A AU2013270464 A AU 2013270464A AU 2013270464 A AU2013270464 A AU 2013270464A AU 2013270464 A1 AU2013270464 A1 AU 2013270464A1
Authority
AU
Australia
Prior art keywords
compound
pct
disease
heteroaryl
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2013270464A
Other versions
AU2013270464B2 (en
Inventor
Brian Bear
Peter Grootenhuis
Sara Hadida-Ruah
Mark Miller
Jinglan Zhou
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vertex Pharmaceuticals Inc
Original Assignee
Vertex Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2009231993A external-priority patent/AU2009231993B2/en
Application filed by Vertex Pharmaceuticals Inc filed Critical Vertex Pharmaceuticals Inc
Priority to AU2013270464A priority Critical patent/AU2013270464B2/en
Publication of AU2013270464A1 publication Critical patent/AU2013270464A1/en
Application granted granted Critical
Publication of AU2013270464B2 publication Critical patent/AU2013270464B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

The present invention relates to modulators of ATP-Binding Cassette ("ABC") transporters or fragments thereof, including Cystic Fibrosis Transmembrane Conductance Regulator ("CFTR"), compositions thereof, and methods therewith. The present invention also relates to methods of treating ABC transporter mediated diseases using such modulators. -107-

Description

WO 2009/123896 PCT/US2009/038203 PYRIDYL DERI VA TIES AS CFTR MOD ULA TORS TECHNICAL FIELD OF THE INVENTION 10011 The present invention relates to modulators of ATP-Binding Cassette ("ABC") transporters or fragments thereof, including Cystic Fibrosis Transmembrane Conductance Regulator ("CFTR"), compositions thereof, and methods therewith. The present invention also relates to methods of treating ABC transporter mediated diseases using such modulators. BACKGROUND OF THE INVENTION [0021 ABC transporters are a family of membrane transporter proteins that regulate the transport of a wide variety of pharmacological agents, potentially toxic drugs, and xenobiotics, as well as anions. ABC transporters are homologous membrane proteins that bind and use cellular adenosine triphospliate (ATP) for their specific activities. Some of these transporters were discovered as multidrug resistance proteins (like the MDRI -P glycoprotein, or the multidrug resistance protein, MRP1), defending malignant cancer cells against chemotherapeutic agents. To date, 48 ABC Transporters have been identified and grouped into 7 families based on their sequence identity and function. [0031 ABC transporters regulate a variety of important physiological roles within the body and provide defense against harmful environmental compounds. Because of this, they represent important potential drug targets for the treatment of diseases associated with defects in the transporter, prevention of drug transport out of the target cell, and intervention in other diseases in which modulation of ABC transporter activity may be beneficial. [0041 One member of the ABC transporter family commonly associated with disease is the cAMP/ATP-mediated anion channel, CFTR. CFTR is expressed in a variety of cells types, including absorptive and secretory epithelia cells, where it regulates anion flux across the membrane, as well as the activity of other ion channels and proteins. In epithelia cells, normal functioning of CFTR is critical for the maintenance of electrolyte transport throughout the body, including respiratory and digestive tissue. CFTR is composed of approximately 1480 amino acids that encode a protein made up of a tandem repeate of transmembrane domains, each containing six transmembrane helices and a nucleotide binding domain. The two transmembrane domains are WO 2009/123896 PCT/US2009/038203 linked by a large, polar, regulatory (R)-domain with multiple phosphorylation sites that regulate channel activity and cellular trafficking. 10051 The gene encoding CFTR has been identified and sequenced (See Gregory, R. J. et al. (19901) Nature 347:382-386; Rich, D. P. et al. (1990) Nature 347:358-362), (Riordan, J. R. et al. (1989) Science 245:1066-1073). A defect in this gene causes mutations in CFTR resulting in Cystic Fibrosis ("CF"), the most common fatal genetic disease in humans. Cystic Fibrosis affects approximately one in every 2,500 infants in the United States. Within the general United States population, up to 10 million people carry a single copy of the defective gene without apparent ill effects. In contrast, individuals with two copies of the CF associated gene suffer from the debilitating and fatal effects of CF, including chronic lung disease. 10061 In patients with cystic fibrosis, mutations in CFTR endogenously expressed in respiratory epithelia leads to reduced apical anion secretion causing an imbalance in ion and fluid transport. The resulting decrease in anion transport contributes to enhanced mucus accumulation in the lung and the accompanying microbial infections that ultimately cause death in CF patients. In addition to respiratory disease, CF patients typically suffer from gastrointestinal problems and pancreatic insufficiency that, if left untreated, results in death. In addition, the majority of males with cystic fibrosis are infertile and fertility is decreased among females with cystic fibrosis. In contrast to the severe effects of two copies of the CF associated gene, individuals with a single copy of the CF associated gene exhibit increased resistance to cholera and to dehydration resulting from diarrhea - perhaps explaining the relatively high frequency of the CF gene within the population. [0071 Sequence analysis of the CFTR gene of CF chromosomes has revealed a variety of disease causing mutations (Cutting, G. R. et al. (1990) Nature 346:366-369; Dean, M. et al. (1990) Cell 61:863:870; and Kerem, B-S. et al. (1989) Science 245:1073-1080; Kerem, B-S et al. (1990) Proc. Natl. Acad. Sci. USA 87:8447-8451). To date, > 1000 disease causing mutations in the CF gene have been identified (11 tf-,"ww, euctc). The most prevalent mutation is a deletion of phenylalanine at position 508 of the CFTR amino acid sequence, and is commonly referred to as AF508-CFTR. This mutation occurs in approximately 70% of the cases of cystic fibrosis and is associated with a severe disease. [0081 The deletion of residue 508 in AF508-CFTR prevents the nascent protein from folding correctly. This results in the inability of the mutant protein to exit the endoplasmic reticulum ("ER"), and traffic to the plasma membrane. As a result, the number of channels present in the membrane is far less than observed in cells expressing wild-type CFTR. In addition to impaired trafficking, the mutation results in defective channel gating. Together, the reduced 2-~ WO 2009/123896 PCT/US2009/038203 number of channels in the membrane and the defective gating lead to reduced anion transport across epithelia leading to defective ion and fluid transport. (Quinton, P. M. (1990), FASEB J. 4: 2709 2727). Studies have shown, however, that the reduced numbers of AF508-CFTR in the membrane are functional, albeit less than wild-type CFTR. (Dalemans ei al. (1991), Nature Lond. 354: 526 528; Denning et al., supra; Pasyk and Foskett (1995), J. Cell. Biochem. 270: 12347-50). In addition to AF508-CFTR, other disease causing mutations in CFTR that result in defective trafficking, synthesis, and/or channel gating could be up- or down-regulated to alter anion secretion and modify disease progression and/or severity. [0091 Although CFTR transports a variety of molecules in addition to anions, it is clear that this role (the transport of anions) represents one element in an important mechanism of transporting ions and water across the epithelium. The other elements include the epithelial Na channel, ENaC, Na7/2CV/K co-transporter, Na '-K -ATPase pump and the basolateral membrane K- channels, that are responsible for the uptake of chloride into the cell. [00101 These elements work together to achieve directional transport across the epithelium via their selective expression and localization within the cell. Chloride absorption takes place by the coordinated activity of ENaC and CFTR present on the apical membrane and the Na -K'-ATPase pump and Cl- channels expressed on the basolateral surface of the cell. Secondary active transport of chloride from the luminal side leads to the accumulation of intracellular chloride, which can then passively leave the cell via Cf channels, resulting in a vectorial transport. Arrangement of Na7/2C /K7 co-transporter, Na*-K -ATPase pump and the basolateral membrane Ki channels on the basolateral surface and CFTR on the luminal side coordinate the secretion of chloride via CFTR on the luminal side. Because water is probably never actively transported itself, its flow across epithelia depends on tiny transepithelial osmotic gradients generated by the bulk flow of sodium and chloride. [0011] In addition to Cystic Fibrosis, modulation of CFTR activity may be beneficial for other diseases not directly caused by mutations in CFTR, such as secretory diseases and other protein folding diseases mediated by CFTR. These include, but are not limited to, chronic obstructive pulmonary disease (COPD), dry eye disease, and Sjigren's Syndrome. [00121 COPD is characterized by airflow limitation that is progressive and not fully reversible. The airflow limitation is due to mucus hypersecretion, emphysema, and bronchiolitis. Activators of mutant or wild-type CFTR offer a potential treatment of mucus hypersecretion and impaired mucociliary clearance that is common in COPD. Specifically, increasing anion secretion across CFTR may facilitate fluid transport into the airway surface liquid to hydrate the mucus and WO 2009/123896 PCT/US2009/038203 optimized periciliary fluid viscosity. This would lead to enhanced mucociliary clearance and a reduction in the symptoms associated with COPD. Dry eve disease is characterized by a decrease in tear aqueous production and abnormal tear film lipid, protein and mucin profiles. There are many causes of dry eye, some of which include age, Lasik eye surgery, arthritis, medications, chemical/thermal burns, allergies, and diseases, such as cystic fibrosis and Sj5grens's syndrome. Increasing anion secretion via CFTR would enhance fluid transport from the corneal endothelial cells and secretory glands surrounding the eye to increase corneal hydration. This would help to alleviate the symptoms associated with dry eye disease. SjC3grens's syndrome is an autoimmune disease in which the immune system attacks moisture-producing glands throughout the body, including the eye, mouth, skin, respiratory tissue, liver, vagina, and gut. Symptoms, include, dry eye, mouth, and vagina, as well as lung disease. The disease is also associated with rheumatoid arthritis, systemic lupus, systemic sclerosis, and polymypositis/dermatomyositis. Defective protein trafficking is believed to cause the disease, for which treatment options are limited. Modulators of CFTR activity may hydrate the various organs afflicted by the disease and help to elevate the associated symptoms. [0013] As discussed above, it is believed that the deletion of residue 508 in AF508-CFTR prevents the nascent protein from folding correctly, resulting in the inability of this mutant protein to exit the ER, and traffic to the plasma membrane. As a result, insufficient amounts of the mature protein are present at the plasma membrane and chloride transport within epithelial tissues is significantly reduced. In fact, this cellular phenomenon of defective ER processing of ABC transporters by the ER machinery, has been shown to be the underlying basis not only for CF disease, but for a wide range of other isolated and inherited diseases. The two ways that the ER machinery can malfunction is either by loss of coupling to ER export of the proteins leading to degradation, or by the ER accumulation of these defective/misfolded proteins [Aridor M, et al., Nature Med., 5(7), pp 745- 751 (1999); Shastry, B.S., et al., Neurochem. International, 43, pp 1-7 (2003); Rutishauser, J., et al., Swiss Med Wkly, 132, pp 211 -222 (2002); Morello, JP et al., TIPS, 21L pp. 466- 469 (2000); Bross P., et al., Human Mut., 14, pp. 186-198 (1999)]. The diseases associated with the first class of ER malfunction are cystic fibrosis (due to misfolded AF508-CFTR as discussed above), hereditary emphysema (due to al-antitrypsin; non Piz variants), hereditary hemochromatosis, coagulation-fibrinolysis deficiencies, such as protein C deficiency, Type I hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type 1 chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo Hurler, mucopolysaccharidoses (due to lysosomal processing enzymes), Sandhof!"Tay-Sachs (due to p-hexosaminidase), Crigler-Najjar type II (due to UDP-glucuronyl-sialyc--transferase). - 4- WO 2009/123896 PCT/US2009/038203 polyendocrinopathy/hyperinsulemia, diabetes niellitus (due to insulin receptor), Laron dwarfism (due to growth hormone receptor), myleoperoxidase deficiency, primary hypoparathyroidism (due to preproparathyroid hormone), melanoma (due to tyrosinase). The diseases associated with the latter class of ER malfunction are glycanosis CDG type 1, hereditary emphysema (due to al antitrypsin (PiZ variant), congenital hyperthyroidism, osteogenesis imperfecta (due to Type I, II, IV procollagen), hereditary hypofibrinogenemia (due to fibrinogen), ACT deficiency (due to al antichymotrypsin), diabetes insipidus (DI), neurophyseal DI (due to vasopvessin hormone/V2 receptor), neprogenic DI (due to aquaporin II), Charcot-Marie Tooth syndrome (due to peripheral myelin protein 22), Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease ( due to PAPP and presenilins), Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders such as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and myotonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease (due to prion protein processing defect), Fabry disease (due to lysosomal c-galactosidase A), Straussler-Scheinker syndrome, chronic obstructive pulmonary disease (COPD), dry eye disease, and Sj~gren's Syndrome. [00141 In addition to up-regulation of CFTR activity, reducing anion secretion by CFTR modulators may be beneficial for the treatment of secretory diarrheas, in which epithelial water transport is dramatically increased as a result of secretagogue activated chloride transport. The mechanism involves elevation of cAMP and stimulation of CFTR. [00151 Although there are numerous causes of diarrhea, the major consequences of diarrheal diseases, resulting from excessive chloride transport are common to all, and include dehydration, acidosis, impaired growth and death. 100161 Acute and chronic diarrheas represent a major medical problem in many areas of the world. Diarrhea is both a significant factor in malnutrition and the leading cause of death (5,000,000 deaths/year) in children less than five years old. [00171 Secretory diarrheas are also a dangerous condition in patients of acquired immunodeficiency syndrome (AIDS) and chronic inflammatory bowel disease (IBD). Sixteen million travelers to developing countries from industrialized nations every year develop diarrhea, with the severity and number of cases of diarrhea varying depending on the country and area of travel.
WO 2009/123896 PCT/US2009/038203 [00181 Diarrhea in barn animals and pets such as cows, pigs and horses, sheep, goats, cats and dogs, also known as scours, is a major cause of death in these animals. Diarrhea can result from any major transition, such as weaning or physical movement, as well as in response to a variety of bacterial or viral infections and generally occurs within the first few hours of the animal's life. [00191 The most common diarrheal causing bacteria is enterotoxogenic E-coli (ETEC) having the K99 pilus antigen. Common viral causes of diarrhea include rotavirus and coronavirus. Other infectious agents include cryptosporidium, giardia lamblia, and salmonella, among others. [00201 Symptoms of rotaviral infection include excretion of watery feces, dehydration and weakness. Coronavirus causes a more severe illness in the newborn animals, and has a higher mortality rate than rotaviral infection. Often, however, a young animal may be infected with more than one virus or with a combination of viral and bacterial microorganisms at one time. This dramatically increases the severity of the disease. [00211 Accordingly, there is a need for modulators of an ABC transporter activity, and compositions thereof, that can be used to modulate the activity of the ABC transporter in the cell membrane of a mammal. [00221 There is a need for methods of treating ABC transporter mediated diseases using such modulators of ABC transporter activity. [00231 There is a need for methods of modulating an ABC transporter activity in an ex vivo cell membrane of a mammal. 100241 There is a need for modulators of CFTR activity that can be used to modulate the activity of CFTR in the cell membrane of a mammal. [00251 There is a need for methods of treating CFTR-mediated diseases using such modulators of CFTR activity. [00261 There is a need for methods of modulating CFTR activity in an ex vivo cell membrane of a mammal. SUMMARY OF THE INVENTION [00271 It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are useful as modulators of ABC transporter activity, particularly CFTR activity. These compounds have the general formula 1: - 6- WO 2009/123896 PCT/US2009/038203 1 O0 N 1 (R)x- B - W(Rw),)w N 4 A RN 3 1 or a pharmaceutically acceptable salt thereof-, wherein RN, ring A, ring B, W, R , R1, n, w, and x are described below. 100281 These compounds and pharmaceutically acceptable compositions are useful for treating or lessening the severity of a variety of diseases, disorders, or conditions, including, but not limited to, cystic fibrosis, hereditary emphysema, hereditary hemochromatosis, coagulation fibrinolysis deficiencies, such as protein C deficiency, Typc I hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type I chylonicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, mucopolysaccharidoses, Sandhof/Tay-Sachs, Crigler-Najjar type II, polyendocrinopathy/hyperinsuilemia, diabetes mellitus, laron dwarfism, myleoperoxidase deficiency, primary hypoparathyroidism, melanoma, glycanosis CDG type 1, hereditary emphysema, congenital hyperthyroidisin, osteogenesis imperfecta, hereditary hypofibrinogenemia, ACT deficiency, diabetes insipidus (di), neurophyseal di, neprogenic DI, Charcot-Marie Tooth syndrome, Perlizaeus-Merzbacher disease., neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders asuch as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and myotonic dystrophy, as well as spongiform enceplialopathies, such as hereditary Creutzfeldt-Jakob disease, Fabry disease, StraussIer-Scheinker syndrome, COPD, dry-eye disease, and Sj5gren's disease. DETAILED DESCRIPTION OF THE INTENTION [00291 General Description of Compounds of the Invention: [00301 The present invention relates to compounds of formula 1: (R0 - N 4(W (R"')), N 4 A .FN 3 or a pharmaceutically acceptable salt thereof, wherein: WO 2009/123896 PCT/US2009/038203 [00311 RN is 1-1, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocycloalkyl; [00321 A is an optionally substuted 3-7 membered monocyclic ring; [00331 B is optionally fused to a 5-7 membered ring selected from the group consisting of cycloaliphatic, aryl, heterocyclic, and heteroaryl; 100341 R1 is halo, alkyl, OH, alkoxy, thioalkyl, trifluoromethoxy; or J [00351 two R' on adjacent atoms, taken together, form ; [00361 wherein J is selected from the group consisting of CH2, CF2, or C(CH 3
)
2 ; [00371 W is independently a bond or an optionally substituted (Ci-C6) alkylide ne chain wherein up to two methylene units of W are independently replaced by --- CO--, -0--, -S-, -S02-, or NR'-; [00381 R' is independently H, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocycloalkyl; [00391 Rw is independently H, halo, CN, NO 2 , N(R)2, CF 3 , OCF 3 , OH, OR, C(O)N(R) 2 , C(O)R, CO2R, -O(Cl1 -C6)alkylidene-OR, -O(ClI -C6)alkylidene-N(R) 2 , -O(Cl-C6)alkylidene heterocycloalkyl, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, 2 or heteroaryl, wherein, when substituted, R" is substituted with up to two R 100401 R 2 is halo, CN, NO2, CF 3 , OCF 3 , OR., OC(O)R. OC(O)N(R) 2 , SR, S(O)R, SO 2 R, SO2N(R)2, SO 3 R, C()R0, CO 2 R, C(O)N(R)2, N(R)2, NRC(O)R, NRCO 2 R, NRC(O)N(R) 2 ,
NRSO
2 R, B(OH) 2 , or NRSO 2
N(R)
2 ; [00411 R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; [00421 n is 1 or 2; [00431 w is an integer from 0 to 4 inclusive; and [00441 x is an integer from 0 to 5 inclusive; provided that when W is a bond and R" is cycloaliphatic, heterocycloaliphatic, aryl, or heteroaryl, -W-Rw is attached to the 3- or 4- position of the pyridyl ring. -8- WO 2009/123896 PCT/US2009/038203 [00451 Compounds and Definitions: [00461 Compounds of this invention include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. [00471 The term "ABC-transporter" as used herein means an ABC-transporter protein or a fragment thereof comprising at least one binding domain, wherein said protein or fragment thereof is present in vivo or in vitro. The term "binding domain" as used herein means a domain on the ABC-transporter that can bind to a modulator. See, e.g., Hwang, T. C. et al., J. Gen. Physiol. 1998): 111(3), 477-90. [00481 The term "CFTR" as used herein means cystic fibrosis transmembrane conductance regulator or a mutation thereof capable of regulator activity, including, but not limited to, AF508 CFTR and G55 1D CFTR (see, e.g., htr;./iwwwsgenesickksson.ca/eft, for CFTR mutations). [00491 The term "modulating" as used herein means increasing or decreasing by a measurable amount. [00501 For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 7 5 th Ed. Additionally, general principles of organic chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5 th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference. [00511 As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted." In general, the term "substituted", whether preceded by the term "optionally" or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to -9- WO 2009/123896 PCT/US2009/038203 allow for their production, detection, and preferably their recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40'C or less, in the absence of moisture or other chemically reactive conditions, for at least a week. [00521 The term "aliphatic" or "aliphatic group" or alkyll", as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle" "cycloaliphatic" or "cycloalkyl"), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-8 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodiments aliphatic groups contain 1-4 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbocycle" or "cycloalkyl") refers to a monocyclic C 3
-C
8 hydrocarbon or bicyclic Cs-C 12 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule wherein any individual ring in said bicyclic ring system has 3-7 members. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. [00531 The term "heteroaliphatic", as used herein, means aliphatic groups wherein one or two carbon atoms are independently replaced by one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon. Heteroaliphatic groups may be substituted or unsubstituted, branched or unbranched, cyclic or acyclic, and include "heterocycle", "heterocyclyl", "heterocycloaliphatic", or "heterocyclic" groups. [00541 The term "heterocycle", "heterocyclyl", "heterocycloaliphatic", or "heterocyclic" as used herein means non--aromatic, monocyclic, bicyclic, or tricyclic ring systems in which one or a plurality of ring members is an independently selected heteroaton. In some embodiments, the "heterocycle", "heterocyclyl", "heterocycloaliphatic", or "heterocyclic" group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from the group consisting of oxygen, sulfur, nitrogen, and phosphorus, and each ring in the system contains 3 to 7 ring members. - 10- WO 2009/123896 PCT/US2009/038203 [00551 The term "heteroatom" means one or more of boron, oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR_ (as in N-substituted pyrrolidinyl)). [00561 The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation. [00571 The term "alkoxy", or "thioalkyl", as used herein, refers to an alkyl group, as previously defined, attached to the principal carbon chain through an oxygen ("alkoxy") or sulfur ("thioalkyl") atom. 100581 The terms "haloaliphatic" and "haloalkoxy" means aliphatic or alkoxy, as the case may be, substituted with one or more halogen atoms. The term "halogen" means F, Cl, Br, or L Examples of haloaliphatic include -CHF 2 , -CH 2 F, -CF 3 , -CF 2 -, or perhaloalkyl, such as, -CFCF3. [00591 The term "arvyl" used alone or as part of a larger moiety as in "aralkyl", "aralkoxy", or "aryloxyalkyl", refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term "aryl" may be used interchangeably with the term "aryl ring". The term "arvyl" also refers to heteroaryl ring systems as defined hereinbelow. 100601 The term "heteroaryl"., used alone or as part of a larger moiety as in "heteroaralkyl" or "heteroarylalkoxy", refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, and wherein each ring in the system contains 3 to 7 ring members. The term "heteroaryl" may be used interchangeably with the term "heteroaryl ring" or the term "heteroaromatic". [00611 An aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) group may contain one or more substituents. Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group are selected from the group consisting of halogen; -R'; -OR'; -SR'; 1,2-methylene-dioxy; 1,2 ethylenedioxy; phenyl (Ph) optionally substituted with R"; -O(Ph) optionally substituted with R";
-(CH
2 )p 2 (Ph), optionally substituted with R'; -CH-=CH(Ph), optionally substituted with R'; -NO 2 ; CN; -N(R 0 )2; -NR 0
C(O)R
0 ; -NR C(O)N(R 0 )2; -NR'CO 2 R-; -NR 0
NR
0 C(O)R"; NR 0
NR
0 C(O)N(R')2; -NRONR 0
CO
2
R
0 ; -C(O)C(O)R; -C(O)CH2C(O)R; -C0 2 R: -C(O)R 0 ; - 11 - WO 2009/123896 PCT/US2009/038203
C(O)N(R
0 )2; -OC(O)N (R )2; -S(O) 2
R
0 ; -SO 2
N(R)
2 ; -S(O)RO; -NR'SO 2
N(R')
2 ; -NR 0
SO
2 R; -C(=S)N(R')2; -Q=NH)-N(R) 2 ; and -(CH2)-2NHC(O)R' wherein each independent occurrence of R' is selected from the group consisting of hydrogen, optionally substituted C 1
.
6 aliphatic, an unsubstituted 5-6 membered heteroaryl or heterocyclic ring, phenyl, -O(Ph), and -C-1 2 (Pl), or, notwithstanding the definition above, two independent occurrences of R', on the same substituent or different substituents, taken together with the atom(s) to which each R' group is bound, form a 3 8-membered cycloalkyl, heterocyclyl, aryl, or heteroaryl ring having 0-3 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. Optional substituents on the aliphatic group of R are selected from the group consisting of NH2, NH(Ci 4 aliphatic), N(C 1 4 aliphatic) 2 , halogen, C1 4 aliphatic, OH, O(Cy 4 aliphatic), NO 2 , CN. CO2.H, C0 2 (Cp 4 aliphatic), O(haloCt 4 aliphatic), and haloCp 4 aliphatic, wherein each of the foregoing C 1 ,aliphatic groups of R' is unsubstituted. 100621 An aliphatic or heteroaliphatic group, or a non-aromatic heterocyclic ring may contain one or more substituents. Suitable substituents on the saturated carbon of an aliphatic or heteroaliphatic group, or of a non-aromatic heterocyclic ring are selected from the group consisting of those listed above for the unsaturated carbon of an aryl or heteroaryl group and additionally include the following: =0, =S, :=NNHR', =NN(R*) 2 , =NN-{C(O)R', =NNHCO 2 (alkyl),
=NTNH-SO
2 (alkyl), and =NR*, where each R* is independently selected from the group consisting of hydrogen and an optionally substituted C 1
.
6 aliphatic. Optional substituents on the aliphatic group of R* are selected from the group consisting of NH?, NH(CI 4 aliphatic), N(CI 4 aliphatic) 2 , halogen, C4 aliphatic, OH, O(C 1 4 aliphatic), NO 2 , CN, CO2, C0 2 (Cp 4 aliphatic), O(halo Cp 4 aliphatic), and halo(C 1 4 aliphatic), wherein each of the foregoing C 1
.
4 aliphatic groups of R* is unsubstituted. [00631 Optional substituents on the nitrogen of a non-aromatic heterocyclic ring are selected from the group consisting of --- R, N(R') 2 , -C(O)R , -CO 2 R , -C(O)C(O)Ri, -C(O)CH 2 C(O)R SO 2 R, -SO 2 N(Rt) 2 , -C(=S)N(R)2, -C(=NH)-N(R') 2 , and -NRSO 2 R; wherein R is hydrogen, an optionally substituted C 1 .6 aliphatic, optionally substituted phenyl, optionally substituted -O(Ph), optionally substituted -CH 2 (Ph), optionally substituted -(CH 2 )p 2 (Ph); optionally substituted CH=CH(Ph); or an unsubstituted 5-6 membered heteroaryl or heterocyclic ring having one to four heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R-, on the same substituent or different substituents, taken together with the atom(s) to which each Ri group is bound, form a 3 8-membered cycloalkyl, lieterocyclyl, aryl, or heteroaryl ring having 0-3 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. Optional substituents on the -12 - WO 2009/123896 PCT/US2009/038203 aliphatic group or the phenyl ring of R+ are selected from the group consisting of NHI- 2 , NH(C 1 aliphatic), N(C 1
-
4 aliphatic) 2 , halogen, C 1 4 aliphatic, OH, O(C 1
.
4 aliphatic), NO 2 , CN, C0 2 H,
CO
2
(C
1 4 aliphatic), O(halo C14 aliphatic), and halo(C 1 4 aliphatic), wherein each of the foregoing
C
4 aliphatic groups of R is unsubstituted. [00641 The term "alkylidene chain" refers to a straight or branched carbon chain that may be fully saturated or have one or more units of unsaturation and has two points of attachment to the rest of the molecule. The term "spirocycloalkylidene" refers to a carbocyclic ring that may be fully saturated or have one or more units of unsaturation and has two points of attachment from the same ring carbon atom to the rest of the molecule. [00651 As detailed above, in some embodiments, two independent occurrences of R' (or R-, or any other variable similarly defined herein), are taken together with the atom(s) to which each variable is bound to form a 3-8-membered cycloalkyl, heterocyclyl, aryl, or heteroaryl ring having 0-3 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. Exemplary rings that are formed when two independent occurrences of R (or RN, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound include, but are not limited to the following: a) two independent occurrences of R' (or R , or any other variable similarly defined herein) that are bound to the same atom and are taken together with that atom to form a ring, for example, N(R) 2 , where both occurrences of R" are taken together with the nitrogen atom to form a piperidin-1-yl, piperazin- -yl, or morpholin-4-yl group; and b) two independent occurrences of R' (or RV, or any other variable similarly defined herein) that are bound to different atoms and are taken together with both of those atoms to form a ring, for
OR
0 H | example where a phenyl group is substituted with two occurrences of OR" , these two occurrences of R" are taken together with the oxygen atoms to which they are bound to form a fused 6-membered oxygen containing ring: 0. It will be appreciated that a variety of other rings can be formed when two independent occurrences of R' (or RI, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound and that the examples detailed above are not intended to be limiting. [00661 Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double -13 - WO 2009/123896 PCT/US2009/038203 bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 1 C- or 14 C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays. 100671 Description of Exemplary Compounds: 100681 In one embodiment, the present invention relates to a compound of formula 1: (R) i O (RI)x+B 0 __(W (R*)n),. A RN 3 1 100691 or a pharmaceutically acceptable salt thereof, wherein: [00701 RN is H, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocycloalkyl; [00711 A is an optionally substuted 3-7 membered monocyclic ring; [00721 B is optionally fused to a 5-7 membered ring selected from the group consisting of cycloaliphatic, aryl, heterocyclic, and heteroaryl; [00731 R' is halo, alkyl, 01-, alkoxy, trifluoromethoxy; or I0 [00741 two R on adjacent atoms, taken together, form [00751 wherein J is selected from the group consisting of CH 2 , CF 2 . or C(CHl 3
)
2 ; [00761 W is independently a bond or a (CI-C6) alkylidene chain wherein up to two methylene units of W are independently replaced by -CO-, -0-, -S-, -SO2-, or -- NR'-; [00771 R' is independently H, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocyclic; [00781 Rw is independently H, halo, CN, NO 2 , N(R)2, CF 3 , OCF 3 , OH, OR, C(O)R, CO 2 R,
C(O)N(R)
2 , -O(Cl-C6)alkylidenc-OR, -O(Cl-C6)alkylidene-N(R)2, -O(Cl-C6)alkylidene - 14 - WO 2009/123896 PCT/US2009/038203 heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, R' is substituted with up to two R; 100791 R 2 is halo, CN, NO2, CF3, OCF 3 , OR., OC(O)R, OC(O)N(R)2, SR, S(O)R, SO,2R, SO2N(R)2, SO 3 R, C(O)R, CO 2 R, C(O)N(R2, N(R), NRC(O)R, NRCO 2 R, NRC(O)N(R)2,
NRSO
2 R, B(OR)2, or NRSO2N(R)2; [00801 R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; [00811 n is 1 or 2; [00821 w is an integer from 0 to 4 inclusive; and [00831 x is an integer from 0 to 5 inclusive; [00841 provided that when W is a bond and Rw is cycloaliphatic, heterocycloaliphatic, aryl, or heteroary], -W-R' is attached to the 3- or 4- position of the pyridyl ring. [00851 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein A is selected from the group consisting of: q (R~ (R3 (R3) (R") (R)(3 a b C d and e, wherein [00861 R 3 is alkyl, alkaryl, aryl, or heteroaryl; and (R3) [00871 q is an integer from 0 to 4 inclusive. In a further embodiment, A is . In a further embodiment, A is . 100881 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein x is 2. [00891 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein R' is halo. In another embodiment, R' is chloro. In another embodiment, R' is alkyl. In another embodiment, R' is methyl. In another embodiment, R1 is OH. In another embodiment, R' is alkoxy. In a further embodiment, R 1 is methoxy. In another -'15- WO 2009/123896 PCT/US2009/038203 embodiment, R 1 is thioalkyl. In another embodiment, R 1 is thiomethyl. In a further embodiment, R is trifluoromethoxy. 100901 In another embodiment, the present invention relates to a compound of formula I and / J the attendant definitions, wherein two RI on adjacent atoms, taken together, form ; wherien J is selected from the group consisting of CH 2 , CF 2 , or C(CH3)2. In a further embodiment, J is CH 2 . In a further embodiment, J is CF2. 100911 In another embodiment, the present invention relates to a compound of formula I and N N the attendant definitions, ' wherein RN is 1- or alkyl. In a further embodiment, RN is H-. In a further embodiment, RN is akyl [00921 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein w is 0. In a further embodiment, w is 1. In a further embodiment, W is 2. [00931 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein n is 1. In a further embodiment, n is 2. [00941 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein W is a bond. In a further embodiment, W is an optionally substituted (Ci-C6) alkylidene chain. In a further embodiment, W is -CH2-. In a further embodiment, W is -NH-. In a further embodiment, W is -0-. In a further embodiment, W is -CO-. In a further embodiment, W is -OCH-. [00951 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein R"' is H. In a further embodiment, R" is OH. In a further embodiment, R" is heterocyclic. In a further embodiment, R" is aryl. In a further embodiment, R" is phenyl. In a further embodiment, RI is heteroaryl. In a further embodiment, R' is pyridyl. In a further embodiment, Rw is alkoxy. In a further embodiment, RW is cycloalkyl. In a further embodiment, R"' is cyclohexyl. [00961 In another embodiment, the present invention relates to a compound of formula I and the attendant definitions, wherein --- W-R' is acyclic. In another embodiment, -W -RI is an aryl, heteroaryl, cycloaliphatic, or heterocycloaliphatic ring. In a further embodiment, --- W(R"'), is selected from the following: -16- WO 2009/123896 PCT/US20091'038203 0 H O H 0,-- N -F, -CH 3 , -CH_ 2 CfH 3 , -CN, -CE 3 , -CONI-1 2 . -CI-1 2 C1-Jl(CH_ 3
)
2 , 1 5 H NC 0 N 0 ~~0 OH, 0~ H2N-" 1-1 NN 0 0H ,O 1 ,O 0 0 N N 0o ~ N 1 " N ~~~0 ii ~ - I 70 WO 2009/123896 PCT/US2009/038203 00 c Ni OH H H 2 N * 07H [0097] In a. further embodiment, -W(R"),, selected frorn the following:, HO O N0 OB(OH)2 0HN N H N02, H [00981 In another embodiment, the present invention relatest to a compound having formula Ta: 0 - (W(R'N H 3 Ia [00991 or a pharmaceutically acceptable salt thereof, wherein: [001001 J is CH 2 or CF 2 ; - 18 - WO 2009/123896 PCT/US2009/038203 [001011 W is independently a bond or a (Ci-C6) alkylidene chain wherein up to two methylene units of W are independently replaced by -CO-, -O-, or -NR ; 1001021 W is independently H or alkyl; 1001031 Rw is independently H, halo, CN., N(R)2, CF 3 , OH, CO 2 R, C(O)N(R) 2 , -- O(Cl- C6)alkyliden e-OR, -O(C I -C6)alkyidene-N(R) 2 , -O(C I -C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, RI is substituted with up to two Ri; [001041 R2 is halo, OR, CO 2 R, C(O)N(R) 2 , SO 2
N(R)
2 , B(OR) 2 , orN(R) 2 ; [001051 R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; [001061 n is I or 2; and [001071 w is an integer from 0 to 4 inclusive; [001081 provided that when W is a bond and R" is cycloaliphatic, heterocycloaliphatic, aryl, or heteroaryl, -W-R"' is attached to the 3- or 4-- position of the pyridyl ring. 1001091 In another embodiment, the present invention relates to a compound of formula la and the attendant definitions, wherein J is CH2. In a further embodiment, J is CF2. [001101 In another embodiment, the present invention relates to a compound of formula la and the attendant definitions, wherein --W-RI is acyclic. In another embodiment, the present invention relates to a compound of formula la and the attendant definitions, wherein -W-R" that is an aryl, heteroaryl, cycloaliphatic, or heterocycloaliphatic ring. [001111 In another embodiment, the present invention relates to a compound of formula la and the attendant definitions, wherein -W(R"), is selected from the following: OH OH 0 N -F, -CH 3 , -CH 2
CH
3 , -- CN, -- CF 3 , -- CONH 2 , -CH2CH(CH 3
)
2 , ------ - 9 I 0 N cr cr,-j 1 0 OH -19- WO 2009/123896 PCT/US20091'038203 1-0 0 N>0 0 -- H 2 N - , 01 N N, HN0 0 Fl0 Y N I> \ >N , 0 "o 0 0 N OH, OH, O H OH 00 0 N N 1-1 N ~,o 0 N - o 0 N N 0 ci NH 0 i O ci c
H
2 N - 20 - WO 2009/123896 PCT/US2009/038203 N 0 . H 0N [00112] In a further embodiment, -- W(R") selected from the following: / HO 0 H2NO B(OH) 2 H 0 !H2 H 2
~N
2 or 0 NHH 0 NH [00113] In another embodiment, the present invention relates to a compound having formula Ib: (Rik-- W(R*), H Ib [0)0114] or a pharmaceutically acceptable salt thereof, wherein: [00115] R 1 is halo, alkyl, OH, alkoxy, thioalkyi, trifluoromethoxy; or [00116] two R on adjacent atoms, taken together, form; [00117] wherein J is selected from the group consisting of CH2, CF 2 , or C(CH3)2; [00118] W is -NR', -CO-, or -CH 2 -; [00119] R' is independently H- or alkyl; -21- WO 2009/123896 PCT/US2009/038203 [001201 Rw is independently H, halo, CN, N(R-) 2 , CF 3 . OHf, COR, C(O)N(R)2, -O(C C6)alkylidene-OR, -O(Cl-C6)alkylidene-N(R), -O(CI-C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, R' is substituted with up to two R2 [001211 R is halo, OR, CO 2 R, C(O)N(R) 2 , SO 2
N(R)
2 , B(OR) 2 , or N(R)2; [00122] R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; [001231 n is I or 2; and [001241 x is an integer from 0 to 5 inclusive. [001251 In another embodiment, the present invention relates to a compound of formula J lb and the attendant definitions, wherein two R' on adjacent atoms, taken together, form 0 and J is CH2. In another embodiment, J is CF 2 . [00126] In another embodiment, the present invention relates to a compound of formula lb and the attendant definitions, wherein --W(R"), is selected from the following:
-CH
3 , -CH 2
CH
3 , -CH 2
CH(CH
3
)
2 , OI9 OH 0 N Ks 0 N K 0OHN\ OH, H2N Oq O0, OH, OH, -22 - WO 2009/123896 PCT/US2009/038203 O OC N N N OH, 0 H N 0 N O NN ON1 O 111 N,N NHN H NO :J N NH NH NN N, , 0 ,OH 501 ,CI 0 0N N0 N ~/N 5' ,B(OH ~ HNN 0 I
~NH
2 N /5 0 0, NH Q ,or [001271 In another emnbodimnent, the present invention relates to a compound having formula 1c: NV 0 N N H Ic - 23 - WO 2009/123896 PCT/US2009/038203 [001281 or a pharmaceutically acceptable salt thereof, wherein: [001291 R 1 is halo, alkyl, OH, alkoxy, thioalkyl, trifluoromethoxy; or J two R' on adjacent atoms, taken together, form O wherein J is selected from the group consisting of CH 2 , CF 2 , or C(CH 3
)
2 ; [001301 W is -NR', -CO--, or -CH 2 -; [001311 R' is independently -1 or alkyl; [001321 Rw is independently H, halo, CN, N(R-) 2 , CF 3 , OHI, CO2R, C(O)N(R)2, -O(Cl C6alklidene-OR, -O(Cl-C6)alkylidene-N(R) 2 , -O(CI-C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, R" is substituted with up to two R2 [001331 R is halo, OR, CO 2 R, C(O)N(R) 2 , SO 2
N(R)
2 , B(OR) 2 , or N(R)2; [001341 R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; [001351 n is I or 2; and [001361 x is an integer from 0 to 5 inclusive. [001371 In another embodiment, the present invention relates to a compound of formula Ic J and the attendant definitions, wherein two R on adjacent atoms, taken together, form 0 and J is CH 2 . In another embodiment, J is CF 2 . [001381 In another embodiment, the present invention relates to a compound of formula Ic and the attendant definitions, wherein --W(R") is selected from the following:
-CH
3 , -CH 2
CH
3 , -CH2CH(CH 3
O
2 , OH >0~~~~ 0~O~ N~O 0 N 0 NN OH, O0HN -24 - WO 2009/123896 PCT/US2009/038203 H2N OO O11 N N N 0 N OH H, OH, 00 N0 , 0O ~ N 0 N HH OH 0 0,, 01O 0 -N A N' / N 00 K-i N\ N N H NHy H 0 B(OH)2 0 H N INN-!, r - , NH 2 / I NH 0 NH or [00139] In another embodiment, the present invention relates to a compound of Table 1. -25 - WVO 2009/123896 PCT/US20091'038203 [0)01401 Table 1. r~ I ii H '-1 7 77 f '1'Ti EJ ------------------ ------------------------------------------- ' .. K -26- WO 2009/123896 PCT/US20091'038203 ---------------------------- 7 ------------------- ------------------------ ------------ -----------------I ----------------- T7 2 2' -- --- - -- --- --- - -- --- - -- --- -- -- --- --- --- --- -- --- --- ---- --- --- --- --- --- --- --- --- ---- --- -- --- --- --- --- --- ---- --- --- --- --- --- --- --- - N j [ ~ ? KN _ I2.
WO 2009/123896 PCT/US20091'038203 ' 2 1 i i '1t F-f-. --- -- --- -- - - -- -- --- -- -- -- --- -- --- -- - - - - --- - - - - -- -3- -- 1' 77 WO 2009/123896 PCT/US20091'038203 LINI -------------------- TU------------------- ------------------- T--------------------I; ------------------- 7------------ I N i:2f. 43~44 C" 0H~3 -, N -29 WO 2009/123896 PCT/US20091'038203 7-7I K N X., Il ------------------- ------------ 1 --------- ------------------------
---------
-3 WO 2009/123896 PCT/US20091'038203 H 7 J, 1 ~ Y T ii f ' -- - - - - - - -- - - - - - - - -- - - - - - - -- - - - - - - - -- - - - - - - -- - - - - - - - -- - - - - - - -- - - - - - - - -- - - - - - - ~ 7 7 - 3- WO 2009/123896 PCT/US2009/038203 [04]Inaohe mo iet th prsn.neto eaes oapamcuia acetbecie InL anterebdiet th hamcetcaomoito fute1omrssa TKi additional agent selected from the group consisting of a nmcolytic agent, bronchodialator, an anti biotic, an anti-infective agent, an anti-inflammatory agent, CFTRc corrector, and a nutritional agent. [001421 In another embodiment, the present invention relates to a method of modulating ABC transporters in a membrane of a cell, comprising the step of contacting said cell with a compou nd of the present invention. In another embodiment, the ABC transporter is CFTR. [001431 In another embodiment, the present invention relates to a method of treating a condition, disease, or disorder in a patient implicated by ABC transporter activity, comprising the step of administering to said patient a compound of the present invention, In another embodiment, the ABC transporter is CFTR. In another embodiment, said condition, disease, or disorder is selected from cystic fibrosis, hereditary emphysema, hereditary henmochronmatosis, coagulation fibrinolysis deficiencies, such as protein C deficiency, Type 1 hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type 1 chylomicroniemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, mucopolysaccharid oses, Sandhof/Tay-Sachs, Crigler-Najjar type II, polyendocrinopathy/hyperinsulemia, diabetes mellitus, laron dwarfism, myleoperoxidase WO 2009/123896 PCT/US2009/038203 deficiency, primary hypoparathyroidism, melanoma, glycanosis CDG type 1, hereditary emphysema, congenital hyperthyroidism, osteogenesis imperfecta, hereditary hypofibrinogenemia, ACT deficiency, diabetes insipidus (di), neurophyseal di, neprogenic DI, Charcot.-Marie Tooth syndrome, Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders asuch as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and myotonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease, Fabry disease, Straussler-Scheinker syndrome, COPD, dry-eye disease, and Sjbgren's disease. 1001441 In another embodiment, the present invention relates to a kit for use in measuring the activity of a ABC transporter or a fragment thereof in a biological sample in vitro or in vivo, comprising: (i) a first composition comprising a compound of the present invention; and (ii) instructions for: a) contacting the composition with the biological sample; and b) measuring activity of said ABC transporter or a fragment thereof. [001451 General Synthetic Schemes [001461 Compounds of formula I can be prepared by well-known methods in the art. Illustrated below are exemplary methods for the preparation of compounds of formula 1. Schemes I below illustrates an exemplary synthetic method for compounds of formula 1. SYNTHETIC SCHEMES 1001471 Compounds of the invention may be prepared by known methods and as illustrated in Schemes I-IV. - 33 - WO 2009/123896 PCT/US20091'038203 [001481 Scheme 1. Preparation of cyclopropyl acids 111 a) o C 2 Me b) OF 0 0 C) ) P -0 N ON00 0 0 - OH 00 X== Br or 1; [001491 a) Pd(PPh,4, CO), 'IeOl-l; b) LiAl-1 4 ; C) SOCd 2 ; d) NaCN; e) CIFCl-CH?Br, NatOJ-, AT; f) CICH 2
CH
2 B3r, NaOH;- g) aG AT. [001501 Scheme H. Preparation of amino pyridyl building blocks C) 0C) I tOH a) b) I) N. - / N Rw 0e) O.(H f)
H
2 N NF1 2 N N RWH 2 N N R X = C1, Br, or 1; HO! HN0OMe 1001511 a) CiCO;IBu, !Et 3 N ; b) /c) RwM' (M= Li, MgBr); d) NH;i, EtOI; e) NaBH 4 , MeOl-I; 1), E1 3 Si, TFA. -341 - WO 2009/123896 PCT/US20091'038203 [0)01521 Scheme Ill. Amide coupling 0J ) 0 N. N 0) 0 Jz-Y
H
2 N N [001531 a) SOCI 2 , DMF; b) Pyridine. ~ R%11; c) HA~TU Et 3 N. DMF,
H
2 N YN -: ''R~ or alternatively: [001541 Scheme IV. Derivatization of Arnides 1001551 Scgherme Iva N N:~ a) ,0 ~ N~ -A- ) - 0 b o r c) J -- NF.IRvw \
-
0 X=Ci, Br, or 1 [001561 a)l RCH 2 ZnIY,, Idppf) 2 PdCi 2 , TH (Y= Cl.n=!; Y=Br, n=1);b,) R NH,, (dppf)2PdC11 2 .Xantplios, K t OBu, dioxane. Et; c) R~NEJ 2
.
WO 2009/123896 PCT/US2009/038203 [001571 Scheme IVb W H H YN N a) N NJ -',I J --CN a ) .A b) 0 OH e) c) H d) H [0 8 aN N b NOi d NpN-ln e 00 R I-:rI llo 0 N 2 0: 0 OR [001 581 a:) R'MgBr; b) \a.BH 4 , MeOH;c) Et 3 SiH, TF'A d,) ROH,p-7LOH. tolue-ne; e,)1) MsCl, DIEA; 2) R<NH, DIEA. [001591 One of skill in the art will readily appreciate that synthetic routes suitable for various substituents of the present invention are such that the reaction conditions and steps. [001601 Uses, Formulation and Administration [001611 Pharmaceutically ageptable compositions 1001621 As discussed above, the present invention provides compounds that are useful as modulators of ABC transporters and thus are useful in the treatment of disease, disorders or conditions such as Cystic fibrosis, Hereditary emphysema, Hereditary hemochromatosis, Coagulation -Fibrinolysis deficiencies, such as Protein C deficiency, Type I hereditary angioedema, Lipid processing deficiencies, such as Familial hypercholesterolemia, Type 1 chylomicronemia, Abetalipoproteinemia, Lysosomal storage diseases, such as I-cell disease/Pseudo-Hurler, Mucopolysaccharidoses, Sandh of/Tay-Sachs, Crigler-Najjar type II, Polyendocrinopathy/Hyperinsulem ia, Diabetes mellitus, Laron dwarfism, Myleoperoxidase - 36 - WO 2009/123896 PCT/US2009/038203 deficiency, Primary hypoparathyroidisn, Melanoma, Glycanosis CDG type 1, Hereditary emphysema, Congenital hyperthyroidism, Osteogenesis imperfecta, Hereditary hypofibrinogenemia, ACT deficiency. Diabetes insipidus (DI), Neurophyseal DI, Neprogenic DI, Charcot-Marie Tooth syndrome, Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders asuch as Huntington, Spinocerebullar ataxia type I, Spinal and bulbar muscular atrophy, Dentatorubal pallidoluysian, and Myotonic dystrophy, as well as Spongiform encephalopathies, such as Hereditary Creutzfeldt-Jakob disease (due to Prion protein processing defect), Fabry disease,Straussler-Scheinker syndrome, COPD, dry-eye disease, and Sjdgren's disease. 1001631 Accordingly, in another aspect of the present invention, pharmaceutically acceptable compositions are provided, wherein these compositions comprise any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle. In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. [001641 It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof. [001651 As used herein, the term pharmaceuticallyy acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A pharmaceuticallyy acceptable salt" means any non-toxic salt or salt of an ester of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof. As used herein, the term "inhibitorily active metabolite or residue thereof' means that a metabolite or residue thereof is also an inhibitor of an ATP-Binding Cassette Transporters. [001661 Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et aL describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19., incorporated herein by reference. Pharmaceutically acceptable salts of the WO 2009/123896 PCT/US2009/038203 compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2 hydroxy-ethanesulfonate, lactobionate, lactate, laurate. lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphtlialenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pirate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and
N'(C-,.
4 alkyl) 4 salts. This invention also envisions the quaternization of any basic nitrogen containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. [001671 As described above, the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other components) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, - 38 - WO 2009/123896 PCT/US2009/038203 lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. 1001681 Uses of Compounds and Pharmaceutically Acceptable Compositions 1001691 In yet another aspect, the present invention provides a method of treating a condition, disease, or disorder implicated by ABC transporter activity. In certain embodiments, the present invention provides a method of treating a condition, disease, or disorder implicated by a deficiency of ABC transporter activity, the method comprising administering a composition comprising a compound of formula (I) to a subject, preferably a mammal, in need thereof. [001701 In certain preferred embodiments, the present invention provides a method of treating cystic fibrosis, hereditary emphysema (due to al-antitrypsin; non Piz variants), hereditary hemochromatosis, coagulation--fibrinolysis deficiencies, such as protein C deficiency, Type 1 hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type I chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo Hurler, mucopolysaccharidoses (due to lysosomal processing enzymes), Sandhof/T'ay-Sachs (due to p1-hexosaminidase), Crigler-Najjar type II (due to UDP-glucuronyl--sialyc-transferase), polyendocrinopathy/hyperinsulemia, diabetes mellitus (due to insulin receptor), Laron dwarfism (due to growth hormone receptor), myleoperoxidase deficiency, primary hypoparathyroidi sm (due to preproparathyroid hormone), melanoma (due to tyrosinase). The diseases associated with the latter class of ER malfunction are glycanosis CDG type 1, hereditary emphysema (due to Ul - 39 - WO 2009/123896 PCT/US2009/038203 antitrypsin (PiZ variant), congenital hyperthyroidism, osteogenesis imperfecta (due to Type I, II, IV procollagen), hereditary hypofibrinogenemia (due to fibrinogen), ACT deficiency (due to al antichymotrypsin), diabetes insipidus (DI), neurophyseal DI (due to vasopvessin hormone/V2 receptor), neprogenic DI (due to aquaporin II), Charcot-Marie Tooth syndrome (due to peripheral mvelin protein 22), Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease ( due to pAPP and presenilins), Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders such as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluvsian, and myotonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease (due to prion protein processing defect), Fabry disease (due to lysosomal a-galactosidase A), Straussler- Scheinker syndrome, chronic obstructive pulmonary disease (COPD), dry eye disease, and Sj~gren's Syndrome, comprising the step of administering to said mammal an effective amount of a composition comprising a compound of formula (11), or a preferred embodiment thereof as set forth above. [001711 According to an alternative preferred embodiment, the present invention provides a method of treating cystic fibrosis comprising the step of administering to said mammal a composition comprising the step of administering to said mammal an effective amount of a composition comprising a compound of formula (I), or a preferred embodiment thereof as set forth above. 1001721 According to the invention an "effective amount" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of cystic fibrosis, hereditary emphysema (due to al-antitrypsin; non Piz variants), hereditary hemochromatosis, coagulation-fibrinolysis deficiencies, such as protein C deficiency, Type 1 hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type I chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, mucopolysaccharidoses (due to lysosomal processing enzymes), Sandhof/Tay-Sachs (due to p-hexosaminidase), Crigler-Najjar type II (due to UDP glucuronyl-sialyc-transferase), polyendocrinopathy/hyperinsulemia, diabetes mellitus (due to insulin receptor), Laron dwarfism (due to growth hormone receptor), myleoperoxidase deficiency, primary hypoparathyroidism (due to preproparathyroid hormone), melanoma (due to tyrosinase). The diseases associated with the latter class of ER malfunction are glycanosis CDG type 1, hereditary emphysema (due to al-antitrypsin (PiZ variant), congenital hyperthyroidism, osteogenesis imperfecta (due to Type 1, II, IV procollagen), hereditary hypofibrinogenemia (due to - 40 - WO 2009/123896 PCT/US2009/038203 fibrinogen), ACT deficiency (due to al-antichymotrypsin), diabetes insipidus (DI), neurophyseal DI (due to vasopvessin hormone/V2-receptor), neprogenic DI (due to aquaporin II), Charcot-Marie Tooth syndrome (due to peripheral myelin protein 22), Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease ( due to [3APP and presenilins), Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders such as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and notonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease (due to prion protein processing defect), Fabry disease (due to lysosomal a-galactosidase A), Straussler-Scheinker syndrome, chronic obstructive pulmonary disease (COPD), dry eye disease, and Sj~gren's Syndrome. [001731 The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of one or more of cystic fibrosis, hereditary emphysema (due to al-antitrypsin; non Piz variants), hereditary hemochromatosis, coagulation -fibri nolysis deficiencies, such as protein C deficiency, Type I hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type I chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, mucopolysaccharidoses (due to lysosomal processing enzymes), Sandh of/Tay-Sachs (due to P-hexosaminidase), Crigler-Najjar type II (due to UDP glucuronyl-sialye-transferase), polyendocrinopathy/hyperinsulemia, diabetes mellitus (due to insulin receptor), Laron dwarfism (due to growth hormone receptor), myleoperoxidase deficiency, primary hypoparathyroidism (due to preproparathyroid hormone), melanoma (due to tyrosinase). The diseases associated with the latter class of ER malfunction are glycanosis CDG type 1, hereditary emphysema (due to ctl-antitrypsin (PiZ variant), congenital hyperthyroidism, osteogenesis imperfecta (due to Type I, II, IV procollagen), hereditary hypofibrinogenemia (due to fibrinogen), ACT deficiency (due to al-antichymotrypsin), diabetes insipidus (DI), neurophyseal DI (due to vasopvessin hormone/V2-receptor), neprogenic DI (due to aquaporin II), Charcot-Marie Tooth syndrome (due to peripheral myelin protein 22), Perlizaeus-Merzbacher disease, neurodegenerative diseases such as Alzheimer's disease ( due to PAPP and presenilins), Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear play, Pick's disease, several polyglutamine neurological disorders such as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and myotonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease (due to prion protein processing defect), Fabry disease (due to lysosomal a-galactosidase A), Straussler-Scheinker -41 - WO 2009/123896 PCT/US2009/038203 syndrome, chronic obstructive pulmonary disease (COPD), dry eye disease, and Sjgren's Syndrome. 1001741 The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human. 1001751 The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. 1001761 Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyiformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, -42 - WO 2009/123896 PCT/US2009/038203 the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. 1001771 Injectable preparations, for example. sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending inedium. For this purpose any bland fixed oil can be employed including synthetic mono-- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. 1001781 The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. [001791 In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues. [001801 Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. -43 - WO 2009/123896 PCT/US2009/038203 [001811 Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethyicellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar--agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as tale, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents. [001821 Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. 1001831 The active compounds can also be in microencapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a - 44 - WO 2009/123896 PCT/US2009/038203 composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. 1001841 Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms are prepared by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. [001851 As described generally above, the compounds of the invention are useful as modulators of ABC transporters. Thus, without wishing to be bound by any particular theory, the compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder where hyperactivity or inactivity of ABC transporters is implicated in the disease, condition, or disorder. When hyperactivity or inactivity of an ABC transporter is implicated in a particular disease, condition, or disorder, the disease, condition, or disorder may also be referred to as a "ABC transporter-mediated disease, condition or disorder". Accordingly, in another aspect, the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where hyperactivity or inactivity of an ABC transporter is implicated in the disease state. 1001861 The activity of a compound utilized in this invention as a modulator of an ABC transporter may be assayed according to methods described generally in the art and in the Examples herein. [00187] It will also be appreciated that the compounds and pharmaceutically acceptable compositions of the present invention can be employed in combination therapies, that is, the compounds and pharmaceutically acceptable compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the WO 2009/123896 PCT/US2009/038203 desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder (for example, an inventive compound may be administered concurrently with another agent used to treat the same disorder), or they may achieve different effects (e.g., control of any adverse effects). As used herein, additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition, are known as appropriate for the disease, or condition, being treated". [00188] In one embodiment, the additional agent is selected from a mucolytic agent, bronchodialator, an anti-biotic, an anti-infective agent, an anti-inflammatory agent, a CFTR modulator other than a compound of the present invention, or a nutritional agent. [001891 In another embodiment, the additional agent is a compound selected from gentamicin, curcumin, cyclophosphamide, 4-phenylbutyrate, miglustat, felodipine, nimodipine, Philoxin B, geniestein, Apigenin, cAMP/cGMP modulators such as rolipram, sildenafil, milrinone, tadalafil, amrinone, isoproterenol, albuterol, and almeterol, deoxyspergualin, HSP 90 inhibitors, HSP 70 inhibitors, proteosome inhibitors such as epoxomicin, lactacystin, etc. [001901 In another embodiment, the additional agent is a compound disclosed in WO 2004028480, W0 2004110352, WO 2005094374, WO 2005120497, or WO 2006101740. [001911 In another embodiment, the additiona agent is a benzo(c)quinolizinium derivative that exhibits CFTR modulation activity or a benzopyran derivative that exhibits CFTR modulation activity. 1001921 In another embodiment, the additional agent is a compound disclosed in US7202262, US6992096, US20060148864, US20060148863, )S20060035943, ljS20050164973, W02006110483, W02006044456, W02006044682, W02006044505, W02006044503, WO2006044502, or W02004091502. [001931 In another embodiment, the additional agent is a compound disclosed in WO2004080972, W02004111014, W02005035514, W02005049018, W02006002421, WO2006099256, W02006127588, or WO2007044560. 1001941 In another embodiment, the an additional agent selected from compounds disclosed in U.S. Patent Application Serial No. 11/165,818, published as U.S. Published Patent Application No. 2006/0074075, filed June 24, 2005, and hereby incorporated by reference in its entirety. In another embodiment, the additional agent is N-(5-hydroxy-2 ,4-ditert-butyl-phenyi)-4 oxo-iH-quinoline-3-carboxamide. These combinations are useful for treating the diseases - 46 - WO 2009/123896 PCT/US2009/038203 described herein including cystic fibrosis. These combinations are also useful in the kits described herein. 1001951 The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. [001961 The compounds of this invention or pharmaceutically acceptable compositions thereof may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Accordingly, the present invention, in another aspect, includes a composition for coating an implantable device comprising a compound of the present invention as described generally above, and in classes and subclasses herein, and a carrier suitable for coating said implantable device. In still another aspect, the present invention includes an implantable device coated with a composition comprising a compound of the present invention as described generally above, and in classes and subclasses herein, and a carrier suitable for coating said implantable device. Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6.099,562; 5,886,026; and 5,304,121. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition. [001971 Another aspect of the invention relates to modulating ABC transporter activity in a biological sample or a patient (e.g., in vitro or in vivo), which method comprises administering to the patient, or contacting said biological sample with a compound of formula I or a composition comprising said compound. The term "biological sample", as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen., tears, or other body fluids or extracts thereof. [001981 Modulation of ABC transporter activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, the study of ABC transporters in biological and pathological phenomena; and the comparative evaluation of new modulators of ABC transporters. - 47 - WO 2009/123896 PCT/US2009/038203 [001991 In yet another embodiment, a method of modulating activity of an anion channel in vitro or in vivo, is provided comprising the step of contacting said channel with a compound of formula (I). In preferred embodiments, the anion channel is a chloride channel or a bicarbonate channel. In other preferred embodiments, the anion channel is a chloride channel. [00200] According to an alternative embodiment, the present invention provides a method of increasing the number of functional ABC transporters in a membrane of a cell, comprising the step of contacting said cell with a compound of formula (I). The term "functional ABC transporter" as used herein means an ABC transporter that is capable of transport activity. In preferred embodiments, said functional ABC transporter is CFTR. [002011 According to another preferred embodiment, the activity of the ABC transporter is measured by measuring the transmembrane voltage potential. Means for measuring the voltage potential across a membrane in the biological sample may employ any of the known methods in the art, such as optical membrane potential assay or other electrophysiological methods. [002021 The optical membrane potential assay utilizes voltage-sensitive FRET sensors described by Gonzalez and Tsien (Se, Gonzalez, J. E. and R. Y. Tsien (1995) "Voltage sensing by fluorescence resonance energy transfer in single cells" Biophys J 69(4) 1272-80, and Gonzalez, J. E. and R. Y. Tsien (1997) "Improved indicators of cell membrane potential that use fluorescence resonance energy transfer" Chem Biol 4(4): 269-77) in combination with instrumentation for measuring fluorescence changes such as the Voltage/Ion Probe Reader (VIPR) (See, Gonzalez, J. E., K. Oades, et al. (1999) "Cell-based assays and instrumentation for screening ion-channel targets" DgDiscovToday 4(9): 431-439). [002031 These voltage sensitive assays are based on the change in fluorescence resonant energy transfer (FRET) between the membrane-soluble, voltage-sensitive dye, DiSBAC 2 (3), and a fluorescent phospholipid, CC2-DMPE, which is attached to the outer leaflet of the plasma membrane and acts as a FRET donor. Changes in membrane potential (Vm,) cause the negatively charged DiSBAC 2 (3) to redistribute across the plasma membrane and the amount of energy transfer from CC2-DMPE changes accordingly. The changes in fluorescence emission can be monitored using VIPR'Fm II, which is an integrated liquid handler and fluorescent detector designed to conduct cell-based screens in 96- or 384-well microtiter plates. [002041 In another aspect the present invention provides a kit for use in measuring the activity of a ABC transporter or a fragment thereof in a biological sample in vitro or in vivo comprising (i) a composition comprising a compound of formula (I) or any of the above embodiments; and (ii) instructions for a) contacting the composition with the biological sample and -48 - WO 2009/123896 PCT/US2009/038203 b) measuring activity of said ABC transporter or a fragment thereof. In one embodiment, the kit further comprises instructions for a) contacting an additional composition with the biological sample; b) measuring the activity of said ABC transporter or a fragment thereof in the presence of said additional compound, and c) comparing the activity of the ABC transporter in the presence of the additional compound with the density of the ABC transporter in the presence of a composition of formula (I). In preferred embodiments, the kit is used to measure the density of CFTR. [002051 In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner. EXAMPLES 1002061 Preparation of 1-benzo[L131dioxol-5-vi-cyclopropanecarboxylic acid CI B0 O 50% NaOH (aq) o OH [002071 A mixture of 2-(benzo[d][1,3]ldioxol-5-yl)acetonitrile (5.10 g 31.7 mmol), I bromo-2-chloro-ethane (9.00 mL 109 mmol), and benzyltriethylammonium chloride (0.181 g, 0.795 mmol) was heated at 70 'C and then 50% (wt./,wt.) aqueous sodium hydroxide (26 mL) was slowly added to the mixture. The reaction was stirred at 70 'C for 24 hours and then heated at 130 'C for 48 hours. The dark brown reaction mixture was diluted with water (400 mL) and extracted once with an equal volume of ethyl acetate and once with an equal volume of dichloromethane. The basic aqueous solution was acidified with concentrated hydrochloric acid to pI less than one and the precipitate filtered and washed with 1 M hydrochloric acid. The solid material was dissolved in dichloromethane (400 n.) and extracted twice with equal volumes of I M hydrochloric acid and once with a saturated aqueous solution of sodium chloride. The organic solution was dried over sodium sulfate and evaporated to dryness to give a white to slightly off-white solid (5.23 g, 80%) ESI-MS n/z caic. 206.1, found 207.1 (M+1Y. Retention time 2.37 minutes. 'H NMR (400 MHz, DMSO-d 6 ) 6 1.07-1.11 (m, 2H), 1.38-1.42 (m, 2H), 5.98 (s, 2H), 6.79 (in, 2H)1, 6.88 (im., 1H). 12.26 (s, 1-I). [002081 Preparation of 1-(2,2-difluoro-benzo[1,3dioxol-5-cv)-celonro !anecarboxylic acid -49- WO 2009/123896 PCT/US2009/038203 F O Pr Pd 1
JPF'
3
)
4 F CO 2 Me LiA!H 4 F *OH __ _ F 1 ! CO/CH30H F O- F O C; NaC:N CICHCH 2 Br C GN NaOH F CXH -NaGH F: 0 [002091 Step a: 2,2-Difluoro-benzo[1,3]dioxole-5-carboxylic acid methyl ester [002101 A solution of 5-bromo-2,2-difluoro-benzo[1,3]dioxole (11.8 g, 50.0 mmol) and tetrakis(triphenviphosphine)paliadium (0) [Pd(PPh3)4, 5.78 g, 5.00 mmol] in methanol (20 nL) containing acetonitrile (30 mL) and triethylamine (10 mL) was stirred under a carbon monoxide atmosphere (55 PSI) at 75 C (oil bath temperature) for 15 hours. The cooled reaction mixture was filtered and the filtrate was evaporated to dryness. The residue was purified by silica gel column chromatography to give crude 2,2-difluoro-benzo [1,3] dioxole-5-carboxylic acid methyl ester (11.5 g), which was used directly in the next step. [002111 Step b: (2,2-Difluoro-benzo[1,31dioxol-5-yl)-methanol [002121 Crude 2,2-difluoro-benzo1,3 ldioxole-5-carboxylic acid methyl ester (11.5 g) dissolved in 20 mitL of anhydrous tetrahydrofuran (THF) was slowly added to a suspension of lithium aluminum hydride (4.10 g, 106 mmol) in anhydrous THF (100 mL) at 0 C. The mixture was then warmed to room temperature. After being stirred at room temperature for 1 hour, the reaction mixture was cooled to 0 "C and treated with water (4.1 g), followed by sodium hydroxide (10% aqueous solution, 4.1 mL). The resulting slurry was filtered and washed with THF. The combined filtrate was evaporated to dryness and the residue was purified by silica gel column chromatography to give (2,2-difluoro-benzo[1,3]dioxol-5-vl)-methanol (7.2 g, 38 mmol, 76 % over two steps) as a colorless oil. 1002131 Step c: 5-Chloromethyl-2,2-difluoro-b enzo[1,3]dioxole 1002141 Thionyl chloride (45 g, 38 mmol) was slowly added to a solution of (2,2-difluoro benzo[ 1,3ldioxol-5-yl)-methanol (7.2 g, 38 mmol) in dichloromethane (200 mL) at 0 C. The resulting mixture was stirred overnight at room temperature and then evaporated to dryness. The residue was partitioned between an aqueous solution of saturated sodium bicarbonate (100 mL) and - 50- WO 2009/123896 PCT/US2009/038203 dichloromethane (100 mL). The separated aqueous layer was extracted with dichloromethane (150 mL) and the organic layer was dried over sodium sulfate, filtrated, and evaporated to dryness to give crude 5-chloromethyl-2,2--difluoro-benzo[1,3]dioxole (4.4 g) which was used directly in the next step. [002151 Step d: (2,2-Difluoro-beiizo[1,3]dioxol-5-yl)-acetonitrile [002161 A mixture of crude 5-chloromethyl-2,2-difluoro-benzo[1,3]dioxole (4.4 g) and sodium cyanide (1.36 g, 27.8 mmol) in dimethylsulfoxide (50 mL) was stirred at room temperature overnight. The reaction mixture was poured into ice and extracted with ethyl acetate (300 mL). The organic layer was dried over sodium sulfate and evaporated to dryness to give crude (2,2 difluoro-benzo[1,3]dioxol-5-yl)-acetonitrile (3.3 g) which was used directly in the next step. 1002171 Step e: 1-(2,2-Difluoro-benzo[1,3]dioxol-5-yI)-cyclopropanecarbonitrile 1002181 Sodium hydroxide (50% aqueous solution, 10 mL) was slowly added to a mixture of crude (2,2-di fluoro-benzo[1,3]dioxol-5-yl)-acetonitrile, benzyltriethylammonium chloride (3.00 g, 15.3 mnol), and 1-bromo-2-chloroethane (4.9 g, 38 mmol) at 70 C. The mixture was stirred overnight at 70 'C before the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and evaporated to dryness to give crude 1-(2,2-difluoro-benzo[1,3]dioxol-5-yl)-cyclopropanecarbonitrile, which was used directly in the next step. 1002191 Step f: 1-(2,2-Difltoro-benzo[1,31dioxol-5-yl)-cyclopropanecarboxylic acid 1002201 1-(2,2-Difluoro-benzo[1,3]ldioxol-5-yl)-cyclopropanecarbonitrile (crude from the last step) was refluxed in 10% aqueous sodium hydroxide (50 mL) for 2.5 hours. The cooled reaction mixture was washed with ether (100 nL) and the aqueous phase was acidified to pH 2 with 2M hydrochloric acid. The precipitated solid was filtered to give I-(2,2-difluoro-benzo[i.3]dioxol 5-yl)-cyclopropanecarboxylic acid as a white solid (0.15 g, 1.6% over four steps). ESI-MS m/z calc. 242.04, found 241.58 (M+1.; 1H NIR (CDCl 3 ) 6 7.14-7.04 (in, 211), 6.98-6.96 (m, 11H), 1.74 1.64 (m, 2H). 1.26-1.08 (m, 2H). 1002211 The following Table 2 contains a list of carboxylic acid building blocks that were commercially available, or prepared by the methods described above. [002221 Table 2: Carboxylic acid building blocks. Name Structure -51- WO 2009/123896 PCT/US20091'038203 carboxylic acid / 0 1 (3 methoxyphertyicyciopropane-1- Hy ~ carboxviic acid ~~ 1 (4- nithoxyphecnybcyciopropan c-I -OH carboxylic acid -~'0 (trifluiorornetlhcxy)pheInvl cyclopropaie.- 1- 0N carboxylic acid 03> !H 0 Hx I I-phecnyclopropane- I -carboxylic acid 1 (4-cliIorophecnyi)cyclopr-opane- I 1 -OH ca-boxylic acid 0 1 O3H4 dirnctlioxyphenivi'cvclopropanecarboxylic 'N ~ 0 acid 0 0 acid o OH I p etliycl ~hjy opropinecarboxylic i - 52 0 WO 2009/123896 PCT/US2009/038203 [002231 Preparation of (6-aminopyridin-3-vl)(2-methoxvphenyl)methanone yOMe
CO
2 H 1) CICO 2 1Bu/TEA Br C N H.HCI C N O1 n-BuLi N2) NN 0 OMeO OMeO
NH
3 /EtOH N CI N NH 2 [002241 Step a: 6-chloro-N-methoxy-N-methyl-nicotinamide [002251 To a solution of 6-chloro-nicotinic acid (94.5 g, 0.6 mol) in dichloromethane (1000 mL) was added N-methyl morpholine (181.8 g, 1.8 mol) followed by iso-butyl chloroformate (81.9 g, 0.6 mol) at -25 C under N 2 atmosphere. The mixture was stirred for 15 minutes and then 0,N-dimethyl hydroxylamine hydrochloride (64.35 g, 0.66 mol) was added. The mixture was stirred for 30 min at -25 C and warmed to room temperature slowly. The mixture was stirred for an additional hour at room temperature and a saturated solution of NaHC0 3 (800 mL) was added. The separated aqueous layer was extracted with dichloromethane (500 mL x 3). The combined organic layers were washed with water and brine. The solvent was evaporated to give the crude product, which was purified by column chromatography to give the pure product 6-chloro-N methoxy-N-methyl-nicotinamide as a white solid (56 g, 46.6%). [002261 Step b: (6-chloropyridin-3-yl)-(2-methoxyphenyl)-methanone [002271 To a solution of 1-bromo-2-methoxy-benzene (49.2 g, 0.263 mol) in dry THF (500 mL) was added dropwise n-BuLi (2.5 M in hexane, 105 mL, 0.263 mol) at -78 'C under N 2 atmosphere. The mixture was stirred at this temperature for 1 hr and then a solution of 6-chloro-N methoxy-N-methyl-nicotinamide (50 g, 0.25 mol) in THF (100 mL) was added dropwise. The reaction mixture was warmed to room temperature, stirred overnight and then was quenched with a saturated solution of NH 4 Cl (300 mL). The separated aqueous layer was extracted with ethyl acetate (3 X 300 mL). The combined organic layers were washed with brine, dried over Na2SO 4 , evaporated in vacuo to give the crude product that was purified by column chromatography to yield (6-chloropyridin-3-yl)-( 2 -methoxyphenyl)-methanone as a white solid (31 g, 47.6 %). [002281 Step c: (6-Amino-pyridin-3-yl)-(2-methoxyphenyl)-methanone - 53 - WO 2009/123896 PCT/US2009/038203 [002291 A solution of 6-chloro-pyridin-3-yl)-(2-methoxy-phenyl)-methanone (30 g, 0.121 mol) in ethanol (150 ml) and anhydrous ammonia (100 ml) was placed in a bomb and heated atl 45 'C for 16 hours. The solvents were removed in vacuo and the residue was purified by column chromatography to give (6-Amino-pyridin-3-yl)-(2-methoxyphenyl)-methanone (18 g, 65.2%). 'H NMR (DMSO-d6): 6: 8.12 (s, 1 H,), 7.68 (d, J= 8.8 Hz, 1 H), 7.47 (t, J= 7.2, 1 1), 7.21 (d, J=- 7.2, 1 H), 7.13 (d, J= 7.2, 1 H), 7.03 (d, 1 H), 6.96 (br, 2 H), 6.44 (d, J= 8.8, 1 H), 3.69 (s, 3 H). MS (ESI) m-/e- (M+-H-) 229.27. 1002301 Preparation of 5-(2-mnethoxybenzvl)pvridin-2-anine OMe O OMe OH Me NaBH 4 /MEOH EtSHITFA * I | NN NH 1002311 Step a: (6-aninopyridin-3-yl)-(2-methoxyphenvl)-nethanol 1002321 To a solution of (6-amino-pyridin-3-yl)-(2-methoxy-phenyl)-metianone (14 g, 61.4 mol) in CH 3 0H (150 mL) was added NaBH 4 (3.48 g, 92.1 mmol) portion-wise at 0 C. After the addition was completed, the reaction was warmed to room temperature and stirred at room temperature for 1 hr. The solvent was removed in vacuo and water (100 mL) was added to the residue. The mixture was extracted with dichloromethane (100 mL X 3). The combined organic layers were washed with saturated solution of NaIC0 3 and brine, dried over Na 2 SO4, filtered and concentrated in vacuo to give a crude product that was used directly in the next step with no further purification. [002331 Step b: 5-(2-methoxy-benzyl)-pyridin-2-ylanine [002341 A mixture of the crude compound from previous step, Et 3 SiH (57 g, 491 mmol) and TFA (112 g, 982 mmol) in CH 2 C2 (150 mL) was stirred at room temperature overnight. The excess Et3SiH, TFA and CH2Ci 2 were removed under reduced pressure. The residue was stirred in water; the solid precipitate was filtered, washed with diethyl ether and petroleum ether to give 5-(2 methoxv-benzvl)-pyridin-2-ylamine as TFA salt. (5 g, 24.8% fbr 2 steps). H1-1 NMR (DMSO-d 6 ): 6 13.5 (br, 1 H), 7.93 (brs, 2 H), 7.75-7.73 (m, 2 H), 7.22 (t, J= 8.0 Hz, 1 H), 7.19 (d, J= 7.2 Hz, I H), 6.98 (d. J= 8.0 Hz,I H), 6.90-6.86 (m. 2 H), 3.77 (s, 2 H). 3.76 (s. 3 H). MS (ESI) n/c (M+H-) 215.27. [002351 Preparation of 1-(benzodl[L31 dioxol-5-vl)-N-(pyridin-2 yl)cycopropanecarboxam ide -54 - WO 2009/123896 PCT/US2009/038203 1) SOC1 2 H DMF N OH 2) pyridine O O 0 0 N
H
2 N N 1002361 1-Benzo[i.3]dioxol-.5-yl-cyclopropanecarboxylic acid (41 mg, 0.20 mmol) was placed in an oven-dried flask under nitrogen. Thionyl chloride (0.3 nL) and N,A dintethylformamide (0.03 mL.) were added and the solution was allowed to stir for 10 minutes at room temperature. The excess thionyl chloride was removed under vacuum and the resulting solid was suspended in anhydrous pyridine (1 mL). This solution was then slowly added to a solution of pyridin-2-amine (19 mg, 0.20 mmol) in anhydrous pyridine (1 mL). The resulting mixture was allowed to stir for 15 hours at 110 C. The crude product was then filtered, evaporated to dryness, dissolved in N,N-dimethylformamide (1 mL) and purified by reverse-phase preparative liquid chromatography utilizing a gradient of 0-99% acetonitrile in water containing 0.05% trifluoroacetic acid to yield the pure product (5.9 mg, 0.021 mmol, 10%). ESI-MS in/z caic. 282.1, found 283.1 (M+-1). Retention time 2.13 minutes. [002371 Preparation of 1-(2,2-difluorobenzo[d [1,31dioxo-5-vl)-iN-(5-methlpyridin.
2-yl)cyclopropanecarboxamide 1) SOCl 2 --- DMF H OH 2) pyridine F a F a 0
H
2 N N [002381 1 -(2,2-Difluorobenzo[d][1 ,3]dioxol-5-yl)cyclopropanecarboxylic acid (2.32 g, 9.58 mmol) was placed in an oven-dried flask under nitrogen. Thionyl chloride (3 mL) and AN dimethylformamide (0.3 mL) were added and the solution was allowed to stir for 30 minutes at room temperature. The excess thionyl chloride was removed under vacuum and the resulting solid was suspended in anhydrous dichloromethane (10 mL). This solution was then slowly added to a solution 5-methylpyridin-2-amine (0.798 g, 7.38 mmol) in anhydrous dichloromethane (10 mL) containing triethylamine (4.11 mL, 29.5 nmmol). The resulting mixture was allowed to stir for 15 hours at room temperature. The crude product was then washed two times with a IM aqueous solution of hydrochloric acid, two times with a saturated aqueous solution of sodium bicarbonate, and finally two times with a saturated aqueous solution of sodium chloride. The organic layer was dried over sodium sulfate, evaporated to near dryness, and then purified on 120 g of silica gel - 55 - WO 2009/123896 PCT/US2009/038203 utilizing a gradient of 0-30% ethyl acetate in hexanes to yield the pure product (1.20 g, 3.60 mmol, 48.7%). ESI-MS m/z calc. 332.1, found 333.1 (M+1) Retention time 1.48 minutes. 1002391 The compounds listed in Table 3 below were made in a manner analogous to that described above from the appropriate reactants: [002401 Table 3. Naie Structure V7 H (benzo[d[1,3]dioxol-5-yl)-N-(6- N7 NH O,--N0 imethylpyridin-2 yl)cyclopropanecarboxamide 1-(benzo[d][1,3idioxol-5-yl)-N-(5- H methylpyridin-2- N yi)cyclopropanecarboxamide I-(benzo[d]i 1,3]idioxol-5-yl)-N-(5- H cyanopyridin-2- N yl)cyclopropanecarboxamide HN 1-(benzo[d]i1,3]dioxol-5-yl)-N-(4- H ii N N ethylpyridin-2 yl)cyclopropanecarboxamide 1-(benzo[d][1,3idioxol-5-yl)-N-(4- H N N methylpyridin-2 yilccopropanecarboxamide 0 ~ 0 6-(1-(benzod[i,3]dioxol-5- H ylcy clopropanecarboxamido)nicotinamide N N~ 0O-O NH 2 0 1-(ez ]1,3]idioxol-5-yl)-N-(5- H (trifluoromethyl)pyridin-2- N~ F yl)cyclopropanecarboxamideC - 56 - WVO 2009/123896 PCT/US20091'038203 1bczojd]F1,3dioxo[-5--l)Nx ( H mn thylpyridii-2-K 0 y i)ccopropanecarboxamide0 (benzo[d_ , doo5yI~T6 H ethyipyridiin-2-N x ~cx ciopropanecarboxamnide0 1 (bejizo~di[13doo--l--5 fluoiopyridirt-2-K 0 l" N yI)&ccpropanecarboxamide 0 F N5 - (2 - reth oxyb enzy i~pyridi n- 2-y1) -I H rn mtloxyplinyl)cylopropaniecarboxamiide -~ 0 N-(5-(3-iniethoxybenizyI)pyr-idini-2-yi)-i- 0 0 (2 N N I -i oxyphe-nyi)cyclopropai ecarboxar-nide 0 N y -(3(benzyioxy)pyr-idini-2-yl)-i1-
H
phi-ni cyclopropanecarboxar-nideN 1 (4- metloxyphenyl)-N-(5 -methiyipyridin- H N N ) v~cyciopropan 'ccar-boxarnide 0 ilthcnvicyclopropanecar-boxarnideN Cr 0 i-{4benz ll1,3 lix l5yl----3 H nzodiN N (benzyioxy~pyridin-2 yi)cyciopropanecarboxarnide - 57 - WO 2009/123896 PCT/US2009/038203 N-(4-ethylpyridin-2-yi)-1-(4- H N N methoxyphenyl)cyclopropanecarboxamide 0 X 0 * 1-(4-methoxyphenyl)-N-(6-methylpyridin- H N N 2 -vl)cyclopropane'carboxamide N 0 ~ 0 I N-(6-ethlvpyridin-2-yl)-1-(4- H 1-(benzo[d][1,3]dioxol--5-yl)-N-(3-ethyi-6- H N N methylpyridin-2 yl)cyclopropanecarboxamide I -(benzo[d] [1,3]dioxol-5-yl)-N-(5- H fluoropyridin -2- (H N N ylbcyclopropanecarboxamide 1-(benzo[d][i,3]dioxol-5-yl)-N(5-N 7 N N methylpyridin-2 yl)cyclopropanecarboxamide [002411 Preparation of N-(5-(2-methoxybenzoyllvridin2yl)-1-(4 meth oxyphenyl)cyclopropanecarboxamide 0 0 H OH HATU LNI 0 2 DMF, Et 3 N 0 O1 OH2N D 1002421 1-(4-Methoxyphenyl)cyclopropanecarboxylic acid (19.2 mg, 0.100 mmol) and (6 aminopyridin-3-yl)(2-methoxvphenyi)methanone (22.8 g, 0.100 mmol) were dissolved in N,-AT dintethylforniamide (DMF, 0.7 ml) containing triethylamine (0.042 mL, 0.300 mmol). 0-(7 Azabenzotriazol---yl)-N,,N',N'-tetramethyluronium hexafluorophosphate (HATU., 38.0 mg, 0.100 mmol) was added and the solution was allowed to stir for 72 hours at 70 'C. The mixture was then purified by reverse phase preparative liquid chromatography utilizing a gradient of 0-99% - 58 - WO 2009/123896 PCT/US2009/038203 acetonitrile in water containing 0.05% trifluoroacetic acid to yield the pure product (4.2 mg, 0.010 nmmol, 10%). ESI-MS m/z caic. 402.2, found 403.5 (M+1) Retention time 3.50 minutes. 1002431 The compounds listed in Table 4 below were made in a manner analogous to that of (5 -(21-ethox ybenzoyl)pyridin-2-yl)-1-(4-methoxyphenylcyclopropanecarboxamide from the appropriate reactants: [002441 Table 4. Name Structure NI -(5-(2-methoxvbenzvl)pyridin-2-yl)-I-(4- H N N 0 methoxyphenyl)cvclopropanecarboxamide N 00 I -(benzo[d] [1,3]dioxol-5-yl)-N-(5-(2- H Nv~~ methoxybenzyl)pyridin-2- N ylcyclopropanecarboxamide 0 011 I 1-(beinzo[d][1,3dioxol-5-l)N-(5-(2- 0 methoxybenzoyl)pyridin-2- 0 ylcyclopropanecarboxamide N N H N-(5-(2-methoxybenzoyl)pyridin-2-yl)-i-(4 methoxyphenvl)cvclopropanecarboxamide MeO N N H (-ho enyi)N-(5-(2-moxybenzylp)pridin-2-v -p toylcyclopropanecarboxamide 0 N N I I chitoropheniyi)-N-(5 -(2-meith-oxyben ny7lpyridin- 0 N N H - 59 - WO 2009/123896 PCT/US20091'038203 1--(3,4.-dirnethoxypheniyl).-N-(5 -(2-- 0 rn mthoxybenzvi)pyridin-2- N )ylyclopropanecai-boxanlde H I (3-niethoxyphenyl)-N-(5-(2 0 methoxybenzvi)pyridin-2-)o yI)&ccpropanecarboxamide -o N N 0H N-5 (-methoxvbenzvi)pyricin-2'--yl)- 1--(4--o (tifuronehxypenyl)cyclopropanecarboxamide F 3 0 ~N. N N H N-'(.i5-(2-methoxybetizoyi)pyr-idin-2 -ylp- I- 0o i lthcnvicyclopropanecar-boxarnide - o N,. 0N N' H -. N .(5--(2--etlionxybenzoyI)pyridin--2--y)- I-p- 0 o0 toly ley clopropan ecarboxarnide x o N:. N N H I (I -4c]hiorophcnyi>N-(5-(2- 0 methoxybenzoyi)pyridin--2- C H I ( " -cifluorob-enzo [dl [1,31 dioxo-5 -Ni)-.N-(5-(2- 0 methoxybenzoyi)pyridln-2- FO x . N x bcyciopropanccarboxarnide N0 1 (3,4-dim-eth oxyphei y]Y-N-(5 -(2-0 0 1m- tbcoxybenzoyh1,pyridin--2- -0 . ,N yl )cyclopropaiiecarboxamiide 0- N N N H - 60 - WO 2009/123896 PCT/US20091'038203 N.-(5-('2--metioxybenzoyI)pyridin--2--y)-i-(2- 0 0 rn tboxy lienyl)cyclopropaniecarboxamide -P 0 N N H N-(5 (2'-nethoxybenizoyI)pyridii-2 -yb-i -(4- 0 (thyithllio)pleiybcYciopropainecarboxarnide N N N-(5-(2-r-nethoxyben-zoyi~pyr-idini-2-yi)-i-(4-9 9 (tiluorometlioxy)pheniyi)cvclopropanecarboxamide- 0 0 N N H [002451 Preparation of 3-(2-(1-(2, -difluorobenzo Id] IE31 dioxol-5 !Lcycloplroanecarboxanido)5=rinethylpyI i==Ihnoc acid) CH; Pd(PPh 3 ) N% ~( ~HO' BI o~ 0K 2 K03 0 J N
H
2 0 2 0-PTSCI
CH
3 ReO 3 N "'0 prd 0H 2 C1 2 I 2-aminoethanol N 0 0 H 2 1N 111 0
FX
0 I a F 0 F I-A0 F " 00 N 01OH H O [0)02461 Step a: tert-butyl 3-(3-mnethylpyridin-4-y1)beiizoate [0)02471 4-Chioro-3-rnethyipyridine (2100 g,15.7 rnmoi), 3-(et butoxycarbonyilplienyiboronic acid (5.42 g, 24.4. mmoi), a 2M I aqueous solution of potassium - 61 - WO 2009/123896 PCT/US2009/038203 carbonate (31.4 niL, 62.8 mrol), and palladium tetrakis(triphenylphosphine) (Pd(PPh 3
)
4 , 0.906 g, 0.784 mmol) were suspended in 1,2-dimethoxyethane (DME, 150 mL). The resulting mixture was stirred and heated to 80 'C for 60 hours. The crude reaction mixture was cooled to room temperature and then the layers were separated. The organic layer was evaporated to dryness and then purified on 120 g of silica gel utilizing a gradient of 0-70% ethyl acetate in hexanes to yield the pure product as a pale yellow oil (3.02 g, 1 .2 mmol, 71.4%). [00248] Step b: 4-(3-(tert-Butoxvcarbonyl)phenyl)-3-nethylpyridine 1-oxide [00249] tert-Butyl 3-(3-rnethylpyridin-4-yl)benzoate (0.500 g, 1.86 mmol) was dissolved in a mixture of dichloromethane (0.800 mL) and 30% hydrogen peroxide (0.421 mL). Methyltrioxorhenium (V11) (2.3 mg, 0.92 mmol) was added and the reaction mixture was stirred vigorously for 5 hours. The layers were then separated and the organic layer was treated with sodium sulfite, and then dried over sodium sulfate. The crude product filtered, evaporated to dryness, and used without further purification. ESI-MS m/z calc. 285. 1, found 286.1 (M+i). Retention time 1.22 minutes. [00250] Step c: tert-Butyl 3-(2-aniiio-5-methylpyridin-4-yl)benzoate [00251] 4-(3-(tert-Butoxycarbonyl)pienyl)-3-methylpyridine I -oxide (0.467 g, 1.64 mmol) was dissolved in a mixture of pyridine (0.53 mL) and acetonitrile (15 mL) under an atmosphere of argon. 4-Methylbenzene-i-sulfonyl chloride (0.406 g, 2.13 mmol) was added and the reaction mixture was stirred at 75 'C for 72 hours. Ethanolamine (7 mL) was then added and the reaction mixture was allowed to stir for 5 minutes at room temperature. The crude product was partitioned between dichloromethane and a saturated aqueous solution of sodium bicarbonate. The layers were separated and the organic layer was washed with a saturated aqueous solution of sodium chloride. The organic layer was dried over sodium sulfate and then purified on 40 g of silca. gel utilizing a gradient of 0-100% ethyl acetate in hexanes to yield the pure product (0. 169 g, 0.594 mmol, 36.3%). ESI-MS m/z cale. 284.2, found 285.1 (M+1) Retention time of 1.28 minutes. ifH NMR (400.0 MHz, DMSO-d 6 ) 7.94 - 7.91 (m, 11H), 7.84 (s, 11H), 7.79 (d, J = 1.7 Hz, 1Hf), 7.62 7.56 (m, 2H), 6.33 (s, 1H), 5.78 (s, 2H), 1.99 (s, 3H), 1.55 (s, 9H). [00252] Step d: tert-Butyl 3-(2-(1-(2,2-difluorobeizo[d][1,3]dioxo-5 yl)cyclopropanecarboxamido)-5-nethylpyridin-4-yl)benzoate [002531 1 -(2,2-Difluorobenzo[d][1 ,3]dioxol-5-yl)cyclopropanecarboxylic acid (144 mg, 0.593 mmol) was placed in an oven-dried flask under nitrogen. Thionyl chloride (1 mL) and NN dimethylformamide (0.1 mL) were added and the solution was allowed to stir for 30 minutes at - 62 - WO 2009/123896 PCT/US2009/038203 room temperature. The excess thionyl chloride was removed under vacuum and the resulting solid was suspended in 2 mL of anhydrous dichloromethane. This solution was then slowly added to a solution of tert-butyl 3-(2-amino-5-methylpyridin-4-yi)benzoate (129 mg, 0.454 mmol) in 5 mL of anhydrous dichloromethane containing triethylamine (0.165 mL, 1.19 mmol). The resulting mixture was allowed to stir for 15 hours at room temperature. The crude product was evaporated to dryness and then purified on 12 g of silica gel utilizing a gradient of 0-40% ethyl acetate in hexanes to yield the pure product as a yellow solid (162 mg, 0.319 mmol, 70.3%). ESI-MS n/ calc. 508.2, found; 509.1 (M+1)> Retention time 2.22 minutes. H NMR (400 MHz, DMSO-d 6 ) 6 8.85 (s, 1H), 8.18 (s, 1H), 7.98 - 7.96 (m, iH), 7.89 (s, IH), 7.82 (d, J = 1.6 Hz, 1H), 7.64 - 7.62 (1, 2HE), 7.57 (d, J = 1.6 Hz, 1H). 7.42 (d, J = 8.3 Hz, 1H), 7.34 (dd, J = 1.7, 8.3 Hz, 1H), 2.14 (s., 3H). 1.55 (s., 9H). 1.51 - 1.49 (m, 21H) 1.19 - 1.15 (m, 2H). [002541 Step e: 3-(2-(1-(2,2-Difluorobeiizo[d][1,3]dioxo-5 yl)cyclopropanecarboxamido)-5-nethylpyridin-4-yl)benzoic acid [002551 tert-Butyl 3 -(2-( 1-.(2,2-difluorobenzoid ][1,3]dioxol-5 yl)cyclopropanecarboxamido)-5-inethylpyridin-4-yl)benzoate (40. mg, 0.079 minol) was dissolved in dichloromethane (1 mL). Trifluoroacetic acid (0.500 nL) was added and the reaction mixture was stirred at room temperature for 1 hour. The crude reaction mixture was evaporated to dryness to yield the pure product. ESI-MS m/z cale. 452.1, found; 453.0 (M+i) Retention time 1.64 minutes. 11- NMR (400 MHz, DMSO-dr) 6 1H NMR (400.0 MHz, DMSO-d6) d 9.01 (s, 1H), 8.22 (s, 1H), 8.03 - 8.00 (,in 1H), 7.89 (d, J = 2.5 Hz, 2H), 7.68 - 7.64 (m, 2H), 7.58 (d. J = 1.6 Hz, 1H), 7.43 (d, J = 8.3 Hz. 1H). 7.35 (dd, J= 1.7, 8.3 Hz. 1H), 2.18 (s, 3H), 1.54 - 1.51 (m, 2H), 1.21.- 1.17 (m, 21-1). [002561 Preparation of 1-(benzo[dl[1,31dioxol-5-vl)-N-(6-(evcloh exvlmeth Vllpyridin 2-vi)cylevelopanecarboxamide 1 SOC:B 2 , MFr ZnB1 2 . Iyr e --- --- -- -- --- -- 0n< N' N Br P(dPPT)C:' 2 , VHF 0 N N "~ 'J-I
H
2 N N Br 1002571 Step a: 1-(Benzo Id]11,31 dioxol-5-yI)-N-(6-bromopyridin-2 yl)cyclopropanecarboxamide -63 - WO 2009/123896 PCT/US2009/038203 [002581 To 1-(benzo[d][1,3]dioxol-5-yl)cyclopropanecarboxylic acid (2.4 g, 11.6 mmol) in thionyl chloride (2.5 nl, 34.7 nmol) was added N,N-dinethylformamide (0.1 mL). The reaction mixture was stirred at room temperature for 30 minutes before it was evaporated to dryness to yield the acid chloride. [002591 To 6-bromopyridin-2-amine (2.0 g, 11.6 nmol) in pyridine (10 ml,) was added the acid chloride. The reaction was heated to 100 'C for 12 hours. The reaction was diluted with dichloromethane (30 mL) and washed with I N NaOH (3 x 20 niL). The organics were dried over Na2SO4 and evaporated to dryness. The crude material was purified by silica gel chromatography elutingg with 0-50% ethyl acetate in hexanes) to yield the product (2.9g, 8.0 mmol, 70%). ESI-MS n/z cale. 360.01, found 361.1 (M-1). Retention time 3.59 minutes. [002601 Step b: 1 -(Benzo[d] [1,3]dioxol-5-yI)-N-(6-(cyclohexylmethyl)pyridin-2 yl)cyclopropanecarboxamide [002611 To a 0.5 M solution of (cyclohexylmethyl)zinc(II) bromide in THF (0.8 nL, 0.4 nimol) was added [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium(Il) (8 mg, 0.01 nimol) and the reaction was stirred under nitrogen for 20 minutes. 1-(Benzold[,3]dioxol-5-yl)-N-(6 bromopyridin-2-yl)cvclopropanecarboxamide (36 mg, 0.1 mmol) was added and the reaction was irradiated in the microwave for 10 minutes at 150 'C. The reaction was quenched with a saturated ammonium chloride solution (2 nL) and a saturated ethylenediamine tetraacetic acid disodium salt solution (2 mL). The mixture was stirred for 30 minutes before it was extracted with dichloromethane (3 x 4 mL). The organics were dried over Na 2
SO
4 and evaporated. The crude product was dissolved in DMSO (1 mL) and purified by reverse-phase preparative liquid chromatography utilizing a gradient of 0-99% acetonitrile in water containing 0.05% trifluoracetic acid to yield the pure product (20 mg, 0.05 mmol, 50%). ESI-MS m/z calc. 378.19, found 379.1 (M+-1); retention time 3.55 minutes. [002621 Preparation of 1-(benzo[dl[1,31dioxol-5-yl)-NV-('6-isobutylpyridin-2 yl)cyclopropanecarboxamide
K
0 0 0 : N N H [002631 1-(Benzo [d] [1,3]idioxol-5 -yl)-N-(6-isobutylpyridin-2 yl)cyclopropanecarboxaniide was prepared from I -(benzo [d] [ 1,3 ] dioxol-5-yl)-V-(6-bromopyridin 2-yl)cyclopropanecarboxamide and isobutylzinc(ll) chloride in a manner analogous to that of 1 (benzo[d][i,3]dioxol-5-.yl)-N-(6-.(cyclohexylmethyl)pyridin-2-yl)cyclopropanecarboxamide. -64 - WO 2009/123896 PCT/US2009/038203 [002641 Preparation of N-(6-(beiizylanino)-5-nethylpyridin-2-yl)-1-(2,2 difluorobenzoldl13l1dioxol-5-y)velopropanecarboxamide F 1.SOd- 2 , DMF - 0) P-> 2N~r. 2. ,N, DCI XIF OH N CI - N N N . OH
-
H2N
H
2 N N:' I CI 1 [002651 Step a: N-(6-chloro-5-methylpyridin-2-yl)-1-(2,2-difluorobenzo[d][1,3]dioxol 5-yl)cyclopropanecarboxamide 1002661 To 1-(2 ,2-difluorobenzo[d] [1,3]dioxol-5-yl)cyclopropanecarboxylic acid (18.8 g, 78 mmol) in thionyl chloride (17 mL, 233 mmol) was added NN-dimethylformamide (0.2 mL, 2.6 mmol). The reaction mixture was stirred at room temperature for two hours. Excess thionyl chloride and N,N-dimethlviformrnamide were removed in vacuo and the resulting acid chloride was used directly in next step. [002671 To a solution of 6-chloro-5-methlipyridin-2-amine (11.1 g, 78 nmol) and triethylamine (22.0 niL, 156 mmol) in dichloromethane (100 mL) was added a solution of 1-(2,2 difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarbonyI chloride (20.3 g, 78 mmol) in dichloromethane (50 mL). The resulting reaction mixture was allowed to stir at room temperature for eighteen hours. The reaction mixture was then washed with IN aqueous NaO-1 (2 x 200 mL), 1 N aqueous HCI (1 x 200 niL) and saturated aqueous NaHC0 3 (1 x 200 nL). The organics were dried over sodium sulfate and evaporated to yield the product (26.9 g, 73.3 mmol, 94% 2--step). ESI-MS n calc. 366.06, found 367.3(M1-1). Retention time 2.19 minutes. 1- NMR (400 MHz, DMSO-d 6 ) 6 9.30 (s, 11H), 7.89-7.87 (in, H-1), 7.78-7.76 (m, 111), 7.54-7.53 (n, 1i), 7.41-7.39 (m, 1H), 7.33-7.30 (in, 1H), 2.26 (s, 3H), 1.52-1.49 (n, 2H), 1.19-1.16 (in, 2H). 1002681 Step b. N-(6-(benzylamino)-5-methylpyridin-2-yl)-1-(2,2 difluorobeiizo[d[11,3]dioxol-5-y l)cyclopropanecarboxamide [002691 To benzylamine (0.5 nL, 4.6 niiol) was added N-(6-chlioro-5-methylpyridin-2 yl)-I -(2,2-difliuorobeinzo[d] [1,3]dioxol-5-yl)cyclopropanecarboxamide (37 ng, 0. 1 mmol) and the reaction mixture was irradiated in the microwave at 170 'C for 60 minutes. The crude product purified by reverse-phase preparative liquid chromatography utilizing a gradient of 0-99% acetonitrile in water containing 0.05% trifluoracetic acid to yield the pure product. ESI-MS mlz calc. 437.4, found 438.3 (M+i1); retention time 1.83 minutes. -65 - WO 2009/123896 PCT/US2009/038203 [002701 Preparation of 1-(benzo[dl[1,31dioxol-5-vl)--N(6-(2 methoxyphenylanin o)pyridin-2-vIlevelopropanecarboxamide 0 0N B r -NH 2 N H N r (DPPF) 2 PdC2.CH 2
C
2 0 N N N XANTPHOS, KBuO Dioxane, EtN [002711 To 1-(benzo[d][1,3]dioxol-5-yl)-N-(6-bromopyridin-2 yl)cyclopropanecarboxamide (72 mg, 0.2 mmol), XANTPHOS (7.00 mg, 0.008 mmol), K'BuO (31 mg, 0.28 mmol), (DPPF)2PdCl 2 .CH2Cl2 (33.00 mg, 0.24 mmol), and 2-methoxyaniline (30 mg, 0.24 mmol), 1,4-dioxane (0.400 mL) and triethylamine (0.200 mL) were added. The reaction mixture was heated to 150 'C in a microwave reactor for 10 minutes. The resulting material was cooled to room temperature. The solvent was evaporated under reduced pressure. The resulting mixture was dissolved in dichloromethane and washed with H,0. The organic layer was dried over anhydrous Na2SO 4 and evaporated under reduced pressure. The crude product was purified by column chromatography on silica gel to yield 1-(benzoid][i,3]dioxol-5-yi)-N-(6-(2 methoxyphenylamino)pyridin-2-yl)cyclopropanecarboxamide which was then treated with HCl in MeOH to form the ICI salt (2.4 mg, 0.0055 mmol, 2.7%). ESI-MS n/z calc. 403.1, found 404.5 (M+--1) retention time 3.01 minutes. [002721 Preparation of 1-(benzoldl11,31dioxol-5-y)-N-(6-((2 methoxyphenyl)(methl)amino)pyridin-2-yI)cvclopro panecarboxamide 0 0 N N N [002731 1-(Benzo[d][1,3]dioxol-5-yl)-N-(6-((2-methoxypheinyl)(ntethyl)amino)pyridin-2 yl)cyclopropanecarboxamide was synthesized using the procedure of 1-(benzo[d][1,3]dioxol-5--yl) N-(6-(2-methoxyphenvlamino)pyridin-2-yl)cyclopropanecarboxamide by reacting 2-methoxy-N methylaniline with 1-(benzo [d] [1,3]dioxol-5-yl)-N-(6-bromopyridin-2 yl)cyclopropanecarboxamide. [002741 Preparation of 1-(benzoldl11,31dioxol-5-y)-N-(5-benzoylpyridi-2 yl)cyclopropanecarboxamide -66- WO 2009/123896 PCT/US2009/038203 N MgBr N -- N 0 N N H H [002751 To a solution of -(benzo[d]l[1,3]ldioxol-5-yl)-N-(5-cyanopyridin-2 yl)cyclopropanecarboxamide (61.00 mg, 0.20 mmol) in THF (1 mL) at 0 C, a solution of phenyimagnesium bromide (133 pL, 3 M, 0.40 mmol) in ether was slowly added and the reaction mixture was allowed to warm up to room temperature and stirred at room temperature for eighty hours. Hydrochloric acid (IM) was added to the reaction material. The product was extracted using ethyl acetate. The organic layer was dried over anhydrous Na 2
SO
4 and evaporated under reduced pressure. The crude product was dissolved in DMSO (I mL), filtered and purified by reverse phase preparative HPLC to yield 1 -(benzo[d] [1,3] dioxol-5-vi)-N-(5 -benzoylpyridin-2 yl)cyclopropanecarboxamide as a TFA salt (18 mg, 0.035 mmol, 18%). ESI-MS m 1 z calc. 386.1, found 387.1 (NI-1) _; retention time 3.45 minutes. [002761 Preparation of I (benzo [d [131 dioxol-5~vl)-N-(6-(4-mnethoxvbenzy)lpyridin 2-yl)cyclopropanecarboxamide 0 N N Br C--O- H (DPPF) 2 PdC!.C01 0 N N H [002771 To (DPPF)2PdCl 2 .CH2Cl 2 (17 mg, 0.02 mmol), a solution of (4 methoxybenzyl)zinc(II) chloride (1.60 mL, 0.5 M, 0.80 mmol) in THF was added under nitrogen atmosphere and stirred at room temperature for five minutes. To this, a solution of I (benzo[d][1,3]dioxol-5-yl)-N-(6-bromopyridin-2-yl)cyclopropanecarboxamide (72 mg, 0.20 mmol) in THF (800 pL) was added slowly under nitrogen atmosphere. The reaction was heated to 150 'C in a microwave reactor for 10 minutes. The resulting material was cooled to room temperature. Na2EDTA and saturated aqueous NH 4 CI were added to the reaction material and stirred at room temperature for 30 minutes. The product was extracted using dichloromethane. The organic layer was dried over anhydrous Na 2
SO
4 and evaporated under reduced pressure. The crude product was dissolved in DMSO (1 mL), filtered and purified by reverse phase preparative 1- PLC to yield 1 (benzo[d][1,3]dioxol-5-yl)-N-(6-(4-methoxybenzyl)pyridin-2 -yl)cyclopropanecarboxamide as a TFA salt (27 mg, 0.052 mmol, 26%). ESI-MS m/z calc. 402.2, found 403.0 (M-1) retention time 3.24 minutes. 67' WO 2009/123896 PCT/US2009/038203 [002781 Preparation of 1-(benzo[dl[1,31dioxol-5-vl)-N-(6-(2-chilorobenzyl)pvridiii-2 yl)cycopropanecarboxam ide HCI [002791 1-(Benzo[d][1,3 ]dioxol-5-l)-V-(6-(2-chliorobenzyi)pyridin-2 ylicyclopropanecarboxamide was synthesized using the procedure of 1-(benzoid][1,3]dioxol-5-yl) N-(6-(4-nethoxvbenzyl)pyridin-2-yl)cyclopropanecarboxanide by reacting (2 chlorobenzyl)zinc(T) chloride with 1-(benzo[d] [1,3] dioxol-5-yl)-N-(6-bromopyridin-2 yl)cyclopropanecarboxamide. [002801 Preparation of 1-(benzo[d1[1,31dioxol-5-vl)-N-(6-(2-methvlbenzyl)pyridin-2 yl)cyclopropanecarboxamide H [002811 1 -(Benzo[d] [1,3 ]dioxol-5 -yl)-N-(6-(2 -methylbenzyi)pyridin-2 yl)cyclopropanecarboxamide (VR T-810)61) was synthesized using the procedure of 1 (benzo[d][1,3]dioxol-5-yl)-N-(6-(4-methoxybenzyl)pyridin-2-y)cyclopropanecarboxanide by reacting (2-methylbenzyl)zinc(II) chloride with 1-(benzo[d][1,3]dioxol-5-yl)-N-(6-bromopyridin-2 yl)cyclopropanecarboxamide. [002821 Preparation of 1-(benzo[l[1,31dioxol-5-yl)-N-(5-beiizylpyridin-2 yLecyloproanecarboxamide i) SOCl 2 , DMF B Br O N N OH ii) Pyridine H (DPPF) 2 PdOl 2
.H
2 Cl 2 , THF Br
H
2 N N [002831 Step a: 1-(Benzo[d][1,3]dioxol-5-yl)-N-(5-bromopyridinl-2 yl)cyclopropanecarboxamide - 68 - WO 2009/123896 PCT/US2009/038203 [002841 To 1-(benzo[d][1,3]dioxol-5-yl)cyclopropanecarboxylic acid (9.50 g, 46.00 nmmol), thionyl chloride (10.00 mL, 138.00 mmol) and DMF (4 drops) were added and the reaction mixture was stirred and heated at 60 C for thirty minutes. The excess thionyl chloride was evaporated under reduced pressure. A portion of the acid chloride (14.4 mmol) was dissolved in pyridine (10 mL) and was slowly added to 5-bromopyridin-2-amine (14.4 mmol) in pyridine (10 mL). The reaction mixture was stirred at 110 C for one hour and thirty minutes. The pyridine was evaporated under reduced pressure. The resulting mixture was dissolved in dichloromethane (100 mL) and washed with I N NaOlH (3 x 25mL). The organic layer was dried over anhydrous Na 2
SO
4 and evaporated under reduced pressure. The crude product was purified by column chromatography on silica gel to yield 1.-(benzo[di[i,3]dioxol-5-yi)-N-(5-bromopyridin-2 yl)cyclopropanecarboxamide (4.20 g, 11.7 mmol, 81%). [002851 Step b: 1-(Beizo[d][1,3]dioxol-5-yl)-N-(5-benzylpyridin-2 yl)cyclopropanecarboxamide [002861 A solution of benzylzinc(II) bromide (1.60 mL, 0.5 M, 0.8 mmol) in THF and (DPPF)2PdCl2.C-1 2 Cl2 (17 mg, 0.02 mmol) was stirred at room temperature for 20 minutes under nitrogen atmosphere. To this, a solution of 1 -(benzo[d]i ,3]dioxol-5-yl)-N-(5-bromopyridin-2 yl)cyclopropanecarboxamide (72 mg, 0.20 mmol) in THF (800 pL) was added and the reaction mixture was heated to 150 'C in a microwave reactor for 10 minutes. The resulting material was cooled to room temperature. Na 2 EDTA and saturated aqueous NH 4 Cl were added to the reaction material and stirred at room temperature for 20 minutes. The product was extracted using dichloromethane. The organic layer was dried over anhydrous Na 2
SO
4 and evaporated under reduced pressure. The crude product was dissolved in DMSO (1 mL), filtered and purified by reverse phase preparative HPLC to yield 1 -(benzo[dl [1,3dioxol-5-yl)-N-(5-benzylpyridin-2 yl)cyclopropanecarboxamide as a TFA salt (15 mg, 0.031 mmol, 15%). ESI-MS m 1 z calc. 372.1, found 373.1 (M-1) ; retention time 7.14 minutes. 1002871 Preparation of 1-(benzo[dl[1,31dioxol-5-vl)-N-(5-(2,6-dichlorobenzyl)pyridin 2-yl)cyclopropanecarboxamide 0 N N CI [002881 1-(Benzo[d][1,3]dioxol-5-vl)-V-(5-(2,6-dichliorobenzyl)pyridin-2 ylfcyclopropanecarboxamide was synthesized using the procedure of 1-(benzo'd][1,3]dioxol-5-yl)- -69- WO 2009/123896 PCT/US20091038203 A(5 -ben zy lpyridini-2-yl)cyciopropaniecarboxami de by reacting 1 -(benzo[l [1 ,3 idioxoi-5 -vi)-N-(5 bromopyridin-2-yi.)cyciopropan-ecar-boxan-ide with (2,6-dichiorobenzyi)zinc(II) chloride. 1002891 Prep~aration of 1-(Benzo IdI 11,31 dioxol=5-y)-N4-5-(cyclohexylmethvl)py1ridin 2-vl)cyclopropanecarboxanile 1002901 1 KBe-nzo [dl [1,3 ]dioxoi-5 -y1>)-N-5-(cycioh exyi thyl~pyridint-2 yi)cyclopropanecarboxamide was synthesized using the procedure of i-(benzo-id][13]dioxol--5--y} NV-( 5-benizylpyridini-2-ylbcyclopr-opanecarboxanmide by reacting I -(benizo[d][ 13]dioxo.-5-y)-/(5 bromiopyridini-2 -yl,)cyciopropainecarboxamnide with (cyclohexyimnethyl)zinc(ii) bromide. [002911 Preparation of 1-{benzoldll ,3dioxob--yl)-,-(5-isobuty lpyridin-2 Il)ejeloprop~anecarboxarnide 0 N 1 N H [002921 I ->Benizo[edi [1,3 ]dioxoi-5-yi)-NV-(5-isobutyipyridin-2 yi)cyciopr-opanecarboxamn-ide- was synthesized using the procedure of 1 4benzo [dl[153Id ioxo]-5 -yi) NV-(5-benizylpyridin--2--yl)cyclopropanecarboxamide by reacting l--(benzo[d][1,3)idioxol--5-yli)-N,\-(5 bromyopyridin-2-yl)cyciopropaniecarboxamide wifli isobutyizinc(II) bromide. [002931 Preparation of I -(benizo [dl[1,31 dioxol-5-vI)-jN45-4-methoxybenv prdn 2-Acoroa ecarboxaiie 0N N 0 H [002941 1 -(B enzo[d] [ 1,3 ]dioxoi-5-yi)-,V-(5- 4-n-ethoxybenzyIlpvridin -2 yficyclopropanearboxamide was synthesized -using the procedure of I -(beizo 'd][1,3]dioxol-5-yl)- AL(5 -ben zy lpyridini-2-yl)cyciopropaniecarboxami de by reacting -(beuizo [dl [I , dioxoi-5-y)-V-(5 brornopyridin-2-yi'cyciopropan-ecar-boxan-ide with (4-methoxybenzy]bzi nc(1 chloride. 1002951 Pre )aration of-1= benizoM 1 3 dioxol-5=vI =jNr 5-2-chlorobenzvl )vridin-2 Ifc~lopQoanecarboxamile - 70 - WO 2009/123896 PCT/US2009/038203 0N - NC!I H [00296] 1-(Benzo[d][1,3 ]dioxol-5-vl)-V-(5-(2-chliorobenzyi)pyridin-2 ylicyclopropanecarboxamide was synthesized using the procedure of 1-(benzoid][1,3]dioxol-5-yl)- N-(5-benzylpyridin-2-yl)cyclopropanecarboxami de by reacting 1 -(benzo[d] [1,3]dioxol-5-yl)-N-(5 bromopyridin-2-yl)cyclopropanecarboxanide with (2-chlorobenzyl)zinc(II) chloride. [002971 Preparation of 1-(benzo Idl 11,3] dioxol-5-yl)-N-(5-(2 niethoxypheivilamino)pyridiii-2-vlclopropanecarboxaniide Pd,(dba) N NHN- Cs 2
CO
3 0 NH. XANTPHOS O |{ N- Dioxane I IH OKEN P O [002981 1--(Benzo[d] [1,3 ]dioxol-5-vl)-N-(5-iodopyridin-2-.yl)cyclopropanecarboxamide (816 mg, 0.200 rnmol) was suspended in 1,4-dioxane (0.4 mL) and triethylamine (0.2 mL) containing cesium carbonate (91.2 ng, 0.0280 mniol), tris(dibenzylideneatcetone)dipalladium (0) (1.8 ng, 0.0020 mmol), 2-methoxyaniline (0.027 nL, 0.24 mmol), and (9,9-dimethyl-9H xanthene-4,5-diyl)bis(diphenylphosphine) (XANTPHOS, 2.3 ng, 0.0040 mmol). The reaction mixture was heated to 100 'C for 15 hours. The crude product was then filtered, evaporated to dryness, dissolved in ,N--dimethylforrnamide (1 mL) and purified by reverse-phase preparative liquid chromatography utilizing a gradient of 0-99% acetonitrile in water containing 0.05% trifluoroacetic acid to yield the pure product (2.6 mg, 0.0049, 2.4%). ESI-MS m/Z calc. 403.2, found 404.5 (M+ 1.) Retention time 2.73 minutes. [002991 Preparation of 1-(benzoldli1,31dioxol-5-yi)-N-(5-(hydroxv(2 methoxyphenvl)methvl)pyridin-2-vl)cvelopropanecarboxamide -71- WO 2009/123896 PCT/US2009/038203 .NH H 0 / NaBH 4 0 MeOH NO< OH [003001 1-(Benzo[d][1,3 ]dioxol-5-vl)-V-(5-(2-methoxybenzoyi)pyridin-2 yl)cyclopropanecarboxamide (0.50 g, 1.2 mmol) was suspended in 25 mL of methanol. Sodium borohydride (0.454 g, 1 2.0 mmol) was added in small portions. After stirring for one hour the reaction mixture was evaporated to dryness and purified on 40g of silica gel utilizing a gradient of 15-70% ethyl acetate in hexanes to yield the pure product as a white solid (0.2125 g, 0.5078 mmol, 42%). ESI-MS m/z cale. 418.6, found 419.3 (M+1)+. Retention time 2.59 minutes. 1003011 Preparation of 1-(benzo[dl[1,31dioxol-5-vl)-N-(5-((3-hydroxypropoxy)(2 rn eth oxyphenyl)methvl)pyridin-2-vlcvclopropan ecarboxa mide HH N 0' S HO MOH pTsOH O 'r 1 O----- O 0 N Toluene -- OH f -0,,,. 0 [003021 1-(Benzo[d][1,3 ]dioxol-5-yi)--(5-(hydroxy(2-methoxyphenyl)methyl)pyridin-2 yl)cyclopropanecarboxamide (41.8 mg, 0.100 mmol) was suspended in 2 mL of toluene contaning 4-methylbenzenesulfonic acid hydrate (pTsOH, 23.8 mg, 0. 125 mmol) and propane-1,3-diol (9.89 mg, 0.130 mmol). The reaction mixture was then heated to 140 'C for 5 minutes in a microwave reactor. The crude product was then filtered, evaporated to dryness, dissolved in NNT dimethylformamide (1 nL.) and purified by reverse-phase preparative liquid chromatography utilizing a gradient of 0-99% acetonitrile in water containing 0.05% trifluoroacetic acid to yield the pure product (26.5 mg, 0.0556 mmol, 55.6%). ESI-MS m/z calc. 476.5, found 477.3 (M+1). Retention time 2.71 minutes. [00303] Preparation of 1-(enzoidl[1I,31dioxol-5-vl)-N-5-(2 (dimethylamino)ethoxy)(2-methoxyphenvl)methyl)pvridin-2-yl)cyclopropanecarboxamide - 72
-
WO 2009/123896 PCT/US20091038203 0 OMe S0 N N DD'2 H [003041 1 -( Benizo [di[1 ,3 dioxoi-5 -yi)-N-(5-('2-(ditnethylamnino)elioxy)(2 metlioxypheniyi)n-et-iyi'pyridin-2-yi'cyciopropan-ecar-boxan-ide was prepared from I (benzo[df][i .3]dioxoi--5-.yi)-- N4-5-ThIydroxy(2- -methoxyphenyl)methyi)pyridin--2yi)cyclopropanecarboxarnide and 2-(dimethilamino)ethanol in a manner analogous to that of I (benizo[l[i ,3]dioxoi-5-vi)- 'N(5-((3 -hydroxypfropoxy)('2-methioxyphenyl)inethyi'pyr-idini-2 yi)cyci opropai ecarbox amidec. 1003051 Prep~aration of 1-(benizo [dl[1,31dioxol-5---v)=,N=(5-((2-methoxyphenyl)(2 (piperidin-1 -vI)etlioxv')rnethyl)p!vridin-2-yi)cvclqpropaiiecarboxarnide fIN 0) OMe -~ N N H [003061 1 -(B enzo[d] [1,3 ]dioxoi-5-vi)-iV-(54'(2-n-thoxyphenvd)(24p(iperidin-i yi)etlioxy )methvl)pyridin-2 -yl)cyc lopr-opanecarboxamide was prepared from I (bnz[li,3]dioxoi-5-yi)-N-(5-(hlydroxy (2 -mettoxyp11ei nethvi" yridin-2 yi)cyciopropanecarboxamnide and 2-(piperidin- I-vi)ethanoI in a manner analogous to that of I (benzo [d] [1,3-]dioxoi-5--yi)-N,-(5- ((3 '-hydiroxyp~ropoxy)(2--methoxyphenyl)mthyl)pyridin-2yl)cyclopropanecarboxamide. [003071 Preparation of I -4Beizo [d [1,31 diqxoI-5-j-vN-(5-(( rn th xypeny)(popov~mtyjpjyridin-Z-yI)cv clopropanccarboxarnide 0 Ome 0 N N
H
- 73 - WO 2009/123896 PCT/US2009/038203 [003081 1 -(Benzo[d][1,3]dioxol-5-yl)-N-(5-((2-m ethoxyphenyl)(propoxy)nethyl)pyridin 2-yl)cyclopropanecarboxamide was prepared from 1-(benzo[d][1,3]dioxol-5-yl)-N-(5-(hydroxy(2 methoxyphenyl)methyl)pyridin-2--yl)cyclopropanecarboxamide and propan-- 1-ol in a manner analogous to that of 1-(benzo[d][1,3]dioxol-5-y)-N-(5-((3 -hydroxvpropoxy)(2 methoxyphenyl)methyl)pyridin-2-yl)cyclopropanecarboxamide. [003091 Preparation of 1-(benzoldl[1,31dioxol-5-yl)-N-(5-((2 (idiisopropylamino)ethoxy)(2-methoxyphenyl)nethyl)pyridiii-2-yl)cyclopro anecarboxamide fN 0 OMe 0 N N H [003101 1 -(Benzo[d][ 1,3 ]dioxol-5-yl)-N-(5-((2-(diisopropyl]amino)ethoxv)(2 methoxyphenyl)methyl)pyridin-2-yl)cyclopropanecarboxamide was prepared from I (benzo[d][1,3] dioxol-5--yi)-N-(5-(hydroxy(2--methoxyphenyl)methyl)pyridin-2 yl)cyclopropanecarboxamide and 2-(diisopropylamino)ethanol in a manner analogous to that of 1 (benzo[d] [1,3]dioxol-5-yl)-N-(5-((3 -hydroxypropoxy)(2-nethoxyphenyl)methyli)pyridin-2 yl)cyclopropanecarboxamide. [003111 Preparation of 1-(benzoldl1 1 ,3dioxol-5-y)-N-(5-((2-methoxyphenyl)(2 niorpholinoethoxy)niethvl)pyridii-2-vil)eveoropanecarboxamide rO N 0 OMe 0 NN H 1003121 1-(Benzo[d][1,3]dioxol-5 -yl)--N-(5--((2-methoxyphenyi)(2 morpholinoethoxy)methiyl)pyridin-2-yl)cyclopropanecarboxamide was prepared from 1 (benzo[d][1,3]dioxol-5-yl)-N-(5-(iydroxy(2-methoxyphenyl)methyl)pyridin-2 yl)cyclopropanecarboxamide and 2-morpholinoethanol in a manner analogous to that of 1 (benzo[d][1,3]dioxol-5-yl)-N-(5--((3--hydroxypropoxy)(2--methoxyphenyl)methvl)pyridin-2 yl)cyclopropanecarboxamide. - 74 - WO 2009/123896 PCT/US2009/038203 [003131 Preparation of I -(benzo [d [1,31 dioxol-5-vl)-N-(5-((2-hydroxvethoxv)(2 methoxyphenyl) ethyljpyridin-2-yllevelopropanecarboxamide 0 OMO 0 3 N N H [003141 1 -(Benzo[d] [1,3 ]dioxol-5 -yl)-N-(5-((2-hydroxyethoxy)(2 rnethoxypienyl)methyl)pyridin-2-yl)cyclopropanecarboxamide was prepared from 1 (benzo [d] [1,3] dioxol-5-yi)-N-(5-(hydroxy(2-methoxyphenyl)nethyl)pridin-2 yicyclopropanecarboxamide and ethane- 1,2-diol in a manner analogous to that of 1- (benzo[dII[1 ,3 Idioxol-5-yl)-N-(5-( (3- hydroxypropoxy)(2-methoxyphenvl)methyl)pyridin-2 yi)cyclopropanecarboxamide. [003151 Preparation of 1-(benzoldtl[1,31dioxo-5-yl)-N-(5-((dimethylamino)(2 methoxyphenyl)methyl)pyridin-2-yl)cyclopropanecarboxamide H H N N OH CH 3
SO
2 C I DIEA 0 S=O O O 0 O" H
(CH
3
)
2 NH, N ---------- O---- OM N N , 0 0" [003161 Step a: (6-(i-(benzo[d][1,31dioxol-5-yl)eyclopropanecarboxamido)pyridin-3 yl)(2-methoxyphenyl)methyl methanesulfonate [003171 Methanesulfonyl chloride (56 pl, 0.72 mmol) was added slowly to the mixture of 1- benzo[1,3 ]dioxol-5-yl-cvclopropanecarboxylic acid {5-[hydroxy-(2-methoxy-phenvl)-methyl] pyridin-2-yl}-amide (50 mg, 0.12 mmol) and DIEA (24pl, 0.14 mmol) in dichloromethane (2 ml) at 0" C. The reaction mixture was stirred at ambient temperature for 3h. Solvents were evaporated and the residue was used for the next step without any additional purification. - 75 - WO 2009/123896 PCT/US2009/038203 [003181 Step b: 1-(benzo d][1,3]dioxol-5-yl)-N-(5-((dimethylamino)(2 methoxyphenyl)rn ethyl)pyridin-2-yl)cyclopropanecarboxamide 1003191 (6-(I-(Benzo[d] [1,3idioxol-5-yl)cyclopropanecarboxamido)pyridin-3-y)(2 methoxyphenyl)methyl metianesulfonate (0.12 mmol) was dissolved in NNP (2 ml) following by addition of 2M solution of dimethylamine in THF (600 pl, 0.12 mmol) and DIEA (21 II, 0.12 mmol). The reaction mixture was maintained at ambient temperature for 16 h. The resulting mixture was subjected to HPLC purification to provide the trifluoroacetic salt of target material (55 mg, 82%) as a colorless solid. 1 H-NMR (DMSO): 6 8.43 (s, 1-1), 6 840 (d, J=2 -Hz, 1H-), 6 8.06 (d, J=8.7 Hz, 1H1), 6 7.96 (dd, .J= 2 Hz, l8.7 Iz, 1H), 6 7.56 (bd, J=6.5 -Iz, 11-1), 6 7.39 (bt, J=6.5 Hz, 1H), 6 7.05-7.11 (m, 3H), 6 6.96 (s, 2H), 6 6.05 (s, 2H), 6 5.68 (bm, IH), 6 3.85 (s. 3H), 6 2.75 (s, 6H), 6 1.42-1.45 (in, 21-1), 1.13-1.16 (in, 21-1). [003201 Preparation of 1-(benzo dl[11,3dioxol-5-yl)-N-(5-(2 methoxyphenyl)(methylamino)methvl)pyridin-2-yI)cvclopropanecarboxamide HN OMe H [003211 1-(benzo[d][1,3]dioxol-5-yl)-N-(5-((2 methoxyphenyl)(methylamino)methyl)pyridin-2-yl)cyclopropanecarboxamide was prepared from (6-1- .(benzo[d][1,3]dioxol.-5-vl)cvclopropanecarboxamido)pyridin-3 -yl)(2--methoxyphenyl)methyl methanesulfonate and methyl amine in a manner analogous to that of 1-(benzo[d][1,3]dioxol-5-yl) N-(5-((dimethyl amino)(2-methoxyphenyi)methyl)pyridin-2-yl)cyclopropanecarboxamide. [003221 Preparation of 1-(benzoldl 1,31dioxol-5-1L-N-(5-((ethylinethlamino)2 methoxy hen y methyll vridin-2-vI evelopropanecarboxamide N OMe
K
0 DKI N N H [003231 1 -(benzo[d] [1,3] dioxol-5-yl)-N-(5 -((ethyl(methyl)amino)(2 methoxyphenyl)nethyl)pyridin-2-yl)cyc lopropanecarboxamide was prepared from (6-(1 (benzo [d] [1,3 ] dioxol-5-yl)cyclopropanecarboxamido)pyridin-3 -yl)(2-methoxyphenyl)methy methanesulfonate and N-methylethanamine in a manner analogous to that of I -76- WO 2009/123896 PCT/US20091038203 (benzo [d] [ 1,3 idioxoi-5 -vi) -N--(5-(diethylamino)("2-methioxyphenyi)metiyl)pyri dini-2.
yi)cyci opropai ecarbox amidec. 1003241 Preparation of 1-(benzoIdj 111,31 dioxo=5-jl)-N-( 5-((2 methoxyphenylh'pvrrolidin-J ~yI)nethv1,)p r-idini-2-vlkcvclopropaneearhboxamide 0 N OMe 0 N N H [003251 1 4beizo [d] [1,31 di oxoi-5 -yl)-,N-(5-((2 -meihoxypheniyl )(Pyrrolidinl- I yi)methvl)pyridini-2 -yl)cyclopr-opanecarboxamide was prepared from (61 1 -(be-nzol-dl [1,3 ]dioxol-5 yi)cyciopropanecarboxamnido) yrid in-3 -yfi(2-m-ethoxypi enyi)m-nethyi meth anesui fon ate and pyrrolidine in a. manner analogrous to that of i-(benzo[d][i1,3]dioxol--5-yvl)-N-.(5-((dim-tliyamino)(2methoxy lhenyl)metliylPpyridin-2-vl)cyciopropaniecarboxamide. [003261 Preparation of I 4benzo [dl 11,31 dioxo1-5-yI)--45-((diethylamiiio o}2 rn eth oxjyphe l t) ~prdin-2-vlcclppaecro me N OMe H [003271 1 -(b rnzo[dl [1,31 dioxoi-5-y)-N-'5 -((diethiylamino)(2 meith-oxyphenyi)mnethyi)pyidin-2-yJ)cvciopropan-carboxamide was prepared from (64(1 (benzo [d] [1,31 dioxoi.-5--yi)cyclopropanecarboxamidiolpyridin--3--y)(2-mthoxyphenyi)methyl methanesulfonate and cliethvlamine in a manner analogous to that of I -(benzoi-dj[i,3ildioxol-5-yl) -N-(5-((dinietliylamino)(2-m thtoxyph mi)metliyl)pyridini-) -yi)cyclopr-opanecarboxamnide. [003281 Preparation of _I_-nzoldji3 dixo1-_---yI-N--(2 rnethoxypllenyl)(piperidin-1-yl)rnethyl)pyridin-2=I)eclopropanecarboxarnide N 0 \ /0 ? N N - 77 - WO 2009/123896 PCT/US20091038203 [003291 1 -(b rnzo[dl [1 ,3]dioxoi-5-yi)-N-(5-((2-myethioxyphenyhl(iperidin- 1 yli-methyi.)pyr-idi-2-"-yi)evciopropanecarboxaimide was prepared from (641 -(b enzo[d1 [I,3 ]dioxol-5 yi)cyclopropanecarboxamnio)pyridin--3 -- y)(2-methoxyphenyl)methyl methanesulfonate and piperidine in a manner analogous to that of 1(be-nzol-d-fIi ,3]Idioxol-5-yi)-.N45-((dimetliylaminlo)(2' mnetlioxypheniyi)niethiyh'pyridin-2-yl)cyeiopropanie arboxamide. [003301 Prprto f1{ezl 1~1doo==I=-5(()=3j-hydxypyrro idin 1=y)(2ni thoVhenv)nethI)wri in-2:yI)cjclojrojanecar oxamide OH N 0> Z - ! 0 /0 1003311 1 4Be-nzo-d-Il [1,31 dioxol-5 -yi)-.N45-(((R)-3 -lw droxypyrrolidin-1I-vil)( metlioxyphenyi)methyi)pyridini-2-yl)cyeiopropanieearboxami de was prepared from (6-( 1 (be-nzo [d [I.3 ] dioxoi-5-yF~cyciopr-opanecarb~oxamido'pyridin-3 -yi)(2-methoxyphenyl)methyl , metlianesulfonate and (R)- 1-hydroxypyrroi dine in a manner analogous to that of I (benizo[di [1 ,3 dioxoi-5 -yi -. N-(5-(d'iethiylamino)(2-m ethioxyphenyi)rnethiyl)pyri dini-2.
yi)eycliopropai ecarbox amidec. 1003321 P1re ~aration of-1= benizo d 1 3 1dioxo=5=--.N- 5= (2= methoxyphenyi)(morphlolino)metliyl)pvridin-2=yl1)cveopr-opanecarboxamide N OMe O C N 'N [003331 1 -(Benzo [d] [1, ,3 ] dioxo -5-yi"- N--(5- ((2 meth oxyphei yP,(m-orpholiino)methyi'ppyridin--2-yP~cyciopr-opanecarboxamide was prepared from (6 (1 4benzo-d-Il [1,3 Idioxol-5-yi)eyelopropaneearboxamnido)pyridini-3 -yl)(2'-metliox-ypheiinyimethyl methanesulfonate and morplioline in a manner analogous to that of I -('benzo[dli[i,3idioxoi-5-yi)-N (5-((dimethyiamin o)2-methoxyph-enyi'jmethyhpyridin-2-yvecicopropanecarboxam-ide.
WO 2009/123896 PCT/US20091038203 [003341 Preparation of 1 -(betizo [dl[1.31 dioxol-5-yl)-N-(5-((4-hvdroxyri ciridin-1 yl)(2-m ethoxy phejyl)mthly -idin-2- Icclop opan ecar-boxamide OHI NOMe 00 O -'' N N H 1003351 1-(be-nzo [d 1 1l[,3]dioxol-5--yi)-.N-(5 -((4-hydroxyp[iperidin- l-vl)( 2
.
metlioxyphenyi)methyi)pyridini-2-yl)cyciopropanie arboxami de was prepared from (6-( 1 (benzo [d] [1.3] dioxoi-5 -yll)cyclopropaneeart~oxamido)pyridin-3 "-yl)(2/--methoxyphenyi)methyl metlianesulfonate and 4-iy droxypiperi dine in a manner analogous to that of I -(benzol-d][11,3 ldioxol 5-yi)--(-.5-((dimetiylamino)(2 -ti thoxyphenvylrnethbyl)pyridini-2.-yi)eyclopr-opanecarboxanmide [003361 Preparation of 1-((6-(4b(henzol dj [1,31 dioxol-5 yl)cyclopropanecarboxanido)py1ridi-3-yI)(2-methoxypjhenyl)rnethyl)pyErrolidine2 carboxarnide
H
2 N N OMe
K
0 I~~ N ~N H [003371 1.-((6--41 -4Benzo [d-1 1,3] dioxol--5-yl)cyelopropaneearboxamido)pyricdin-.3 -yl)( 2
-
metliox-yphenii,)methvl)pyrrolidine-2-carboxarniide was prepared from (6-( 1 -(benzo-d-I 1 1,3 1dioxol 5 -Yb~cye] opropanecarbox amnido)pyri din -3 -y 1)(2-met] oxyphen vlin ethyl mnetlianesuifonate and pyrrolidine-2-ecarboxamide in a manner analogous to that of 1-(benzo[d][i,3)]dioxo-5--yl)-.N--(15- ((dimetliylamnino)(2 -mnethoxypheniyllmetlhvl)pyridini-'--y1)cyclopropaniecarboxamide. [003381 Preparation of 1 (benzo [d] [1,31 dioxol-5-1I)-N,- 45- -S)-2 (-n eth oxvrnethvl~hyrrolidin-i-yl)(2-m ethoxyph envl)methvl pyidin-2 3:l)cycloprop~anecarboxarnide -0 N OMe 0 NN H - 79 - WO 2009/123896 PCT/US20091038203 [003391 1 -(B tizo[di [1,3 ]dioxoi-5-yi)-N-(5-(((S)-2-(mefiioxynetliyl) yrroidin-- i-l)(2 methoxyphenyi)m ethyl) yyridin-2-yi)cvciopropane-Carboxamide was prepared from (64(1 (benzo [l] [1,3-]dioxoi.-5--yi)cyclopropaniecarboxamiio)pyridin--3 -- y)(2-methioxyphenyi)methyl methanesulfonate and piperidine in a manner analogous to that of I -benzol-d-Il1,3ldioxol-5-i)N (5-((dimyethiyiamino)(2 -mj~ethioxypliinethvi"pyridini-2-yi)cyclopropanecarboxamiide. [003401 Prepgration of _I_-nzoldji3 dixo1-_-yI)--N-{-(26 dhlnethlrnorh olino)(J2-methoxyihenvi)nethy)pyridin-2:yl)ce loiooDanecarboxarnide N OMe 011 N N H [003411 1 -(B enzo [d] [ 1,31 IdIoxoi-5 -yi)-N-(5-("(2,6-dimyetiylnmorp~hoiino"(2 methoxyphenyi)m ethyl) yridin-2-yI)ecicopropane-carboxamide was prepared from (64(1 (benzo [l] [1,3-]dioxoi.-5--yl)cyclopropaniecarboxamidiolpyridin--3 -- y)(2-methioxyphenyi)methyl methanesulfonate and 2,6-dimethyimorpholine in a manner analogous to that of I (benzo [dl [1,3 ]di oxoi-5 -yi)-,N-(5-((ditnethylainino"(2-mnethboxyphenyi'rmethiybpyridin -2 yhcyclopropanecarL oxamide. [003421 Preparation of 1=(benzoj dJ t,3J dioxo1-5y1i)-N-(5=(J2 (niethoxyrnethyvl)jprrolidin-1 -yI)(2-rethoxyphienI)rn ethyl)pyidin-2 y1)cyclo tQroE~ anectarboxam ide __0 N OMe 0i 0 N N
H
1003431 1 -("Benzo[d] [1,3 ]dioxoi -5-vi -N--(5 -((2-(mietboxyethiy])pyrroi din- 1 -yi)("2 metlioxypheniyl)nmeth-vi)pyvridin-2-yi'cyciopropan iccar-boxanmide was prepared from (6-( 1 (benzo[d][i .3]dioxol--5-.yl)cyclopropaneart~oxamido)pvridin-3 "-yl)(,2--methoxyphenyi,)methyl methanesulfonate and 2-(methioxymiethbyl)pyrroiidine in a manner analogous to that of I (benzo [dl [1,3 ]dioxoi-5 -vi)-N-(5-((dim-ethi,am-ino)(2-mei-thoxyphenvi)methyi'pyr-idin--2 yi)cyclopropanecarboxamide. - 80 - WO 2009/123896 PCT/US20091038203 [003441 Preparation of 1 -4beiizo [dl[1.31 dioxolb5-yl)-N- 5-((4- 2 hydrxyehvl~ijprazid-y)(2- etoxypenj lehly -idiln-2 Il)cyclopropanecarboxainid e OHI (N: N OMe 0 N 'N 0- H [003451 1 KBenj-zo [dl[1,3 ]dioxoi-5~-yi)-N-(5 -((4-(2"I-1 ydroxyethyl~piperazin-- I -yi)( 2 methioxyphenyi)methyl)pyridin--2--yl)cyclopropanecarboxamidewas prepared from (6-(11-. (benizo [d] [1,31 dioxoi-5-yI)cyclopropaniecar-boxamidopyridin-. -vI)('-)methoxvphenyl)metliyI metliesulfonate and 2-4piperazin-l -yi~ethanol in a manner analogous to that of I (be-nzo [dl [1,3 Idi oxoi-5 -yl)-.N-(54((dim-ethyiamii-no)(2--methoxy ph nyi~rnthyi)pyridin-2 yIhcyclopropaneccarL oxamide. [003461 Prepar-ation oflb(beiizoj d][1j,3J dioxoI-5-yl1)-N-(5-(4-(2ni etlloxjethyl) pi peraziin-I -11IX2-ni etioxvph eni)reth yi) pyridi n-2 y1)cyclopropanecarboxalnid e N 0 Me 0 D!IZ N N H [003471 1 -(B izo [di [1,3 ]dioxoi-5 -yi)-N-(5-'' ((42-hydroxyethiyi'piperazin- I-yli(2 methoxyphenyi)rnethyl) yridin-2-yJ)cvciopropan-carboxamidewas prepared from(-(1 (benzo [d] [1,3-]dioxoi.-5-yi)cyclopropaniecarboxamidio)pyridin-3--.yI)(2-methioxyphenyi)methyl rnethaniesulfonate and i-(2-nethioxyethiyi)piperazine in a manner analogous to that of 1 (benzo [di [1,3 idi oxoi-5 -yi)-N-(5-((diinethylarnino"(2-mnethboxv Iienyi'methiy])pyridin -2 yi~cyclopropanecarboxarnide.
WO 2009/123896 PCT/US2009/038203 [003481 Set forth below is the characterizing data for compounds of the present invention prepared according to the above examples. Table 5. 1 403.1 3.48 2 311.1 2.26 3 403.: 3.24 5 387.1 3.44 6 326.3 2.33 7 474.3 2.94 8 389.3 3.2 9 472.3 2.88 10 419.3 2.91 11532.3 2.39 12 433.3 3.3 13 389.1 2.89 14 387.1 3.45 15 390.3 2.93 16 379.3 8.16 17 351.1 3.5 18 546.5 2.59 19 297.1 2.67 20 446.3 2.8 21 297.3 2.09 22 545.5 2.74 23 516.3 2.96 24 441.3 8.81 - 82 - WO 2009/123896 PCT/US2009/038203 Cmd LCIMS LC R 25 269.1 2.82 26 311.3 2.4 27 417.3 3.5 28 403.5 3.57 29 339.3 6.6 30 457.5 3.79 31 373.3 3.33 32 486.3 2.94 33 461.5 3.6 35 403.3 3.15 36 516.3 2.97 37 419.3 3.69 38 283.1 2.68 39 531.5 2.61 40 333.1 1 .48 42 297.3 2.18 43 325.3 2.13 44 419.3 2.57 45 515.5 2.75 46 393.1 3.3 47 301.3 3.05 49 488.3 2.8 50 453 1.63 51 516.3 3.01 52 404.5 2.73 - 83 - WO 2009/123896 PCT/US2009/038203 Cmd LCIMS LC R 53 432 3 2.74 54 283 1 2.61 55 488.3 2.76 56 339.2 3.19 57 463.5 2.66 58 502.3 2.75 59 407.5 3.39 60 403.5 3.11 61 267.1 1.6 63 373.1 7.14 64 404.5 3.01 66 477.3 2.71 67 379.1 3.56 68 453.1 3.69 70 387.3 3.7 71 530.5 2.53 73 438.3 1 .83 74 418.3 2.87 75 297.1 2.48 76 407.5 3.69 77 373.1 3.5 78 407 3.6 - 84-3 WO 2009/123896 PCT/US2009/038203 1003491 Assays fbr Detecting and Measuring AF508-CFTR Correction Properties of Compounds 1003501 Membrane potential optical methods for assaying AF508-CFTR modulation fiQ.p.rtietsof compounds. [003511 The assay utilizes fluorescent voltage sensing dyes to measure changes in membrane potential using a fluorescent plate reader (e.g., FLIPR III, Molecular Devices, Inc.) as a readout for increase in functional AF508-CFTR in NIH 3T3 cells. The driving force for the response is the creation of a chloride ion gradient in conjunction with channel activation by a single liquid addition step after the cells have previously been treated with compounds and subsequently loaded with a voltage sensing dye. [003521 Identification of Correction Compounds [003531 To identify small molecules that correct the trafficking defect associated with AF508-CFTR; a single-addition HTS assay format was developed. Assay Plates containing cells are incubated for ~2-4 hours in tissue culture incubator at 37oC, 5%C02, 90% humidity. Cells are then ready for compound exposure after adhering to the bottom of the assay plates. [003541 The cells were incubated in serum-free medium for 16-24 hrs in tissue culture incubator at 37oC, 5%C02, 90% humidity in the presence or absence (negative control) of test compound. The cells were subsequently rinsed 3X with Krebs Ringers solution and loaded with a voltage sensing redistribution dye. To activate AF508-CFTR, 10 pM forskolin and the CFTR potentiator, genistein (20 pM), were added along with Ci-free medium to each well. The addition of Cl--free medium promoted Cl- efflux in response to AF508-CFTR activation and the resulting membrane depolarization was optically monitored using voltage sensor dyes. [003551 Identification of Potentiator Compounds [003561 To identify potentiators of AF508-CFTR, a double-addition HTS assay format was developed. This HTS assay utilizes fluorescent voltage sensing dyes to measure changes in membrane potential on the FLIPR III as a measurement for increase in gating (conductance) of AF508 CFTR in temperature-corrected AF508 CFTR N-I 3T3 cells. The driving force for the response is a Cf ion gradient in conjunction with channel activation with forskolin in a single liquid addition step using a fluoresecent plate reader such as FLIPR III after the cells have previously been treated with potentiator compounds (or DMSO vehicle control) and subsequently loaded with a redistribution dye. [003571 Solutions: -85- WO 2009/123896 PCT/US2009/038203 [003581 Bath Solution #1: (in mM) NaCl 160, KCI 4.5, CaCl 2, MgCL 2 1, HEPES 10, p1 7.4 with NaOH. 1003591 Chloride-free bath solution: Chloride salts in Bath Solution i are substituted with gluconate salts. [003601 Cell Culture [003611 NIH3T3 mouse fibroblasts stably expressing AF508-CFTR are used for optical measurements of membrane potential. The cells are maintained at 37 "C in 5% CO2 and 90 % humidity in Dulbecco's modified Eagle's medium supplemented with 2- mM glutamine, 10 % fetal bovine serum, I X NEAA, f-ME, 1 X pen/strep, and 25 mM HEPES in 175 cm 2 culture flasks. For all optical assays, the cells were seeded at -20,000/well in 384--well matrigel--coated plates and cultured for 2 hrs at 37 'C before culturing at 27 'C for 24 hrs. for the potentiator assay. For the correction assays, the cells are cultured at 27 'C or 37 'C with and without compounds for 16 - 24 hours. [003621 Electrophysiological Assays for assaying AF508-CFTR modulation properties of compounds. [003631 L.Ussing Chamber Assay [003641 Ussing chamber experiments were performed on polarized airway epithelial cells expressing AF508-CFTR to further characterize the AF508-CFTR modulators identified in the optical assays. Non-CF and CF airway epithelia were isolated from bronchial tissue, cultured as previously described (Galietta, L.J.V., Lantero, S., Gazzolo, A., Sacco, 0., Romano, L., Rossi, G.A., & TA Zegarra-Moran, 0. (1998) in Vitro Cell. Dev. Biol. 34, 478-481), and plated onto Costar@R Snapwell" filters that were precoated with NIH3T3-conditioned media. After four days the apical media was removed and the cells were grown at an air liquid interface for >14 days prior to use. This resulted in a monolayer of fully differentiated columnar cells that were ciliated, features that are characteristic of airway epithelia. Non-CF HBE were isolated from non-smokers that did not have any known lung disease. CF-HBE were isolated from patients homozygous for AF508-CFTR. [003651 HBE grown on Costar) SnapwelTM cell culture inserts were mounted in an Using chamber (Physiologic Instruments, Inc., San Diego, CA), and the transepithelial resistance and short-circuit current in the presence of a basolateral to apical Cl- gradient (Isc) were measured using a voltage-clamp system (Department of Bioengineering, University of Iowa, IA). Briefly, HBE were examined under voltage-claip recording conditions (Vhold = 0 mV) at 37 "C. The basolateral solution contained (in mM) 145 NaCl, 0.83 K 2 HP0 4 , 3.3 KH 2
PO
4 , 1.2 MCl2, 1.2 - 86 - WO 2009/123896 PCT/US2009/038203 CaCl 2 , 10 Glucose, 10 HEPES (pH adjusted to 7.35 with NaOI-1) and the apical solution contained (in mM) 145 NaGluconate, 1.2 MgCl 2 , 1.2 CaCl2, 10 glucose, 10 HEPES (pH adjusted to 7.35 with NaOH). 1003661 Identification of Correction Compounds [003671 Typical protocol utilized a basolateral to apical membrane Cl- concentration gradient. To set up this gradient, normal ringer was used on the basolateral membrane, whereas apical NaCl was replaced by equimolar sodium gluconate (titrated to pH 7.4 with NaOH) to give a large Cl- concentration gradient across the epithelium. All experiments were performed with intact monolayers. To fully activate AF508-CFTR, forskolin (10 pM), PDE inhibitor, IBMX (100 pM) and CFTR potentiator, genistein (50 pM) were added to the apical side. 1003681 As observed in other cell types, incubation at low temperatures of FRT cells and human bronchial epithelial cells isolated from diseased CF patients (CF-HBE)expressing AF508 CFTR increases the functional density of CFTR in the plasma membrane. To determine the activity of correction compounds, the cells were incubated with test compound for 24-48 hours at 37 0 C and were subsequently washed 3X prior to recording. The cAMP- and genistein-mediated Isc in compound-treated cells was normalized to 37 0 C controls and expressed as percentage activity of CFTR activity in wt-HBE. Preincubation of the cells with the correction compound significantly increased the cAMP- and genistein-mediated Isc compared to the 37 0 C controls. 1003691 Identification of Potentiator Compounds 1003701 Typical protocol utilized a basolateral to apical membrane Cl concentration gradient. To set up this gradient, normal ringers was used on the basolateral membrane, whereas apical NaCl was replaced by equirnolar sodium gluconate (titrated to pH 7.4 with NaOH) to give a large Cl- concentration gradient across the epithelium. Forskolin (10 pM) and all test compounds were added to the apical side of the cell culture inserts. The efficacy of the putative AF508-CFTR potentiators was compared to that of the known potentiator, genistein. 1003711 2. Patch-clamp Recordings 1003721 Total Cl current in AF508-NIH3T3 cells was monitored using the perforated patch recording configuration as previously described (Rae, J., Cooper, K., Gates, P., & Watsky, M. (1991) .J. Neurosci. Methods 37, 15-26). Voltage-clamp recordings were performed at 22 'C using an Axopatch 200B patch-clamp amplifier (Axon Instruments Inc., Foster City, CA). The pipette solution contained (in mNJ) 150 N-methyl-D-glucamine (NMDG)-Cl, 2 MgCl2, 2 CaCl 2 , 10 EGTA, 10 HEPES, and 240 pg/mi amphotericin-B (p- adjusted to 7.35 with HCI). The extracellular - 87 - WO 2009/123896 PCT/US2009/038203 medium contained (in mM) 150 NMDG-Cl, 2 MgC 2 , 2 CaC 2 , 10 HEPES (pH adjusted to 7.35 with HCI). Pulse generation, data acquisition, and analysis were performed using a PC equipped with a Digidata 1320 A/D interface in conjunction with Clampex 8 (Axon Instruments Inc.). To activate AF508-CFTR, 10 pM forskolin and 20 pM genistein were added to the bath and the current-voltage relation was monitored every 30 sec. 1003731 Identification of Correction Compounds 1003741 To determine the activity of correction compounds for increasing the density of functional AF508-CFTR in the plasma membrane, we used the above-described perforated-patch recording techniques to measure the current density following 24-hr treatment with the correction compounds. To fully activate AF508-CFTR, 10 pM forskolin and 20pM genistein were added to the cells. Under our recording conditions, the current density following 24-hr incubation at 27 C was higher than that observed following 24-hr incubation at 37 'C. These results are consistent with the known effects of low-temperature incubation on the density of AF508-CFTR in the plasma membrane. To determine the effects of correction compounds on CFTR current density, the cells were incubated with 10 pM of the test compound for 24 hours at 37C and the current density was compared to the 27C and 37 0 C controls (% activity). Prior to recording, the cells were washed 3X with extracellular recording medium to remove any remaining test compound. Preincubation with 10 PM of correction compounds significantly increased the cAMP- and genistein-dependent current compared to the 37 0 C controls. [003751 Identification of Potentiator Compounds [003761 The ability of AF508-CFTR potentiators to increase the macroscopic AF508 CFTR Cl- current (IAF508) in NIH3T3 cells stably expressing AF508-CFTR was also investigated using perforated-patch-recording techniques. The potentiators identified from the optical assays evoked a dose-dependent increase in IAF 5 0 8 with similar potency and efficacy observed in the optical assays. In all cells examined, the reversal potential before and during potentiator application was around -30 mV, which is the calculated Eci (-28 mV). [003771 Cell Culture [003781 NIH3T3 mouse fibroblasts stably expressing AF508-CFTR are used for whole cell recordings. The cells are maintained at 37 'C in 5% CO 2 and 90 % humidity in Dulbecco's modified Eagle's medium supplemented with 2 mM glutamine, 10 % fetal bovine serum, 1 X NEAA, f3-ME, I X pen/strep, and 25 mM HEPES in 175 cm 2 culture flasks. For whole-cell recordings, 2,500 - 5,000 cells were seeded on poly-L-lysine-coated glass coverslips and cultured - 88 - WO 2009/123896 PCT/US2009/038203 for 24 - 48 hrs at 27 'C before use to test the activity of potentiators; and incubated with or without the correction compound at 37 'C for measuring the activity of correctors. [003791 3.Single-channel recordings [003801 Gating activity of wt-CFTR and temperature-corrected AF508-CFTR expressed in NIH3T3 cells was observed using excised inside-out membrane patch recordings as previously described (Dalemans, W., Barbry, P., Champigny, G., Jallat, S., Don, K., Dreyer, D., Crystal, R.G., Pavirani, A., Lecocq, J-P., Lazdunski, M. (1991) Nature 354, 526 - 528) using an Axopatch 200B patch clamp amplifier (Axon Instruments Inc.). The pipette contained (in mM): 150 NMDG, 150 aspartic acid, 5 CaCl 2 , 2 MgCl 2 , and 10 HEPES (p-I adjusted to 7.35 with Tris base). The bath contained (in mM): 150 NMDG-CI, 2 MgCl2, 5 EGTA, 10 TES, and 14 Tris base (p1- adjusted to 7.35 with 1-IC). After excision, both wt- and AF508-CFTR_ were activated by adding 1 mM Mg-ATP, 75 nM of the catalytic subunit of cAMP-dependent protein kinase (PKA; Promega Corp. Madison, WI), and 10 mM NaF to inhibit protein phosphatases, which prevented current rundown. The pipette potential was maintained at 80 mV. Channel activity was analyzed from membrane patches containing < 2 active channels. The maximum number of simultaneous openings determined the number of active channels during the course of an experiment. To determine the single-channel current amplitude, the data recorded from 120 see of AF508-CFTR activity was filtered "off-line" at 100 Hz and then used to construct all-point amplitude histograms that were fitted with multigaussian functions using Bio-Patch Analysis software (Bio-Logic Comp. France). The total microscopic current and open probability (P,) were determined from 120 sec of channel activity. The Po was determined using the Bio-Patch software or from the relationship P 0 = I/i(N), where I = mean current, i = single channel current amplitude, and N = number of active channels in patch. [003811 Cell Culture [003821 NIH3T3 mouse fibroblasts stably expressing AF508-CFTR are used for excised membrane patch-clamp recordings. The cells are maintained at 37 'C in 5% CO 2 and 90 % humidity in Dulbecco's modified Eagle's medium supplemented with 2 mM glutamine, 10 % fetal bovine serum, I X NEAA, f-3-NE, I X pen/strep, and 25 mM H-EPES in 175 cm 2 culture flasks. For single channel recordings, 2,500 -- 5,000 cells were seeded on poly-L--lysine-coated glass coverslips and cultured for 24 -- 48 hrs at 27 'C before use. [003831 The compounds of Table 1 were found to exhibit Correction activity as measured in the assay described above. - 89 - WO 2009/123896 PCT/US2009/038203 [003841 Compounds of the invention are useful as modulators of ATP binding cassette transporters. Using the procedures described above, the activities, i.e., EC50s, of compounds of the present invention have been measured and are shown in Table 6. 1003851 Table 6. ~C50 E C50 3ms++ < 2 05 flinnedEC5G'nd~x cc 1 +- ++ 2 +++ 3 +++ ++ 4 4+ +++ 5 ++++ 6 + ++ 7 +++++ 8 ++4 ++ 9 ++++ 10 ++ ++ 11 ++++ 13 ++-++ 14 ++ +++4 15 ++ + 16 +++ ++++ 17 +++ 18 +++4 ++ 19 + ++ 20 ++ ++ 21 + 4+++ 22 +++ ++ -90 - WO 2009/123896 PCT/US20091'038203 .. .. .. ... .. ... .. ... .. ... .. . ........... 2 .tc t v t .. ... .... .... .... ... .... .... .... .... ... .... .... .... .. X ... ... .. .. .. ........... 3. .......... .... ................... + ......................... 2 4............ 2 5 +.. .. . . . . . .. . . .. .. . . . . . .. . . . . .. . .. . .. . . . . . .. . . . . .. .. . . . .. . . . . .. . . . . . 2 ..... ..... ... .. ........... ........... .......... 27-X +++** ............... 28.... .+ ... .. 2.. . ... ..... .. .. ..... ...... .... 3 0 ... .............. ................. ................ 3 1.. .. .. ... ......... .. .. . 32....... 3 3. ....... ......... ....... .. .. ... . 3 4. . .. .......... .............. .............. ............... ........................... 3 5...... ...... +.+ ......... .... 36............... +4+d 37................. .. ......0 .. .... .. 23 .. ... 45 + + 46 ++.4-. 47 + +4+. 48 + 4-4. 31 + +91 + WO 2009/123896 PCT/US20091'038203 .. .. .. ... .. ... .. ... .. ... .. . .......... 2. .tc t v t .. .... .... ..... .... .... ..... .... .... ..... .... .... ... X ... ... .. 4.. . .......................... 9. .... ...................... ................ 5 0.............. 5 1.. . . .. . . . . . .. . . . . .. . . . . .. . . . . .. . . .. . .. . . . . .. . . . . .. . . . . . .. . . . . .. . . . . . 5 2 ... ........ ... .......... ...... .. ............ 53-X +++** ............... 5 4.... . . ....... . 5 5.. ... ..... .. .. ..... ...... .... 5 6 ... ........ ..................... .. ...... .....-. .... 5. .... .... ....... .. ... 58....... .. ........ ... 9. .... ....... ............. 6 0. . ... ......... .............. ............... .............. ........................... 6 1.. .. .... .... X .. .... ... . .... 62............... i* d: 63............... 64................. 6 5.. .. .. .. .. .. .. . 6 6.............. 6 7 ... ... .... .. ............ .. ..... ..... - 6 8.... .. ....... 69 .. . 70 +- + 51 ++4-+ 72 ++44. 73 ++ .. 74 + + + + 552 WO 2009/123896 PCT/US20091'038203 .. .. .. ... .. ... .. ... .. ... .. . .......... 2. .tc t v t .. .... .... ..... .... .... ..... .... .... ..... .... .... ... X ... ... .. .. .. ............... ........... 5. ..... ................. +................ .. . .. . . .. . . .. . . 7 7 + + +.. .. . . .. . . . .. .. . . . . ... . . . . .. . .. . .. . . . . .. . . . . . .. .. . . . .. . . .. . . . . . . . . 7 8 .... ........ .. ..... ..... ..... ..... ..... ..... .... -X-XK ~ ~~~ ~~ 93+-* ................

Claims (38)

1. A compound of formula I: (R )x N 4(W (R")n). N1 4 A RN 3 or a pharmaceutically acceptable salt thereof, wherein: RN is H, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocycloalkyl; A is an optionally substuted 3-7 membered monocyclic ring; B is optionally fused to a 5-7 membered ring selected from the group consisting of cycloaliphatic, aryl, heterocyclic, and heteroaryl; R 1 is halo, aikyl, OH, alkoxy, thioalkyl, trifluoromethoxy; or J two Ri on adjacent atoms, taken together, form ; wherein J is selected from the group consisting of CH 2 , CF 2 , or C(C-1 3 ) 2 ; W is independently a bond or a (C -C6) alkylidene chain wherein up to two methviene units of W are independently replaced by -CO-, -0-, -S- -S02-, or -NR-; R' is independently H, alkyl, aryl, heteroaryl, aralkyl, cycloalkyl, or heterocyclic: Rw is independently H, halo, CN, NO2, N(R) 2 , CF3, OCF 3 , OH, OR, C(O)R, CO 2 R, C(O)N(R) 2 , -O(Cl-C6)alkvlidene-OR, -O(CI -C6)alkylidene-N(R) 2 , -O(Cl C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl. aryloxy., heterocyclic, or heteroaryl, wherein, when substituted, R* is substituted with up to two R2 R2 is hliio, CN, NO 2 , CF 3 , OCF 3 , OR, OC(O)R, OC(O)N(R)2, SR, S(O)R, SO2R, SO 2 N(R) 2 , SO 3 R, C(O)R, CO 2 R, C(O)N(R) 2 , N(R) 2 , NRC(O)R, NRCO2R, NR-C(O)N(R)2, NRSO 2 R., B(OR) 2 ., or NRSO 2 N(R) 2 ; R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; - 94 - WO 2009/123896 PCT/US2009/038203 n is I or 2; w is an integer from 0 to 4 inclusive; and x is an integer from 0 to 5 inclusive; provided that when W is a bond and R" is cycloaliphatic, heterocycloaliphatic, aryl, or heteroaryl, -W-R" is attached to the 3- or 4- position of the pyridyl ring.
2. The compound of claim 1, wherein A is selected from the group consisting of: (R) (R 3 /(R3) (R 3 ) (R) a b c d and e, wherein R is alkyl, alkaryl, aryl, or heteroaryl; and q is an integer from 0 to 4 inclusive. (R )3
3. The compound of claim 2, wherein A is
4. The compound of claim 2, wherein A is
5. The compound of claim 1, wherein R is selected from the group consisting of chloro. methyl, OH, methoxy, thionethyl, and trifluoromethoxy. 0
6. The compound of claim 1, wherein two R on adjacent atoms, taken together, form wherien J is selected from the group consisting of CHF, CF, or C(CI-1) 2 .
7. The compound of claim 6, wherein J is C112.
8. The compound of claim 6, wherein J is CF 2 .
9. The compound of claim 1, wherein RN is H or alkyl.
10. The compound of claim 1, wherein W is a bond.
11. The compound of claim 1, wherein W is an optionally substituted (Cl -C6) alkylidene chain. - 95 - WO 2009/123896 PCT/US2009/038203
12. The compound of claim 1, wherein W is selected from the group consisting of -CH 2 -, -N-i-, -0-, -CO-, and -OCH2-.
13. The compound of claim 1, wherein R' is selected from the group consisting of H, OH, heterocyclic, aryl, phenyl, heteroaryl, pyridyl, alkoxy, cycloalkyl, and cyclohexyl.
14. The compound of claim 1, wherein -W-R' is acyclic.
15. The compound of claim 1, wherein -W-Rw is an aryl, heteroaryl, cycloaliphatic, or heterocycloaliphatic ring.
16. The compound of claim 1, wherein -W(R"), is selected from the following: 0 H OH 0 N -F, -CH 3 , -CH 2 CH 3 , -CN, -CF 3 , -CONH 2 , -CH 2 CH(CH 3 ) 2 , "'o 0 0 N 0 11 01 N x~~ N> A K HN 0 N 0 O O HN 0 O0 0 , OH, OH, OH, OH -96- WO 2009/123896 PCT/US20091'038203 Ic0 0 ~ N N I 0 N CN K N A 0 N -" 0 0 011 N 1l~ NN 00 NH2 NOH 'Ncl6 o0,, 0 H N 0: 0 / ~ B(0H) 2 0-, 0 HNJN N2 ,or SN 0 WO 2009/123896 PCT/US2009/038203 H 0 N I7. The compound of claim 1, wherein -W(R")n selected from the following: HO 0 H2NO !s~ ~B(OH) 2 H HN 2 N O NH
18. The compound according to claim 1, having formula 1a: O ON N J\ (01 W(R-),), 0 N!Z H 3 Ia or a pharmaceutically acceptable salt thereof, wherein: J is CH2 or CF2; W is independently a bond or a (Ci-C6) alkylidene chain optionally substituted with R 2 wherein up to two methylene units of W are independently replaced by -CO-, -0-, or R' is independently H or alkyl; Rw is independently 1-1, halo, CN, N(R) 2 , CF 3 , 0H, CO 2 R, C(O)N(R) 2 , -O(Cl C6)alkylidene-OR, -O(Cl-C6)alkylidene-N(R) 2 , -O(CI-C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, R" is substituted with up to two R2; R 2 is halo, OR, CO 2 R, C(O)N(R) 2 , SO 2 N(R)2, B(OR) 2 , or N(R) 2 ; R is independently H, alkyl, cycloalkl, heterocyclic, aryl, or heteroaryl; n is I or 2; and w is an integer from 0 to 4 inclusive; provided that when W is a bond and RI is cycloaliphatic, heterocycloaliphatic, aryl, or heteroaryl, -W-R" is attached to the 3- or 4- position of the pyridyl ring. - 98 - WO 2009/123896 PCT/US2009/038203
19. The compound of claim 18, wherein J is CI2.
20. The compound of claim 18, wherein J is CF2.
21. The compound of claim 18, wherein -W-R" is acyclic.
22. The compound of claim 18, wherein --- W -R" is an aryl, heteroaryl, cycloaliphatic, or heterocycloaliphatic ring.
23. The compound of claim 18, wherein -W(R")n is selected from the following: 0 H OH 0 N -F, -CH3[,, -CH12CH1, -CN, -CF3, -CONH2l, -CH2ICH(CH3l)2, , 01 ~ N H2N N 0 N 0 0 N -99- N 2 N A 0 OH"HN 0N NN 1? H I o NN O 0H, OH, O H, O H 0 0 N N N 111 0 N - 99 - WO 2009/123896 PCT/US2009/038203 0 0 NH B(OH) O H HNo N 00 H 0~ N.N 24O The compound of claim 18, wherein -W(Rw)a selected from the following: H, HO O N. H2N B(OH)2 O N 0orN NHH 2am or F12 NH2 0 HH -0 HO 0 tooH WO 2009/123896 PCT/US2009/038203 Ib or a pharmaceutically acceptable salt thereof, wherein: R 1 is halo, alkyl, OH, alkoxy, thioalkyl, trifluoromethoxy; or J two Ri on adjacent atoms, taken together, form wherein J is selected from the group consisting of CH2, CF 2 , or C(CH 3 ) 2 ; W is -NR', -CO-, or -Cl- 2 -; R' is independently H or alkyl; Rw is independently H, halo, CN, N(R) 2 , CF3, OH, CO2R, C(O)N(R)2, -O(Cl C6)alkylidene-OR, -O(Cl-C6)alkyiidene--N(R)2, -- O(Cl-C6)alkylidene-heterocyclic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, R'' is substituted with up to two R 2 ; R 2 is halo, OR, CO 2 R, C(O)N(R) 2 , SO 2 N(R) 2 , B(OR) 2 , or N(R) R is independently H, alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl; n is I or 2; w is an integer from 0 to 4 inclusive, and x is an integer from 0 to 5 inclusive. J
26. The compound of claim 25, wherein two R- on adjacent atoms, taken together, form and J is CH2. J
27. The compound of claim 25, wherein two R' on adjacent atoms, taken together, form o and J is CF 2 .
28. The compound of claim 25, wherein -- W(R'), is selected from the following: - 101 - WO 2009/123896 PCT/US20091'038203 -CH 3 , -CH 2 CH 3 , -CH 2 CH(CH 3 ) 2 , OH 011 N0 AN OH 0AHN\ 0OHOH N I N A2 0 yq (N AN OH OH, 0 H,2 0: 0 O N) 0N I-1 NA CI NHNN NN .102- WO 2009/123896 PCT/US2009/038203 H N 0 Si, , Nhox; o NHH2Q ,or
29. The compound according to claim I having formula C: H or a pharmaceutically acceptable salt thereof, wherein: R' is halo, ai k l, OH, alkox , t ioalkyl, trifluoromethoxC; or J\ two R on adjacent atoms , taken together, form wherein , is selected from the group consisting of CH, iF, Or C(CH 3 ) 2 ; R2 is -hal, O-, or ---C(S, R is independently H or akyl; R is independently H, halo, N(R), M, OH, C0 2 R, C(OWI(R) 2 . -O(Ci C6)alkylidenc-OR, -O(C 1-C6(ilak di den-e-N(R)' 2 , -O(C I-C6)aikyiide-ne-heterocyciic, or an optionally substituted aliphatic, cycloaliphatic, aryl, aryloxy, heterocyclic, or heteroaryl, wherein, when substituted, Rw' is substituted with up to two R; s halo, OR, CO 2 R, C(O)N(R) 2 SON(R) 2 , (Ro ( h * is independently H. alkyl,. cycloalk-yl, heterocyclic, aryl, or heteroaryl; n is I or 2; w is an integer from 0 to 4 inclusive; and x is an integer from 0 to 5 inclusive. -103- WO 2009/123896 PCT/US2009/038203 /C)3 J
30. The compound of claim 29, wherein two R on adjacent atoms, taken together, form and J is CH2. J
31. The compound of claim 29, wherein two R on adjacent atoms, taken together, form o and J is CF 2 .
32. The compound of claim 29, wherein -- W(RW), is selected from the following: -CH 3 , -CH2CH 3 , -CH2?CH(CH3) -1O OH ' -O 0 OH,0 OHN) NN H NN O OHOH OH OH' OHNOO 'N'N I O N 0N OH, OH, OH, N 0 'N, 0 C N 0 N> Y N OH, 0I K0 "0 ) 0) 0 N C~7 N> I 'N1 N. .104- WO 2009/123896 PCT/US2009/038203 N NN O0 NH CI NH B(OH) 2 H , r NI NH H 2 ,or 0
33. The compound of claim 1, wherein the compound is selected from Table 1.
34. A pharmaceutical composition comprising (i) a compound of claim 1; and (ii) a pharmaceutically acceptable carrier.
35. The composition of claim 34, further comprising an additional agent selected from the group consisting of a mucolytic agent, bronchodialator, an anti-biotic, an anti-infective agent, an anti-inflammatory agent, CFTR corrector, and a nutritional agent.
36. A method of modulating ABC transporters in a membrane of a cell, comprising the step of contacting said cell with a compound according to claim 1.
37. The method of claim 36, wherein the ABC transporter is CFTR.
38. A method of treating a condition, disease, or disorder in a patient implicated by ABC transporter activity, comprising the step of administering to said patient a compound of any one of claims 1 to 33.
39. The method of claim 38, wherein the ABC transporter is CFTR.
40. The method of claim 38, wherein said condition, disease, or disorder is selected from cystic fibrosis, hereditary emphysema, hereditary hem ochromatosis, coagulation-fibrinolysis deficiencies, such as protein C deficiency, Type 1 hereditary angioedema, lipid processing deficiencies, such as familial hypercholesterolemia, Type I chylomicronemia, abetalipoproteinemia, lysosomal storage diseases, such as I-cell disease/pseudo-Hurler, m ucopolysacchiaridoses, Sandhof/Tay-Sachs, Crigler-Najjar type II, polyendocrinopathy/hyperinsulemia, diabetes mellitus, laron dwarfism, -105- WO 2009/123896 PCT/US2009/038203 myleoperoxidase deficiency, primary hypoparathyroidism, melanoma, glycanosis CDG type 1, hereditary emphysema., congenital hyperthyroidism, osteogenesis imperfecta, hereditary hypofibrinogenemia, ACT deficiency., diabetes insipidus (di), neurophyseal di, neprogenic DI, Charcot-Marie Tooth syndrome, Perlizaeus-Merzbaclier disease, neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear plasy, Pick's disease, several polyglutamine neurological disorders asuch as Huntington, spinocerebullar ataxia type I, spinal and bulbar muscular atrophy, dentatorubal pallidoluysian, and myotonic dystrophy, as well as spongiform encephalopathies, such as hereditary Creutzfeldt-Jakob disease, Fabry disease, Straussler-Scheinker syndrome, COPD, dry-eye disease, and Sjbgren's disease.
41. A kit for use in measuring the activity of a ABC transporter or a fragment thereof in a biological sample in vitro or in vivo, comprising: (i) a first composition comprising a compound according to claim 1; and (ii) instructions for: a) contacting the composition with the biological sample; b) measuring activity of said ABC transporter or a fragment thereof. -106-
AU2013270464A 2008-03-31 2013-12-10 Pyridyl derivatives as CFTR modulators Ceased AU2013270464B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013270464A AU2013270464B2 (en) 2008-03-31 2013-12-10 Pyridyl derivatives as CFTR modulators

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US61/040,776 2008-03-31
AU2009231993A AU2009231993B2 (en) 2008-03-31 2009-03-25 Pyridyl derivatives as CFTR modulators
AU2013270464A AU2013270464B2 (en) 2008-03-31 2013-12-10 Pyridyl derivatives as CFTR modulators

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2009231993A Division AU2009231993B2 (en) 2008-03-31 2009-03-25 Pyridyl derivatives as CFTR modulators

Publications (2)

Publication Number Publication Date
AU2013270464A1 true AU2013270464A1 (en) 2014-01-09
AU2013270464B2 AU2013270464B2 (en) 2016-05-26

Family

ID=49883392

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013270464A Ceased AU2013270464B2 (en) 2008-03-31 2013-12-10 Pyridyl derivatives as CFTR modulators

Country Status (1)

Country Link
AU (1) AU2013270464B2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11155533B2 (en) 2017-10-19 2021-10-26 Vertex Pharmaceuticals Incorporated Crystalline forms and compositions of CFTR modulators
US11179367B2 (en) 2018-02-05 2021-11-23 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for treating cystic fibrosis
US11253509B2 (en) 2017-06-08 2022-02-22 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11414439B2 (en) 2018-04-13 2022-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11426407B2 (en) 2014-10-06 2022-08-30 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US11434201B2 (en) 2017-08-02 2022-09-06 Vertex Pharmaceuticals Incorporated Processes for preparing pyrrolidine compounds
US11453655B2 (en) 2016-12-09 2022-09-27 Vertex Pharmaceuticals Incorporated Modulator of the cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11465985B2 (en) 2017-12-08 2022-10-11 Vertex Pharmaceuticals Incorporated Processes for making modulators of cystic fibrosis transmembrane conductance regulator
US11517564B2 (en) 2017-07-17 2022-12-06 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0081756B1 (en) * 1981-12-14 1985-05-15 MEDEA RESEARCH S.r.l. New compounds with antiinflammatory and antitussive activity, process for their preparation and relative pharmaceutical compositions
TW200307539A (en) * 2002-02-01 2003-12-16 Bristol Myers Squibb Co Cycloalkyl inhibitors of potassium channel function
WO2007056341A1 (en) * 2005-11-08 2007-05-18 Vertex Pharmaceuticals Incorporated Heterocyclic modulators of atp-binding cassette transporters
AU2005210474B2 (en) * 2004-01-30 2011-07-07 Vertex Pharmaceuticals Incorporated Modulators of ATP-Binding cassette transporters

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11426407B2 (en) 2014-10-06 2022-08-30 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator
US11453655B2 (en) 2016-12-09 2022-09-27 Vertex Pharmaceuticals Incorporated Modulator of the cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
US11253509B2 (en) 2017-06-08 2022-02-22 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11517564B2 (en) 2017-07-17 2022-12-06 Vertex Pharmaceuticals Incorporated Methods of treatment for cystic fibrosis
US11434201B2 (en) 2017-08-02 2022-09-06 Vertex Pharmaceuticals Incorporated Processes for preparing pyrrolidine compounds
US11155533B2 (en) 2017-10-19 2021-10-26 Vertex Pharmaceuticals Incorporated Crystalline forms and compositions of CFTR modulators
US11465985B2 (en) 2017-12-08 2022-10-11 Vertex Pharmaceuticals Incorporated Processes for making modulators of cystic fibrosis transmembrane conductance regulator
US11179367B2 (en) 2018-02-05 2021-11-23 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions for treating cystic fibrosis
US11414439B2 (en) 2018-04-13 2022-08-16 Vertex Pharmaceuticals Incorporated Modulators of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator

Also Published As

Publication number Publication date
AU2013270464B2 (en) 2016-05-26

Similar Documents

Publication Publication Date Title
AU2009231993B2 (en) Pyridyl derivatives as CFTR modulators
US9751890B2 (en) Heteroaryl derivatives as CFTR modulators
AU2007351454B2 (en) Azaindole derivatives as CFTR modulators
AU2013270464B2 (en) Pyridyl derivatives as CFTR modulators
US8563573B2 (en) Azaindole derivatives as CFTR modulators
AU2015228930B2 (en) Heteroaryl derivatives as CFTR modulators
AU2013231026B2 (en) Azaindole derivatives as CFTR modulators
AU2013205162B2 (en) Heteroaryl derivatives as CFTR modulators

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired