NZ792410A - Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator - Google Patents

Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator

Info

Publication number
NZ792410A
NZ792410A NZ792410A NZ79241017A NZ792410A NZ 792410 A NZ792410 A NZ 792410A NZ 792410 A NZ792410 A NZ 792410A NZ 79241017 A NZ79241017 A NZ 79241017A NZ 792410 A NZ792410 A NZ 792410A
Authority
NZ
New Zealand
Prior art keywords
compound
chosen
mmol
groups
pharmaceutically acceptable
Prior art date
Application number
NZ792410A
Inventor
Alexander Russell Abela
Timothy Alcacio
Corey Anderson
Minson Baek
Brett Bradley Busch
Thomas Cleveland
Bryan A Frieman
Peter Grootenhuis
Ruah Sara Sabina Hadida
Robert M Hughes
Original Assignee
Vertex Pharmaceuticals Incorporated
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Publication of NZ792410A publication Critical patent/NZ792410A/en

Links

Abstract

Compounds of Formula (I), pharmaceutically acceptable salts thereof, deuterated derivatives of any of the foregoing, and metabolites of any of the foregoing are disclosed. Pharmaceutical compositions comprising the same, methods of treating cystic fibrosis using the same, and methods for making the same are also disclosed. same are also disclosed.

Description

nds of Formula (I), pharmaceutically acceptable salts thereof, deuterated derivatives of any of the foregoing, and metabolites of any of the foregoing are disclosed. Pharmaceutical compositions comprising the same, methods of treating cystic fibrosis using the same, and methods for making the same are also disclosed.
NZ 792410 Modulator of Cystic Fibrosis Transmembrane Conductance Regulator, Pharmaceutical Compositions, Methods of Treatment, and Process for Making the Modulator This application is a divisional application of New Zealand patent application 752486, which is the national phase entry in New Zealand of PCT international application (published as content of each of which are incorporated herein by reference.
Disclosed herein is a modulator of Cystic Fibrosis Transmembrane tance Regulator (CFTR), pharmaceutical compositions ning the modulator, s of treatment of cystic fibrosis, and a process for making the modulator.
Cystic is (CF) is a recessive c disease that affects approximately 70,000 children and adults worldwide. Despite progress in the treatment of CF, there is no cure.
In patients with CF, ons in CFTR endogenously expressed in respiratory epithelia lead to reduced apical anion secretion causing an imbalance in ion and fluid transport. The ing decrease in anion transport contributes to enhanced mucus accumulation in the lung and accompanying microbial infections that ultimately cause death in CF patients. In addition to respiratory disease, CF patients lly suffer from gastrointestinal problems and pancreatic insufficiency that, if left untreated, result in death.
In addition, the majority of males with cystic fibrosis are infertile, and ity is reduced among females with cystic fibrosis.
Sequence analysis of the CFTR gene has revealed a variety of disease causing mutations (Cutting, G. R. et al. (1990) Nature 346:366-369; Dean, M. et al. (1990) Cell :870; and Kerem, B-S. et al. (1989) Science 73-1080; Kerem, B-S et al. (1990) Proc. Natl. Acad. Sci. USA 7-8451). To date, greater than 2000 mutations in the CF gene have been identified; currently, the CFTR2 database contains information on only 322 of these identified mutations, with sufficient evidence to define 281 mutations as disease g. The most prevalent disease-causing mutation is a on of phenylalanine at position 508 of the CFTR amino acid sequence, and is commonly referred to as the F508del mutation. This mutation occurs in approximately 70% of the cases of cystic fibrosis and is associated with severe disease.
The deletion of residue 508 in CFTR prevents the nascent protein from folding correctly. This results in the inability of the mutant protein to exit the endoplasmic lum (ER) and traffic to the plasma membrane. As a result, the number of CFTR channels for anion transport present in the membrane is far less than observed in cells sing wild-type CFTR, i.e., CFTR having no mutations. In addition to impaired trafficking, the mutation results in defective channel gating.
Together, the reduced number of channels in the membrane and the defective gating lead to d anion and fluid transport across epitheha. (Quinton, P. M. (1990), FASEB J. 4: 2709-2727). The channels that are defective because of the F508del mutation are still functional, albeit less onal than wild-type CFTR channels. ans et al. (1991), Nature Lond. 354: 526-528; Pasyk and Foskett (1995), J. Cell.
Biochem. 270: 12347-50). In addition to F508del, other disease causing mutations in CFTR that result in defective trafficking, synthesis, and/or channel gating could be upor down-regulated to alter anion secretion and modify disease progression and/or severity.
CFTR is a cAMP/ATP-mediated anion channel that is sed in a variety of cell types, including absorptive and secretory lia cells, where it tes anion flux across the membrane, as well as the activity of other ion channels and proteins. In epithelial cells, normal functioning of CFTR is critical for the nance of electrolyte transport throughout the body, including respiratory and digestive tissue.
CFTR is composed of approximately 1480 amino acids that encode a protein which is made up of a tandem repeat of transmembrane domains, each containing six transmembrane helices and a nucleotide binding domain The two transmembrane domains are linked by a large, polar, regulator}' (R)-domain with multiple phosphory lation sites that regulate channel activity and cellular trafficking.
Chloride ort takes place by the coordinated activity of ENaC and CFTR present on the apical membrane and the Na+-K+-ATPase pump and CT channels expressed on the basolateral surface of the cell. Secondary' active ort of chloride from the luminal side leads to the accumulation of intracellular chloride, which can then ely leave the cell via Cl" channels, resulting in a vectorial transport. Arrangement of Na+/2C17K+ co-transporter, Na+-K+-ATPase pump and the basolateral membrane K+ channels on the basolateral surface and CFTR on the luminal side coordinate the secretion of chloride via CFTR on the luminal side. e water is probably never actively transported itself, its flow across epithelia depends on tiny transepithelial osmotic gradients ted by the bulk flow of sodium and chloride.
Accordingly, there is a need for novel treatments of CFTR ed diseases.
SUBSTITUTE SHEET (RULE 26) sed herein are novel compounds, including compounds of Formulae I- IV and pharmaceutically acceptable salts thereof. For example, compounds of Formula I can be depicted as: 9 o x ...A., N S,R’r L-ArV or a pharmaceutically acceptable salt thereof, wherein: - one of Y1 and Y2 is N and the other is CH; - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, wherein each R is ndently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is ndently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is ndently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently a halogen; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8.
SUBSTITUTE SHEET (RULE 26) Also disclosed herein are pharmaceutical compositions comprising at least one of the novel compounds disclosed herein and/or at least one pharmaceutically acceptable salt thereof, which compositions may further include at least one additional active pharmaceutical ingredient and/or at least one carrier. Also disclosed are methods of treating the CFTR-mediated disease cystic fibrosis comprising stering at least one of the novel compounds disclosed herein and/or at least one pharmaceutically acceptable salt thereof, optionally as part of a pharmaceutical composition comprising at least one additional component, to a subject in need thereof.
Also disclosed are methods of treating the CFTR-mediated disease cystic fibrosis sing administering at least one of the novel compounds disclosed herein and/or at least one pharmaceutically acceptable salt thereof, -(2.2- difluorobenzo[d][l,3]dioxolyl)-N-(l-(2,3-dihydroxypropyl)fluoro(l-hydroxy- ylpropanyl)-lH-mdolyl)cyclopropanecarboxamide (Compound II), andN- [2,4-bis(l,l-dimethylethyl)hydroxyphenyl]-l,4-dihydrooxoquinoline carboxamide (Compound III), optionally as part of at least one pharmaceutical ition comprising at least one additional component, to a t in need thereof.
Brief Description of the Drawings shows the structures of non-limiting examples of novel compounds disclosed herein. is a X-ray Powder ctogram (“XRPD”) of a spray dried dispersion (SDD) of 50% Compound 1 in HPMCAS-HG. is a spectrum showing a modulated differential scanning calorimetry (MDSC) spectrum of a spray dried dispersion (SDD) of 50% Compound 1 in HPMCAS-HG. is a entative list of CFTR genetic mutations. is an XRPD of a sample of the sodium salt of nd 1 prepared as reported in the e of the sodium salt of Compound 1.
Definitions As used herein, the term “alkyl” refers to a saturated, branched or unbranched aliphatic hydrocarbon containing carbon atoms (such as, for example, 1, 2, SUBSTITUTE SHEET (RULE 26) 3, 4, 5, 6, 7, 8, 9, 10,11, 12, 13,14, 15,16,17,18,19, or 20 carbon atoms). Alkyl groups may be substituted or unsubstituted.
The term “alkoxy” as used herein refers to an alky l or cycloalkyl covalently bonded to an oxygen atom. Alkoxy groups may be substituted or unsubstituted.
As used herein, “cycloalkyl” refers to a cyclic, bicyclic, tricyclic, or clic non-aromatic hydrocarbon groups having 3 to 12 s (such as, for example 3-10 carbons). “Cycloalkyf’ groups encompass monocyclic, bicyclic, tricyclic, bridged, fused, and spiro rings, including mono spiro and dispiro rings. Non-limiting examples of cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, norbomyl, and o[2.0.2.1]heptane. Cycloalkyl groups may be substituted or unsubstituted.
“Substituted,” whether preceded by the term “optionally” or not, tes that at least one hydrogen of the “substituted” group is replaced by a substituent. Unless otherwise indicated, an “optionally substituted” group may have a suitable 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 tuent chosen from a ied group, the substituent may be either the same or different at each position.
As used herein, “deuterated derivative(s)” means the same chemical structure, but with one or more en atoms replaced by a deuterium atom.
As used herein, “CFTR” means cystic fibrosis transmembrane conductance regulator.
As used herein, “mutations” can refer to mutations in the CFTR gene or the CFTR protein. A “CFTR gene on” refers to a on in the CFTR gene, and a “CFTR protein mutation” refers to a mutation in the CFTR protein. A genetic defect or mutation, or a change in the nucleotides in a gene in general results in a mutation in the CFTR protein translated from that gene, or a frame shift(s).
The term “F508del” refers to a mutant CFTR protein which is lacking the amino acid phenylalanine at on 508.
As used herein, a patient who is “homozygous” for a particular gene mutation has the same mutation on each allele.
SUBSTITUTE SHEET (RULE 26) As used , a patient who is ‘‘heterozygous” for a particular gene mutation has this mutation on one allele, and a different mutation on the other .
As used herein, the term “modulator” refers to a nd that increases the activity of a biological compound such as a protein. For example, a CFTR modulator is a compound that increases the activity of CFTR. The increase in activity resulting from a CFTR modulator includes but is not d to compounds that correct, potentiate, stabilize and/or amplify CFTR.
As used herein, the term “CFTR corrector” refers to a compound that facilitates the processing and trafficking of CFTR to se the amount of CFTR at the cell surface. Compounds of Formulae (I), (It), (III), (IV), and (V), and Compound II, and their pharmaceutically acceptable salts thereof disclosed herein are CFTR correctors.
As used herein, the term “CFTR potentiator” refers to a compound that ses the channel activity of CFTR protein located at the cell surface, resulting in ed ion transport. Compound III disclosed herein is a CFTR potentiator.
As used herein, the term “active pharmaceutical ingredient” (“API”) refers to a biologically active compound.
As used herein, the term “pharmaceutically acceptable salt” refers to a salt form of a compound of this disclosure wherein the salt is nontoxic. Pharmaceutically acceptable salts of the compounds of this disclosure include those derived from suitable inorganic and organic acids and bases. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, etal. describe pharmaceutically acceptable salts in detail in J. ceutical Sciences, 1977, 66, 1-19.
As used herein, the term “amorphous” refers to a solid material having no long range order in the position of its les. Amorphous solids are generally supercooled liquids in which the les are arranged in a random manner so that there is no well-defined arrangement, e.g., molecular packing, and no long range order.
Amorphous solids are generally isotropic, i.e. t similar properties in all directions and do not have definite melting points. For e, an amorphous material is a solid material having no sharp characteristic crystalline peak(s) in its X-ray power diffraction (XRPD) pattern (i.e., is not crystalline as determined by XRPD). Instead, one or several SUBSTITUTE SHEET (RULE 26) broad peaks (e.g., halos) appear in its XRPD pattern. Broad peaks are characteristic of an amorphous solid. See, US 2004/0006237 for a comparison of XRPDs of an amorphous material and crystalline material.
As used herein, the term "substantially amorphous" refers to a solid material having little or no long range order in the position of its molecules. For example, substantially amorphous materials have less than 15% crystallinity (e g., less than 10% crystallinity or less than 5% crystallinity). It is also noted that the term 'substantially amorphous' includes the descriptor, hous', which refers to als having no (0%) crystallinity.
As used herein, the term "dispersion" refers to a disperse system in which one substance, the dispersed phase, is distributed, in discrete units, throughout a second nce (the continuous phase or vehicle). The size of the sed phase can vary considerably (e.g. colloidal particles of nanometer dimension, to multiple microns in size). In general, the dispersed phases can be solids, liquids, or gases. In the case of a solid dispersion, the dispersed and continuous phases are both solids. In pharmaceutical ations, a solid dispersion can e a crystalline drug (dispersed phase) in an amorphous polymer (continuous phase); or alternatively, an amorphous drug (dispersed phase) in an amorphous polymer (continuous phase). In some embodiments, a solid dispersion includes the polymer tuting the dispersed phase, and the drug constitute the continuous phase. Or, a solid dispersion includes the drug constituting the dispersed phase, and the polymer constituting the continuous phase.
The terms “patient"’ and “subject” are used interchangeably and refer to an animal including humans.
The terms "effective dose" and "effective amount" are used interchangeably herein and refer to that amount of a compound that produces the desired effect for which it is administered (e.g., improvement in CF or a symptom of CF, or lessening the severity of CF or a symptom of CF). The exact amount of an effective dose will depend on the purpose of the ent, and will be ainable by one skilled in the art using known techniques (see, e.g., Lloyd (1999) The Art, e and Technolog}' of Pharmaceutical Compounding).
SUBSTITUTE SHEET (RULE 26) As used herein, the terms "treatment," "treating," and the like generally mean the improvement of CF or its symptoms or lessening the ty of CF or its symptoms in a subject. “Treatment,” as used herein, includes, but is not limited to, the following: increased growth of the subject, increased weight gain, reduction of mucus in the lungs, improved pancreatic and/or liver function, reduction of chest infections, and/or reductions in coughing or shortness of breath. ements in or lessening the severity of any of these symptoms can be readily assessed according to standard methods and techniques known in the art.
As used herein, the term “in combination with,” when referring to two or more compounds, , or additional active pharmaceutical ingredients, means the administration of two or more compounds, agents, or active pharmaceutical ingredients to the patient prior to, concurrent with, or subsequent to each other.
The terms “about” and “approximately”, when used in connection with doses, amounts, or weight percent of ients of a composition or a dosage form, include the value of a ied dose, amount, or weight percent or a range of the dose, amount, or weight percent that is recognized by one of ordinary skill in the art to provide a pharmacological effect equivalent to that obtained from the specified dose, amount, or weight percent.
Each of Compounds of Formulae (I), (II), (III), (IV), and (V), and nds II, III, IV, and ceutically able salts thereof described, and their deuterated derivatives herein independently can be administered once daily, twice daily, or three times daily. In some embodiments, at least one compound chosen from Compounds of Formulae (I), (II), (III), (IV), and (V), and pharmaceutically acceptable salts thereof, and their deuterated derivatives is administered once daily. In some embodiments, at least one compound chosen from Compounds of Formulae (I), (II), (III), (IV), and (V), and pharmaceutically acceptable salts thereof, and their deuterated derivatives are stered twice daily. In some embodiments, at least one compound chosen from Compound II and pharmaceutically acceptable salts thereof is stered once daily. In some embodiments, at least one compound chosen from nd II and pharmaceutically acceptable salts thereof is stered twice daily. In some ments, at least one compound chosen from Compound III and pharmaceutically acceptable salts thereof is administered once daily. In some embodiments, at least one SUBSTITUTE SHEET (RULE 26) compound chosen from Compound III and ceutically acceptable salts thereof is administered twice daily. In some ments, at least one compound chosen from nd IV and pharmaceutically acceptable salts thereof is administered once daily.
In some embodiments, at least one compound chosen from Compound IV and pharmaceutically acceptable salts thereof is administered twice daily. In some embodiments, a deuterated derivative of Compound II, III, and/or IV or a pharmaceutically acceptable salt thereof is employed in any one of these embodiments.
In some embodiments, 10 mg to 1,500 mg of a compound disclosed herein, a pharmaceutically acceptable salt thereof, or a deuterated derivative of such compound or salt are administered daily.
One of ordinary skill in the art would recognize that, when an amount of “a compound or a pharmaceutically acceptable salt thereof’ is disclosed, the amount of the pharmaceutically acceptable salt form of the compound is the amount equivalent to the concentration of the free base of the compound. It is noted that the disclosed amounts of the compounds or their pharmaceutically able salts thereof herein are based upon their free base form. For example, “10 mg of at least one compound chosen from compounds of Formula (I) and ceutically acceptable salts thereof’ includes 10 mg of a compound of Formula (I) and a concentration of a pharmaceutically acceptable salt of compounds of Formula (I) equivalent to 10 mg of nds of Formula (I).
As stated above, disclosed herein are compounds of Formula (I): 0 0. X (R2)P / N Y' N R1 -r (R3)q and pharmaceutically acceptable salts f, wherein: - one of Y1 and Y2 is N and the other is CH; - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; SUBSTITUTE SHEET (RULE 26) - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is ndently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, and wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl , OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - r is 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8.
Also disclosed herein are compounds of Formula (II): 0 Q XV// H X(R2)p R1 -r (R3)q (r4), and pharmaceutically acceptable salts thereof, - X is chosen from O, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 lkyl groups optionally substituted with one or more substituents each independently SUBSTITUTE SHEET (RULE 26) chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl , and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups ally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - r is 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1,2, 3, 4, or 5; and - qis 0, 1,2, 3, 4, 5, 6, 7, or 8.
Encompassed within the scope of ae (I) and (II) are compounds 0 0 0 0 ^ /NR'2 \^NRs comprising an - or n group (where R’ is H or C1-C4 alkyl), i.e., wherein X is chosen from NH and N(Ci-C4 alkyl) groups. Non-limiting examples of such compounds include compounds having the following structure: mO \\ // N rvs-.N W^o O F SUBSTITUTE SHEET (RULE 26) and pharmaceutically able salts thereof, either as a isomeric mixture or enantioenriched (e.g., >90% ee, >95% ee, or >98% ee) isomers.
Also disclosed herein are compounds of a (III): o 0 0V/ H X(R2)p N N N (R3)q (R4)r (III) and pharmaceutically acceptable salts thereof, wherein: - R1 is chosen from -(CR2)k(CR2)ra(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally tuted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, ns, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1,2, 3, 4, 5, 6, 7, or 8.
SUBSTITUTE SHEET (RULE 26) In some embodiments, in compounds of Formula (I), (II), (III), and pharmaceutically acceptable salts f, if R2 is cyano, then R2 is meta or para relative to the sulfur atom.
In some embodiments, in compounds of Formula (I), (II), (III), and pharmaceutically able salts thereof: - each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally tuted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and ns, and - each R is independently chosen from H and OH; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, and halogens; - R4 is F; - k is 0; - p is 0,1, or 2; - q is 0,1,2, 3, or 4; - r is 0; and - m and n are not 0 at the same time.
In some embodiments, in compounds of Formula (I), (II), (III), and pharmaceutically acceptable salts thereof: - R1 is chosen from (CR2)m-R'ng A groups, wherein Ring A is chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and ns, and - m is 1 or 2.
In some embodiments, in compounds of Formula (I), (II), (III), and pharmaceutically acceptable salts thereof, each R3 is a methyl group and q is 3 or 4.
Also disclosed herein are compounds of Formula (IV): ° °v ,° ¥ r^X^N'" K X<R2)P Ring A r N N N SUBSTITUTE SHEET (RULE 26) and pharmaceutically acceptable salts thereof, wherein: - Ring A is chosen from CN-Cio cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens; and - each R2 is independently chosen from C1-C2 alkyl groups, OH, F, Cl, and Ci- C2 alkoxy groups; - m is 1 or 2; and - p is 0, 1, or 2. In some ments, p is 0 or 1. In some embodiments, p is 0.
Also disclosed herein are compounds of Formula V: 0 0 Ov H X(r2)p J(t0 N\ XX Ring A ' N N N and pharmaceutically acceptable salts thereof, wherein: - Ring A is chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, nated C1-C2 alkyl groups, and halogens; and - each R2 is independently chosen from C1-C2 alkyl groups, OH, F, Cl, and Ci- C2 alkoxy groups; - m is 1 or 2; and - p is 0, 1, or 2.
In some embodiments, in compounds of Formula (I), (II), (III), (IV), (V), and ceutically acceptable salts f, each R2 is independently chosen from CH3, OH, F, and OCH3. In some embodiments, p is 0 or 1. In some embodiments, p is 0.
In some embodiments, in compounds of Formula (I), (II), (III), (IV), (V), and pharmaceutically acceptable salts thereof, Ring A is a cyclopropyl group substituted SUBSTITUTE SHEET (RULE 26) with a halogenated Ci alkyl group or a halogenated C2 alky] group. In some embodiments, Ring A is a cyclopropyl group substituted with a CF3 group.
In some embodiments, in compounds of Formula (I), (II), (III), (IV), (V), and pharmaceutically acceptable salts thereof, m is 1, Ring A is a cyclopropyl group substituted with a CF3 group, p is 0 or 1, and R2, if t, is a methyl group, a hydroxy group, or a methoxy group. In some embodiments, m is 2, Ring A is a cyclopropyl group substituted with a CF3 group, and p is 0.
In some embodiments, in compounds of Formula (I), (II), (III), (IV), (V), and pharmaceutically acceptable salts f, m is 2, Ring A is a C3 cycloalkyl group substituted with a CF3 group, p is 0 or 1, and R2, if present, is a methyl group, a hydroxy group, or a methoxy group. In some embodiments, m is 2, Ring A is a cyclopropyl group substituted with a CF3 group, and p is 0.
In some ments, m is 2, Ring A is a cyclopropyl group substituted with aCF3 group, and p is 0.
In some embodiments, in compounds of Formula (I), (II), (III), (IV), (V), and pharmaceutically acceptable salts thereof, Ring A is chosen from C5 oalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens. In some embodiments, Ring A is a C5 bicycloalkyl group optionally substituted with a halogen.
In some embodiments, in compounds of Formula (I), (II),(III), (IV), (V), and pharmaceutically acceptable salts thereof, Ring A is chosen from C7 bicycloalkyl groups and ycloalkyl groups optionally substituted with one or more substituents each ndently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens. In some embodiments, Ring A is an unsubstituted C7tricycloalkyl group.
Also disclosed herein are compounds having a formula chosen from any one of the formulae depicted in and pharmaceutically acceptable salts thereof.
Also sed herein are Compounds 1-5, 8, 10-16,18-30, 32, 33, 35-37, 39- 60, 63, and 64, and pharmaceutically acceptable salts thereof.
Also disclosed herein are Compounds 9, 31, 34, 38, 61, 62, and 65, and pharmaceutically acceptable salts thereof.
Also sed herein are Compounds 6, 7, and 17, and pharmaceutically acceptable salts thereof.
SUBSTITUTE SHEET (RULE 26) Also disclosed herein are ated derivatives of any one of Compounds 1- , 8,10-16,18-65, and pharmaceutically acceptable salts thereof.
Also disclosed herein are a compound having the following formula: rf^rVS"Ph O—P N N N-A(S) and pharmaceutically acceptable salts thereof Also sed herein are a compound having the following formula: O o n 0H (s)\^ 0 N N and pharmaceutically acceptable salts thereof.
Also disclosed herein are a compound having the following formula: 0 // N N N M OH and pharmaceutically acceptable salts thereof.
Also disclosed herein are a nd having the following formula: n-n^n^n H ,0 m.
SUBSTITUTE SHEET (RULE 26) and pharmaceutically acceptable salts thereof.
Also disclosed herein are a compound having the following formula: 0 0 o F QXr N N N"\S) ^ and pharmaceutically acceptable salts thereof.
Also disclosed herein are a compound having the following a: .s\\// N"N^N N k(S) and pharmaceutically acceptable salts thereof.
Also disclosed herein are a compound having the following formula: p // N N nA(S) and pharmaceutically acceptable salts thereof.
In some embodiments, at least one novel nd (and/or at least one pharmaceutically acceptable salt thereof and/or at least one ated derivative of such compound or salt) can be administered in combination with at least one additional active pharmaceutical ingredient. In some embodiments, at least one additional active ceutical ingredient is chosen from: (a) Compound II: SUBSTITUTE SHEET (RULE 26) FX J0 OH F 0 ^ 0 F N / NH; OH and pharmaceutically acceptable salts f.
A chemical name for Compound II is (//)-!-(2.2-diriuorobenzo|d|| 1.3 |dio\olyl)-/V- (l-(2,3-dihydroxypropyl)fluoro(l-hydroxymethylpropanyl)-lH-indol yl)cyclopropanecarboxamide; (b) Compound III: H and pharmaceutically acceptable salts thereof A chemical name for Compound III is ,V-(5-hydroxy-2.4-di-/m-biityl-phenyl)oxolH-quinolinecarboxamide ; and (c) Compound IV: 0^,0 H Ka ^ 0 H nV'N^X^ and pharmaceutically acceptable salts f A chemical name for Compound IV is l-(2,2-difluorobenzo[d][l,3]dioxol yl)cyclopropanecarboxamido)methylpyridinyl)benzoic acid.
Suitable pharmaceutically acceptable salts are, for example, those sed in S. M. Berge, etal. J. Pharmaceutical Sciences, 1977, 66, 1-19. For example, Table 1 of that article provides the following pharmaceutically acceptable salts: SUBSTITUTE SHEET (RULE 26) Table 1: Acetate Iodide Benzathine Benzenesulfonate Isethionate Chloroprocaine Benzoate Lactate Choline Bicarbonate Lactobionate Diethanolamine Bitartrate Malate Ethylenediamine Bromide Maleate Meglumine Calcium e Mandelate Procaine Camsylate Mesylate Aluminum Carbonate bromide Calcium Chloride Methylnitrate Lithium Citrate Methylsulfate Magnesium Dihydrochloride Mucate Potassium Edetate Napsylate Sodium Edisylate Nitrate Zinc Estolate Pamoate (Embonate) Esylate Pantothenate Fumarate Phosphate/diphosphate Gluceptate lacturonate Gluconate Salicylate Glutamate te Glycollylarsanilate Subacetate Hexylresorcinate Succinate Hydrabamine Sulfate Hydrobromide e Hydrochloride Tartrate Hy droxy naphthoate te Triethiodide Non-limiting examples of pharmaceutically acceptable salts derived from appropriate acids include: salts formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, or oric acid; salts formed with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or c acid; and salts formed by using other methods used in the art, such as ion exchange. Non-limiting examples of pharmaceutically acceptable salts include adipate, te, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, e, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, ophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, ionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nate, nitrate, oleate, oxalate, SUBSTITUTE SHEET (RULE 26) palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, e, tartrate, thiocyanate, ptoluenesulfonate , undecanoate, and valerate salts. ceutically acceptable salts derived from appropriate bases e alkali metal, alkaline earth metal, ammonium, and N+(Ci.4alkyl)4 salts. This disclosure also envisions the quatemization of any basic nitrogen-containing groups of the compounds disclosed herein. Suitable non-limiting es of alkali and alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium. Further non-limiting examples of pharmaceutically acceptable salts include ammonium, quaternary7 ammonium, and amine s formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate. Other suitable, non-limiting examples of pharmaceutically acceptable salts include besylate and glucosamine salts.
In some embodiments, at least one compound chosen from the novel nds disclosed herein, pharmaceutically acceptable salts thereof, and deuterated derivatives of the foregoing is administered in combination with at least one compound chosen from Compound II, pharmaceutically acceptable salts thereof, and ated derivatives of the foregoing. In some embodiments, at least one compound chosen from the novel compounds disclosed herein, pharmaceutically acceptable salts f, and deuterated tives of the ing is administered in combination with at least one compound chosen from Compound III and pharmaceutically acceptable salts thereof. In some embodiments, at least one compound chosen from the novel compounds disclosed herein, pharmaceutically able salts thereof, and deuterated derivatives of the foregoing is stered in combination with at least one compound chosen from Compound IV and pharmaceutically acceptable salts thereof. In some embodiments, at least one compound chosen from the novel compounds sed herein, pharmaceutically acceptable salts, and deuterated tives of the foregoing thereof is administered in combination with Compounds II or a pharmaceutically acceptable salt or deuterated tive thereof and at least one compound chosen from Compound III, pharmaceutically acceptable salts thereof, and deuterated derivatives of any of the ing. In some embodiments, at least one nd chosen from the novel compounds disclosed herein, pharmaceutically acceptable salts, and deuterated derivatives of any of the foregoing thereof is administered in combination with at least one compound chosen from Compound III, pharmaceutically acceptable salts thereof, SUBSTITUTE SHEET (RULE 26) and deuterated tives of any of the foregoing and at least one compound chosen from Compound IV, pharmaceutically acceptable salts thereof, and deuterated derivatives of any of the foregoing.
Any of the novel compounds disclosed herein, such as for example, compounds of Formula (I), (II), (III), (IV), (V), and their pharmaceutically acceptable salts thereof, and ated derivatives of such compounds and salts can be comprised in a single pharmaceutical composition or separate pharmaceutical compositions in combination with other additional active pharmaceutical ingredient(s) (e g., Compound II, III, or IV, or its ceutically acceptable salt thereof, or a ated tive of such Compound or salt). Such pharmaceutical compositions can be administered once daily or multiple times daily, such as twice daily. In some embodiments, the disclosure features a pharmaceutical composition comprising at least one compound chosen from any of the nds disclosed herein and pharmaceutically acceptable salts thereof, and at least one pharmaceutically able carrier.
In some embodiments, the disclosure features a pharmaceutical composition comprising at least one compound chosen from the novel compounds disclosed herein and pharmaceutically acceptable salts thereof, at least one compound chosen from Compound II and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier.
In some embodiments, the disclosure features a pharmaceutical ition comprising at least one compound chosen from the novel compounds disclosed herein and pharmaceutically acceptable salts thereof, at least one compound chosen from Compound III and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier.
In some embodiments, the disclosure features a pharmaceutical composition comprising at least one compound chosen from the novel compounds disclosed herein and pharmaceutically able salts thereof, at least one compound chosen from Compound II and pharmaceutically acceptable salts thereof, at least one compound chosen from Compound III and pharmaceutically acceptable salts thereof, and at least one ceutically acceptable carrier.
SUBSTITUTE SHEET (RULE 26) In some embodiments, the sure es a pharmaceutical composition comprising at least one compound chosen from the novel compounds disclosed herein and pharmaceutically acceptable salts f, at least one compound chosen from Compound III and pharmaceutically acceptable salts thereof, at least one compound chosen from Compound IV and pharmaceutically acceptable salts thereof, and at least one pharmaceutically acceptable carrier.
In some embodiments, pharmaceutical compositions disclosed herein comprise at least one additional active pharmaceutical ingredient. In some embodiments, the at least one additional active pharmaceutical ingredient is a CFTR tor. In some embodiments, the at least one additional active pharmaceutical ingredient is a CFTR corrector. In some embodiments, the at least one additional active pharmaceutical ingredient is a CFTR iator. In some embodiments, the pharmaceutical composition comprises (i) a compound of Formulae (I), (II), (III), (IV), or (V), or a pharmaceutically acceptable salt thereof, or a deuterated tive of such nd or salt; and (ii) at least two additional active pharmaceutical ingredients, one of which is a CFTR corrector and one of which is a CFTR potentiator.
In some embodiments, at least one additional active pharmaceutical ingredient is selected from mucolytic , bronchodialators, antibiotics, anti-infective agents, and anti-inflammatory agents.
A pharmaceutical composition may further comprise at least one pharmaceutically acceptable carrier. In some embodiments, the at least one pharmaceutically acceptable carrier is chosen from pharmaceutically acceptable vehicles and pharmaceutically acceptable adjuvants. In some embodiments, the at least one pharmaceutically acceptable is chosen from ceutically acceptable fillers, disintegrants, surfactants, binders, lubricants.
It will also be iated that a ceutical ition of this sure, including a pharmaceutical composition comprising combinations described previously , can be employed in combination therapies; that is, the compositions can be administered concurrently with, prior to, or subsequent to, at least one additional active pharmaceutical ingredient or medical procedures.
SUBSTITUTE SHEET (RULE 26) Pharmaceutical compositions comprising these combinations are useful for treating cystic fibrosis.
As described above, pharmaceutical compositions disclosed herein may ally further comprise at least one pharmaceutically acceptable carrier. The at least one pharmaceutically acceptable carrier may be chosen from adjuvants and vehicles.
The at least one pharmaceutically acceptable carrier, as used herein, includes any and all solvents, diluents, other liquid vehicles, dispersion aids, suspension aids, surface active agents, isotonic agents, thickening agents, emulsifying agents, preservatives, solid binders, and lubricants, as suited to the particular dosage form desired. Remington: The Science and Practice of Pharmacy, 21st edition, 2005, ed. D.B. Troy, Lippincott Williams & Wilkins, Philadelphia, wA Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York discloses various earners used in formulating ceutical compositions and known ques for the preparation f. Except insofar as any conventional carrier is incompatible with the compounds of this disclosure, such as by ing any undesirable biological effect or otherwise cting in a rious manner with any other component(s) of the pharmaceutical composition, its use is plated to be within the scope of this disclosure. Non-limiting es of le pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, um stearate, in, serum proteins (such as human serum albumin), buffer substances (such as phosphates, glycine, sorbic acid, and potassium sorbate), partial glyceride mixtures of saturated vegetable fatty acids, water, salts, and electrolytes (such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, and zinc salts), dal silica, magnesium trisilicate, polyvinyl pyrrohdone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars (such as lactose, glucose and sucrose), starches (such as com starch and potato starch), ose 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, com oil and soybean oil), glycols (such as propylene glycol and polyethylene glycol), esters (such as ethyl oleate and ethyl laurate), agar, buffenng agents (such as magnesium hydroxide and aluminum hydroxide), alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl l, phosphate buffer solutions, non-toxic SUBSTITUTE SHEET (RULE 26) compatible lubricants (such as sodium lauryl sulfate and magnesium stearate), ng agents, releasing agents, coating agents, sweetening agents, flavoring agents, perfuming , preservatives, and antioxidants.
It will also be appreciated that a pharmaceutical composition of this disclosure, including a pharmaceutical composition comprising any of the combinations described previously, can be employed in combination therapies; that is, the compositions can be administered concurrently with, prior to, or subsequent to, at least one active pharmaceutical ingredients or medical procedures.
In some embodiments, the methods of the disclosure employ stering to a patient in need thereof at least one compound chosen from any of the compounds disclosed herein and ceutically acceptable salts thereof, and at least one compound chosen from Compound II, Compound III, Compound IV, and pharmaceutically acceptable salts of any of the foregoing.
Any le pharmaceutical compositions known in the art can be used for the novel compounds sed herein, Compound II, Compound III, Compound IV, and pharmaceutically acceptable salts thereof. Some exemplary ceutical itions for Compound 1 and its pharmaceutically acceptable salts are described in the Examples. Some exemplary pharmaceutical compositions for Compound II and its pharmaceutically acceptable salts can be found in 2014/015841, all of which are incorporated herein by reference. Some exemplary pharmaceutical compositions for Compound III and its ceutically acceptable salts can be found in 2012/027731, and Some exemplary pharmaceutical compositions for Compound IV and its ceutically able salts can be found in reference.
In some embodiments, a pharmaceutical composition comprising at least one compound chosen from the novel compounds disclosed herein and pharmaceutically acceptable salts thereof is administered with a pharmaceutical composition comprising Compound II and Compound III. Pharmaceutical compositions comprising nd II and Compound III are disclosed in PCT Publication No.
SUBSTITUTE SHEET (RULE 26) incorporated herein by reference. An exemplary embodiment is shown in the following Table: Table 2. Exemplary Tablet Comprising 100 mg of Compound II and 150 mg of Compound III. ient Amount per tablet (mg) nd II SDD (spray dried dispersion) Intra-granular 125 (80 wt % Compound II, 20 wt % HPMC) Compound III SDD (80 wt % Compound III, 19.5 wt% HPMCAS-HG; 187.5 0.5 wt% sodium lauryl sulfate) Microcrystalline cellulose 131.4 Croscarmellose Sodium 29.6 Total 473.5 Extra-granular Microcrystalline cellulose 112.5 Magnesium Stearate 5.9 Total 118.4 Total uncoated Tablet 591.9 Film coat Opadry 11,1 Total coated Tablet 609.6 In some embodiments, a pharmaceutical composition comprising at least one nd chosen from the novel compounds disclosed herein and ceutical salts thereof is administered with a pharmaceutical composition comprising Compound III. ceutical compositions comprising Compound III are disclosed in PCT Publication No. embodiment is shown in the following Table: Table 3: Ingredients for Exemplary Tablet of Compound III.
Tablet Formulation Percent Dose Dose Batch %Wt./Wt. (mg) (g) SUBSTITUTE SHEET (RULE 26) Compound III SDD (80 wt % Compound III, 19.5 wt% HPMCAS-HG; 0.5 wt% sodium lauryl ____________sulfate)____________ 34.09% 187.5 23.86 Microcrystalline cellulose 30.51% 167.8 21.36 Lactose 30.40% 167.2 21.28 Sodium croscarmellose 3.000% 16.50 2.100 SLS 0.500% 2.750 0.3500 Colloidal silicon dioxide 0.500% 2.750 0.3500 Magnesium te 1.000% 5.500 0,7000 Total 100% 550 70 Additional pharmaceutical compositions comprising Compound III are disclosed in PCI Publication No.
Exemplary mini-tablets (~2 mm diameter, ~2 mm thickness, each mini-tablet weighing about 6.9 mg) was formulated to have approximately 50 mg of Compound III per 26 mini-tablets and approximately 75 mg of Compound III per 39 mini-tablets using the amounts of ingredients recited in Table 4, below.
Table 4: ients for mini-tablets for 50 mg and 75 mg potency Tablet Formulation Percent Dose Dose (mg) Dose (mg) Batch %Wt./Wt. 50 mg potency 75 mg potency (g) nd III SDD 35 62.5 93.8 1753.4 (80 wt % nd III, 19.5 wt% HPMCASHG ; 0.5 wt% sodium lauryl sulfate)_________ ol 13.5 24.1 36.2 675.2 Lactose 41 73.2 109.8 2050.2 Sucralose 2.0 3.6 5.4 100.06 Croscarmellose 6.0 10.7 16.1 300.1 sodium Colloidal silicon 1.0 1.8 2.7 50.0 dioxide Magnesium stearate 1.5 2.7 4.0 74.19 Total 100 178.6 268 5003.15 In some embodiments, the pharmaceutical compositions are a tablet. In some embodiments, the tablets are suitable for oral administration.
These combinations are useful for treating cystic fibrosis.
SUBSTITUTE SHEET (RULE 26) In some embodiments, sed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising stering an effective amount of at least one pharmaceutical composition of this disclosure to the t, such as a human, n said patient has cystic fibrosis and is chosen from patients with F508dellmm\mA function (MF) genotypes, patients with F508del/F508del genotypes, patients with /oO-Vt/e//gating genotypes, and patients with /o/Wc/e//residual function (RF) genotypes.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic on is any CF-causing mutation, and is ed to be and/or is responsive to any combinations of (i) the novel compounds disclosed herein, such as Compound 1, and (ii) Compound II, and/or Compound III and/or Compound IV genotypes based on in vitro and/or clinical data.
] Patients with an F508del/mimma[ function genotype are defined as patients that are heterozygous F508del-CFTR with a second CFTR allele containing a mutation that is predicted to result in a CFTR n with minimal function and that is not expected to respond to Compound II, Compound III, or the combination of nd II and Compound III. These CFTR mutations were defined using 3 major sources: • biological plausibility for the mutation to respond (i.e., mutation class) • evidence of clinical severity on a population basis (per CFTR2 patient registry; accessed on 15 February 2016) o average sweat chloride >86 mmol/L, and o prevalence of pancreatic insufficiency (PI) >50% • in vitro testing o mutations resulting in baseline chloride transport <10% of wild-type CFTR were considered minimal function o mutations resulting in chloride transport <10% of ype CFTR following the addition of nd II and/or Compound III were considered nonresponsive.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic is in a t comprising administering an effective amount of a pharmaceutical composition of this disclosure to the t, such as a human, wherein the patient possesses a CFTR genetic mutation G551D. In some embodiments, the patient is homozygous forthe G551D genetic SUBSTITUTE SHEET (RULE 26) mutation. In some embodiments, the patient is heterozygous for the G551D genetic on. In some embodiments, the patient is heterozygous for the G551D genetic mutation, having the G551D mutation on one allele and any other CF-causing mutation on the other allele. In some embodiments, the patient is heterozygous for the G55 ID genetic mutation on one allele and the other CF-causing genetic mutation on the other allele is any one of F508del, G542X, N1303K, W1282X, R117H, R553X, 1717-1G->A, 621+1G->T, 2789+5G->A, 3849+10kbC->T, , G85E, 3120+1G->A, AI507, 1898+1G->A, 3659delC, R347P, R560T, R334W, A455E, 2184delA, or 711+1G->T.
In some embodiments, the patient is heterozy gous for the G551D genetic mutation, and the other CFTR genetic mutation is F508del. In some embodiments, the t is heterozygous for the G551D genetic mutation, and the other CFTR genetic mutation is R117H.
In some embodiments, disclosed herein is a method of treating, lessening the ty of, or symptomatically ng cystic fibrosis in a t comprising administering an effective amount of a pharmaceutical composition of this sure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation F508del. In some embodiments, the patient is homozygous for the F508del genetic mutation. In some ments, the patient is heterozygous for the F508del genetic mutation wherein the patient has the F508del c mutation on one allele and any CF-causing genetic mutation on the other allele. In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any CF-causing mutation, ing, but not limited to G551D, G542X, N1303K, W1282X, R117H, R553X, 1717-1G->A, 621+1G->T, 2789+5G->A, 3849+10kbC->T, R1162X, G85E, 3120+1G->A, AI507,1898+1G->A, lC, R347P, R560T, R334W, A455E, 2184delA, or 711+1G->T. In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic on is G551D. In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is R117H.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or matically treating cystic fibrosis in a patient sing administering an effective amount of a pharmaceutical composition of this sure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, , S1255P, G1349D, S549N, S549R, SUBSTITUTE SHEET (RULE 26) S125IN, E193K, F1052V, G1069R, R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N, D1152H, 1717-1G->A, 621+1G->T, G->A, G->A, 711+1G->T, 2622+lG->A, 405+lG->A, 406-lG->A, 4005+lG->A, 1812-1G->A, 1525-1G->A, ->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, G->A, 3849+1 OkbC->T, 3272-26A->G, 711+5G->A, 3120G->A, 1811+1.6kbA- >G, ->G, 1898+3A->G, 1717-8G->A, 1342-2A->C, 405+3A->C, 1716G/A, 1811+1G->C, 1898+5G->T, 3850-3T->G, IVS14b+5G->A, 1898+1G->T, T- >C, 621+3A->G, 1949del84, 3141del9, 3195del6, 3199del6, 3905InsT, TT->A, A1006E, A120T, A234D, A349V, A613T, C524R, D192G, D443Y, D513G, D836Y, D924N, D979V, E116K, E403D, E474K, E588V, E60K, E822K, F1016S, F1099L, F191V, F311del, F311L, F508C, F575Y, G1061R, G1249R, G126D, G149R, G194R, G194V, G27R, G314E, G458V, G463V, G480C, G622D, G628R, G->A), G91R, G970D, , H1085P, H1085R, H1375P, H139R, H199R, H609R, H939R, I1005R, I1234V, I1269N, I1366N, I175Y, I502T, I506S, I506T, I601F, I618T, I807M, I980K, L102R, , L1335P, L138ins, L1480P, L15P, L165S, L320V, L346P, L453S, L571S, L967S, M1101R, M152V, MIT, M1V, M265R, M952I, M952T, P574H, P5L, P750L, P99L, Q1100P, Q1291H, Q1291R, Q237E, Q237H, Q452P, Q98R, R1066C. R1066H, R117G, R117L, R117P, R1283M, R1283S, R170H, R258G, R31L, R334L, R334Q, R347L, R352W, R516G, R553Q, R751L, R792G, R933G, S1118F, S1159F, S1159P, S13F, S549R(A->C), S549R(T->G), S589N, S737F, S912L, T1036N, T1053I, , T604I, V1153E, V1240G, V1293G, V201M, V232D, V456A, V456F. V562I, W1098C, , W1282R, W361R, W57G, W57R, Y1014C, Y1032C, Y109N, Y161D, Y161S, Y563D, Y563N, Y569C, and Y913C.In some embodiments, the patient has at least one combination mutation chosen from: G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, S125IN, E193K, F1052V, G1069R, R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N, D1152H, 1717-1G-+A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, 2622+lG->A, 405+1G-+A, 406-lG->A, 4005+1G-+A, 1812-1G->A, 1525-1G->A, 712-1G->T, 1248+1G-+A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-1G-+A, 2789+5G-+A, 3849+lOkbC- >T, 6A->G, 711+5G->A, 3120G-+A, 1811+1.6kbA->G, 711+3A->G, 1898+3A- SUBSTITUTE SHEET (RULE 26) >G, 1717-8G->A, 1342-2A->C, 405+3A->C, 1716G/A, 1811+1G->C, 1898+5G->T, 3850-3T->G, IYS14b+5G->A, 1898+1G->T, 4005+2T->C, and 621+3A->G In some embodiments, the patient has at least one combination mutation chosen from: l84, l9, 3195del6, l6, 3905InsT, 4209TGTT->A, A1006E, A120T, A234D, A349V, A613T, C524R, D192G, D443Y, D513G, D836Y, D924N, D979V, E116K, E403D, E474K, E588V, E60K, E822K, F1016S, F1099L, F191V, l, F311L, F508C, F575Y, G1061R, G1249R, G126D, G149R, G194R, G194V, G27R, G314E, G458V, G463V, G480C, G622D, G628R, G628R(G->A), G91R, G970D, H1054D, H1085P, , H1375P, H139R, H199R, H609R, H939R, I1005R, I1234V, I1269N, I1366N, I175V, I502T, I506S, I506T, I601F, I618T, I807M, I980K, L102R, L1324P, L1335P, L138ins, L1480P, L15P, L165S, L320V, L346P, L453S, L571S, L967S, M1101R, M152V, MIT, M1V, M265R, M952I, M952T, P574H, P5L, P750L, P99L, Q1100P, Q1291H, Q1291R, Q237E, Q237H, Q452P, Q98R, R1066C. R1066H, R117G, R117L, R117P, R1283M, R1283S, R170H, R258G, R31L, R334L, R334Q, R347L, R352W, R516G, R553Q, R751L, R792G, R933G, S1118F, S1159F, S1159P, S13F, S549R(A->C), S549R(T->G), S589N, S737F, S912L, T1036N, T1053I, T1246I, T604I, V1153E, V1240G, V1293G, V201M, V232D, V456A, V456F, V562I, , W1098R, W1282R, W361R, W57G, W57R, , Y1032C, Y109N, Y161D, Y161S, Y563D, Y563N, Y569C, and Y913C.
In some embodiments, the patient has at least one combination mutation chosen from: D443Y;G576A;R668C, F508C;S1251N, G576A; R668C, G970R; M470V, R74W;D1270N, R74W;Y201M, and R74W;V201M;D1270N.
In some ments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising SUBSTITUTE SHEET (RULE 26) administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, n the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, , S1255P, G1349D, S549N, S549R, S1251N, E193K, F1052V and G1069R. In some embodiments, this disclosure provides a method of treating CFTR comprising administering a compound of Formula (I), (II), (III), (IV), (V), or a pharmaceutically acceptable salt thereof to a patient possessing a human CFTR mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R and S125 IN. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a ceutical ition of this disclosure to the patient, such as a mammal, n the patient possesses a CFTR genetic mutation selected from E193K, F1052V and G1069R. In some embodiments, the method produces an se in chloride transport relative to baseline chloride transport of the t of the patient.
In some embodiments, sed herein is a method of treating, lessening the severity of, or symptomatically ng cystic fibrosis in a patient comprising administering an ive amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR c mutation selected from R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N andD1152H. In some embodiments, the method produces an increase in chloride transport above the baseline chloride transport of the patient.
] In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic on selected from 1717-1G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, 2622+lG->A, ->A, 406-lG->A, 4005+lG->A, 1812-1G->A, 1525-1G->A, 712-1G->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, 2789+5G->A, 3849+1 OkbC->T, 6A->G, 711+5G->A, 3120G->A, 1811+1.6kbA- >G, 711+3A->G, 1898+3A->G, 1717-8G->A, 1342-2A->C, 405+3A->C, 1716G/A, 1811+1G->C, 1898+5G->T, 3850-3T->G, +5G->A, 1898+1G->T, 4005+2T->C and 621+3A->G. In some embodiments, disclosed herein is SUBSTITUTE SHEET (RULE 26) a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising stering an effective amount of a pharmaceutical ition of this sure to the patient, such as a mammal, wherein the patient possesses a CFTR c mutation selected from 1717-1G->A, 1811+1.6kbA->G, 2789+5G->A, 3272-26A->G and 3849+1 OkbC->T. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically ng cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from 2789+5G->A and 3272- 26A->G.
In some ments, disclosed herein is a method of treating, ing the ty of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the t, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, , E193K, F1052V, , R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N, , 1717-1G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, 2622+lG->A, 405+lG->A, 406-lG->A, 4005+lG->A, 1812-1G->A, 1525-1G->A, 712-1G->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, 2789+5G->A, 3849+1 OkbC->T, 3272-26A->G, 711+5G->A, 3120G->A, 1811+1.6kbA- +3A->G, 1898+3A->G, 1717-8G->A, 1342-2A->C, 405+3A->C, A, 1811+1G->C, 1898+5G->T, 3850-3T->G, IVS14b+5G->A, 1898+1G->T, 4005+2T->C and 621+3A->G, and a human CFTR mutation selected from F508del, R117H, and G551D.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, S1251N, E193K, F1052V and G1069R, and a human CFTR mutation selected from F508del, R117H, and G551D. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically ng cystic fibrosis in a patient SUBSTITUTE SHEET (RULE 26) comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient ses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R and S125 IN, and a human CFTR mutation selected from F508del, R117H, and G551D. In some embodiments, disclosed herein is a method of ng, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a ceutical composition of this disclosure to the patient, such as a mammal, wherein the t possesses a CFTR genetic mutation selected from E193K, F1052V and G1069R, and ahuman CFTR mutation selected from F508del, R117H, and G55 ID. In some embodiments, the method produces an increase in chloride transport relative to baseline de transport of the patient.
In some ments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composihon of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N and , and ahuman CFTR mutation selected from F508del, R117H, and G55 ID. In some embodiments, the method produces an increase in chloride transport which is above the baseline de ort of the patient.
In some embodiments, sed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, G->A, 405+lG->A, 406-lG->A, 4005+lG->A, 1812-1G->A, G->A, 712-1G->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, 2789+5G->A, 3849+10kbC->T, 3272-26A->G, 711+5G->A, 3120G->A, 1811+T6kbA- >G,711+3A->G, A->G, G->A, 1342-2A->C, 405+3A->C, 1716G/A, 1811+1G->C, 1898+5G-+T, 3850-3T-+G, IVS14b+5G->A, 1898+1G->T, 4005+2T-+C and 621+3A->G, and a human CFTR mutation selected from l, R117H, and G551D. In some embodiments, disclosed herein is a method of treating, lessening the SUBSTITUTE SHEET (RULE 26) severity of, or symptomatically treating cystic is in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from 1717-1G->A, .6kbA->G, 2789+5G->A, 3272-26A->G and 3849+1 OkbC->T, and a human CFTR mutation selected from F508del, R117FI, and G551D. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this sure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from 2789+5G->A and 3272-26A->G, and a human CFTR mutation selected 08del, R117H.
In some ments, disclosed herein is a method of treating, lessening the ty of, or matically treating cystic fibrosis in a t comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic on selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, S125IN, E193K, F1052V, G1069R, R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N, D1152H, 1717-1G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, 2622+lG->A, 405+lG->A, 406-lG->A, 4005+lG->A, 1812-1G->A, 1525-1G->A, 712-1G->T, 1248+1G->A, G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, 2789+5G->A, 3849+1 OkbC->T, 3272-26A->G, ->A, 3120G->A, 1811+1.6kbA- >G,711+3A->G, A->G, 1717-8G->A, 1342-2A->C, 405+3A->C, A, 1811+1G->C, 1898+5G->T, 3850-3T->G, IVS14b+5G->A, 1898+1G->T, 4005+2T->C and 621+3A->G, and a human CFTR mutation selected from F508del, R117H, and G551D.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the t possesses a CFTR c mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, S1251N, E193K, F1052V and . In some embodiments, disclosed herein is a method of treating, lessening the severity of, or matically treating cystic fibrosis SUBSTITUTE SHEET (RULE 26) in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R and SI25IN. In some embodiments, disclosed herein is a method of ng, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from E193K, F1052V and G1069R. In some embodiments, the method produces an increase in de transport relative to baseline chloride transport of the patient.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or matically ng cystic fibrosis in a patient sing administering an effective amount of a pharmaceutical composition of this sure to the t, such as a mammal, wherein the patient possesses a CFTR genetic mutation ed from R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N andD1152H. In some embodiments, the method produces an increase in chloride transport which is above the baseline chloride transport of the t.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising stering an ive amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G->A, 621+1G->T, 3120+1G->A, G->A, 711+1G->T, G->A, 405+lG->A, 406-lG->A, 4005+1G-+A, 1812-1G-+A, 1525- 1G->A, 712-1G->T, 1248+1 G->A, 1341+1G->A, 3121-1G-+A, 4374+1 G->T, 3850-1G-+A, 2789+5G-+A, 0kbC->T, 3272-26A->G, 711+5G- >A, 3120G->A, 1811+1.6kbA->G, 711+3A-+G. 1898+3A->G, 1717-8G-+A. 1342-2A-+C, 405+3A-+C, 1716G/A, 1811+1G-+C, 1898+5G-+T, 3850-3T-+G, +5G->A, 1898+1G->T, 4005+2T->C and 621+3A->G. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically ng cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from 1717-1G-+A, SUBSTITUTE SHEET (RULE 26) 1811+1.6kbA->G, 2789+5G->A, 3272-26A->G and 3849+1 OkbC->T. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic is in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic on selected from 2789+5G->A and 3272-26A->G.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, G1349D, S549N, S549R, S125IN, E193K, , G1069R, R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, , D1152H, 1717-1G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, 2622+lG->A, 405+lG->A, 406-lG->A, 4005+lG->A, G->A, G->A, 712-1G->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, 3850-lG->A, 2789+5G->A, 3849+1 OkbC->T, 3272-26A->G, 711+5G->A, 3120G->A, 1811+1.6kbA- >G,711+3A->G, 1898+3A->G, G-+A, 1342-2A->C, 405+3A-+C, 1716G/A, 1811+1G->C, 1898+5G->T, 3850-3T-+G, +5G->A, 1898+1G->T, 4005+2T-+C and 621+3A->G, and a human CFTR mutation selected from F508del, R117H, and G551D, and one or more human CFTR ons selected from F508del, R117H, and G551D.
In some ments, disclosed herein is a method of ng, ing the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, S1255P, , S549N, S549R, S1251N, E193K, F1052V and G1069R, and one or more human CFTR mutations selected from l, R117H, and G55 ID. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising stering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the t possesses a CFTR genetic mutation selected from G178R, G551S, G970R, G1244E, SUBSTITUTE SHEET (RULE 26) S1255P, G1349D, S549N, S549R and S125 IN, and one or more human CFTR mutations selected from F508del, R117H, and G551D. In some embodiments, disclosed herein is a method of ng, lessening the severity of, or matically treating cystic fibrosis in a patient comprising administenng an effective amount of a pharmaceutical ition of this disclosure to the patient, such as a mammal, n the patient possesses a CFTR genetic mutation selected from E193K, F1052Y and G1069R, and one or more human CFTR mutations selected from F508del, R117H, and G551D. In some embodiments, the method produces an increase in chloride transport relative to baseline chloride transport of the patient.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the t, such as a mammal, n the t possesses a CFTR genetic mutation selected from R117C, D110H, R347H, R352Q, E56K, P67L, L206W, A455E, D579G, S1235R, S945L, R1070W, F1074L, D110E, D1270N and D1152H, and one or more human CFTR ons ed from F508del, R117H, and G551D. In some embodiments, the method produces an se in chloride transport which is above the baseline chloride transport of the patient.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a , n the patient possesses a CFTR genetic mutation selected from 1717-1G->A, 621+1G->T, 3120+1G->A, 1898+1G->A, 711+1G->T, G->A, 405+lG->A, 406-lG->A, 4005+lG->A, 1812-1G->A, 1525-1G->A, 712-1G->T, 1248+1G->A, 1341+1G->A, 3121-1G->A, 4374+lG->T, G->A, 2789+5G->A, 3849+10kbC->T, 3272-26A->G, 711+5G->A, 3120G->A, 1811+1.6kbA- >G,711+3A->G, 1898+3A->G, 1717-8G->A, 1342-2A->C, 405+3A->C, 1716G/A, G->C, 1898+5G-+T, 3850-3T-+G, IVS14b+5G->A, 1898+1G->T, 4005+2T-+C and 621+3A->G, and one or more human CFTR mutations selected from F508del, R117H, and G551D. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR SUBSTITUTE SHEET (RULE 26) genetic mutation selected from 1717-1G->A, 1811+1.6kbA->G, 2789+5G->A, 3272- 26A->G and 3849+10kbC->T, and one or more human CFTR mutations selected from F508del, R117H, and G551D. In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient sing administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a , wherein the patient possesses a CFTR genetic mutation ed from 2789+5G->A and 3272-26A->G, and one or more human CFTR mutations selected from F508del, R117H, and G551D.
In some ments, the patient is heterozygous having one CF-causing mutation on one allele and another CF-causing on on the other allele. In some ments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any CF-causing mutation, including, but not limited to F508del on one CFTR allele and a CFTR mutation on the second CFTR allele that is associated with minimal CFTR function, residual CFTR function, or a defect in CFTR channel gating activity.
In some embodiments, the CF-causing mutation is selected from Table 5A.
In some embodiments, the t is heterozy gous having one CF-causing mutation on one CFTR allele selected from the mutations listed in the table from and another CF-causing mutation on the other CFTR allele is selected from the CFTR mutations listed in Table 5A.
Table 5A.
Y122X L218X Q220X C276X Q290X SUBSTITUTE SHEET (RULE 26) G330X W401X Q414X S434X S466X S489X Q493X W496X Q525X G542X Q552X R553X E585X G673X R709X K710X L732X R764X R785X R792X E822X W846X R851X Q890X S912X W1089X Y1092X El KMX R1158X R1162X S1196X W1204X S1255X W1282X Q1313X 621+lG^T 711+lG^T 711+5G^A 712-lG^T 405+3A^C 406-1G^ A SUBSTITUTE SHEET (RULE 26) 621+lG^T 1248+lG^A 1341+lG^A 1717-lG^A 1811+1.6kbA-»G 1811+lG^C 1812-lG^A 1898+lG^A 2622+IG^A 3120+lG^A 3120G^A 3850-lG^A 4005+1G^ A 4374+1 G^T 663delT 2183AA^G 3659delC 394delTT 3905insT 2184delA 1078delT 1154insTC 2183delAA^G 2143delT 1677delTA 3876delA 2307insA 4382delA 4016insT 2347delG 3007delG 574delA 2711delT 3791delC le22-23 457TAT—>-G 2043delG 2869insG 3600+2insT SUBSTITUTE SHEET (RULE 26) 3737delA 4040delA 541delC T338I R347P L927P S341P L467P 07del V520F A559T R560T R560S A561E Y569D L1065P R1066C R1066M L1077P H1085R M1101K N1303K 2789+5G^A 3849+lOkbC 3272-26A^G D110E D110H R117C L206W R347H R352Q A455E SUBSTITUTE SHEET (RULE 26) D579G E831X S945L S977F F1052V R1070W F1074L D1152H D1270N R117H G178R S549N S549R G551D G551S G1244E S1251N S1255P G1349D Table SB: CFTR Mutations Criteria Mutation Truncation S4X C276X G542X R792X El KMX mutations G27X Q290X G550X E822X R1158X • %PI >50% Q39X G330X Q552X W846X R1162X and/or W57X W401X R553X Y849X S1196X SwCT >86 E60X Q414X E585X R851X W1204X mmol/L R75X S434X G673X Q890X L1254X • no fulllength E92X S466X Q685X S912X S1255X protein Q98X S489X R709X Y913X W1282X Y122X Q493X K710X W1089X Q1313X E193X W496X L732X Y1092X E1371X L218X C524X R764X W1098X Q1382X Q220X Q525X R785X R1102X Q1411X Splice ons 185+1G 711+5G—>A. G^A 2622+1G- 3121- • %PI >50% T A and/or 296+1G- 712-lG^T 1717-lCr^A 2790- 3500- SwCF >86 A 1G—>C 2A^G mmol/L 405+1G- 1248+1G- 1811+1G—>C 3040G^C 3600+2insT • no or little A A (G970R) mature 405+3A- 1249- 1811+1.6kbA 3850- mRNA C IG^A G IG^A 406- 1341+1G- 1812-lG^A 3120G^A 4005+1G- IG^A A A SUBSTITUTE SHEET (RULE 26) Criteria Mutation ^ 1525- 1898+1G—>A G- 4374+1G- T 2A^G A T 711+1G—> 1525- G^C 3121- T 1G-^A 2A^G Small (<3 182delT lA 1782delA 2732ms A 3876delA nucleotide) 306insA 1138insG 1824delA sG 3878delG insertion/deleti 365- 1154insTC 2043 delG 2896insAG 3905insT on (ins/del) 366msT frameshift 394delTT 1161delC 2143delT 2942insT 4016insT mutations A 1213delT 2183AA^Ga 2957delT 4021dupT • %PI >50% and/or 444delA 1259insA 2184delA 3007delG 4040delA SwCr >86 457TAT^ sTA 2184ms A 3028delA 4279insA mmol/L G • garbled 541delC 1471 del A 2307insA 3171delC 4326delTC and/or 574delA 1497delGG 2347delG 3659delC truncated 663delT lG 2585delT 3737delA protein 935delA 1609del CA 2594delGT 3791delC 1078delT 1677delTA 2711delT 3821delT Non-small (>3 CFTRdele2,3 1461ins4 2991del32 nucleotide) CFTRdele22,23 1924del7 3667ins4 insertion/deleti 124del23bp 2055del9^A 4010del4 on (ins/del) 852del22 2105- 4209TGTT^AA frameshift 2117del 13 ins AGAAA mutations 991del5 272 Idelll • %PI >50% and/or SwCF >86 mmol/L • garbled and/or protein SUBSTITUTE SHEET (RULE 26) Criteria Mutation Class IL III, IV A46Db V520F Y569Db N1303K mutations not G85E A559Tb L1065P responsive to R347P R560T R1066C Compound III R560S L1077Pb alone or i in , , combination 15 07del A561E M1101K Compound II or Compound • %PI>50% and/or SwCl >86 mmol/L • Not responsive in vitro to Compound III alone or combinatio n with Compound II or Compound Note: %PI: percentage of F508del-CFTR heterozygous patients in the CFTR2 patient registry who are atic insufficient; SwCF: mean sweat chloride of F508del-CFTR heterozy gous patients in the CFTR2 patient registry a Also known as lAA^G. b Unpublished data.
Table 5B above includes certain exemplary CFTR minimal function mutations, which are able by an FDA-cleared genotyping assay, but does not include an exhaustive list.
] In some embodiments, sed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a patient with F508del/MF (F/MF) pes (heterozygous for F508del and an MF mutation not ed to respond to CFTR modulators, such as Compound III); with F508del/F508del (F/F) genotype (homozygous for F508del); and/or with F508delfgaXmg (F/G) genotypes (heterozygous for F508del and a gating mutation known to be CFTR modulator- SUBSTITUTE SHEET (RULE 26) WO 64632 responsive (e.g., Compound Ill-responsive). In some embodiments, a patient with UMY (F/MF) genotypes has a MF mutation that is not expected to respond to Compound II, Compound III, and both of Compound II and Compound III. In some embodiments, a patent with FSOSdellMF (F/MF) genotypes has any one of the MF mutations in Table 5B.
In some ments, the patient is heterozygous for l, and the other CFTR genetic mutation is any CF-causing mutation, including truncation mutations, splice mutations, small (<3 nucleotide) insertion or deletion (ins/del) frameshift mutations; non-small (>3 nucleotide) insertion or deletion el) frameshift mutations; and Class II, III, IV mutations not responsive to Compound III alone or in combination with Compound II or Compound IV.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is a truncation mutation. In some specific embodiments, the truncation on is a truncation mutation listed in Table 5B.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is a splice on. In some specific embodiments, the splice mutation is a splice mutation listed in Table 5B.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is a small (<3 nucleotide) insertion or deletion (ins/del) frameshift mutation. In some specific ments, the small (<3 nucleotide) insertion or deletion (ins/del) frameshift on is a small (<3 nucleotide) insertion or deletion (ins/del) frameshift mutation listed in Table 5B.
] In some embodiments, the patient is heterozygous for F508del, and the other CFTR c on is any CF-causing mutation expected to be and/or is responsive to, based on in vitro and/or clinical data, any combination of (i) a novel compound chosen from those disclosed herein (e.g., compounds of Formula (I), (II), (III), (IV), or (V), and pharmaceutically acceptable salts f, and their deuterated derivatives), and (ii) Compound II, and/or Compound III, and/or Compound IV.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any CF-causing mutation expected to be and/or is sive, based on in vitro and/or clinical data, to the triple combination of a novel compound chosen from those disclosed herein (e.g., compounds of Formula (I), (II), (III), (IV), or SUBSTITUTE SHEET (RULE 26) (V), and pharmaceutically acceptable salts thereof, and their deuterated derivatives), and Compound II, and Compound III.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic on is a non-small (>3 nucleotide) insertion or deletion el) frameshift mutation. In some specific embodiments, the non-small (>3 nucleotide) ion or deletion (ins/del) hift mutation is anon-small (>3 nucleotide) insertion or deletion (ins/del) frameshift mutation listed in Table 5B.
In some embodiments, the t is heterozygous for F508del, and the other CFTR genetic mutation is a Class II, III, IV mutations not responsive to Compound III alone or in combination with Compound II or Compound IV. In some specific ments, the Class II, III, IV mutations not sive to Compound III alone or in ation with Compound II or Compound IV is a Class II, III, IV mutations not responsive to Compound III alone or in combination with Compound II or Compound IV listed in Table 5B.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any mutation listed in Table 5B.
In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any mutation listed in Table 5A, 5B, and In some embodiments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any mutation listed in Table 5A. In some ments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any mutation listed in Table 5B. In some ments, the patient is heterozygous for F508del, and the other CFTR genetic mutation is any mutation listed in In some embodiments, the patient is homozygous for F508del.
In some embodiments, the patient is heterozygous having one CF-causing mutation on one CFTR allele selected from the mutations listed in the table from and another CF-causing mutation on the other CFTR allele is selected from the CFTR ons listed in Table 5B. ts with an /oWC/efgating mutation genotype are defined as patients that are heterozygous F508del-CFTR with a second CFTR allele that contains a mutation associated with a gating defect and clinically demonstrated to be responsive to SUBSTITUTE SHEET (RULE 26) Compound III. Examples of such mutations include: G178R, S549N, S549R, G551D, G551S, G1244E, S1251N, S1255P, and G1349D.
Patients with an /oti/C/e/tiesidual function pe are defined as patients that are heterozygous F508del-CFTR with a second CFTR allele that contains a mutation that results in reduced protein ty or function at the cell e which can produce partial CFTR activity. CFTR gene ons known to result in a residual function phenotype include in some embodiments, a CFTR residual function mutation selected from 2789+5G^ A, 3849+lOkbC^T, 3272-26A^ G, 711+3A^ G, E56K, P67L, R74W, D110E, Dll OH, R117C, L206W, R347H, R352Q, A455E, D579G, E831X, S945L, S977F, F1052V, R1070W, F1074L, D1152H, D1270N, E193K, and K1060T. In some embodiments, the CFTR residual function on is selected from R117H, S1235R, I1027T, R668C, G576A, M470V, L997F, R75Q, R1070Q, R31C, D614G, G1069R, R1162L, E56K, A1067T, E193K, or K1060T. In some embodiments, the CFTR residual function mutation is selected from R117H, S1235R, I1027T, R668C, G576A, M470V, L997F, R75Q, R1070Q, R31C, D614G, G1069R, R1162L, E56K, or A1067T.
In some embodiments, disclosed herein is a method of treating, lessening the severity of, or symptomatically treating cystic fibrosis in a t comprising administering an effective amount of a pharmaceutical composition of this disclosure to the patient, such as a mammal, wherein the patient possesses a CFTR genetic mutation selected from the mutations listed in In some embodiments, the composition disclosed herein is useful for treating, lessening the severity of, or symptomatically treating cystic is in patients who exhibit residual CFTR activity in the apical ne of respiratory and nonrespiratory epithelia. The presence of residual CFTR activity at the epithelial surface can be readily ed using methods known in the art, e.g., standard electrophysiological, biochemical, or histochemical techniques. Such methods fy CFTR activity using in vivo or ex vivo electrophysiological techniques, measurement of sweat or ry CF trations, or ex vivo biochemical or histochemical techniques to monitor cell surface density. Using such methods, residual CFTR activity can be readily detected for patients that are heterozygous or homozygous for a variety of ent mutations, including patients heterozy gous for the most common on.
SUBSTITUTE SHEET (RULE 26) F508del, as well as other mutations such as the G551D mutation, or the R117H mutation. In some embodiments, itions disclosed herein are useful for treating, lessening the severity of, or symptomatically treating cystic fibrosis in patients who exhibit little to no residual CFTR activity. In some embodiments, compositions disclosed herein are useful for treating, lessening the severity of, or symptomatically treating cystic fibrosis in patients who t little to no residual CFTR activity in the apical membrane of respiratory epithelia.
In some embodiments, the compositions disclosed herein are useful for treating or ing the severity of cystic fibrosis in patients who exhibit residual CFTR activity using pharmacological methods. Such methods increase the amount of CFTR present at the cell surface, thereby inducing a to absent CFTR ty in a patient or augmenting the existing level of residual CFTR activity in a patient.
In some embodiments, the compositions disclosed herein are useful for treating or ing the severity of cystic fibrosis in patients with certain genotypes exhibiting residual CFTR activity.
In some embodiments, itions disclosed herein are useful for treating, lessening the severity of, or symptomatically treating cystic fibrosis in patients within certain clinical phenotypes, e.g., a mild to te clinical phenotype that typically correlates with the amount of residual CFTR ty in the apical membrane of epithelia. Such phenotypes include patients exhibiting pancreatic sufficiency.
In some embodiments, the compositions disclosed herein are useful for treating, lessening the severity of, or symptomatically treating ts diagnosed with pancreatic sufficiency, idiopathic pancreatitis and ital bilateral absence of the vas deferens, or mild lung disease wherein the patient exhibits residual CFTR activity.
In some embodiments, this sure relates to a method of augmenting or inducing anion channel activity in vitro or in vivo, comprising contacting the channel with a composition disclosed herein. In some embodiments, the anion channel is a chloride channel or a bicarbonate channel. In some embodiments, the anion channel is a de channel.
The exact amount of a pharmaceutical composition required will vary from subject to subject, depending on the s, age, and general ion of the t, SUBSTITUTE SHEET (RULE 26) the severity of the disease, the particular agent, its mode of administration, and the like.
The compounds of this sure may be formulated in dosage unit form for ease of stration 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 nds and compositions of this disclosure 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 ic compound ed; the specific composition employed; the age, body , general health, sex and diet of the patient; the time of administration, route of stration, 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 s well known in the medical arts. The term “patient”, as used herein, means an animal, such as a mammal, and even further such as a human.
In some embodiments, the disclosure also is directed to methods of treatment using isotope-labelled compounds of the afore-mentioned compounds, which have the same structures as disclosed herein except that one or more atoms therein have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally (isotope ed). Examples of isotopes which are commercially available and suitable for the disclosure include isotopes of en, carbon, nitrogen, oxygen, phosphorus, fluorine and ne, for e 2H, 'H, 13C, 14C, 15N, 180,170,31P, 32P, 35S, 18F and j6Cl, respectively.
The isotope-labelled compounds and salts can be used in a number of beneficial ways. They can be suitable for medicaments and/or various types of assays, such as substrate tissue distribution assays. For example, tritium (3H)- and/or carbon-14 (14C)-labelled compounds are particularly useful for various types of assays, such as substrate tissue distribution , due to relatively simple preparation and excellent detectability. For example, deuterium (2H)-labelled ones are therapeutically useful with potential therapeutic advantages over the non-2H-labelled nds. In general, deuterium ( H)-labelled compounds and salts can have higher metabolic stability as ed to those that are not isotope-labelled owing to the kinetic isotope effect SUBSTITUTE SHEET (RULE 26) described below. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which could be desired. The isotope-labelled compounds and salts can usually be prepared by carrying out the procedures disclosed in the synthesis s and the d description, in the example part and in the preparation part in the present text, ing a non-isotope-labelled nt by a readily available isotope-labelled reactant.
In some embodiments, the isotope-labelled compounds and salts are deuterium (2H)-labelled ones. In some specific embodiments, the isotope-labelled compounds and salts are deuterium (2H)-labelled, wherein one or more hydrogen atoms therein have been replaced by deuterium. In chemical structures, ium is represented as “2H” or "D." The deuterium (2H)-labelled compounds and salts can manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect.
The primary kinetic isotope effect is a change of the rate for a chemical reaction that s from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange. Exchange of a r isotope usually results in a lowering of the ground state energy for a al bond and thus causes a reduction in the rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi-product reaction, the product distribution ratios can be d substantially. For explanation: if deuterium is bonded to a carbon atom at a nonexchangeable position, rate differences of kM/ko = 2-7 are typical. For a further discussion, see S. L. Harbeson and R. D. Tung, Deuterium In Drug Discovery and Development, Ann. Rep. Med. Chem. 2011, 46, 403-417; and T.G. Gant “Using deuterium in drug ery: leaving the label in the drug" J. Med. Chem. 2014, 57, 3595-3611, relevant portions of which are independently incorporated herein by reference.
The concentration of the isotope(s) (e.g., deuterium) incorporated into the isotope-labelled compounds and salt of the disclosure may be defined by the isotopic enrichment . The term “isotopic enrichment factor” as used herein means the ratio between the isotopic abundance and the natural abundance of a ied isotope. In some embodiments, if a tuent in a compound of the disclosure is denoted deuterium, such compound has an isotopic enrichment factor for each ated SUBSTITUTE SHEET (RULE 26) deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium oration), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
When discovering and developing therapeutic agents, the person skilled in the art attempts to optimize pharmacokinetic parameters while retaining desirable in vitro properties. It may be able to assume that many compounds with poor cokinetic profiles are tible to oxidative metabolism.
One of ordinary skill in the art would understand that deuteration of one or more metabohcally labile positions on a compound or active metabolite may lead to improvement of one or more superior DMPK properties while maintaining biological activity as compared to the corresponding hydrogen analogs. The superior DMPK property or properties may have an impact on the exposure, half-life, clearance, lism, and/or even food requirements for optimal absorption of the drug product.
Deuteration may also change the metabolism at other non-deuterated positions of the deuterated compound.
In some embodiments, the disclosure es deuterated derivatives of the novel compounds sed herein and of their pharmaceutically acceptable salts. Nonlimiting examples of ated nds are disclosed in In some embodiments, Compound IIF as used herein includes the deuterated nd disclosed in U.S. Patent No. 8,865,902 (which is incorporated herein by reference), and CTP-656.
In some embodiments, Compound IIP is: .0 OH QP // D HN HN V \ fD D D D SUBSTITUTE SHEET (RULE 26) Exemplary embodiments of the sure include: The novel compounds disclosed herein (e.g., compounds of ae (I) - (V), pharmaceutically acceptable salts thereof, and deuterated derivatives of any of the foregoing, including the compounds in and those specifically ed herein) can be prepared by suitable methods known in the art. For example, they can be prepared in accordance with procedures described in WO2016/057572 and by the exemplary syntheses bed below in the es. For example, deuterated derivatives of the novel compounds of Formulae (I) - (V) and pharmaceutically acceptable salts thereof can be prepared in a r manner as those for compounds of Formulae (I) - (V) and pharmaceutically acceptable salts thereof by employing intermediates and/or reagents where one or more hydrogen atoms are replaced with deuterium. For example, see T.G. Gant “Using deuterium in drug discovery: leaving the label in the drug” J. Med. Chem. 2014, 57, 3595-3611, the relevant portions of which are incorporated herein by reference.
In some embodiments, compounds of Formulae (III), (IV) and (V) and pharmaceutically acceptable salts thereof, and deuterated derivatives of any of the foregoing are prepared as depicted in Schemes 1-2, wherein the variables therein are each and independently are as those for Formula (I), (II), (III), (IV), or (V) above, and wherein each Ra is independently chosen from C1-C4 alkyl groups; and each Xa is independently chosen from F or Cl. Suitable condition(s) known in the art can be employed for each step depicted in the s. In some embodiments, each Xa for Formulae B, C, D, F, B-l, C-l, D-l, and F-l in Schemes 2-4 is independently Cl. In some embodiments, each Xa for Formulae D, L, 0, and P in Scheme 6 is ndently In some embodiments, as shown in Scheme 1, the methods comprise reacting a nd of a (F) or a salt thereof with a compound of Formula (G) or a salt thereof to generate a compound of Formula (Ilia), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the ing.
SUBSTITUTE SHEET (RULE 26) Scheme 1 HN"^R3)q 9 QD 9 o.p yl ^ 97 Yi ^ 1 n*mA (G) n.nA rW/ n Yz Xa RXy Y _// i' n ^Pr3), (R4) r (F) (R4)r (Ilia) Any suitable conditions, such as those for a philic on of amine, known in the art can be used. In some embodiments, the reaction depicted in Scheme 1 is performed in the presence of a base, such as a metal carbonate (e.g., Na2C03 or k2co3).
In some embodiments, nds of Formula (Ilia), pharmaceutically acceptable salts thereof, or deuterated derivatives of any of the foregoing, wherein Y2 is N and Y1 is CH in each of Formulae (F), (G) and (Ilia), are prepared by the methods in Scheme 1.
In some embodiments, a salt of a compound of Formula (G) is ed In some embodiments, an HC1 salt of a compound of Formula (G) is employed.
A compound of Formula (F) or a salt thereof and a compound of Formula (G) or a salt thereof can be prepared by any suitable method known in the art, for e, those in WO2016/57572 and those in the exemplary ses described below in the Examples.
In some embodiments, as shown in Scheme 2, a compound of Formula (F), a ceutically able salt thereof, or a deuterated derivative of any of the foregoing is prepared by a method that comprises reacting a compound of Formula (D) or a salt thereof with a compound of Formula (E) or a salt thereof. In some embodiments, compounds of Formula (D), salts thereof, or deuterated tives of any of the foregoing are prepared by a method that comprises reacting a compound of Formula (A) or a salt thereof with a compound of Formula (B) or a salt thereof to generate a compound of a (C) or a salt thereof; and hydrolyzing the -C(0)0Ra of compound of Formula (C) to generate a compound of Formula (D) or a salt thereof.
Any suitable conditions known in the art can be used for steps (a), (b), and (c) of Scheme 2 below, such as those for a coupling reaction between carboxylic acid and SUBSTITUTE SHEET (RULE 26) sulfonamide or those for an acylation of sulfonamide for step (a), those for ysis of ester for step (b), and those for a nucleophilic reaction of amine for step (c).
In some embodiments, step (a) of Scheme 2 below is performed in the ce of a base. In some specific embodiments, step (a) is performed in the presence of a non-nucleophihc base. In some embodiments, in step (a), the reaction of a compound of Formula (D) or a salt thereof with a compound of Formula (E) or a salt f comprises reacting a compound of Formula (D) or a salt thereof with a coupling reagent, such as carbonyl diimidazole (GDI), and subsequently with a nd of Formula (E) or a salt thereof in the presence of a base, such as a non-nucleophilic base.
In some embodiments, a compound of a (D) or a salt thereof is reacted with GDI prior to the reaction with a compound of Formula (E) or a salt thereof, and then subsequently with a compound of Formula (E) or a salt thereof in the presence of a base, such as DBU (l,8-Diazabicyclo(5.4.0)undecene).
In some embodiments, step (b) of Scheme 2 below is med in the presence of a base. In some ments, step (b) is performed in the presence of an aqueous base, such as aqueous hydroxide. In some embodiments, step (b) is med in the presence of an aqueous metal hydroxide, such as aqueous NaOH.
In some embodiments, step (c) of Scheme 2 below is performed in the presence of a base. In some embodiments, step (c) is performed in the presence of a metal carbonate (e.g., Na2C03 or K2CO3).
SUBSTITUTE SHEET (RULE 26) Scheme 2 IT 0 Y1^ 'ORa RVN. Xa Y2xa -N'NA Yi NH (B) z Xa (A) 'r (C) (R4)r SOpNHp Y1^ V OH N-N^ 2 N-NA r (E) R'-r Xa (a) (R4)r (R4) r (F) (D) In some embodiments, disclosed herein is a method of preparing a compound of the following formula: PF3 P 0 o N" 'Ph b—f n N N-AlS) or a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing. The method comprises reacting a compound of Formula (F-l) or a salt thereof with a nd of Formula (G-l) or a salt thereof, n Xa is F or Cl, as shown in Scheme 3: SUBSTITUTE SHEET (RULE 26) Scheme 3 HNA(S) pF3 0 0 0 0 o 0 XM:s:n, AN:s'Th (G-1) N Ph H b-A N ■na*®) b->N Nx: xa (F-1) Compound (1) ] Any suitable conditions, such as those for a nucleophilic reaction of amine, known in the art can be used. In some ments, the reaction depicted in Scheme 3 is performed in the presence of a base, such as a metal carbonate (e.g., Na2C03 or K2co3).
In some embodiments, a salt of nd of Formula (G-1) is employed. In some embodiments, a HC1 salt of a compound of Formula (G-1) is employed.
] A compound of Formula (F-1) or a salt thereof and a nd of Formula (G-1) or a salt thereof can be prepared by any suitable method known in the art, for example, those in WO2016/57572 and those in the exemplary syntheses described below in the Examples.
In some embodiments, as shown in Scheme 4, a compound of Formula (F-1) or a salt thereof, or a deuterated derivative of any of the foregoing is prepared by a method that comprises reacting a compound of Formula (D-l) or a salt thereof with a compound of Formula (E-l) or a salt thereof. In some embodiments, nds of Formula (D-l) or salts thereof, or their deuterated derivatives are prepared by a method that comprises reacting a compound of Formula (A-l) or a salt thereof with a compound of Formula (B-l) or a salt thereof to generate a compound of formula (C-1) or a salt thereof; and hydrolyzing the -C(0)0Ra of compound of Formula (C-1) or salt thereof to generate a nd of a (D-l) or a salt thereof. Any suitable conditions known in the art can be used for steps (a-l), (b-l), and (c-1) of Scheme 4 below, such as those for a coupling reaction between carboxylic acid and sulfonamide or those for an ion of sulfonamide for step (a-l), those for hydrolysis of ester for step (b-l), and those for a nucleophilic reaction of amine for step (c-1).
SUBSTITUTE SHEET (RULE 26) In some embodiments, step (a-1) of Scheme 4 below is performed in the presence of a base. In some embodiments, step (a-1) of Scheme 4 below is performed in the presence of a non-nucleophilic base. In some embodiments, in step (a-1), the reaction of a compound of Formula (D-l) or a salt thereof with a compound of Formula (E-l) or a salt f comprises reacting a compound of Formula (D-l) or a salt thereof with a coupling reagent, such as carbonyl diimidazole (GDI), and subsequently with a compound of Formula (E-l) or a salt thereof in the presence of abase, such as anonnucleophilic base. In some embodiments, (i) a compound of Formula (D-l) or a salt thereof is reacted with GDI prior to the reaction with a nd of Formula (E-l) or a salt thereof, and then subsequently (ii) the reaction product of step (i) is reacted with a nd of Formula (E-l) or a salt thereof in the presence of a base, such as DBU (l,8-Diazabicyclo(5.4.0)undecene).
In some ments, step (b-1) of Scheme 4 below is performed in the presence of a base. In some ments, step (b-1) is performed in the presence of an aqueous base, such as aqueous hydroxide. In some embodiments, step (b-1) is performed in the presence of an aqueous metal hydroxide, such as aqueous NaOH.
In some embodiments, step (c-1) of Scheme 4 below is performed in the presence of a base. In some ments, step (c-1) is med in the presence of a metal carbonate (e.g., Na2CC>3 or K2CO3).
SUBSTITUTE SHEET (RULE 26) Scheme 4 'ORa 0 F3C ■cf3 O^N. Xa N Xa ORa NH (B-D N. ^5 n Xa step (c-1) (A-1) (C-1) pF3 o cf3 PhSOoNHs OH (E-1) v4 ------------- ► \ N, - H step(b-1) ^ Xa step (a-1) b-AN^N^2 Xa (D-1) (F-1) HN'AiS' pF3 0 0 0 XN:s:ph \ J s' N' 'N A(S Compound (1) In Scheme 4, Ra is chosen from C1-C4 alkyl groups; and each Xa is independently chosen from F or Cl.
In some embodiments, s of preparing a compound of Formulae (I) and (II), wherein X is NH or N(Ci-C4 alkyl) or a pharmaceutically able salt thereof, or a deuterated derivative of any of the foregoing, comprise reacting a compound of Formula (L) or a salt thereof with NR*3 where R* is H or C1-C4 alkyl, as depicted in Schemes 5 and 6: SUBSTITUTE SHEET (RULE 26) Scheme 5 0 0 II 11 Y1 ^ yi^ rI^'N^Y^N-A ^ N-NA NR*3 R-tw Y ^R3), VKyr (I: where X is NR*) (r! (L) Scheme 6 0 0 .s\V/ N'S N H l>3)P H t-(r2)p NR*3 Nv ■v rs RXw N ^Ir3), ^R3), (r4) (II: where X is NR*) (L: where Y2 is CH) Any le conditions known in the art can be used for the sulfoxamination reaction, for example, for those for electrophilic additions by amines. In some embodiments, the sulfoxamination reaction is performed in the presence of a chlorinating or oxidizing agent, such as N-chlorosuccinimide (NCS).
In some ments, a compound of Formula (L) or a salt thereof is ed by a method comprising oxidizing the sulfur unit of the H group of a compound of Formula (M) or salt thereof as shown in Scheme 7 below: SUBSTITUTE SHEET (RULE 26) Scheme 7 0 0 yi^ 'OH yl ^ NH2 yi% N'N^ N'N^'y2^xa RXw i 'Xa R'tV rV Y (r4) (r4)lKjr (p) (r4), <°> 0 0 II II yl^ K's1Qhr2)p Y1^ Y2' R,-tw Y ^R3). ^R3), (R4)r Any suitable conditions known in the art can be used for the oxidation on. In some embodiments, the oxidation is performed in the presence of a peroxycarboxylic acid, such as meta-Chloroperoxybenzoic acid (m-CPBA).
In some embodiments, a compound of Formula (M) or a salt thereof is prepared by a method comprising reacting a compound of Formula (O) with a compound of Formula (G) or a salt f. Any suitable conditions known in the art can be used.
In some embodiments, a compound of Formula (0) or a salt thereof is prepared by a method comprising reacting a nd of Formula (P) or salt thereof ;Hr2) with a phenyl disulfide of a (Q): p. In some embodiments, a compound of Formula (P) or a salt thereof is prepared by amidating the -C(0)0H group of a compound of Formula (D) or salt thereof. Any le conditions known in the art can be used.
SUBSTITUTE SHEET (RULE 26) onal embodiments include: 1. A compound of Formula I: 0 O X \(r2)„ R1 -r (R3), (R4), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein: - one of Y1 and Y2 is N and the other is CH; - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl , nated C1-C2 alkyl groups, and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally tuted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; SUBSTITUTE SHEET (RULE 26) - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8. 2. A compound of a II: 0 a x¥ \<R2)p / N N N R1 -r (R3)q (r4), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalky] groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is ndently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and SUBSTITUTE SHEET (RULE 26) - qis 0, 1,2, 3, 4, 5, 6, 7, or 8. 3. A nd of Formula III: 0 o. o \(R2)p N N N * (R3)q (R4)r (HI), a pharmaceutically acceptable salt thereof, or a ated derivative of any of the foregoing, wherein: - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and ns, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8.
SUBSTITUTE SHEET (RULE 26) 4. A compound according to any of embodiments 1-3, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein if R2 is cyano, then said R2 is meta or para relative to the sulfur atom.
. A compound according to any of embodiments 1-3, a ceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein: - each Ring A is independently chosen from C3-C10 lkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, and - each R is independently chosen from H and OH; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, and halogens; - R4 is F; - kis 0; - p is 0, 1, or 2; - qis 0, 1, 2, 3, or 4; - r is 0; and wherein m and n are not 0 at the same time. 6. A compound according to embodiment 5, a pharmaceutically acceptable salt thereof, or a deuterated tive of any of the ing, wherein: - R1 is chosen from (CR2)m-Ring A groups, wherein Ring A is chosen from C3-C10 cycloalkyl groups groups ally substituted with one or more substituents each ndently chosen from C1-C2 alkyl groups, nated C1-C2 alkyl groups, and halogens, - m is 1 or 2. 7. A compound according to embodiment 6, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein each R3 is a methyl group and q is 3 or 4. 8. A compound according to embodiment 7 having Formula IV: SUBSTITUTE SHEET (RULE 26) l0 0 0 ¥ H X(R2)p Ring AAx ' N N (IV), a pharmaceutically acceptable salt thereof, or a deuterated tive of any of the foregoing, wherein: - Ring A is chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens; and - each R2 is independently chosen from C1-C2 alkyl groups, OH, F, Cl, and Ci- C2 alkoxy groups; - m is 1 or 2; and - p is 0, 1, or 2. 9. A compound according to embodiment 8, wherein p is 0 or 1.
. A compound according to embodiment 8, wherein p is 0. 11. A nd according to embodiment 8 having Formula V: ° °v PV H r!,p Jrk0 XX XX ' N N N fS) Ring A SUBSTITUTE SHEET (RULE 26) a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, - Ring A is chosen from C3-C10 lkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, nated C1-C2 alkyl groups, and halogens; and - each R2 is independently chosen from C1-C2 alkyl , OH, F, Cl, and Ci- C2 alkoxy groups; - m is 1 or 2; and - p is 0, 1, or 2. 12. A compound according to any one of embodiments 1-11, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein each R2 is independently chosen from CH3, OH, F, and OCH3. 13. A compound according to embodiment 12, a ceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein p is 0 or 1. 14. A compound according to embodiment 13, a pharmaceutically acceptable salt thereof, or a ated derivative of any of the foregoing, wherein p is 0.
. A compound according to embodiment 11, a pharmaceutically acceptable salt f, or a deuterated derivative of any of the foregoing, wherein Ring A is a cyclopropyl group substituted with a halogenated Ci alkyl group or a halogenated C2 alkyl group. 16. A compound ing to embodiment 15, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein Ring A is a cyclopropyl group substituted with a CF3 group. 17. A compound according to embodiment 11, a pharmaceutically acceptable salt f, or a deuterated derivative of any of the foregoing, wherein m is 1, Ring A is a cyclopropyl group substituted with a CF3 group, p is 0 or 1, and R2, if present, is a methyl group, a hydroxy group, or a methoxy group.
SUBSTITUTE SHEET (RULE 26) 18. A compound according to embodiment 11, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, n m is 2, Ring A is a C3 cycloalkyl group tuted with a CF3 group, p is 0 or 1, and R2, if present, is a methyl group, a hydroxy group, or a methoxy group. 19. A compound according to embodiment 17 or 18, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein m is 2, Ring A is a cyclopropyl group substituted with a CF3 group, and p is 0.
. A compound according to ment 11, a pharmaceutically acceptable salt thereof, or a deuterated tive of any of the foregoing, wherein Ring A is chosen from C5 bicycloalkyl groups optionally substituted with one or more substituents each ndently chosen from Ci-C2 alkyl groups, halogenated Ci-C2 alkyl , and halogens. 21. A compound according to embodiment 20, a pharmaceutically able salt thereof, or a ated derivative of any of the foregoing, n Ring A is a C5 bicycloalkyl group optionally substituted with a halogen. 22. A compound according to embodiment 11, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein Ring A is chosen from C? bicycloalkyl groups and Cytricycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens. 23. A compound according to embodiment 22, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein Ring A is an unsubstituted Cy tricycloalkyl group. 24. A compound having a formula chosen from any one of the formulae depicted in a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing.
. A compound ing to embodiment 1 having the following formula: SUBSTITUTE SHEET (RULE 26) OS N N N'A(S) a pharmaceutically able salt thereof, or a deuterated derivative of any of the foregoing. 26. A nd according to embodiment 1 having the following formula: 0 0 0 0H oS V N N \(S) a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing. 27. A compound according to embodiment 1 having the following formula: 0 n'A(s) a pharmaceutically acceptable salt thereof, or a ated derivative of any of the foregoing. 28. A compound according to embodiment 1 having the following formula: SUBSTITUTE SHEET (RULE 26) HO KJ ,0 N a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing. 29. A compound according to embodiment 1 having the following formula: 0 Oo F rfVWs 0^/' N N N a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the . A nd having the following formula: r''V^N'®V^ N (S) a pharmaceutically acceptable salt f, or a deuterated derivative of any of the foregoing. 31. A compound having any one of the following formulae: SUBSTITUTE SHEET (RULE 26) |\L. ./x. 0 // N N N*A(S) F or P QO\'// 0~fJ NN. N'xfS) a pharmaceutically acceptable salt thereof, or a ated derivative of any of the foregoing. 32. A compound according to embodiment 1 having the following formula: 0 N cd3 F (S) F 6D or a pharmaceutically acceptable salt thereof. 33. A compound having the following formula: /VVA°"fn ysv^ HO 0-// 'N-m- -M N N R F DDD or a pharmaceutically able salt thereof.
SUBSTITUTE SHEET (RULE 26) 34. A compound according to embodiment 1 having the following formula: Q 0. 0 0 OH or a pharmaceutically acceptable salt thereof.
. A pharmaceutical composition comprising at least one compound chosen from compounds of any one of embodiments 1-34, a pharmaceutically able salt thereof, or a ated derivative of any of the foregoing, and optionally one or more of: (a) Compound II: Fx0-^^ OH F 0 F N / N a pharmaceutically able salt thereof, or a deuterated derivative of any of the foregoing; (b) Compound III: a pharmaceutically acceptable salt f, or a deuterated derivative of any of the foregoing; and (c) a pharmaceutically acceptable carrier.
SUBSTITUTE SHEET (RULE 26) WO 64632 36. A method of treating cystic fibrosis comprising administering to a patient in need thereof a pharmaceutical composition according to embodiment 35. 37. A method of preparing a compound of Formula : ...A H {R2)p rW/ n Y (R4)r (III) a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, comprising reacting a nd of Formula (F) or a salt thereof with a nd of Formula (G) or a salt thereof to generate said compound of Formula (Ilia) or a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing: P QO HN-0^R3)q o on ,sA'/ ^(O) yi ^ yi ^ 1 N-m^ nonG'/ ^ V2 x" 2 rV n-A i'MJ Y ^R3), (R4)r (F) (R4)r (Ilia) wherein in each of said ae: - one of Y1 and Y2 is N and the other is CH; - each R1 is independently chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups. wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and ns, wherein each R is independently chosen from H, OH, and Ci-C2 alkyl groups optionally substituted with one or more halogens; SUBSTITUTE SHEET (RULE 26) - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH ; - each R4 is independently chosen from halogens; - Xa is chosen from F or Cl; - each k is independently 0 or 1; - each r is independently 0 or 1; - each m is independently 0, 1, 2, or 3; - each n is independently 0 or 1; - each p is independently 0,1, 2, 3, 4, or 5; and - each q is independently 0, 1,2, 3, 4, 5, 6, 7, or 8. 38. The method of embodiment 37, wherein each Y2 is independently N; and each Y1 is independently CH. 39. The method of embodiment 37 or 38, wherein said reacting a compound of Formula (F) or a salt thereof with a compound of Formula (G) or a salt thereof is performed in the presence of a base. 40. The method of any one of embodiments 37-39, wherein a salt of nd of Formula (G) is employed. 41. The method of embodiment 40, wherein said salt of compound of Formula (G) is a HC1 salt of a compound of Formula (G). 42. A method of ing a compound of Formula (F) or a salt thereof: Q 0. o yi^s N" A c H 1r2)p N y2 R1-^ ^ Mr (F) SUBSTITUTE SHEET (RULE 26) or a deuterated derivative of any of the foregoing, comprising reacting a compound of Formula (D) or salt thereof with a compound of Formula (E) or a salt thereof to generate a compound of Formula (F) or a salt thereof: 2 Q 0 0 o Y1^. 's' In l>\ yi^ Rl-t^Y2" H OH Xa R1 /^'N^Y^X3 (E) (R4)r (F) wherein in each of said formulae: - one of Y1 and Y2 is independently N and the other is independently CFI; - each R1 is independently chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+1 groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, n each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is ndently chosen from halogens; - Xa is chosen from F or Cl; - each k is independently 0 or 1; - each r is independently 0 or 1; - each m is independently 0, 1, 2, or 3; SUBSTITUTE SHEET (RULE 26) - each n is independently 0 or 1; - each p is independently 0, 1.2, 3, 4, or 5; and - each q is independently 0, 1.2, 3, 4, 5, 6, 7, or 8. 43. The method of embodiment 42, wherein each Y2 is ndently N; and each Y1 is independently CH. 44. The method of embodiment 42 or 43, wherein said reacting a compound of Formula (D) or a salt thereof with a compound of a (E) or salt thereof is performed in the presence of a base. 45. The method of embodiment 42 or 43, wherein said reacting a compound of Formula (D) or salt f with a compound of Formula (E) or salt thereof comprises reacting a compound of Formula (D-l) with a coupling reagent and uently with a compound of Formula (E-l) in the presence of a base. 46. A method of preparing a compound of the following formula: CFs IX, or a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, comprising reacting a compound of Formula (F-l) or a salt thereof, wherein Xa is chosen from F or Cl, with a compound of Formula (G-l) or a salt thereof to generate said compound or a ceutically acceptable salt thereof, or a deuterated tive of any of the foregoing: HN PF3 PF3 o 0 0 o o 0 XN:s',ph AN:s:Ph (G-1) b-oN N J H H \ N«m°~O n N Xa (F-1) Compound (1) SUBSTITUTE SHEET (RULE 26) 47. The method of embodiment 46, wherein said reacting a compound of Formula (F-l) or a salt thereof with a nd of Formula (G-l) or a salt thereof is performed in the presence of a base. 48. The method of embodiment 46 or 47, wherein a salt of compound of Formula (G-l) is employed. 49. The method of embodiment 48, n said salt of compound of Formula (G-l) is aHCl salt of a compound of Formula (G-l). 50. A method of preparing a compound of Formula (F-l) or a salt thereof: ~a H Nk,,, 0 N' 'Xa (F-1) or a ated derivative of any of the foregoing, comprising reacting a compound of a (D-l) and a compound of Formula (E-l) to generate a compound of Formula (F-l) or a salt thereof: PF3 0 PF3 o o 0 PhS02NH2 "A- XN:s:Ph OH (E-1) \ N.m N Xa \ n-m N Xa (D-1) (F-1) wherein each Xa is ndently chosen from F or Cl. 51. The method of ment 50, wherein said reacting a compound of Formula (D-l) or a salt thereof with a compound of Formula (E-l) or a salt thereof is performed in the ce of a base. 52. The method of embodiment 50, wherein said reacting a compound of Formula (D-l) or a salt thereof with a compound of Formula (E-l) or a salt thereof comprises reacting a compound of Formula (D-l) with a coupling reagent and subsequently with a compound of Formula (E-l) in the presence of a base.
SUBSTITUTE SHEET (RULE 26) 53. A method of preparing a compound of Formula (D) or a salt thereof: ''s-'VY1^ OH R1-// Xa (R4)r or a deuterated derivative of any of the ing, comprising: (i) reacting a compound of Formula (A) or a salt thereof with a compound of Formula (B) or a salt thereof to generate a compound of Formula (C) or a salt thereof: y2^ ORa CI^Y1"" O R' N xa (B) Y1 'V' OR riJHAY* xa (R4)r (C) ; and (ii) hydrolyzing the -C(0)0Ra group of a compound of a (C) to generate a compound of Formula (D) or a salt thereof, wherein in each said formulae: - one of Y1 and Y2 is ndently N and the other is independently CH; - each R1 is ndently chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalky] groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and ns, SUBSTITUTE SHEET (RULE 26) wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; --each Ra is independently chosen from C1-C4 alkyl; - each Xa is independently chosen from F or Cl; - each k is independently 0 or 1; - each r is independently 0 or 1; - each m is independently 0,1, 2, or 3; - each n is independently 0 or 1; - each p is independently 0,1, 2, 3, 4, or 5; and - each q is independently 0, 1,2, 3, 4, 5, 6, 7, or 8. 54. The method of embodiment 53, wherein each Y2 is independently N; and each Y1 is independently CH. 55. The method of ment 53 or 54, wherein the ysis of the -C(0)ORa group is performed in the presence of a base. 56. The method of any one of embodiments 53-55, wherein said reacting a compound of Formula (A) or a salt thereof with a compound of Formula (B) or salt thereof is performed in the presence of a base. 57. The method of any one of ments 53-56, wherein Ra is ethyl or l. 58. A method of preparing a compound of Formula (D-l) or a salt thereof: SUBSTITUTE SHEET (RULE 26) CF3 0 (D-1) or a ated derivative of any of the foregoing, comprising: (i) reacting a compound of a (A-l) or a salt thereof and a compound of Formula (B-l) or a salt thereof to generate a compound of Formula (C-l) or a salt thereof: CFs ORa CFs 0 X* N X* °-v (B-1) "ORa (A-1) (C-1) (ii) hydrolyzing the -C(0)0Ra group of a compound of Formula (C-l) or a salt thereof to generate a compound of Formula (D-1) or a salt thereof, wherein each Ra is independently chosen from C1-C4 alkyl; and each - Xa is independently chosen from F or Cl. 59. The method of embodiment 58, wherein the hydrolysis of the -C(0)0Ra group is performed in the presence of a base. 60. The method of 58 or 59, wherein said reacting a compound of Formula (A-l) or a salt thereof and a nd of Formula (B-l) or a salt thereof is performed in the presence of a base. 61. The method of any one of embodiments 58-60, wherein Ra is ethyl or l. 62. A method of preparing a compound of Formula (I) or a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, sing reacting a compound of Formula (L) or a salt f with NR*3 : SUBSTITUTE SHEET (RULE 26) 0 o v' v2^ II 11 n Yf NR*3 Y R%v RXy ^R3), (R4)r wherein in each of said formulae: - X is NH orN(Ci-C4 alkyl); - one of Y1 and Y2 is independently N and the other is independently CH; - each R1 is independently chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl , halogenated C1-C2 alkyl groups, and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy , halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - R* is H or C1-C4 alkyl.
- Xa is chosen from F or Cl; - each k is independently 0 or 1; - each r is independently 0 or 1; - each m is independently 0, 1, 2, or 3; - each n is independently 0 or 1; SUBSTITUTE SHEET (RULE 26) - each p is independently 0, l, 2, 3, 4, or 5; and - each q is independently 0, 1.2, 3, 4, 5, 6, 7, or 8. 63. Use of at least one compound chosen from nds of any one of embodiments 1-34, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, and optionally one or more of: (a) Compound II: Fxl0 N F 0 ^ 0 F N / N a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing; and (b) Compound III: a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing; for treating cystic fibrosis.
] Methods of Preparing Compounds General Experimental Procedures Reagents and starting materials were obtained by commercial sources unless otherwise stated and were used without purification. Proton and carbon NMR a were acquired on either of a Bruker n DRX 400 MHz FTNMR spectrometer ing at a H and C resonant frequency of 400 and 100 MHz respectively, or on a 300 MHz NMR spectrometer. One dimensional proton and carbon spectra were SUBSTITUTE SHEET (RULE 26) acquired using a broadband observe (BBFO) probe with 20 Hz sample on at 0.1834 and 0.9083 Hz/Pt digital resolution respectively. All proton and carbon spectra were acquired with temperature l at 30°C using standard, usly published pulse sequences and routine processing parameters. Final purity of compounds was determined by reversed phase UPLC using an Acquity UPLC BEH Cig column (50 x 2.1 mm, 1.7 pm particle) made by Waters (pn: 186002350), and a dual nt run from 1-99% mobile phase B over 3.0 minutes. Mobile phase A = H2O (0.05 % CF3CO2H). Mobile phase B = CFF,CN (0.035 % CF3CO2H). Flow rate = 1.2 mL/min, injection volume = 1.5 pL, and column temperature = 60 °C. Final purity was calculated by averaging the area under the curve (AUC) of two UY traces (220 nm, 254 nm). Low-resolution mass spectra were reported as [M+H]+ species obtained using a single quadrupole mass spectrometer equipped with an electrospray ionization (ESI) source capable of achieving a mass accuracy of 0.1 Da and a minimum resolution of 1000 (no units on resolution) across the ion range. Optical purity of methyl (2S)- 2,4-dimethylnitro-pentanoate was determined using chiral gas chromatography (GC) analysis on an Agilent 7890A/MSD 5975C instrument, using a Restek Rt-(3DEXcst (30m x 0.25mm x 0.25um_df) column, with a 2.0 mL/min flow rate (H2 r gas), at an injection temperature of 220°C and an oven temperature of 120°C, 15 minutes.
Example 1: ation of a Spray Dried Dispersion (SDD) of Compound 1 A spray dried dispersion of Compound 1 was prepared using Buchi Mini Spray Dryer B290. HPMCAS-HG (6.0 grams) was dissolved in 200 mL of MeOH (methanol)/DCM (dichloromethane) (1/1), and Compound 1 (6.0 grams) was added and stirred for 30 minutes forming a clear on. The resulting on was spray dried under the following conditions resulting in a 50% Compound 1/50% HPMCAS- HG spray dried sion (Yield: 80%, Solid load: 6%).
Conditions Inlet Temperature (°C) 77 Outlet Temperature (°C) 39 Nitrogen Pressure (PSI) 95 SUBSTITUTE SHEET (RULE 26) WO 64632 Aspirator (%) 100 Pump (%) 30 Rotameter (mm) 60 Filter Pressure (mBar) -50 Condenser Temperature (°C) -10 Powder X-ray Diffraction The powder x-ray diffraction measurements were performed using PANalyticaTs X-pert Pro ctometer at room temperature with copper radiation (1.54060 A). The incident beam optic was comprised of a variable ence slit to ensure a constant illuminated length on the sample and on the diffracted beam side; a fast linear solid state detector was used with an active length of 2.12 degrees 2 theta measured in a scanning mode. The powder sample was packed on the ed area of a zero background silicon holder and spinning was performed to achieve better statistics.
A symmetrical scan was measured from 4-40 degrees 2 theta with a step size of 0.017 degrees and a scan step time of 15.5s. shows the XRPD spectrum of a SDD of 50% Compound 1 in -HG, and shows that Compound 1 is amorphous in the SDD.
Modulated Differential Scanning Calorimetry (MDSC) MDSC was used to determine the glass transition ature of the amorphous material. MDSC was performed using TA Discovery DSC differential scanning calorimeter (TA Instruments, New , DE). The instrument was calibrated with indium. Samples of approximately 1-3 mg were weighed into hermetic pans that were crimped using lids with one hole. The MDSC sample was scanned from -20°C to 210°C at a heating rate of 20C/min with +/- 1°C of modulation within 1 minute. Data was collected and analyzed by TA Instruments Trios Software (TA Instruments, New Castle, DE). shows a MDSC spectrum of a SDD of 50% Compound 1 in HPMCAS-HG, and shows that the SDD has an onset temperature of about 75.6°C, a midpoint temperature of about 82.7°C, and an offset ature of about 89.7°C.
SUBSTITUTE SHEET (RULE 26) WO 64632 Example 2: Synthesis of Compound II: (R)-l-(2,2- Difluorobenzo[d][l,3]dioxolyl)-N-(l-(2,3-dihydroxypropyl)fluoro(lhydroxymethylpropanyl )-lH-indolyl)cyclopropanecarboxamide 02N. '£3 F' N ■OCH2Ph H d CsC03, DMF o2n o2n LiAIH/i. TK)F F N + F N •O -o ■□•V oV 02N OH H2N H2, Pd-C ito:OH toOH SOCI7, DMF -O XV 2) Et3N, CH2CI2 -O OH Step 1: (R)-Benzyl 2-(l-((2,2-dimethyl-l,3-dioxolanyl)methyl) nitro-lH-indolyl)methylpropanoate and ((S)-2,2-Dimethyl-l,3- dioxolanyl)methyl 2-(l-(((R)-2,2-dimethyl-l,3-dioxolanyl)methyl)fluoro nitro-lH-indolyl)methylpropanoate Cesium carbonate (8.23 g, 25.3 mmol) was added to a mixture of benzyl 2-(6- fluoronitro-lH-indolyl)methylpropanoate (3.0 g, 8.4 mmol) and (S)-(2,2- dimethyl-l,3-dioxolanyl)methyl 4-methylbenzenesulfonate (7.23 g, 25.3 mmol) in DMF (N,N-dimethylformamide) (17 mL). The reaction was stirred at 80 °C for 46 hours SUBSTITUTE SHEET (RULE 26) under a nitrogen atmosphere. The e was then partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate. The combined ethyl acetate layers were washed with brine, dried over MgS04, filtered and concentrated.
The crude product, a viscous brown oil which contains both of the products shown above, was taken directly to the next step without further purification. (R)-Benzyl 2-(l- ((2,2-dimethyl-l,3-dioxolanyl)methyl)fluoronitro-lH-indolyl) methylpropanoate, ESI-MS m/z calc. 470.2, found 471.5 (M+l)+. Retention time 2.20 minutes. ((S)-2,2-Dimethyl-l,3-dioxolanyl)methyl 2-(l-(((R)-2,2-dimethyl-1,3- dioxolanyl)methyl)fluoronitro-lH-indolyl)methylpropanoate, ESI-MS m/z calc. 494.5, found 495.7 (M+l)+. Retention time 2.01 minutes.
Step 2: (l-((2,2-dimethyl-1,3-dioxolanyl)methyl)fluoro nitro-lH-indolyl)methylpropan-l-ol The crude reaction mixture obtained in step (A) was dissolved in THE (tetrahydrofuran) (42 mL) and cooled in an ice-water bath. L1AIH4 (16.8 mL of 1 M solution, 16.8 mmol) was added drop-wise. After the addition was te, the mixture was stirred for an additional 5 minutes. The reaction was quenched by adding water (1 mL), 15% NaOEl solution (1 mL) and then water (3 mL). The mixture was filtered over Celite, and the solids were washed with THE and ethyl acetate. The filtrate was concentrated and purified by column chromatography (30-60% ethyl acetatehexanes ) to obtain (l-((2,2-dimethyl-l,3-dioxolanyl)methyl)fluoronitrolH-indolyl )methylpropan-l-ol as a brown oil (2.68g, 87 % over 2 steps). ESI-MS m/'z calc. 366.4, found 367.3 (M+l)+. Retention time 1.68 s, 'll NMR (400 MHz, ?6) 5 8.34 (d, J = 7.6 Hz, 1H), 7.65 (d, J = 13.4 Hz, 1H), 6.57 (s, 1H), 4.94 (t, J = 5.4 Hz, 1H), 4.64 - 4.60 (m, 1H), 4.52 - 4.42(m, 2H), 4.16 - 4.14 (m, 1H), 3.76 - 3.74 (m, 1H), 3.63 - 3.53 (m, 2H), 1.42 (s, 3H), 1.38 - 1.36 (m, 6H) and 1.19 (s, 3H) ppm. (DMSO is dimethylsulfoxide).
] Step 3: (R)(5-amino-l-((2,2-dimethyl-l,3-dioxolanyl)methyl) fluoro- lH-indolyl)methylpropan- l-ol (R)(l-((2,2-dimethyl-l,3-dioxolanyl)methyl)fluoronitro-lH-indol- 2-yl)methylpropan-l-ol (2.5 g, 6.82 mmol) was ved in ethanol (70 mL) and the reaction was flushed withN2. Then Pd-C (250 mg, 5% wt) was added. The reaction was flushed with nitrogen again and then stirred under H2 (atm). After 2.5 hours only partial SUBSTITUTE SHEET (RULE 26) conversion to the t was observed by LCMS. The reaction was filtered through Celite and concentrated. The residue was re-subjected to the conditions above. After 2 hours LCMS indicated complete conversion to product. The reaction mixture was filtered through Celite. The filtrate was concentrated to yield the product (1.82 g, 79 %).
ESI-MS m/z calc. 336.2, found 337.5 (M+l)+. Retention time 0.86 minutes, 'tt NMR (400 MHz, DMSO-<76) 5 7.17 (d, J = 12.6 Hz, 1H), 6.76 (d, J = 9.0 Hz, 1H), 6.03 (s, 1H), 4.79 - 4.76 (m, 1H), 4.46 (s, 2H), 4.37 - 4.31 (m, 06 (dd, J = 6.1, 8.3 Hz, 1H), 3.70 - 3.67 (m, 1H), 3.55 - 3.52 (m, 2H), 1.41 (s, 3H), 1.32 (s, 6H) and 1.21 (s, 3H) Step 4: (R)-l-(2,2-difluorobenzo[d][l,3]dioxolyl)-N-(l-((2,2-dimethyl- l,3-dioxolanyl)methyl)fluoro(l-hydroxymethylpropanyl)-lH-indol yljcyclopi'opanecarboxainide DMF (3 drops) was added to a stirring mixture of 1-(2,2- difluorobenzo[d][l,3]dioxolyl)cyclopropanecarboxylic acid (1.87 g, 7.7 mmol) and thionyl chloride (1.30 mL, 17.9 mmol). After 1 hour a clear solution had formed. The solution was concentrated under vacuum and then toluene (3 mL) was added and the mixture was concentrated again. The toluene step was ed once more and the residue was placed on high vacuum for 10 minutes. The acid de was then dissolved in dichloromethane (10 mL) and added to a mixture of (5-amino-l- ((2,2-dimethyl-l,3-dioxolanyl)methyl)fluoro-lH-indolyl)methylpropan-l-ol (1.8 g, 5.4 mmol) and triethylamine (2.24 mL, 16.1 mmol) in dichloromethane (45 mL).
The reaction was stirred at room temperature for 1 hour. The reaction was washed with IN HC1 on, ted NaHCCL solution and brine, dried over MgSCL and concentrated to yield the product (3g, 100%). ESI-MS m/z calc. 560.6, found 561.7 (M+l)+. Retention time 2.05 minutes, 'll NMR (400 MHz, DMSO-t/6) 8 8.31 (s, 1H), 7.53 (s, 1H), 7,42 - 7.40 (m, 2H), 7,34 - 7.30 (m, 3H), 6.24 (s, 1H), 4.51 - 4.48 (m, 1H), 4.39 - 4.34 (m,2H), 4.08 (dd, J = 6.0, 8.3 Hz, 1H), 3.69 (t, J = 7.6 Hz, 1H), 3.58 - 3.51 (m, 2H), 1.48 - 1.45 (m, 2H), 1.39 (s, 3H), 1.34 - 1.33 (m, 6H), 1.18 (s, 3H) and 1.14 - 1.12 (m, 2H) ppm SUBSTITUTE SHEET (RULE 26) Step 5: (2,2-difluorobenzo[d] [l,3]dioxolyl)-N-(l-(2,3- dihydroxypropyl)fluoro(l-hydroxymethylpropanyl)-lH-indol yl)cyclopropanecarboxamide (2,2-difluorobenzo[d][l,3]dioxolyl)-N-(l-((2,2-dimethyl-l,3- dioxolanyl)methyl)fluoro(l-hydroxymethylpropanyl)-lH-indol yl)cyclopropanecarboxamide (3.0 g, 5.4 mmol) was dissolved in methanol (52 mL).
Water (5.2 mL) was added followed by p-Ts0H.H20 (p-toluenesulfonic acid hydrate) (204 mg, 1.1 mmol). The reaction was heated at 80 °C for 45 s. The on was concentrated and then partitioned between ethyl acetate and ted NaHCO, solution.
The ethyl acetate layer was dried over MgSC^ and concentrated. The residue was purified by column chromatography (50-100 % ethyl acetate - hexanes) to yield the product. (1.3 g, 47 %, ee >98% by SFC). ESI-MS m/z calc. 520.5, found 521.7 (M+l)+.
Retention time 1.69 minutes. 'H NMR (400 MHz, DMSO-e/6) 8 8.31 (s, 1H), 7.53 (s, 1H), 7.42 - 7.38 (m, 2H), 7.33 - 7.30 (m, 2H), 6.22 (s, 1H), 5.01 (d, J = 5.2 Hz, 1H), 4.90 (t, J = 5.5 Hz, 1H), 4.75 (t, J = 5.8 Hz, 1H), 4.40 (dd, J = 2.6, 15.1 Hz, 1H), 4.10 (dd, J = 8.7, 15.1 Hz, 1H), 3.90 (s, 1H), 3.65 - 3.54 (m, 2H), 3.48 - 3.33 (m, 2H), 1.48 - 1.45 (m, 2H), 1.35 (s, 3H), 1.32 (s, 3H) and 1.14-1.11 (m, 2H) ppm.
Example 3: Synthesis of Compound HI: N-(2,4-di-/e/7-butyl hydroxyphenyl)oxo-l,4-dihydroquinolinecarboxamide Part A: Synthesis of 4-oxo-l,4-dihydroquinolinecarboxylic acid Step 1: 2-Phenylaminomethylene-malonic acid diethyl ester A mixture of aniline (25.6 g, 0.275 mol) and diethyl 2- (ethoxymethylene)malonate (62.4 g, 0.288 mol) was heated at 140-150 °C for 2 h. The mixture was cooled to room temperature and dried under reduced pressure to afford 2- phenylaminomethylene-malonic acid diethyl ester as a solid, which was used in the next step without further purification, fa NMR (DMSO-c/r,) 5 11.00 (d, 1H), 8.54 (d, J= SUBSTITUTE SHEET (RULE 26) 13.6 Hz, 1H), 7.36-7.39 (m, 2H), 7.13-7.17 (m, 3H), 4.17-4.33 (m, 4H), .40 (m, Step 2: 4-Hydroxyquinolinecarboxylic add ethyl ester AIL three-necked flask fitted with a mechanical stirrer was charged with 2- phenylarmnomethylene-malonic acid diethyl ester (26.3 g, 0.100 mol), osphoric acid (270 g) and phosphoryl chloride (750 g). The mixture was heated to 70 °C and stirred for 4 h. The mixture was cooled to room temperature and filtered. The residue was treated with aqueous Na2C03 solution, filtered, washed with water and dried. 4- Hydroxyquinolinecarboxylic acid ethyl ester was obtained as a pale brown solid (15.2 g, 70%). The crude product was used in next step without further purification.
Step 3: 4-Oxo-l,4-dihydroquinolinecarboxylic add 4-Hydroxyquinolinecarboxylic acid ethyl ester (15 g, 69 mmol) was suspended in sodium hydroxide solution (2N, 150 mL) and stirred for 2 h at reflux.
After cooling, the mixture was filtered, and the filtrate was ied to pH 4 with 2N HC1. The ing precipitate was collected via filtration, washed with water and dried under vacuum to give 4-oxo-l,4-dihydroquinolinecarboxylic acid as a pale white solid (10.5 g, 92 %). 'H NMR (DMSO-r/tf) 5 15.34 (s, 1 H), 13.42 (s, 1 H), 8.89 (s, 1H), 8.28 (d, ,/= 8.0 Hz, 1H), 7.88 (m, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.60 (m, 1H).
Part B: Synthesis of N-(2,4-di-fe/7-butylhydroxyphenyl)oxo-l,4- dihydroquinolinecarboxamide SUBSTITUTE SHEET (RULE 26) CIC02Me HN03, H2S04 NEt3, DMAP ’OH o 02N' o 0 ch2ci2 O^O KOH, MeOH o2n OH hco2nh4 02N' 'OH Pd-C, EtOH H2N' 'OH Step 1: Carbonic acid -fe/7-butyl-phenyl ester methyl ester Methyl chloroformate (58 mL, 750 mmol) was added dropwise to a solution of 2,4-di-fert-butyl-phenol (103.2 g, 500 mmol), EhN (139 mL, 1000 mmol) and DMAP (3.05 g, 25 mmol) in dichloromethane (400 mL) cooled in an ice-water bath to 0 °C. The mixture was allowed to warm to room temperature while stirring overnight, then filtered through silica gel (approx. 1L) using 10% ethyl acetate - hexanes (~ 4 L) as the eluent. The combined tes were concentrated to yield carbonic acid 2,4-di- fe/7-butyl-phenyl ester methyl ester as a yellow oil (132 g, quant.), 'h NMR (400 MHz, DMSO-c/g) 5 7.35 (d, J = 2.4 Hz, 1H), 7.29 (dd, J = 8.5, 2.4 Hz, 1H), 7.06 (d, J = 8.4 Hz, 1H), 3.85 (s, 3H), 1.30 (s, 9H), 1.29 (s, 9H).
] Step 2: Carbonic acid 2,4-di-to7-butylnitro-phenyl ester methyl ester and Carbonic acid 2,4-di-fc/f-butyliiitro-phenyl ester methyl ester ] To a stirring mixture of carbonic acid 2.4-di-/e/V-butyl-phenyl ester methyl ester (4.76 g, 180 mmol) in cone, sulfuric acid (2 mL), cooled in an ice-water bath, was added a cooled mixture of sulfuric acid (2 mL) and nitric acid (2 mL). The addition was done slowly so that the on ature did not exceed 50 °C. The reaction was allowed to stir for 2 h while warming to room temperature. The reaction mixture was then added to ice-water and extracted into diethyl ether. The ether layer was dried SUBSTITUTE SHEET (RULE 26) (MgS04), concentrated and purified by column chromatography (0 - 10% ethyl acetate - hexanes) to yield a mixture of carbonic acid 2,4-di-rer/-butylnitro-phenyl ester methyl ester and carbonic acid 2,4-di-fert-butylnitro-phenyl ester methyl ester as a pale yellow solid (4.28 g), which was used directly in the next step.
Step 3: -/e/7-butylnitro-phenol and 2,4-Di-te/7-butylnitrophenol The mixture of carbonic acid 2,4-di-/er/-butylnitro-phenyl ester methyl ester and carbonic acid 2,4-di-/erf-butylmtro-phenyl ester methyl ester (4.2 g, 14.0 mmol) was dissolved in MeOH (65 mL) before KOH (2.0 g, 36 mmol) was added. The mixture was stirred at room temperature for 2 h. The reaction mixture was then made acidic (pH 2-3) by adding cone. HC1 and partitioned between water and diethyl ether.
The ether layer was dried (MgSCf). concentrated and purified by column tography (0-5 % ethyl acetate - hexanes) to provide 2.4-di-fcr/-butvlnitrophenol (1.31 g, 29% over 2 steps) and 2,4-di-/ert-butylnitro-phenol. 2.4-Di-/e/7- butylnitro-phenol: 1HNMR(400 MHz, DMSO-c4) 5 10.14 (s, 1H, OH), 7.34 (s, 1H), 6.83 (s, 1H), 1.36 (s, 9H), 1.30 (s, 9H). 2.4-Di-/m-butylnitro-phenol: JH NMR (400 MHz, CDC13) 5 11.48 (s, 1H), 7.98 (d, J = 2.5 Hz, 1H), 7.66 (d, J = 2.4 Hz, 1H), 1.47 (s, 9H), 1.34 (s, 9H).
] Step 4: 5-Amino-2,4-di-to7-butyl-phenol To a refluxing on of 2.4-di-/m-butylnitro-phenol (1.86 g, 7.40 mmol) and ammonium e (1.86 g) in ethanol (75 mL) was added Pd-5% wt. on activated carbon (900 mg). The reaction mixture was stirred at reflux for 2 h, cooled to room temperature and filtered through Celite. The Celite was washed with ol and the combined filtrates were concentrated to yield 5-amino-2,4-di-/ert-butyl-phenol as a grey solid (1.66 g, quant). XH NMR (400 MHz, /6) 5 8.64 (s, 1H, OH), 6.84 (s, 1H), 6.08 (s, 1H), 4.39 (s, 2H, NH2), 1.27 (m, 18H); HPLC ret. time 2.72 mm, 10-99 % CH3CN, 5 min run; ESI-MS 222.4 m/z [M+H]+.
Step 5: N-(5-hydroxy-2,4-di-terf-butyl-phenyl)oxo-lH-quinoline carboxamide SUBSTITUTE SHEET (RULE 26) O OH O HN' OH O o N N H H2N OH H To a suspension of 4-oxo-l,4-dihydroquinolincarboxylic acid (35.5 g: 188 mmol) and HBTU (85.7 g, 226 mmol) in DMF (280 mL) was added Et3N (63.0 mL, 451 mmol) at ambient temperature. The mixture became homogeneous and was allowed to stir for 10 min before 5-amino-2.4-di-/c/7-buty 1-phenol (50.0 g, 226 mmol) was added in small portions. The mixture was allowed to stir overnight at ambient temperature.
The mixture became heterogeneous over the course of the reaction. After all of the acid was consumed (LC-MS is, MH+ 190, 1.71 min), the solvent was removed in vacuo. EtOH (ethyl alcohol) was added to the orange solid material to produce a slurry.
The mixture was stirred on a rotovap (bath temperature 65 °C) for 15 min without placing the system under . The mixture was filtered and the captured solid was washed with hexanes to e a white solid that was the EtOH crystalate. Et20 yl ether) was added to the solid obtained above until a slurry was formed. The mixture was stirred on a rotovapor (bath temperature 25 °C) for 15 min without placing the system under vacuum. The mixture was filtered and the solid captured. This procedure was performed a total of five times. The solid obtained after the fifth precipitation was placed under vacuum overnight to e N-(5-hydro\y-2.4-di-/mbutyl-phenyl )oxo-lH-quinolinecarboxamide (38 g, 52%). HPLC ret. time 3.45 min, 10-99% CH3CN, 5 min run; 1HNMR(400 MHz, DMSO-c/6) 5 12.88 (s, 1H), 11.83 (s, 1H), 9.20 (s, 1H), 8.87 (s, 1H), 8.33 (dd, J = 8.2, 1.0 Hz, 1H), 7.83-7.79 (m, 1H), 7.76 (d, J = 7.7 Hz, 1H), 7.54-7.50 (m, 1H), 7.17 (s, 1H), 7.10 (s, 1H), 1.38 (s, 9H), 1.37 (s, 9H); ESI-MS m/z cak’d 392.21; found 393.3 [M+H]+.
Example 4: Synthesis of Compounds 1-65 Synthetic e 1: Synthesis of N-(benzenesulfonyl)[3-[2-[l- (trifhioromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-ylJpyridinecarboxamide (Compound 1) SUBSTITUTE SHEET (RULE 26) Part A: Synthesis of (4S)-2,2,4-trimethylpyrrolidine hydrochloride 0 o no2 PalataseLipase THF, Base no2 I O i) ,SJ LiAIH4 Raney Ni, H? HN' HN^VS) no2 * ii) Step 1: Synthesis of methyl-2,4-dimethylnitro-pentanoate 0 O 0 0 ydrofuran (THF, 4.5 L) was added to a 20 L glass reactor and stirred under N2 at room temperature. 2-Nitropropane (1.5 kg. 16.83 mol) and 1,8- diazabicyclo[5.4.0]undecene (DBU) (1.282 kg, 8.42 mol) were then charged to the reactor, and the jacket temperature was increased to 50 °C. Once the reactor contents were close to 50 °C, methyl methacrylate (1.854 kg, 18.52 mol) was added slowly over 100 minutes. The reaction temperature was maintained at or close to 50 °C for 21 hours.
The reaction mixture was concentrated in vacuo then transferred back to the reactor and diluted with methyl te/V-butyl ether (MTBE) (14 L). 2 M HCI (7.5 L) was added, and this mixture was stirred for 5 minutes then allowed to . Two clear layers were e - a lower yellow aqueous phase and an upper green organic phase. The aqueous layer was removed, and the organic layer was stirred again with 2 M HCI (3 L). After tion, the HCI washes were ined and stirred with MTBE (3 L) for 5 minutes. The aqueous layer was removed, and all of the organic layers were combined in the reactor and stirred with water (3 L) for 5 minutes. After separation, the organic layers were concentrated in vacuo to afford a cloudy green oil. This was dried with MgSCE and filtered to afford methyi-2,4-dimethylmtro-pentanoate as a clear green oil (3.16 kg, 99% yield). NMR (400 MHz, form-c/) 5 3.68 (s, 3H), 2.56 - 2.35 (m, 2H), 2.11-2.00 (m, 1H), 1.57 (s, 3H), 1.55 (s, 3H), 1.19 (d, 6.8 Hz, 3H).
SUBSTITUTE SHEET (RULE 26) Step 2: sis of methyl (2S)-2,4-dimethylnitro-pentanoate 0 0 / PalataseLipase^ NOo N02 A r was charged with purified water (2090 L; 10 vol) and then potassium phosphate sic (27 kg, 198.4 moles; 13 g/L for water charge). The pH of the reactor contents was adjusted to pH 6.5 (± 0.2) with 20% (w/v) potassium carbonate solution. The reactor was charged with racemic methyl-2,4-dimethylnitropentanoate (209 kg; 1104.6 , and Palatase 20000L lipase (13 L, 15.8 kg; 0.06 vol).
The reaction mixture was adjusted to 32 ± 2 °C and stirred for 15-21 hours, and pH 6.5 was maintained using a pH stat with the automatic addition of 20% potassium ate solution. When the racemic starting material was converted to >98% ee of the S-enantiomer, as determined by chiral GC, external heating was switched off. The reactor was then charged with MTBE (35 L; 5 vol), and the s layer was extracted with MTBE (3 times, 400-1000L). The combined organic extracts were washed with aqueous Na2C03 (4 times, 522 L, 18 % w/w 2.5 vol), water (523 L; 2.5 vol), and 10% aqueous NaCl (314 L, 1.5 vol). The organic layer was concentrated in vacuo to afford methyl (25')-2.4-dimethylnitro-pentanoate as a mobile yellow oil (>98% ee, 94.4 kg; 45 % yield).
Step 3: Synthesis of (3S)-3,5,5-trimethylpyrrolidinone Raney-Ni O H2 (5) O' ■*> ] A 20 L reactor was purged with N2. The vessel was charged sequentially with DI water-rinsed, damp Raney® Ni (2800 grade, 250 g), methyl (2S)-2,4-dimethyl mtro-pentanoate , 9.2 mol), and ethanol (13.9 L, 8 vol). The reaction was stirred at 900 rpm, and the reactor was flushed with H2 and maintained at ~2.5 bar. The reaction mixture was then warmed to 60 °C for 5 hours. The reaction mixture was cooled and filtered to remove Raney nickel, and the solid cake was rinsed with ethanol SUBSTITUTE SHEET (RULE 26) (3.5 L, 2 vol). The ethanolic solution of the product was ed with a second equal sized batch and concentrated in vacuo to reduce to a m volume of ethanol (~1.5 volumes). Heptane (2.5 L) was added, and the suspension was concentrated again to -1.5 volumes. This was repeated 3 times; the resulting sion was cooled to 0-5 °C, fdtered under suction, and washed with heptane (2.5 L). The product was dried under vacuum for 20 minutes then transferred to drying trays and dried in a vacuum oven at 40 °C overnight to afford (3S)-3,5,5-trimethylpyrrolidinone as a white crystalline solid (2.042 kg, 16.1 mol, 87 %). 'H NMR (400 MHz, Chlorofomw/) 8 6.39 (s, 1H), 2.62 (ddq, J = 9.9, 8.6, 7.1 Hz, 1H), 2.17 (dd, J = 12.4, 8.6 Hz, 1H), 1.56 (dd, J = 12.5, 9.9 Hz, 1H), 1.31 (s, 3H), 1.25 (s, 3H), 1.20 (d, J = 7.1 Hz, 3H).
] Step 4: Synthesis of (4S)-2,2,4-trimethylpyrrolidine hydrochloride (S) i) LiAlH4 HN ii) HC1 A glass lined 120 L reactor was charged with lithium ium e pellets (2.5 kg, 66 mol) and dry THF (60 L) and warmed to 30 °C. The resulting suspension was charged with (S)-3,5,5-trimethylpyrrolidinone (7.0 kg, 54 mol) in THF (25 L) over 2 hours while maintaining the reaction temperature at 30 to 40 °C.
After complete addition, the reaction temperature was increased to 60 - 63 °C and maintained overnight. The reaction mixture was cooled to 22 °C, then cautiously quenched with the addition of ethyl acetate (EtOAc) (1.0 L, 10 moles), followed by a mixture of THF (3.4 L) and water (2.5 kg, 2.0 eq), and then a mixture of water (1.75 kg) with 50 % aqueous sodium hydroxide (750 g, 2 equiv water with 1.4 equiv sodium hydroxide relative to aluminum), followed by 7.5 L water. After the addition was complete, the reaction mixture was cooled to room temperature, and the solid was removed by filtration and washed with THF (3 x 25 L). The filtrate and washings were combined and treated with 5.0 L (58 moles) of aqueous 37% HC1 (1.05 equiv.) while maintaining the temperature below 30°C. The ant solution was concentrated by vacuum distillation to a slurry. Isopropanol (8 L) was added and the solution was concentrated to near dryness by vacuum distillation. panol (4 L) was added, and the t was slurried by warming to about 50 T. MTBE (6 L) was added, and the SUBSTITUTE SHEET (RULE 26) slurry was cooled to 2-5 °C. The product was collected by filtration and rinsed with 12 L MTBE and dried in a vacuum oven (55 oC/300 torr/N2 bleed) to afford (4S)-2,2,4- trimethylpyrrolidine*HCl as a white, crystalline solid (6.21 kg, 75% yield). NMR (400 MHz, DMSO-t/6) 6 9.34 (hr d, 2H), 3.33 (dd,J= 11.4, 8.4 Hz, 1H), 2.75 (dd,/ = 11.4, 8.6 Hz, 1H), 2.50-2.39 (m, 1H), 1.97 (dd,J= 12.7, 7.7 Hz, 1H), 1.42 (s,3H), 1.38 (dd, J= 12.8, 10.1 Hz, 1H), 1.31 (s, 3H), 1.05 (d, .7=6.6 Hz, 3H).
Part B: sis of N-(benzenesulfonyl)[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide HO cf3 H O . o , 0"CnAo^ DIAD TFA N. f3c f3c 0 I 1) TFA is k2co3 cr n ci DABCO P-{j 'N a 2) GDI, PhS02NH2, f3c DBU O^rjN-m^m N p-^ri^N f3c k2co3 Synthesis of ng materials: ] Synthesis of ferf-Butyl 2,6-dichloropyridinecarboxylate SUBSTITUTE SHEET (RULE 26) O 1) (Boc)20 2) HCI OH o cr n ci cr n ci A solution of 2,6-dichloropyridinecarboxylic acid (10 g, 52.08 mmol) in THF (210 mL) was treated successively with di-fert-butyl dicarbonate (17 g, 77.89 mmol) and ethylamino)pyridine (3.2 g, 26.19 mmol) and stirred ght at room temperature. At this point, HCI IN (400 mL) was added, and the mixture was stirred vigorously for about 10 s. The product was ted with ethyl acetate (2x300mL), and the combined organic layers were washed with water (300 mL) and brme (150 mL) and dried over sodium sulfate and concentrated under reduced pressure to give 12.94 g (96% yield) of utyl 2,6-dichloropyndinecarboxylate as a colorless oil. ESI-MS m/z calc. 247.02, found 248.1 (M+l)+; Retention time: 2.27 s. 'H NMR (300 MHz, CDC13) ppm 1.60 (s, 9H), 7.30 (d, .7=7.9 Hz, 1H), 8.05 (d, 7=8.2 Hz, 1H).
Synthesis of tert-Butyl 3-o\o-2r3-dihydro-lH-pyrazole-l-carboxylate 1) H2N-NH2 2) (Boc)20 Va pH 0 A SOL reactor was started, and the jacket was set to 20 °C, with stirring at 150 rpm, reflux condenser (10 °C) and nitrogen purge. MeOH (2.860 L) and methyl (E)methoxypropenoate (2.643 kg, 22.76 mol) were added, and the reactor was capped. The reaction was heated to an internal temperature of 40 °C, and the system was set to hold jacket temperature at 40 °C. Hydrazine hydrate (1300 g of 55 %w/w, 22.31 mol) was added portion wise via addition funnel over 30 min. The reaction was heated to 60 °C for 1 h. The reaction mixture was cooled to 20 °C and triethyamine (2.483 kg, 3.420 L, 24.54 mol) was added portion-wise, maintaining reaction temperature <30 °C. A solution of Boc anhydride (di-tert-butyl dicarbonate) (4.967 kg, .228 L, 22.76 mol) in MeOH (2.860 L) was added portion-wise maintaining temperature <45 °C. The reaction mixture was stirred at 20 °C for 16 h. The reaction SUBSTITUTE SHEET (RULE 26) on was partially concentrated to remove MeOH, ing in a clear, light amber oil. The resulting oil was transferred to the 50L reactor, stirred and water (7.150 L) and heptane (7.150 L) were added. The additions caused a small amount of the product to precipitate. The aqueous layer was drained into a clean container, and the interface and heptane layer were filtered to separate the solid (product). The aqueous layer was transferred back to the reactor, and the collected solid was placed back into the r and mixed with the aqueous layer. A dropping funnel was added to the reactor and loaded with acetic acid (1.474 kg, 1.396 L, 24.54 mol) and added dropwise. The jacket was set to 0 °C to absorb the quench exotherm. After the addition was complete (pH=5), the reaction e was stirred for 1 h. The solid was collected by filtration and washed with water (7.150 L), and washed a second time with water (3.575 L). The crystalline solid was transferred into a20L rotovap bulb, and heptane (7.150 L) was added. The mixture was slurried at 45 °C for 30 mins, and 1-2 volumes of solvent were distilled off.
The slurry in the rotovap flask was filtered, and the solids were washed with heptane (3.575 L). The solid was further dried in vacuo (50 °C, 15 mbar) to give /m-butyl 5- oxo-lH-pyrazolecarboxylate (2921 g, 71%) as a coarse, lline solid. 1H N\1 R (400 MHz, DMSO-d6) 5 10.95 (s, 1H), 7.98 (d, /= 2.9 Hz, 1H), 5.90 (d, J= 2.9 Hz, 1H), 1.54 (s, 9H).
Synthesis of 2-[l-(trifluoromethyl)cyclopropyl]ethanol F F F LAH HO F F F To a solution of lithium aluminum hydride (293 mg, 7.732 mmol) in THF (10.00 mL) in an th, 2-[l-(trifluoromethyl)cyclopropyl]acetic acid (1.002 g, 5.948 mmol) in THF (3.0 mL) was added dropwise over a period of 30 minutes keeping the reaction temperature below 20 0 C. The mixture was allowed to gradually warm to t ature and was d for 18 h. The mixture was cooled with an ice-bath and sequentially quenched with water (294 mg, 295 pL, 16.36 mmol), NaOH (297 pL of 6 M, 1.784 mmol), and then water (884.0 pL, 49.07 mmol) to afford a granular solid in the mixture. The solid was filtered off using celite, and the precipitate was washed with ether. The filtrate was further dried with MgSCL and filtered and concentrated in SUBSTITUTE SHEET (RULE 26) vacuo to afford the product with al THE and ether. The mixture was taken directly into the next step without further purification.
Step 1: fert-Butyl 3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazole- oxylate 0 HO cf3 0 0=CjnN. DIAD O' /ert-Butyl 5-oxo-lH-pyrazolecarboxylate (1.043 g, 5.660 mmol), 2-[l- (trifluoromethyl)cyclopropyl]ethanol (916 mg, 5.943 mmol), and triphenyl phosphine (1.637 g, 6.243 mmol) were combined in THE (10.48 mL) and the reaction was cooled in an ice-bath. Diisopropyl a/odi carboxyl ate (1.288 g, 1.254 mL, 6.368 mmol) was added dropwise to the reaction mixture, and the reaction was allowed to w arm to room temperature for 16 hours. The mixture was evaporated, and the ing material was partitioned n ethyl acetate (30 mL) and IN sodium hydroxide (30 mL). The organic layer was separated, washed with brine (30 mL), dried over sodium e, and concentrated. The crude material was purified by silica gel chromatography eluting with a gradient of ethyl acetate in hexanes (0- 30%) to give fert-butyl 3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazole-l-carboxylate (1.03 g, 57%). ESI-MS m/z calc. 320.13, found 321.1 (M+l)+; Retention time: 0.72 minutes.
Step 2: 3-[2-[l-(Trifluoromethyl)cyclopropyl]ethoxy]-lH-pyrazole 0^rj oNk TFA N-MLJ F3C F3C /ert-Butyl[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazole-l- carboxylate (1.03 g, 3.216 mmol) was dissolved in dichloromethane (10.30 mL) with trifluoroacetic acid (2.478 mL, 32.16 mmol), and the reaction was stirred at room temperature for 2 hours. The reaction was evaporated, and the ing oil was partitioned between ethyl acetate (10 mL) and a saturated sodium onate solution.
SUBSTITUTE SHEET (RULE 26) The organic layer was separated, washed with brine, dried over sodium sulfate, and evaporated to give 3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]-lH-pyrazole (612 mg, 86%). ESI-MS m/z calc. , found 221.0 (M+l)+; Retention time: 0.5 minutes. ^ NMR (400 MHz, DMSO-d6) 5 11.86 (s, 1H), 7.50 (t,T = 2.1 Hz, 1H), 5.63 (t,J=2.3 Hz, 1H), 4.14 (t, J= 7.1 Hz, 2H), 2.01 (t, J= 7.1 Hz, 2H), 0.96 - 0.88 (m, 2H), 0.88 - 0.81 (m, 2H).
Step 3: ft?/'/-Butyl 2-chloro[3-[2-[l-(trifluoromethyl)cyclopropyl] ethoxy] py razol- 1-yl] py ridinecarboxylate Kk 0 I °N^n K2CO3 cr n ci NH >- DABCO P-^J N Cl /e/7-Butyl 2,6-dichloropyridinecarboxylate (687 mg, 2.770 mmol), 3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]-lH-pyrazole (610 mg, 2.770 mmol), and freshly ground potassium carbonate (459 mg, 3.324 mmol) were combined in anhydrous DMSO (13.75 mL). l,4-diazabicyclo[2.2.2]octane (DABCO (1,4- diazabicyclo[2.2.2]octane), 62 mg, 0.5540 mmol) was added, and the mixture was stirred at room temperature under nitrogen for 16 hours. The reaction mixture was diluted with w ater (20 mL) and stirred for 15 minutes. The resulting solid was collected and washed with water. The solid was dissolved in dichloromethane and dried over magnesium e. The e was filtered and concentrated to give /e/7-butyl 2- chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridine carboxylate (1.01 g, 84%). ESI-MS m/z calc. 431.12, found 432.1 (M+l) +; Retention time: 0.88 minutes.
Step 4: 2-Chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol- l-yl]pyridinecarboxylic acid Vc0 I O L p^J n" c, F3C f3c SUBSTITUTE SHEET (RULE 26) WO 64632 /e/7-Butyl 2-chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol- l-yl]pyridinecarboxylate (1.01 g, 2.339 mmol) and oroacetic acid (1.8 mL, 23.39 mmol) were combined in dichloromethane (10 mL) and heated at 40 °C for 3 h.
The on was concentrated. Hexanes were added, and the mixture was concentrated again to give 2-chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ]pyridinecarboxylic acid (873 mg, 99%) ESI-MS m z calc. 375.06, found 376.1 (M+l)+; Retention time: 0.69 minutes.
Step 5: N-(Benzenesulfonyl)chloro[3-[2-[l- uoromethyl)cyclopropyl] ethoxy]pyrazol-l-yl]pyridinecarboxamide S02NH2 Nv GDI P-QN N Cl P-Q N Cl F ► F F- F F F A solution of 2-chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylic acid (0.15 g, 0.3992 mmol) and carbonyl diimidazole (77 mg, 0,4790 mmol) in THE (2.0 mL) was stirred for one hour, and benzenesulfonamide (81 mg, 0.5190 mmol) and DBU (72 uL. 0.4790 mmol) were added. The reaction was stirred for 16 hours, acidified with 1 M aqueous citric acid, and extracted with ethyl acetate. The combined extracts were dried over sodium e and evaporated. The residue was purified by silica gel chromatography eluting with a gradient of methanol in dichloromethane (0-5%) to give N-(benzenesulfonyl)chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazoll-yl ]pyridinecarboxamide (160 mg, 78%). ESI-MS m/z calc. , found 515.1 (M+l)+; Retention time: 0.74 minutes.
Step 6: N-(Benzenesulfonyl)[3-[2-[l-(trifluoromethyl)cyclopropyl] ethoxy] pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-I-yl] pyridine carboxamide SUBSTITUTE SHEET (RULE 26) F3C- K2C03 P^JN-m^m N A mixture of N-(benzenesulfonyl)chloro[3-[2-[l- (trifluoromethyl)cyclopropyl] ]pyrazol-l-yl]pyridinecarboxamide (160 mg, 0.3107 mmol), (4S)-2,2,4-trimethylpyrrolidine hloride salt (139 mg, 0.9321 mmol), and potassium carbonate (258 mg, 1.864 mmol) in DMSO (1.5 mL) was stirred at 130 °C for 17 hours. The reaction mixture was acidified with 1 M s citric acid and extracted with ethyl acetate. The combined extracts were dried over sodium sulfate and evaporated to yield a crude product that was purified by reverse-phase HPLC utilizing a gradient of 10-99% itrile in 5 mM aqueous HC1 to yield N- (benzenesulfonyl)[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (87 mg, 47%). ESI-MS m/z calc. 591.21, found 592.3 (M+l)+; Retention time: 2.21 minutes, 'll NMR (400 MHz, DMSO-d6) 8 12.48 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 8.04 - 7.96 (m, 2H), 7.81 (d, J= 8.2 Hz, 1H), 7.77 - 7.70 (m, 1H), 7.70 - 7.62 (m, 2H), 6.92 (d, J= 8.2 Hz, 1H), 6.10 (d, .7=2.8 Hz, 1H), 4.31 (t, J= 7.0 Hz, 2H), 2.42 (t, /= 10.5 Hz, 1H), 2.28 (dd, J = .2, 7.0 Hz, 1H), 2.17-2.01 (m, 3H), 1.82 (dd, J= 11.9, 5.5 Hz, 1H), 1.52 (d, 7=9.4 Hz, 6H), 1.36 (t, 7= 12.1 Hz, 1H), 1.01 - 0.92 (m, 2H), 0.92 - 0.85 (m, 2H), 0.65 (d, J = 6.3 Hz, 3H). pKa: 4.95±0.06.
Synthesis of sodium salt of N^benzenesulfonylJ-O-p-ll-tl- oromethy^cyclopropyllethoxylpyrazol-l-yl]-!-!^)-!,!.^- trimethylpyrrolidin-l-yl]pyridinecarboxamide (sodium salt of Compound 1) N-(benzenesulfonyl)[3-[2-[l-(tnfluoromethyl)cyclopropyl]ethoxy]pyrazol- l-yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (1000 mg, 1.679 mmol) was dissolved in ethanol (19.87 ml) under warming, filtered clear through a SUBSTITUTE SHEET (RULE 26) e filter (0.2 |iin). washed with warm ethanol (10 ml) and the warm solution was treated with 1M NaOH (1.679 ml, 1.679 mmol). The solution was evaporated at 30-35 °C, co-evaporated 3 times with ethanol (~20 ml), to give a solid, which was dried ght under vacuum in a drying cabinet at 45 °C with a nitrogen bleed to give 951 mg of a cream colored solid. The solid was further dried under vacuum in a drying cabinet at 45 °C with a nitrogen bleed over the weekend. 930 mg (89%) of the sodium salt of N-(benzenesulfonyl)[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide was obtained as an off-white amorphous solid. ^ NMR (400 MHz, 6) 5 8.15 (d, J= 2.7 Hz, 1H), 7.81 (dd, J= 6.7, 3.1 Hz, 2H), 7.61 (d, /= 7.9 Hz, 1H), 7.39 (dd, ,/ = 4.9, 2.0 Hz, 3H), 6.74 (d,J= 7.9 Hz, 1H), 6.01 (d, /= 2.6 Hz, 1H), 4.29 (X,J= 7.0 Hz, 2H), 2.93 - 2.78 (m, 2H), 2.07 (t, J= 7.1 Hz, 3H), 1.78 (dd, 7= 11.8, 5.6 Hz, 1H), 1.52 (d,/= 13.6 Hz, 6H), 1.33 (t, J= 12.0 Hz, 1H), 1.00 - 0.92 (m, 2H), 0.89 (q, J= 5.3, 4.6 Hz, 2H), 0.71 (d, J= 6.3 Hz, 3H). EST-MS m/z calc. 591.2127, found 592.0 (M+l)+; Retention time: 3.28 minutes. XRPD (see . ate synthesis of 2-Chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylic acid ] Step 1: ethyl 3-hydroxy-lH-pyrazolecarboxylate o o O NH2NH2*xH20 O OH O' OEt EtOH O N k H A mixture of EtOH (20.00 L, 10 vol) and diethyl 2- (ethoxymethylene)propanedioate (2000 g, 9.249 mol, 1.0 equiv) was added under nitrogen purge a to a 50 L r equipped with a reflux condenser (10 °C) and the jacket set to 40 °C. The mixture was stirred, and then hydrazine hydrate (538.9 g of 55 %w/w, 523.7 mL of 55 %w/w, 9.249 mol, 1.00 equiv) was added in portions via an addition funnel. Once the addition was complete, the reaction was heated to 75 °C for 22 h to afford a solution of ethyl 3-hydroxy-lH-pyrazolecarboxylate that was used directly in the next step.
Step 2: l-(tert-butyl) 4-ethyl 3-hydroxy-lH-pyrazole-l,4-dicarboxylate SUBSTITUTE SHEET (RULE 26) 0 0 0 OH Boc20, Eton 0 OH ( )n TEA N N H i The solution of ethyl 3-hydroxy-lH-pyrazolecarboxylate was cooled from 75 °C to 40 °C, then triethylamine (TEA) (46.80 g. 64.46 mL, 462.5 mmol, 0.05 eq.) was added. A solution of Boc ide (2.119 kg, 9.711 mol 1.05 equiv) in EtOH (2.000 L, 1 equiv) was added to the reactor over 35 min. The mixture was d for 4 hours to complete the reaction; then water (10.00 L, 5.0 vol) was added over 15 mins.
The resulting mixture was cooled to 20 °C to complete crystallization of the product.
The crystals were allowed to age for 1 hour, then the mixture was filtered. The solid was washed with a mixture of EtOH (4.000 L, 2.0 vol) and water (2.000 L, 10 vol) The solid was then dried in vacuo to afford l-(tert-butyl)ethylhydroxy-lH-pyrazole- carboxylate (1530 g, 65%) as colorless, fine needle, crystalline solid. 'HNMR (400 MHz, DMSO-d6) 8 11.61 (s, 1H), 8.40 (s, 1H), 4.20 (q, J = 7.1 Hz, 2H), 1.56 (s, 9H), 1.25 (t, J = 7.1Hz,3H).
Step 3: l-(tert-butyl) 4-ethyl 3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole-l,4-dicarboxylate .cf3 HO N.
S' DIAD, PPh3, + NBoc toluene Et02C NBoc Et02C A 5L reactor was started with the jacket set to 40 °C, stirring at 450 rpm, reflux condenser at room temperature and nitrogen purge. The vessel was charged with toluene (1.0L, 10.0 vol), 2-[l-(trifluoromethyl)cyclopropyl]ethanol (100.Og, 648.8 mmol, 1.0 equiv), and l-(tert-butyl) l oxy-lH-pyrazole-l,4-dicarboxylate (166.3 g, 648.8 mmol), and the mixture was stirred. The reaction mixture was charged with triphenyl phosphine (195.7 g, 746.1 mmol, 1.15 equiv), then the r was set to maintain an internal temperature of 40 °C. Diisopropyl azoldicarboxylate (150.9 g, 746.1 mmol, 1.15 equiv) was added into an addition funnel and was added to the SUBSTITUTE SHEET (RULE 26) reaction while maintaining the reaction temperature between 40 and 50 °C (addition was exothermic, exotherm addition controlled), and stirred for a total of 2.5 hours. Once the reaction was deemed complete by HPLC, heptane was added (400 mL, 4 vol), the solution was cooled to 20 °C over 60 minutes, and the bulk of tnphenylphosphine oxide-DIAD complex (TPPO-DIAD) crystallized out. Once at room temp, the mixture was filtered, and the solid was washed with heptane (400 mL, 4.0 vol) and pulled dry.
The filtrate was used in the next step as a solution in toluene-heptane without r purification.
Step 4: ethyl 3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole- 4-carboxylate cf3 cf3 O. O.
NBoc NH Et02C' Et02C' A 500mL reactor was started with the jacket set to 40 °C, stirring at 450 rpm, reflux condenser at room temp, and nitrogen purge. The vessel was charged with a e on consisting of imately 160 mmol, 65.0 g of l-(tert-butyl) 4-ethyl 3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole-l,4-dicarboxylate in 3 vol of toluene red by concentrating a 25% n of filtrate from previous reaction down to 4 volumes in a rotovap). The reaction was set to maintain an internal temperature at 40 °C and KOH (33.1 g, 1.5 eq. of aqueous 45 % KOH solution) w as added in one portion, resulting in a mild exothermic addition, while CO2 was generated upon removal of the ting group. The reaction proceeded for 1.5 hr, monitored by HPLC, with the product partially crystallizing during the reaction. Heptane (160 mL, 2.5 vol) was added to the reaction mixture and the on was cooled to room ature over 30 minutes. The resulting mixture was filtered, and the solid was washed with heptane (80.00 mL, 1.25 vol), pulled dry, then dried in vacuo (55 °C, vacuum). 52.3 g of ethyl 3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole carboxylate was obtained as a crude, colorless solid that was used without further purification.
SUBSTITUTE SHEET (RULE 26) ] Step 5: 3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole carboxylic acid CF3 cf3 KOH, MeOH 0\r^M o. .N NH NH Et02C' H02C' A 500mL reactor was started with the jacket set to 40 °C, stirring at 450 rpm, reflux condenser at room temp, and nitrogen purge. The vessel was charged with methanol (150.0 mL, 3.0 vol), a solution of ethyl 3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazolecarboxylate (50.0 g, 171.1 mmol, 1.0 equiv), and the reaction was stirred to suspend the solids. The r was set to maintain internal temperature at 40 °C. To the mixture was added KOH (96 g of aqueous 45 % KOH, 1.71 mol, 10.0 equiv) in ns maintaining the internal temperature <50 °C. Once addition was complete, the reaction was set to maintain temperature at 50 °C, and the reaction proceeded for 23 hours, monitored by HPLC.
Once complete the reaction was cooled to 10 °C then partially trated on a rotary evaporator to remove most of the MeOH. The resulting solution was diluted with water (250 mL, 5.0 vol) and 2-Me-THF (150 mL, 3.0 vol), and transferred to the r, stirred at room temp, then stopped, and layers were allowed to separate. The layers were tested, with remaining TPPO-DIAD complex in the organic layer and product in the aqueous layer. The aqueous layer was washed again with 2-Me-THF (100 mL, 2.0 vol), the layers separated, and the aqueous layer returned to the reactor vessel. The stirrer was started and set to 450 rpm, and the reactor jacket was set to 0 °C. The pH was adjusted to pH acidic by addition of 6M aqueous HC1 (427mL, 15 equiv) portion wise, maintaining the internal temperature between 10 and 30 °C. The product began to crystallize close to pH neutral and was anied with strong off-gassing, and so the acid was added slowly, and then further added to reach pH 1 once the off-gassing had ended. To the ing sion was added 2-Me-THF (400 mL, 8.0 vol), and the product was allowed to dissolve into the organic layer. Stirring was stopped, the layers were separated, and the aqueous layer was ed to the reactor, stirred and reextracted with 2-Me-THF (100 mL, 2.0 vol). The c layers were combined in the SUBSTITUTE SHEET (RULE 26) reactor and stirred at room temperature, washed with brine (lOOmL, 2 vols), dried over Na2S04: filtered through celite, and the solid was washed with 2-Me-THF (50 mL, 1.0 vol). The filtrate was transferred to a clean rotovap flask, stirred, warmed to 50 °C and e (200 mL, 4.0 vol) added, and then partially concentrated with the addition of heptane (300 mL, 6.0 vol) and then seeded with 50mg of 3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazolecarboxylic acid), and the product crystallized during solvent removal. The lation was stopped when the bulk of the 2-Me-THF had distilled off. The bath heater was turned off, the vacuum removed, and the mixture was allowed to stir and cool to room temperature. The mixture was filtered (slow speed) and the solid wras washed with heptane (100 mL, 2.0 vol), and the solid was collected and dried in vacuo (50 °C, p). 22.47 g of l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazolecarboxylic acid was obtained as an off-white solid. ^ NMR (400 MHz, DMSCM) 5 12.45 (s, 2H), 8.01 (s, 1H), 4.26 (t, J = 7.0 Hz, 2H), 2.05 (t, J= 7.0 Hz, 2H), 0.92 (m, 4H).
Step 6: l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole cf3 cf3 O. O.
NH NH H02C' A mixture of toluene (490.0 mL), l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazolecarboxylic acid (70.0 g, 264.9 mmol), and DMSO (70.00 mL) w?as placed in a reactor and heated to 100 0C with stirring. DBU (approximately 20.16 g, 19.80 mL, 132.4 mmol) was added to the reactor over 15 min. The mixture was stirred for 20 h to complete the reaction and then cooled to 20 °C. The mixture was washed with water (350.0 mL), then 0.5N aq HC1 (280.0 mL), then water (2 x 140.0 mL), and lastly with bnne (210.0 mL). The organic layer was dried with Na2SC>4, and then activated charcoal (5 g, Darco 100 mesh) was added to the stirred slurty. The dried mixture was filtered through , and the solid was washed with toluene (140.0 mL) and then pulled dry. The filtrate was concentrated in a rotovap (50 °C, vac) to afford 3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]-lH- SUBSTITUTE SHEET (RULE 26) pyrazole (30.89 g, 53%) as an amber oil. ^ NMR (400 MHz, DMSO-dg) 5 11.87 (s, 1H), 7.50 (d, J= 2.4 Hz, 1H), 5.63 (d, 7= 2.4 Hz, 1H), 4.23 - 4.06 (m, 2H), 2.01 (t, J= 7.1Hz, 2H), 1.00-0.77 (m, 4H).
] Step 7: ethyl 2-chloro [3- [2- [ 1- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylate cf3 PF3 0 0 OEt 'OEt cr n ci N. xj- °V^N O-fJ N Cl T-— \ NH cat DABCO, K2CO3, DMF A mixture of DMF (180.0 mL), ethyl 2,6-dichloropyridinecarboxylate (approximately 29.97 g, 136.2 mmol), l-(trifluoromethyl)cyclopropyl]ethoxy]- azole (30.0 g, 136.2 mmol), and K2CO3, (325 mesh, approximately 24.48 g, 177.1 mmol) was added to a stirred reactor at 20 °C. DABCO (approximately 2.292 g, .43 mmol) was then added to the reactor, and the mixture was stirred at 20 °C for 1 hour, and then the ature was increased to 30 °C, and the mixture stirred for 24 hours to complete the reaction. The mixture was cooled to 20 °C; then water (360 mL) was added slowly. The mixture was then drained from the reactor and the solid was isolated by filtration. The solid was then washed with water (2 x 150 mL), and then the solid was dried under vacuum at 55 °C to afford ethyl 2-chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylate (51.37 g, 93%) as a fine, beige colored solid. XH NMR (400 MHz, DMSO-c/g) 8 8-44 (d, J=2.9 Hz, 1H), 8.41 (d,7= 8.5 Hz, 1H), 7.75 (d, 7= 8.5 Hz, 1H), 6.21 (d, 7= 2.9 Hz, 1H), 4.34 (m, 4H), 2.09 (t, 7= 7.1 Hz, 2H), 1.34 (t, 7= 7.1 Hz, 3H), 1.00 - 0.84 (m, 4H).
Step 8: 2-Chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol- l-yl]pyridinecarboxylic acid PF3 0 ,cf3 0 OEt OH b-JHl N- -Cl aq NaOH b-01 N- -Cl SUBSTITUTE SHEET (RULE 26) A solution of ethyl ro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylate (50.0 g, 123.8 mmol) in THF (300.0 mL) was prepared in a reactor at 20 °C. EtOH (150.0 mL) was added, followed by aqueous NaOH (approximately 59.44 g of 10 %w/w, 148.6 mmol).
The mixture was stirred for 1 hour to complete the reaction; then aq IN HC1 (750.0 mL) was slowly added. The resulting suspension was stirred for 30 mm at 10 °C, and then the solid was isolated by tion. The solid was washed with water (150 mL then 2 x 100 mL) and then pulled dry by vacuum. The solid was then further dried under vacuum with g to afford 2-chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazoll-yl ]pyridinecarboxylic acid (42.29 g, 91%). 'ff NMR (400 MHz, DMSO-t/,,) 5 13.63 (s, 1H), 8.48 - 8.35 (m, 2H), 7.73 (d, J= 8.4 Hz, 1H), 6.20 (d, J= 2.9 Hz, 1H), 4.35 (t, J = 7.1 Hz, 2H), 2.09 (t, J= 7.1 Hz, 2H), 1.01 - 0.82 (m, 4H).
Synthetic Example 2: Synthesis of Compound 2, (R)-N-(Phenylsulfonyl)- 6-(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)(2,2,4- trimethylpyrrolidin-l-yl)nicotinamide Q o.o 's' K2CO3 H ► 'm N Cl HCI R F • ml P 0.0 N''s'b' 0^N'N^N^N R F y W S^V(R).mi (Phenylsulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH- pyrazol-l-yl)(2,2,4-trimethylpyrrolidin-l-yl)nicotinamide was synthesized in a manner analogous to Compound 1 using N-(benzenesulfonyl)chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (1.5 g, 2.91 mmol), potassium carbonate (2.0 g, 14.56 mmol), (4R)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (1.0 g, 6.7 mmol) inNMP (jV-Methylpyrrolidone) (7.5 mL) and 1,2-diethoxy ethane (1.5 mL) affording N-(benzenesulfonyl)[3-[2-[l- SUBSTITUTE SHEET (RULE 26) WO 64632 (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4R)-2,2,4-trimethylpyrrolidiii-lyl ]pyridinecarboxamide (1.38 g, 79%). ESI-MS m/z calc. 591.2127, found 592.0 (M+l) +; Retention time: 2.3 minutes. ^NMR (400 MHz, DMSO-d6) 5 12.51 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 8.03 - 7.96 (m, 2H), 7.81 (d, J = 8.2 Hz, 1H), 7.76 - 7.69 (m, 1H), 7.66 (dd, J = 8.3, 6.7 Hz, 2H), 6.91 (d, J = 8.2Hz, 1H), 6.11 (d, J = 2.8Hz, 1H), 4.31 (t, J = 7.0 Hz, 2H), 2.41 (t, J = 10.5 Hz, 1H), 2.27 (t, J = 8.7 Hz, 1H), 2.07 (t, J = 7.1 Hz, 3H), 1.82 (dd, J = 11.9, 5.5 Hz, 1H), 1.52 (d, J = 9.4 Hz, 6H), 1.36 (t, J = 12.1 Hz, 1H), 0.99 - 0.92 (m, 2H), 0.88 (tt, J = 3.9, 1.6 Hz, 2H), 0.64 (d, J = 6.3 Hz, 3H).
Synthetic Example 3: Synthesis of Compound 3, (S)-N-((4-Hydroxy methoxyphenyl)sulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH- pyrazol-l-yl)(2,2,4-trimethylpyrrolidin-l-yl)nicotinamide Step A: 2-Chloro-N-((4-hydroxymethoxyphenyl)sulfonyl)(3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)- IH-pyrazol- icotinamide o=s=oI ^ o OH P 0 0's' I OH .0.
GDI In q-Q n' 'Cl✓5 P-^jN N^CI OH R F DBU R F A solution of 2-chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylic acid (0.843 g, 2.24 mmol) and carbonyl diimidazole (434 mg, 2.68 mmol) in THE (2.5 mL) was d for 2.5 hours, and 4-hydroxymethoxybenzenesulfonamide (0.500 g, 2.46 mmol) and DBU (0.5 mL, 3.35 mmol) were added. The reaction was stirred for 21 hours, diluted with ethyl acetate (5 mL) ied with 1 N aqueous hydrochloric acid (10 mL), and extracted with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate and evaporated. The residue was purified by silica gel chromatography SUBSTITUTE SHEET (RULE 26) eluting with a gradient of ethyl e in hexanes (50-100%) to give 2-chloro-N-((4- hydroxymethoxyphenyl)sulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)- lH-pyrazol-l-yl)nicotinamide (906 mg, 72%). ESI-MS m/z calc. 560.07, found 515.1 (M+l) +; Retention time: 0.74 minutes.
Step B: (S)-N-((4-Hydroxymethoxyphenyl)sulfonyl)(3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)(2,2,4- trimethylpyrrolidin-l-yl)nicotinamide 9 QwP N''s5' Ox K2CO3 fr K ■ox N- >5 H -> N Cl 'OH sLfe 'OH HCI F F /—/ A mixture of 2-chloro-N-((4-hydroxymethoxyphenyl)sulfonyl)(3-(2- (l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)nicotinamide (906 mg, 1.62 mmol), (4S)-2,2,4-trimethylpyrrolidine hydrochloride salt (545 mg, 3.64 mmol), and potassium carbonate (1.29 g, 9.33 mmol) in DMSO (5.5 mL) was stirred at 120 0C for 24 hours. The on mixture was d with 15 mL of water and 5 mL of ethyl acetate. The reaction mixture was then acidified with 6 N s hydrochloric acid the layers were separated. The aqueous layer was extracted with 10 mL of ethyl acetate.
The combined extracts were washed with brine, dned over sodium sulfate and evaporated to yield a crude product that was purified by silica gel chromatography utilizing a gradient of ethyl acetate in hexanes to yield (S)-N-((4-hydroxy methoxyphenyl)sulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazoll-yl )(2,2,4-trimethylpyrrolidin-l-yl)nicotinamide (470 mg, 45%) ESI-MS m/z calc. 637.2, found 638.2 ; Retention time: 10.07 minutes.
SUBSTITUTE SHEET (RULE 26) Synthetic Example 4: Synthesis of Compound 4, N-(o-Tolylsulfonyl) [3- [2- [ l-(trifluoromethyl)cyclopropyl]ethoxy] pyrazol- 2- [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(o-tolylsulfonyl)[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide o=s=o OH ,SA'/ I GDI N D-An N- 'ClX H R F P-An N' 'Clyi DBU Fv F To 2-chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (196 mg, 0.5217 mmol) in THF (1.739 mL) was added l,r-carbonyldiimidazole ximately 106.6 mg, 0.6573 mmol) and reaction was stirred for one hour. 2-Methylbenzenesulfonamide (approximately 89.32 mg, 0.5217 mmol) was added, followed by l,8-diazabicyclo(5.4.0)undecene (DBU) ximately 262.2 mg, 257.6 |iL. 1.722 mmol) and reaction was stirred for 3 hours.
The reaction was diluted with ethyl acetate and 1 M s citric acid and the layers were separated. The organic layers were dried and concentrated and resulting solid 2- chloro-N-(o-tolylsulfonyl)[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ]pyridinecarboxamide (approximately 252 mg) was used for next step without characterization.
SUBSTITUTE SHEET (RULE 26) Step B: N-(o-Tolylsulfonyl)[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide o owo O'/ K2C03 % N'S P'0N ^ ^ H1 N Cl ► h F HN R F F F To 2-chloro-N-(o-tolylsulfonyl)[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxainide (approximately 252 mg) and potassium carbonate (392 mg, 2.84 mmol) in 0.4 mL of DMSO was added (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (212 mg, 1.42 mmol) and reaction was d at 130 °C for 16 hours. The reaction was cooled, diluted with ethyl acetate and 1 M s citric acid and the layers were separated. The organics were dried, concentrated and the ing e was purified with silica gel (24 g) eluting with 0-14% methanol in dichloromethane to give N-(o-tolylsulfonyl) [3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (60.6 mg, 19%) 'h NMR (400 MHz, DMSO-d6) 5 12.63 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 8.04 (dd, J = 7.9,1.4 Hz, 1H), 7.81 (d, J = 8.2 Hz, 1H), 7.58 (td, J = 7.5, 1.5 Hz, 1H), 7.50 - 7.40 (m, 2H), 6.93 (d, J = 8.3 Hz, 1H), 6.10 (d, J = 2.7 Hz, 1H), 4.31 (t, J = 7.1 Hz, 2H), 2.64 (s, 3H), 2.39 (d, J = 8.8 Hz, 2H), 2.16 (ddt, J = 11.8, 9.0, 4.5 Hz, 1H), 2.08 (t, J = 7.0 Hz, 2H), 1.82 (dd, J = 11.9,5.6Hz, 1H), 1.52 (s, 6H), 1.35 (t, J= 12.1 Hz, 1H), 1.00-0.93 (m, 2H), 0.92- 0.84 (m, 2H), 0.69 (d, J = 6.2 Hz, 3H). ESI-MS m/z calc. 605.23, found 606.4 (M+l)+; Retention time: 1.92 minutes SUBSTITUTE SHEET (RULE 26) Synthetic Example 5: sis of Compound 5, N-(3- Fluorophenyl)sulfonyl [3- [2- [l-(trifluoromethyl)cyclopropyl] ethoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(3-fhiorophenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide o=s=o OH ,S\\Jt F GDI N O-AjN N- 'ClX H R F p-An n' 'Cl DBU R F To ro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (0.200 g, 0.532 mmol) in THF (1.7 mL) was added 1,1- carbonyldiimidazole (108.8 mg, 0.6707 mmol) and reaction was stirred for 1 hour. 3- Fluorobenzenesulfonamide (93.25 mg, 0.5323 mmol) was added, followed by 1,8- diazabicyclo(5.4.0)undecene (DBU) (267.5 mg, 262.8 pL, 1.757 mmol) and reaction was stirred for 2 hours. The reaction was diluted with ethyl acetate and 1 M aqueous citric acid and layers were separated. The organics were dried and concentrated and resulting solid 2-chloro-N-(3-fluorophenyl)sulfonyl[3-[2-[l- uoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (approximately 259 mg) was used in the next step without characterization.
Step B: N-(3-Fluorophenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]p yrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide AqsP F AqsP F (Ay'n'sVV'F K2CO3 n'n^n^ciH ^ ■*« (Tr N YY Fv F Afe KTo-U >Ms) SUBSTITUTE SHEET (RULE 26) ] To 2-chloro-N-(3-fluorophenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (approximately 259 mg, 0.486 mmol) and potassium carbonate (389.6 mg, 2.819 mmol) in 0.4 mL of DMSO was added (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (211.0 mg, 1.41 mmol) and the reaction was stirred at 130 °C for 16 hours. The reaction was cooled, diluted with ethyl acetate and 1 M aqueous citric acid and the layers were separated. The organics were dried, concentrated and ing the residue was purified on silica gel (24 g) eluting with 0-14% methanol in dichloromethane to give N-(3- phenyl)sulfonyl[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (50.0 mg, 15%) 'll NMR (400 MHz, Methanol-d4) 5 8.23 (d, J = 2.7 Hz, 1H), 7.96 - 7.89 (m, 1H), 7.87 - 7.77 (m, 2H), 7.65 (td, J = 8.1, 5.3 Hz, 1H), 7.46 (tdd, J = 8.5,2.5, 1.0 Hz, 1H), 7.02 (d, J = 8.3 Hz, 1H), 5.95 (d, J = 2.8 Hz, 1H), 4.37 (t, J = 7.0 Hz, 2H), 3.34 (s, 1H), 2.68 (t, J = 10.3 Hz, 1H), 2.56 - 2.48 (m, 1H), 2.28 - 2.16 (m, 1H), 2.10 (t, J = 7.0 Hz, 2H), 1.89 (dd, J = 11.9, 5.7 Hz, 1H), 1.59 (d, J = 9.7 Hz, 6H), 1.48 (t, J = 12.1 Hz, 1H), 1.02 - 0.96 (m, 2H), 0.86 - 0.77 (m, 5H). ESI-MS m/z calc. 609.2, found 610.3 (M+l)+; Retention time: 0.81 minutes Synthetic Example 6: Synthesis of Compound 4S)-3,3-Dideuterio- 2,2-dimethyl(trideuteriomethyl)pyrrolidin-l-yl]-N-(4-hydroxyphenyl)sulfonyl [3- [2- [ l-(trifluoromethyl)cyclopropyl] ethoxy] pyrazol- l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide o=s=oI ^ o OH 0 0.0 ‘OH \W/ GDI %■ P-^Q1 N 'ClNU I H O-An n' Cl 'OH F. F DBU Fv F SUBSTITUTE SHEET (RULE 26) 2-Chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (0.100 g, 0.266 mmol) and GDI (approximately 51.38 mg, 0.3169 mmol) were combined in THF (600.0 pL) and stirred at room temperature for 2 hours. 4-Hydroxybenzenesulfonamide (approximately 50.69 mg, 0.2927 mmol) was added followed by DBU ximately 54.41 mg, 53.45 pL, 0.3574 mmol) and the reaction was stirred for an additional 16 hours at room ature. The reaction mixture was diluted with 10 mL of 1 M aqueous citric acid, and extracted with three 10 mL portions of ethyl acetate. The combined organics were washed with brine, dried over sodium sulfate, and concentrated to give a white solid 2-chloro-N-(4- hy droxypheny l)sulfonyl [3-[2- [ 1 -(trifluoromethy l)cy pyl] ethoxy ] pyrazol-1 - idinecarboxamide (128 mg, 91%) which was used in the next step without further purification. ESTMS m/z calc. 530.1, found 531.0 (M+l)+; Retention time: 0.69 minutes.
Step B: 2-[(4S)-3,3-Dideuterio-2,2-dimethyl (trideuteriomethyl)pyrrolidin- l-yl]-N-(4-hydroxyphenyl)sulfonyl [3- [2-[ 1- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide 9 q„p K2CO3 p^N N^CI ‘OH P-^S1 N' ‘OH R F HN (S)PD V □DO T'd ^ F □ D 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (1.0 g, 1.9 mmol), (S)-2.2-dimethyl(methyl-cA)pyrrolidme-3.3-c/2 hydrochloride salt (0.892 g, .66 mmol) and potassium carbonate (1.55 g, 11.2 mmol) were combined in DMSO (6 mL) and heated to 130 °C for and 16 hours. The reaction was cooled to room temperature, and diluted with water (10 mL). After stirring for 15 minutes ethyl acetate (50 mL) was added to the mixture. The mixture was acidified with 1M aqueous citric acid (pH~3-4) (30 mL) and the layers were separated. The cs were combined, washed with brine, dried over sodium sulfate and concentrated. The crude material obtained was ed by column chromatography (24 g of silica gel) utilizing a gradient SUBSTITUTE SHEET (RULE 26) of 0-30% ethyl acetate in heptane. Individual fractions were analyzed by HPLC and the fractions that met the required purity specifications were combined, evaporated and triturated in a e of 9:1 ethyl acetate/MTBE. The organics were evaporated down to 10% and the solid obtained was filtered and dried ght under high vacuum to afford )-3,3-dideuterio-2,2-dimethyl(trideuteriomethyl)pyrroli dinyl]-N-(4- hy droxypheny l)sulfonyl [3-[2- [ 1 -(trifluoromethy l)cy clopropyl] ethoxy ] pyrazol-1 - yl]pyridinecarboxamide (0.38 g, 32%) ESI-MS m/z calc. 612.2, found 613.7 (M+l)+; ion time: 1.40 minutes.
Synthetic Example 7: Synthesis of Compound 7, N-(Benzenesulfonyl)[(4S)-3,3-dideuterio-2,2-dimethyl (trideuteriomethyl)pyrrolidin-l-yl][3-[2-hydroxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide AqsP k2co3 AQsP ■>> ft HO jX? o HO R F PYJ /S(ddd HCI D A reaction vessel was charged with N-(benzenesulfonyl)chloro[3-[2- hydroxy[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridine carboxamide (0.500 g, 0.942 mmol), (4S)-3,3-dideuterio-2,2-dimethyl (trideuteriomethyl)pyrrolidine (Hydrochloride salt) (320 mg, 2.07 mmol), NMP (3.000 mL) and 1,2-di ethoxy ethane (500.0 pL) under an atmosphere of nitrogen. Potassium carbonate (650.8 mg, 4.709 mmol) was added and the reaction mixture was heated to 130 °C. The reaction mixture was stirred ght. The reaction mixture was cooled and diluted with water (2.000 mL) and adjusted pH to <3 with aqueous HCI (1.3 mL of 6 M, 7.800 mmol), which was added dropwise. The pH was adjusted further with hydrogen chloride (146.0 pL of 6 M, 0.8760 mmol). The aqueous layer was extracted with ethyl acetate (4 mL) twice and the combined organic layers were washed with water twice, brine, and dried over sodium sulfate. The organic layer was then concentrated to a residue which was purified on silica gel utilizing a gradient of 0-60% ethyl acetate in s. This material was then triturated in a e of heptanes and MTBE to yield N-(benzenesulfonyl)[(4S)-3,3-dideuterio-2,2-dimethyl SUBSTITUTE SHEET (RULE 26) (trideuteriomethyl)pyrrolidin-l-yl][3-[2-hydroxy[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl]pyridinecarboxamide (266 mg, 46%) ESI-MS m/z calc. 612.2, found 613.1 (M+l)+; Retention time: 1.67 minutes.!H NMR (400 MHz, DMSO-d6) 5 12.51 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 8.05 - 7.94 (m, 2H), 7.81 (d, J = 8.3 Hz, 1H), 7.72 (d, J = 7.3 Hz, 1H), 7.65 (t, J = 7.6 Hz, 2H), 6.90 (d, J = 8.2 Hz, 1H), 6.12 (d, J = 2.8 Hz, 1H), 5.57 (dd, J = 5.5, 2.7 Hz, 1H), 4.42 - 4.28 (m, 1H), 4.23 - 4.09 (m, 1H), 3.89 (d, J = 4.9 Hz, 1H), 2.39 (d, J = 10.5 Hz, 1H), 2.37 - 2.22 (m, 1H), 2.06 (dd, J = 10.6, 7.0 Hz, 1H), 1.52 (d, J = 9.7 Hz, 6H), 1.04 - 0.83 (m, 4H).
Synthesis of (4S)-3,3-Dideuterio-2,2-dimethyl (trideuteriomethyl)pyrrolidine Hydrochloride 0 to? se D2C. enzyme 'OCD3 'OCD3 CD3 DBU N02 CD3 h2 LiAIH4 HN-AIS) HN CD3 Raney-Ni CD3 OCD3 HCI D ° ■ HCI N02 CD3 D D Step A: Methyl-*/; 4-mcthyl(mcthyl-*/j)nitropcntanoate-3,3-f/2 OCD3 OCD3 cd3 DBU N02 CD3 A 500-mL, three-neck round bottom flask equipped with a magnetic stir bar, a nitrogen line and a J-Kem couple with heating mantle was charged with 2- nitropropane (34.3 g, 385 mmol), thyl rylate (50.0 g, 460 mmol), and was stirred at ambient temperature when l,8-diazabicyclo[5.4.0]undecene (DBU, 1.47 g, 9.62 mmol) was added in one portion. The reaction solution exothermed from 20 to ~40 °C and was allowed to stir without heating or cooling for 16 h. The reaction was only partially completed (HPLC) so the solution was warmed at 80 °C for 4 h. The reaction mixture is d with MTBE (170 mL), washed with 1 M HCI (15 mL), dned over SUBSTITUTE SHEET (RULE 26) magnesium sulfate, filtered and trated (29” Hg at 60 °C) to remove solvent and any residual starting materials to afford product as light yellow oil (75 g, 99%). It was used to the next step without further purification by lation.
Step B: -rfs fV)methyl(methyl-r/i)nitropentanoate-3,3-r/2 D O Palatase D O D D enzyme OCD3 ocd3 NO2 cd3 NO2 cd3 A 5-L, three-neck round bottom flask equipped an overhead mechanical stirrer, a nitrogen line and a J-Kem thermocouple with heating mantle was d with methyl-1/3 yl(methyl-t/3)nitropentanoate-3,3-1/2 (75 g, 380 mmol) and 2000 mL of pH 7.5 Na-phosphate buffer @ 0.8 M. To this was added lipase from Rhizomucor miehei (sigma L4277, palatase from Novozymes) (0.5 vol) and stirred at 30 °C for 25 h. Chiral HPLC (ADH 4.6 x250 mm, 5gm, 1.0 mL/min, 98%Heptane /2% IP A) shows .2 ratio of enantiomers. The reaction mixture was extracted twice with MTBE (1 L each time). The organic included any emulsion formed during the extractions. The combined organics were washed two times with an aqueous solution of sodium bicarbonate (5 vol), brine (5 vol), dried over sodium sulfate and concentrated under vacuum to afford the desired product methyl-^ (,5')methyl(methyl-t/3) mtropentanoate-3,3-1/2 as pale yellow oil (32.5 g, 43% yield).
Step C: 6V)-5,5-Dimethyl(methyl-i/3)pyrrolidinone-4,4-i/2 D _ O H2 D HN \(S) Raney-Ni CD3 (S) OCD3 ------------------► NO2 cd3 D D ] A high-pressure vessel (Parr shaker bottle, 500 mL) was purged with and maintained under N2. The vessel was charged sequentially with deionized water rinsed (3 times) damp Raney®2800 Ni (6.1 g), methyl-tf (5')methYl(niethyl-<A) entanoate-3,3-1/2 (32.5 g, 165 mmol), and ethanol (290 mL). The vessel was sealed and evacuated/backfilled with N2 (3 times). With no stirring, the vessel was then SUBSTITUTE SHEET (RULE 26) evacuated and backfilled with H2 (30 psi). The Parr bottle was shaken while heating the ts to 60 °C, and the H2 pressure was maintained at 30 psi for 8 hours. The vessel was evacuated/backfilled with N2 (3 times) and the contents were d by vacuum filtration (Celite pad; N2 blanket). The flask/filter-pad was washed with ethanol (3 x 50 mL). After the final w ash, the solvent-wet filter-cake was transferred to another receiver and covered with water for al. Note: At no time should the catalyst be fully dried (keep damp throughout the filtration process). The filtrate and washes were combined and concentrated (40 °C/40 torr) to afford (A)-5.5-dimethyl(methyl-r/3)pyrrolidin one-4,4-<f2 as white solid (20 g, 92%).
Step D: (4S)-3,3-Dideuterio-2,2-dimethyl (trideuteriomethyl)pyrrolidine Hydrochloride HN 8,(5) HN'\(S) UAIH4 CD3 HCI D D D “ -HCI A 1-L, three-neck round bottom flask equipped an overhead mechanical stirrer, a nitrogen line and a J-Kem thermocouple was charged with lithium aluminum hydride s (7.6 g, 202 mmol) in THF (80 mL, 4 vol) warmed from 20 - 36 °C (heat of mixing). A solution of (iS)-5,5-dimethyl(methyl-<i3)pyrrolidinone-4,4-(i2 (20. g, 150 mmol) in THF (120 mL, 6 vol) was added to the suspension over 30 minutes while allowing the reaction temperature to rise to ~60 °C. The on temperature was increased to near reflux (~68 °C) and maintained there for 16 h. The reaction mixture was cooled to below 40 °C and diluted with 200 mL (10 vol) of MTBE. The mixture was quenched slowly with ise addition of a saturated s solution of sodium sulfate (1 vol) over 2 h. Note: Vigorous degassing (H2) was observed, the mixture becomes thick then thins, and the dark gray mixture turns white. After the addition was completed, the on mixture was cooled to room temperature. The solid was d by filtration (Celite pad) and washed with ethyl acetate (4 vol). With external cooling and a N2 blanket, the filtrate and washings were combined and treated with drop-wise addition of anhydrous 4 M HCI in dioxane (38 mL, 152 mmol) while maintaining the temperature below 20 °C. After the addition was completed (20 minutes), the resultant suspension was concentrated under vacuum at 45 °C. The SUBSTITUTE SHEET (RULE 26) suspension was lled with heptanes (4 vol) twice during tration. The suspension was cooled to below 30 °C when the solid was collected by filtration under a N2 blanket. The solid was dried under N2 suction and further dried under high vacuum at 45 °C to afford (4S)-3,3-dideuterio-2,2-dimethyl(tndeuteriomethyl)pyrrolidine hydrochloride (17.5 g, 75%). The product is quite hygroscopic so it was manipulated under nitrogen.
Synthetic Example 8: Synthesis of nd 8,6-[3- (Dispiro[2.0.24.l3]heptanylmethoxy)pyrazol-l-yl]-N-(o-tolylsulfonyl)[(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide P O. .0 N'S k2co3 o~fJNk N Cl HCI P 0. 0 0~fj N N 2-Chloro[3-(dispiro[2.0.24.l3]heptanylmethoxy)pyrazol-l-yl]-N-(o- tolylsulfonyl)pyridinecarboxamide (0.170 g, 0.341 mmol) and (4S)-2,2,4- trimethylpyrrolidine (Hydrochloride salt) (0.116 g, 1.02 mmol) were combined and dissolved in DMSO (2 mL). Finely ground potassium carbonate (95 mg, 0.68 mmol) was added. The reaction mixture was sealed and heated overnight to 130 °C. After cooling to room temperature, the on mixture was diluted with ethyl acetate (50 mL) and washed with aqueous citric acid (1 M, 2* 50 mL) and brine (lx 50 mL). The organic layer was dried over sodium sulfate, filtered and trated under reduced pressure. The product was isolated by silica gel column chromatography eluting with a SUBSTITUTE SHEET (RULE 26) 0-20% gradient of methanol in dichloromethane on a 12 gram silica gel column to afford 6-[3-(dispiro[2.0.24.l3]heptanylmethoxy)pyrazol-l-yl]-N-(o-tolylsulfonyl) [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (0.030 g. 15%). ESI-MS m/z calc. 575.26, found 576.36 (M+l)+; Retention time: 2.46 minutes.
Synthetic Example 9: Synthesis of nd 9, N-(Benzenesulfonyl) [4-(hydroxymethyl)-2,2-dimethyl-pyrrolidin-l-yl][3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide O O H H H N :0 N N O ‘O —- 'OH O' OH HO HO o.X./ 4 4 9 ,S N'S F F H °'SiX OH Step A: 2-Hydroxymethylmethylnitro-pentanoic acid methyl ester O 0 ■0"N^0 "OH l,8-Diazabicyclo[5.4.0]undecene (3.6 mL, 24 mmol) was added to 2- mtropropane (26.5 mL, 292 mmol). This mixture was heated to 65 °C and the heat was turned off and methyl 2-(hydroxymethyl)acrylate (25 mL, 243 mmol) was added se. The heat was then turned back on at 80 °C. After heating for Ih the heat was turned off and the on was stirred at room temperature overnight before heating at 80 °C for another 2h. The reaction was diluted with ethyl acetate (250 mL) and washed with 1M hydrogen chloride (2 x 125 mL), aqueous bicarbonate (125 mL) and brine (125 mL). The reaction product mixture was chromatographed on a 330g column of silica gel in 0-60% hexanes:ether eluting at 55-60% to give oxymethylmethylnitro- SUBSTITUTE SHEET (RULE 26) pentanoic acid methyl ester (29.68g, 60%) as alight green oil. ESI-MS m/z calc. , found 206.1 (M+l)+. Retention time: 1.67 minutes. 'H NMR (250 MHz, CDC13) ppm 1.50 - 1.59 (m, 6 H) 1.85 - 1.98 (m, 1 H) 2.10 - 2.23 (m, 1 H) 2.36 - 2.50 (m, 1 H) 2.60 (d, J=5.71 Hz, 1 H) 3.66 - 3.77 (s, 3 H) Step B: 3-Hydroxymethyl-5,5-dimethyl-pyrrolidinone O H N .0 -0'n:0 Hydroxymethylmethylnitro-pentanoic acid methyl ester (4.45g, 21.7 mmol) was added to absolute ethanol (60 mL) followed by Raney Nickel (1.7g, -15% wt). The on was heated at 60 °C under 2 bar of H2 overnight. More Raney Nickel (1,0g, -50% wt) was added and the reaction heated at 60 °C under 5 bar H2 for 3.5 h. At this point, more 2-hydroxymethylmethylnitro-pentanoic acid methyl ester (3.95g, 19.3 mmol) was added and the reaction heated for 72 h refilling H2 to maintain 5 bar.
The reaction was filtered through celite and washed with methanol. The crude reaction was chromatographed on silica gel and eluted with 0-10% dichloromethanemethanol at %, resulting 3-hydroxymethyl-5,5-dimethyl-pyrrolidinone (3.69g, 63%) as a white solid. ‘H NMR (250 MHz, CDC13) 5 ppm 1.31 (d, J=9 01 Hz, 6 H) 1.72 (dd, JM2.52, .33 Hz, 1 H) 2.04 (dd, 1=12.58, 8.84 Hz, 1 H) 2.73 - 2.91 (m, 1 H) 3.31 (d, J=4.72 Hz, 1 H) 3.64 - 3.95 (m, 2 H) 5.93 (br. s., 1 H) Step C: (5,5-Dimethyl-pyrrolidinyl)-methanol N H O N HO HO Lithium aluminum hydride , 103.00 mmol) was suspended in tetrahydrofuran (60 mL). Hydroxymethyl-5,5-dimethyl-pyrrohdmone , 25.77 SUBSTITUTE SHEET (RULE 26) mmol) in tetrahydrofuran (30 mL) was then added dropwise and the reaction was heated at 65°C for 40h. The reaction was diluted with 2-methyl-tetrahydrofuran (125 mL) and then cooled in an ice bath before saturated aqueous Rochelle Salt (200 mL) was added dropwise. The organic layer was extracted with 2-methyl-tetrahydrofuran (2 x 200 mL) and dried over sodium sulfate to give crude imethyl-pyrrolidinyl)-methanol , 104%). NMR (250 MHz, CDC13 5 ppm 1.06 - 1.24 (m, 6 H) 1.29 (dd, J=12.58, 7.20 Hz, 2 H) 1.43 (s, 1 H) 1.68 - 1.89 (bs, 1 H) 2.31 - 2.52 (m, 1 H) 2.83 (dd, J=11.10, 5.49 Hz, 1 H) 3.05 - 3.26 (m, 1 H) 3.48 - 3.71 (m, 1 H) Step D: 4-(tert-Butyl-dimethyl-silanyloxymethyl)-2,2-dimethyl- pyrrolidine H H N N HO O To (5,5-dimethyl-pyrrolidiny])-methanol (3.08g, 23.8 mmol), tert- butyldimethylsilyl chloride (4.3Ig, 28.6 mmol) in acetonitrile (24 mL) was added 1,8- Diazabicyclo[5.4.0]undecene (5.3 mL, 35.7 mmol) The reaction was stirred for 3.5 h. The reaction was diluted with chloroform (250 mL) and washed with water (125 mL) and brine (125 mL) then dried over sodium sulfate. The crude was chromatographed on silica gel and eluted with dichloromethane/methanol, g at 15-35% ol to give 4-(tert-butyl-dimethyl-silanyloxymethyl)-2,2-dimethyl-pyrrolidine , 67%) as ayellow oil after two columns. ESI-MS m/z calc. 243.47, found 244.2 (M+l)+Retention time: 2.52 minutes. LH NMR (250 MHz, CDC13) 5 ppm -0.05 - 0.11 (m, 6 H) 0.89 (s, 9 H) 1.19 (d, M8.02 Hz, 6 H) 1.25 - 1.32 (m, 1 H) 1.74 (dd, M2.63, 8.79 Hz, 1 H) 1.92 (br. s., 1 H) 2.32-2.50 (m, 1 H) 2.81 (dd, J=11.54, 6.37 Hz, 1 H) 3.11 (dd, J=11.48, 7.97 Hz, 1 H) 3.45 - 3.61 (m, 2 H) Step E: N-(Benzenesulfonyl)[4-(hydroxymethyl)-2,2-(limethyl- pyrrolidin- l-yl] [3- [2-[ l-(trifluoromethyl)cyclopropyl] ethoxy] l yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) E tf 0^° NH F' N'S F H Cl xx 1 \P F. %p F ^-0 "N °'SiX N-(benzenesulfonyl)chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-1 -yl]pyridinecarboxainide (25 mg, 0.04855 mmol), tert-butyl-[(5,5-dimethylpyrrolidinyl)methoxy]-dimethyl-silane (approximately 35.45 mg, 0.1456 mmol), and K2CO3 (approximately 33.56 mg, 0.2428 mmol) were combined in DMSO (0.5 mL) and heated at 130 °C for 16 h. The reaction was partitioned between a 1M citric acid solution and ethyl acetate and the organics were separated. The organics were washed with brine, dried over sodium sulfate and evaporated. The crude matenal was purified by silica gel chromatography g with 0-10% methanol in dichloromethane to give N-(benzenesulfonyl)[4-[[tertbutyl (dimethyl)silyl]oxymethyl]-2,2-dimethyl-pyrrolidin-l-yl][3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (15 mg, 43%) ESI-MS m/z calc. 721.2941, found 722.4 (M+l)+; Retention time: 0.97 minutes.
Step F: N-(Benzenesulfonyl)[4-(hydroxymethyl)-2,2-dimethyl- pyrrolidin- l-yl] [3- [2-[ l-(trifluoromethyl)cyclopropyl] ethoxy] l yl]pyridinecarboxamide °'Si OH ] zenesulfonyl)[4-[[tert-butyl(dimethyl)silyl]oxymethyl]-2,2- dimethyl-pyrrolidin-1 -yl] [3-[2-[ 1 luoromethyl)cy clopropy 1] ethoxy |pyra/ol-1 - yl]pyridinecarboxamide (15 mg, 43%) was dissolved in THE (1 mL) and cooled in an SUBSTITUTE SHEET (RULE 26) ice bath. Tetra-n-butylammonium fluoride in THF (300 |iL of 1 M, 0.3000 mmol) was added and the reaction was allowed to warm to room temperature. The reaction mixture was d for 1 h and then partitioned between ethyl acetate and 1M citric acid solution. The organics were washed with brine, dried over sodium sulfate and evaporated. The crude matenal was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give zenesulfonyl)[4- (hydroxymethyl)-2,2-dimethyl-pyrrolidm-l-yl][3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (8.5 mg, 29%) ESI-MS m/z calc. 607.20764, found 608.4 ; ion time: 1.9 minutes.
Synthetic Example 10: Synthesis of nd 10, N-(Benzenesulfonyl)- 6-[3-[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (l-Trifluoromethyl-cyclobutyl)-methanol PH LAH PH F3p O F3C ] 1-Trifluoromethyl-cyclobutanecarboxylic acid (5.0 g, 30. mmol) was dissolved in diethyl ether (60 mL) and cooled to 0 °C. Lithium aluminum hydride (38.66 mL, 1M in diethyl ether) was added se and the solution was allowed to warm to room temperature overnight. The reaction solution was cooled to 0 °C with stirring, and sodium sulfate decahydrate was added, which resulted in gradual evolution of gas.
Portionwise addition was continued until no more bubbling was observed at room temperature. The reaction solution was then filtered over a bed of Celite, washing with diethyl ether. The filtrate was concentrated under reduced pressure to give 5.44 g of a mixture containing the d product and some diethyl ether e (36% by NMR integration). This afforded 1-trifluoromethyl-cyclobutyl-methanol (3.46 g, 78%) as a colorless oil. 'H NMR (250MHz, CDC13) 5 (ppm): 3.82 (s, 2H), 2.39-2.14 (m, 2H), 2.10-1.85 (m,4H). 3-(l-Trifluoromethyl-cyclobutylmethoxy)-pyrazole-l-carboxylic acid tert-butyl ester SUBSTITUTE SHEET (RULE 26) Nk^Boc o Boc OH 0-//- N F3C DIAD, Ph3P f3c 1-Trifluoromethyl-cyclobutyl-methanol (1.50 g, 9.73 mmol) and 3-oxo-2,3- dihydro-pyrazolecarboxylic acid tert-butyl ester (1.63 g, 8.85 mmol) were dissolved in anhydrous tetrahydrofuran (32 mL). The solution was degassed by sonication and flushed with nitrogen gas. nylphosphine (2.55 g, 9.73 mmol) was added, and diisopropyl azodicarboxylate (1.92 mL, 9.73 mmol) was then added dropwise. Upon tion of addition, the on was heated to 50 °C for 16 hours. After cooling to room temperature, the reaction was diluted with ethyl acetate (100 mL) and washed with 1M sodium hydroxide solution (2 x 100 mL), then brine (125 mL). The organics were dried over sodium sulfate, filtered, and concentrated under d pressure. The crude yellow oil was purified by flash chromatography using a 0-10% ethyl acetate in hexanes gradient method to afford rifluoromethyl-cyclobutylmethoxy)-pyrazole-lcarboxylic acid tert-butyl ester (2.48 g, 87%) as an off-white solid. ESI-MS m/z calc. 320.31, found 321.1 (M+l)+. Retention time: 3.74 minutes 3-(l-Trifluoromethyl-cyclobutylmethoxy)-lH-pyrazole hydrochloride N-'m'Boc N"NH o-//- N HCI, dioxane f3c f3c 3-(l-Trifluoromethyl-cyclobutylmethoxy)-pyrazole-l-carboxylic acid tert- butyl ester (2.48 g, 7.74 mmol) was dissolved in 4M hydrogen chloride in dioxane (77 mL). The solution was stirred overnight at room temperature, followed by removal of the volatiles under reduced re to afford the hloride salt of 3-(ltrifluoromethyl-cyclobutylmethoxy )-lH-pyrazole (1.95 g, 98%) as a white powder. ESI- MS m/z calc. 220.20, found 221.2 (M+l)+. Retention time: 2.67 minutes.
SUBSTITUTE SHEET (RULE 26) tyl 2-chloro(3-((l-(trifluoromethyl)cyclobutyl)methoxy)-lH- pyrazol-l-yl)nicotinate .HCI 'OfBu P-Afn cr N ci N. s, 0_N N Cl DABCO, K2CO3 F3C f3c 3-(l-Trifluoromethyl-cyclobutylmethoxy)-lH-pyrazole hydrochloride salt (1.95 g, 7.61 mmol) and 2,6-dichloro-nicotinic acid tert-butyl ester (1.89 g, 7.62 mmol) were dissolved in dimethylformamide (15 mL), and potassium carbonate (4.21 g, 30.5 mmol) was added followed by l,4-diazabicyclo[2.2.2]octane (0.43 g, 3.8 mmol). The reaction was stirred at room temperature ght, then water (150 mL) was added and the aqueous layer was extracted with 4:1 ethyl acetate:hexanes (100 mL). The organic phase was washed with brine (70 mL), dried over sodium sulfate, and trated under d pressure. The crude oil was purified by silica gel chromatography using a 0-10% ethyl acetate in hexanes nt method to afford 2-chloro[3-(ltrifluoromethyl-cyclobutylmethoxy )-pyrazole-l-yl]-nicotinic acid tert-butyl ester (1.94 g, 66%) as a white solid. ESI-MS m/z calc. 431.85, found 432.2 (M+l)+. ion time: 4.61 minutes. 2-Chloro[3-(l-trifluoromethyl-cyclobutylmethoxy)-pyrazole-l-yl]- nicotinic acid 0 0 OfBu TFA OH Ck/'N^N' 'Cl 0--^ N N Cl f3c F3C 2-Chloro[3-(l-trifluoromethyl-cyclobutylmethoxy)-pyrazole-l-yl]- nicotinic acid tert-butyl ester (1.9 g, 4.40 mmol) was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (5.0 mL) was added. The reaction solution was stirred at room temperature overnight, after which the volatiles were removed under reduced pressure to afford 2-chloro[3-(l-tnfluoromethyl-cyclobutylmethoxy)-pyrazole-l-yl]- SUBSTITUTE SHEET (RULE 26) WO 64632 nicotinic acid (1.61 g, 97%) as a white solid. ESI-MS m/z calc. 375.74, found 376.2 (M+l)+. Retention time: 3.57 minutes.
Synthesis ofN-(Benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyri(lmecarboxamide GDI, DBU 9 q 'OH M'S,N ■N"N H 0 0~^ N Cl h2n|-0 'Cl f3c f3c ] To a stirred solution of 2-chloro[3-[[l-(trifluoromethyl)cyclobutyl]meth- oxy]pyrazol-l-yl]pyridinecarboxylic acid (0.150 g, 0.399 mmol) in anhydrous ydrofuran (3.0 mL) was added GDI (78 mg, 0.4810 mmol) in one portion. The solution was stirred at ambient temperature for 2 h. Then solid benzenesulfonamide (76 mg, 0.48 mmol) was added in one portion, followed by DBU (183 mg, 1.20 mmol) and the tea-colored solution was stirred at ambient temperature for an additional 2 h. To the reaction mixture was slowly added citric acid (2.5 mL of 1.0 M, 2.500 mmol), followed by brine (5 mL). After stirring for 10 min, the neous material was extracted with ethyl acetate (3 x 25 mL). The combined organic extracts were washed with brine (10 mL), dried over anhydrous sodium sulfate, ed and concentrated under reduced pressure. After drying under vacuum for 1 h, N-(benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridmecarboxamide (181 mg, 88%) was obtained as white solid. It contained some starting acid impurity, and used in the subsequent step without further purification. ESI-MS m/z calc. 514.0689, found 515.1 (M+l) + ; Retention time: 1.98 minutes sis ofN-(Benzenesulfonyl)[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) o o .HCI Yb'S; 9 q HN'Ais) M-S OS'N'n^n' 'ci 0^,N'N^N✓s N s K2C03 F3c f3c A mixture of zenesulfonyl)chloro[3-[[l-(trifluoromethyl)cyclobutyl]- methoxy]pyrazol-l-yl]pyridinecarboxamlde (0.160 g, 0.311 mmol), (4S)-2,2,4- hylpyrrolidine (Hydrochloride salt) (139 mg, 0.932 mmol) and potassium carbonate (215 mg, 1.554 mmol) was stirred in in anhydrous yl ide (2.7 mL) under an atmosphere of nitrogen at 130 °C for 18 h. The reaction was allowed to cool to ambient temperature and diluted with water (15 mL) and extracted with ethyl acetate (3 x 25 mL). The combined organics successively were washed with aqueous 1 M citric acid (310 pL of 1.0 M, 0.3107 mmol), and brine, dried over anhydrous sodium sulfate, filtered and evaporated to give yellow crude material. It was purified from CombiFlashRf system using 40 g gold silica gel column and during with 0-5 % methanol in methylene chloride (over 45 min). The product came out at 25 min (2.6 % methanol). The desired fractions were combined and concentrated under reduced pressure. Upon further drying overnight under high vacuum, N-(benzenesulfonyl)[3- [[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (HCI salt, 40 mg, 20%) was obtained.
ESI-MS m/z calc. 591.2127, found 592.3 (M+l)+; Retention time: 2.25 minutes. 'H NMR (400 MHz, DMSO-d6) 5 12.49 (s, 1H), 8.22 (d, J = 2.8 Hz, 1H), 8.00 (dd, J = 8.2, 2.1 Hz, 2H), 7.82 (d, J = 8.2 Hz, 1H), 7.72 (tt, J = 8.2, 2.0 Hz, 1H), 7.65 (dt, J = 8.2, 2.0 Hz, 2H), 6.95 (d, J = 8.2 Hz, 1H), 6.18 (d, J = 2.8 Hz, 1H), 4.48 (s, 2H), 2.42 (t, J = 10.5 Hz, 1H), 2.36 - 2.22 (m, 3H), 2.11 (td, J = 12.1, 5.7 Hz, 4H), 1.95 (qd, J = 9.7, 4.3 Hz, 1H), 1.83 (dd, J = 12.0, 5.6 Hz, 1H), 1.54 (s, 3H), 1.51 (s, 3H), 1.37 (t, J = 12.2 Hz, 1H), 0.65 (d, J = 6.3 Hz, 3H).
Synthetic e 11: Synthesis of Compound 11, N-(4-Cyano -phenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-cyanomethyl-phenyl)sulfonyl[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) 0. ,0 0 *Cl XT'A°-<p ‘OH N'CI GDI <u>-u N-N^N^CI ” )Cl.CN F3C f3c A solution of 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (186.4 mg, 0.5 mmol) and l,r-carbonyldiimidazole (97.29 mg, 0.60 mmol) in THF (2.5 mL) was d for 30 minutes, and 4-cyanomethyl-benzenesulfonamide (127.5 mg, 0.65 mmol) and azabicyclo(5.4.0)undecene (DBU) (89.7 pL, 0.60 mmol)were added. After 16 hours the reaction was diluted with 1 M aqueous citric acid and extracted with ethyl e. The combined extracts were dried over sodium sulfate and evaporated. The residue was ed by silica gel chromatography with 0-5% ol in dichloromethane to give 2-chloro-N-(4-cyanomethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (270 mg, 100%) ESI-MS m/z calc. 539.06, found 540.1 (M+l)+; Retention time: 0.73 minutes.
Step B: N-(4-Cyanomethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 Qo O Oo 11 vi'r N'S K2CO3 N'S H N- H N- X N Cl ON (s' V7 n~// "N N N c -4^(s) N f3c HN f3c A mixture of 2-chloro-N-(4-cyanomethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (270 mg, 0.50 mmol), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (168.1 mg, 1.123 mmol), and potassium carbonate (310.4 mg, 2.246 mmol) in DMSO (1.87 mL)was stirred at 130 °C for 15 hours. The reactions were acidified with 1 M aqueous citric acid and extracted with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, and evaporated. The residue was purified by silica gel SUBSTITUTE SHEET (RULE 26) chromatography with 0-5% methanol in dichloromethane to give impure product. The impure product was re-purified using a reverse phase HPLC-MS method using a Luna C18 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00C UO-AX), and a dual gradient run from 1-99% mobile phase B over 15.0 minutes.
Mobile phase A = LLO (5 mM HC1). Mobile phase B = CLLCN. Flow rate = 50 mL/min, and column temperature = 25 °C to provide N-(4-cyanomethylphenyl )sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (150 mg, 48%) ESI-MS m/z calc. 616.21, found 617.3 (M+l)+; Retention time: 2.06 minutes. 'HNMR (400 MHz, DMSO-cfe) 5 12.96 (s, 1H), 8.23 - 8.18 (m, 2H), 8.03 (d, J= 1.6 Hz, 1H), 7.97 (dd, J = 8.0, 1.8 Hz, 1H), 7.88 (d, J= 8.3 Hz, 1H), 6.95 (d, J= 8.3 Hz, 1H), 6.16 (d, T = 2.7 Hz, 1H), 4.43 - 4.32 (m, 2H), 2.67 (s, 3H), 2.27 (d, J= 3.5 Hz, 1H), 2.25 (s, 1H), 2.17 (dd, J = 11.3, 5.7 Hz, 1H), 1.83 (dd, J= 11.9, 5.3 Hz, 1H), 1.52 (d, J=4.4Hz, 6H), 1.36 (s, 1H), 1.09 (dt, J= 5.5, 1.6 Hz, 4H), 0.70 (d, J= 6.0 Hz, 3H).
] Synthetic e 12: Synthesis of Compound 12, N-(2-Methoxy methyl-phenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: ro-N-(2-methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O. ,0 0 Xk i V X0 <Lp-iJ N ClN- GDI DBU <LP^2 N I f3c f3c A on of 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (186.4 mg, 0 5 mmol) and l,r-carbonyldiimidazole (97.29 mg, 0 60 mmol) in THF (2.5 mL) was stirred for 30 minutes, and 2-methoxymethyl-benzenesulfonamide (130.8 mg, 0.65 mmol) and l,8-diazabicyclo(5.4.0)undecene (DBU) (89.7 pL, 0.60 mmol) were added. After 16 hours the reaction was diluted with 1 M aqueous citric acid and extracted with ethyl acetate. The ed extracts were dried over sodium sulfate and SUBSTITUTE SHEET (RULE 26) evaporated. The residue was purified by silica gel tography with 0-5% methanol in dichloromethane to give 2-chloro-N-(2-methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (210 mg, 77%) ESI-MS m/z calc. 544.1, found 545.1 (M+l)': Retention time: 0.73 minutes as a colorless solid.
Step B: N-(2-Methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 op 9 0 OO O K;CO, N-S N-S' N Cl (Si 3c / F3C HN A mixture of ro-N-(2-methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (210 mg, 0.3854 mmol), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (168.1 mg, 1.123 mmol), and potassium carbonate (310.4 mg, 2.246 mmol) in DMSO (1.87 mL) was stirred at 130 °C for 15 hours. The reaction was acidified with 1 M s citric acid and extracted with ethyl acetate. The ed extracts were washed with brine, dried over sodium sulfate, and evaporated. The residue was purified by silica gel chromatography with 0-5% methanol in dichloromethane to give impure product. The impure product was ified using a reverse phase HPLC-MS method using a Luna C18 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00C U0-AX), and a dual gradient run from 1-99% mobile phase B over 15.0 minutes.
Mobile phase A = H20 (5 mM HCI). Mobile phase B = CH3CN. Flow rate = 50 mL/min, and column temperature = 25 °C to provide N-(2-methoxymethylphenyl )sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (95 mg, 39.25%) ESI-MS m/z calc. 621.2, found 622.3 (M+l)+; Retention time: 2.19 minutes. 'H NMR (400 MHz, DMSO-rfe) 8 12.39 (s, 1H), 8.20 (d,T= 2.8 Hz, 1H), 7.78 (t, J= 8.5 Hz, 2H), 7.10 (d, J = 1.4 Hz, 1H), 6.94 (dd, J= 10.1, 8.1 Hz, 2H), 6.15 (d, J= 2.7 Hz, 1H), 4.43 - 4.30 (m, SUBSTITUTE SHEET (RULE 26) 2H), 3.89 (s, 3H), 2.49 - 2.38 (m, 2H), 2.37 (s, 3H), 2.21 (dd, J= 11.2, 6.1 Hz, 1H), 1.85 (dd, J= 11.9, 5.5 Hz, 1H), 1.53 (d, J= 11.0 Hz, 6H), 1.37 (s, 1H), 1.12-1.04 (m, 4H), 0.78 (d, ,7=6.2 Hz, 3H).
Synthetic e 13: Synthesis of Compound 13: N-(2,4- oxyphenyl)sulfonyl [3- [ [ fluoromethyl)cyclopropyl] methoxy] pyrazoll-yl 4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(2,4-dimethoxyphenyl)sulfonyl|3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide i 9 o.
'OH N'3' <LP<2 N'CI GDI <LP-iJ NN- Cl 0 0 DBU I f3c f3c A solution of 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (186.4 mg, 0.5 mmol) and l,r-carbonyldiimidazole (97.29 mg, 0.60 mmol) in THE (2.5 mL) was stirred for 30 minutes, and 2,4-dimethoxybenzenesulfonamide (141.2 mg, 0.65 mmol) and azabicyclo(5.4.0)undecene (DBU) (89.7 pL, 0.60 mmol) were added. After 16 hours the reaction was diluted with 1 M aqueous citric acid and extracted with ethyl acetate. The combined extracts were dried over sodium sulfate and evaporated. The residue was purified by silica gel chromatography with 0-5% methanol in dichloromethane to give 2-chloro-N-(2,4-dimethoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (210 mg, 75%) ESI-MS m/z calc. 560.1, found 561.1 (M+l)+; Retention time: 0.71 minutes as a colorless solid.
Step B: N-(2,4-Dimethoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) 0 oo Ovy. 0 9,0 0 k2co3 N'S I 'SS ^°~Cjn Cl N 0 (s I f3c HN f3c A e of 2-chloro-N-(2,4-dimethoxyphenyl)sulfonyl[3-[[l- (trifluoromethy l)cy clo-propyl] methoxyjpyrazol-1 -y 1] pyridine-3 -carboxamide (210 mg, 0.3744 mmol), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (168.1 mg, 1.123 mmol), and potassium carbonate (310.4 mg, 2.246 mmol) in DMSO (1.87 mL) was stirred at 130 °C for 15 hours. The reactions were acidified with 1 M aqueous citric acid and extracted with ethyl acetate. The combined ts were washed with brine, dried over sodium sulfate, and evaporated. The e was purified by silica gel chromatography with 0-5% methanol in dichloromethane to give impure t. The impure product was re-purified using a reverse phase HPLC-MS method using a Luna C18 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00C U0-AX), and a dual gradient run from 1-99% mobile phase B over 15.0 minutes.
Mobile phase A = H2O (5 mM HCI). Mobile phase B = CH3CN. Flow rate = 50 mL/min, and column ature = 25 °C to provide N-(2,4-dimethoxyphenyl)sulfonyl- 6-[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (110 mg, 46%) ESI-MS m/z calc. 637.2, found 638.3 (M+l)+; Retention time: 2.14 minutes. 1HNMR(400 MHz, DMSO- 4)5 12.34 (s, 1H), 8.20 (d, J= 2.8 Hz, 1H), 7.82 (d,J= 8.8 Hz, 1H), 7.77 (d,J= 8.3 Hz, 1H), 6.92 (d,/= 8.2 Hz, 1H), 6.74 (d, J= 2.3 Hz, 1H), 6.70 (dd, J= 8.8, 2.3 Hz, 1H), 6.15 (d, J= 2.7 Hz, 1H), 4.43 - 4.31 (m, 2H), 3.90 (s, 3H), 3.85 (s, 3H), 2.54 (s, 1H), 2.42 (dd, J= 10.5, 7.0 Hz, 1H), 2.21 (dd, /= 11.6, 5.9 Hz, 1H), 1.85 (dd, J= 11.9, .5 Hz, 1H), 1.55 (s, 3H), 1.52 (s, 3H), 1.38 (s, 1H), 1.09 (dt, J = 5.9, 1.6 Hz, 4H), 0.80 (d, J= 6.3 Hz, 3H).
Synthetic Example 14: Synthesis of Compound 14: N- (Benzenesulfonyl)[3-[[l-(trifhioromethyl)cyclopropyl]methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) WO 64632 ■N'N^cr^ °=U diad' pph^ O—f' HCI F F F- + K2CO3, DABCO N'M^jD^'n^CI HCI F- O-& F F cr n ci N'M^N^CIj(X CDI, DBU o-^ + h2n F- ■o rs^AqsP 'O HN^!i ov-xtol9sP + K2CO3 F F F- Step A: tert-Butyl (trifluoromethyl)cyclopropyl]methoxy]pyrazole- 1-carboxylate DIAD, PPh3 + N..°=u O-A F F F A 5000 mL 3 neck round bottom flask as fitted with a mechanical stirrer, a heating mantle, a J-Kem temperature probe/controller, an on funnel, a water cooled reflux condenser and a nitrogen inlet/outlet. The vessel was charged under a nitrogen atmosphere with tert-bulyl 5-oxo-lH-pyrazolecarboxylate (70 g, 0.3800 mol) and tetrahydrofuran (840 mL, 12 mL/g) which provided a clear pale yellow solution. Stirring was commenced and the pot temperature was recorded at 19 °C. The vessel was then charged with [l-(trifluoromethyl)cyclopropyl]methanol (58.56 g, 0.4180 mol) added neat in one portion followed by triphenylphosphine (109.6 g, 0.4180 mol) added as a solid in one portion. The resulting clear pale yellow solution was then treated with ropyl azodicarboxylate (clear reddish-orange liquid) (82.3 mL, 0.4180 mol) added neat dropwise over 1 hour which resulted in a gradual exotherm to 40 °C and a clear light amber solution. The reaction mixture was then heated to a pot SUBSTITUTE SHEET (RULE 26) temperature of 50 °C and the condition was maintained for 2 hours when analysis by LC/MS indicated te consumption of the starting material. The clear amber reaction mixture was concentrated under reduced pressure and the resulting clear dark amber oil was suspended in toluene (560 mL) and stirred at room temperature for 1 hour during which time a solid (triphenylphosphine oxide MW = 278.28) precipitated. The thick slurry was filtered through a glass frit Buchner funnel and the filter cake was displacement washed with toluene (150 mL) and then pulled for 30 minutes. The clear amber te was concentrated under reduced pressure to provide a clear amber oil. The material was ed by silica gel column flash chromatography (solid load on Celite 1.5 kg RediSep column) eluting with a gradient of 100% hexane to 20% EtOAc in hexane collecting 450 mL fractions. The product elutes around 5% EtOAc in hexane.
The desired fractions were combined and concentrated under reduced re to provide a clear pale yellow oil as the desired product utyl 3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazole-l-carboxylate (81 g, 0.264 mol, 70%).
^ NMR (400 MHz, DMSO-d6) 5 8.10 (d, J = 2.9 Hz, 1H), 6.14 (d, J = 3.0 Hz, 1H), 4.31 (s, 2H), 1.55 (s,9H), 1.07 (dp, J = 4.9, 4H). ESI-MS m/z calc. 306.11914, found 259.0 (M-48)+; Retention time: 1.76 minutes Step B: 3-[[l-(Trifluoromethyl)cyclopropyl]methoxy]-lH-pyrazole ,N'lAo''^ o-//N~nh A 5000 mL 3 neck round bottom flask was fitted with a mechanical stirrer, a g mantle, a J-Kem temperature probe, a water cooled reflux condenser, an addition funnel and a en inlet/outlet. The vessel was charged under a nitrogen atmosphere with tert-butyl (trifluoromethyl)cyclopropyl]methoxy]pyrazole-lcarboxylate (80 g, 0.2612 mol), dichloromethane (320 mL, 4 mL/g) and methyl l (320 mL, 4 mL/g) which provided a clear pale yellow solution. Stirring was commenced and the pot temperature was recorded at 19 °C. The addition funnel was charged with 4 MHC1 in 1,4-dioxane (195.9 mL, 0.7836 mol) which was subsequently added dropwise over 1 hour which resulted in a gradual exotherm to 30 °C. The resulting clear pale yellow solution was heated to a pot temperature of 45 °C and the condition was SUBSTITUTE SHEET (RULE 26) maintained for 1 hour when analysis by LC/MS indicated reaction completion. The reaction mixture was allowed to cool to room temperature and then trated under reduced pressure. The remaining residue was dissolved in tert-butyl methyl ether (640 mL) and then transferred to a separatory funnel and partitioned with 2 M sodium hydroxide solution (391.8 mL, 0.7836 mol). The organic layer was removed and the residual aqueous was extracted with tert-buty l methyl ether (2 x 200 mL). The combined c was washed with saturated sodium chloride solution (500 mL), dried over sodium sulfate (300 g) and then filtered through a glass frit Buchner funnel. The clear pale yellow filtrate was concentrated under reduced pressure to provide a clear light yellow oil which solidified upon standing to provide a white solid (49.5 g, 0.240 mol, 92%) as the d product 3-[[l-(trifluoromethyl)cyclopropyl]methoxy]-lH- pyrazole. 1HNMR(400 MHz,DMSO-d6)8 11.90 (s, 1H), 7.51 (d,J = 2.4Hz, 1H), .67 (d, J = 2.4 Hz, 1H), 4.19 (s, 2H), 1.09 - 0.97 (m, 4H). ESI-MS m/z calc. 206.0667, found 207.0 (M+l)+; Retention time: 1.07 minutes.
] Step C: utyl 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy ] py razol- 1-yl] pyridinecarb oxylate U ♦ F N'NH CW' KyCOs, DABCQ O-f/ N Cl cr n 'ciX F F F' A 5000 mL 3 neck round bottom flask was fitted with a ical stirrer, a cooling bath used as ary containment, a J-Kem temperature probe, a water cooled reflux condenser, an addition funnel and a nitrogen inlet/outlet. The vessel was charged under a nitrogen atmosphere with 3-[[l-(trifluoromethyl)cyclopropyl]methoxy]-lH- pyrazole (45 g, 0.2183 mol) and N,N-dimethylformamide (540 ml, 12 mL/g) which provided a clear pale yellow solution. Stirring was commenced and the pot temperature was recorded at 17 °C. The vessel was then charged with tert-butyl 2,6- dichloropyridinecarboxylate (54 16 g, 0.2183 mol) added as a solid in one portion.
The resulting clear pale yellow solution was then treated with potassium ate (39.22 g, 0.2838 mol) added as a solid in one portion followed by 1,4- diazabicyclo[2.2.2]octane (3.67 g, 4 mol) added as a solid in one portion. The SUBSTITUTE SHEET (RULE 26) resulting pale yellow suspension was allowed to stir at room temperature for 24 hours.
The reaction mixture was cooled to 10 °C with a cmshed ice/water cooling bath. The addition funnel was charged with water (540 mL) added dropwise over 45 s which resulted in a thick suspension and an exotherm to 15 °C. The resulting suspension was continued to stir at 15 °C for 30 minutes and then filtered through a glass frit Buchner funnel. The filter cake was cement washed with water (2 x 500 ml) and then pulled in the Buchner for 2 hours. The material was then allowed to air dry overnight to provide (73 g, 0.175 mol, 80%) of a white granular solid as tert-butyl 2- [3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridine carboxylate. ESI-MS m/z calc. 361.0441, found 361.9 (M+l)+; Retention time: 2.27 minutes.
Step D: 2-Chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid O Jk 0 if 'o 0 N'N' '[s|' 'ClX N-mAA H fY 0 O-// N N Cl F F F F F F A 1000 mL 3 neck round bottom flask as fitted with a mechanical stirrer, a heating mantle, a J-Kem temperature probe/controller, an addition funnel, a water cooled reflux condenser and a en mlet/outlet. The vessel was charged under a nitrogen atmosphere with tert-butyl 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate (70 g, 0.1675 mol) and anol (350 mL) which provided an off-white suspension. Stirring was commenced and the pot ature was recorded at 19 °C. The addition funnel was charged with aqueous 6 M HCI (139.6 mL, 0.8375 mol) which was added se over 10 s which resulted in an exotherm to 30 °C. The resulting suspension was then heated to reflux (pot temperature ~82 °C) Upon heating the suspension turns to a clear pale yellow solution (pot temperature ~75 °C at this point). After stirring at reflux for ~30 minutes a solid began to precipitate. The suspension was continued to stir at reflux for an additional 30 minutes at which point water (210 mL) was added se SUBSTITUTE SHEET (RULE 26) over 15 minutes. The heat was then removed and the suspension was continued to stir and allowed to slowly cool to room temperature. The material was collected by vacuum filtration in a glass frit Buchner funnel and the filter cake was cement washed with 1:1 water/2-propanol (100 mL) followed by water (2 x 100 mL) and then pulled in the Buchner for 30 minutes. The material was further dried in a vacuum oven at 45 °C for 24 hours to provide 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxylic acid (56 g, 0.155 mol. 92%) as a white solid. 'H NMR (400 MHz, DMSCM;) 8 13.64 (s, 1H), 8.44 (d, /= 2.9 Hz, 1H), 8.41 (d, /= 8.4 Hz, 1H), 7.74 (d, J= 8.4 Hz, 1H), 6.24 (d, .7=2.9 Hz, 1H),4.41 (s, 2H), 1.16-1.07 (m, 4H).
ESTMS m/z calc. 41, found 361.9 (M+l)+; ion time: 3.23 minutes Step E: N-(Benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O 1%P ‘OH QwP iT^rnY"! GDI, DBU 0~//N'N'A'N<!^CI + H2N :s:X) N.
F- F- F F F F 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0.4144 mmol) was dissolved in THF (2.000 mL). GDI (approximately 80.64 mg, 0.4973 mmol) was added. The reaction mixture was stirred at room temperature for 1.5 hours. Benzenesulfonamide (approximately 84.68 mg, 0.5387 mmol) was added ed by DBU ximately 126.2 mg, 124.0 pL, 0.8288 mmol). The reaction mixture was allowed to stir at room temperature for another 1.5 hours. The reaction mixture was concentrated to half volume, diluted with dichloromethane and directly injected onto a 12 gram silica gel column and subjected to a 0-10% methanol in dichloromethane gradient; product eluted at 10%. Fractions ning the d product were combined and concentrated. N-(benzenesulfonyl)- 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridine carboxarmde (168 mg, 81%) was ed as a clear colorless oil. ESI-MS m/z calc. 500.05328, found 501.0 (M+l)+; Retention time: 1.92 minutes (3 minute run).
SUBSTITUTE SHEET (RULE 26) Step F: N-(Benzenesulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl ] methoxy] pyrazol- 1-yl] [(4S)-2,2,4-trimethylpy rrolidin- 1-yl] pyridine amide 0 o.XqsP HN^S], V? k?co3 o-v^'n^n^nXs F- F F F F F N-(Benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (168 mg, 0.3354 mmol) and (4S)-2,2,4-trimethylpyrrolidme chloride salt) (approximately 150.6 mg, 1.006 mmol) were combined and dissolved in DMSO (0.5 mL). Finely ground potassium ate (approximately 278.1 mg, 2.012 mmol) was added, and the reaction mixture was allowed to stir at 130 °C overnight. The reaction mixture was diluted with EtOAc (50 mL) and washed with s 1 M citric acid (2x 50 mL) and brme (1 x 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was punfied by silica gel column chromatography: 24 gram silica gel column, 0-5% MeOH/DCM gradient; product eluted at 2.5%. Pure fractions were combined and trated under reduced pressure, and azeotroped with MeOH, to provideN-(benzenesulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide (74.9 mg, 39%). 'h NMR (400 MHz, DMSO-d6) 5 12.51 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 8.05 - 7.95 (m, 2H), 7.82 (d, J = 8.2 Hz, 1H), 7.78 - 7.70 (m, 1H), 7.66 (dd, J = 8 3, 6 7 Hz, 2H), 6 92 (d, J = 8.3 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.43 - 4.30 (m, 2H), 2.40 (t, J = 10.5 Hz, 1H), 2.26 (t, J = 8.6 Hz, 1H), 2.09 (dt, J = 12.3, 6.4 Hz, 1H), 1.82 (dd, J = 12.0, 5.6 Hz, 1H), 1.53 (s, 3H), 1.51 (s, 3H), 1.36 (t, J = 12.1 Hz, 1H), 1.15 -1.04 (m, 4H), 0.64 (d, J = 6.2 Hz, 3H). ESI-MS m/z calc. 577.1971, found 578.3 (M+l)+; Retention time: 2.16 minutes (3 minute run).
SUBSTITUTE SHEET (RULE 26) ] Synthetic Example 15: Synthesis of Compound 15: N-(o-Tolylsulfonyl)- 6- [3- [(2,2,3,3-tetramethylcyclopropyl)methoxy] pyrazol- l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 0 I 0 OH N-nh Ph^P, DIAD NaOH I '0'^' CI^N^CI K2C03, DBU ^'N^N^CI NaOH rf™ N-n'^n^ci + ¥ CPI, DBU p-CS h2n o 0 0 | KyCO-j P~iJ " Cl p~Q NhJOXCIJjT Step A: utyl 3-[(2,2,3,3-teti amethylcyclopropyl)methoxy|pyrazole- 1-carboxylate OH n.naJ< +HO-^j Ph^P, DIAD nj To a degassed solution of PhT (approximately 51.28 g, 195.5 mmol) in toluene (360.0 mL) under nitrogen gas at 0 °C was added DIAD (diisopropylazodicarboxylate) (approximately 39.53 g, 37.86 mL, 195.5 mmol) dropwise. The mixture was stirred at 0 °C for 30 min affording a white slurry. To the mixture was added a solution of (2,2,3,3-tetramethylcyclopropyl)methanol (approximately 29.84 g of 70 %w/w, 162.9 mmol) and tert-butyl 3-hydroxypyrazole-lcarboxylate (30 g, 162.9 mmol) in toluene (600.0 mL) dropwise at ~5 °C over 2 hours.
The e was allowed to warm to ambient ature and stirred for 18 hours. The mixture was heated to 75 °C for a total of 6 hours and then allowed to cool to t temperature. The slurry was diluted with heptane (900.0 mL) and stirred at ambient temperature for 3 hours. The slurry was filtered over celite and the precipitate washed 3X with 100 mL of heptane. The filtrate was concentrated in vacuo affording a thick yellow oil. The crude t chromatographed on a 750 gram silica gel column loading SUBSTITUTE SHEET (RULE 26) WO 64632 with dichloromethane and eluting with a 0-20% hexanes gradient. Collected fractions containing product were concentrated in vacuo affording an off-white solid, tert-butyl 3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazole-l-carboxylate (30.1 g, 63%) was obtained. lH NMR (400 MHz, form-d) 5 7.82 (d, J = 3.0 Hz, 1H), 5.88 (d, J = 2.9 Hz, 1H), 4.30 (d, J = 7.7 Hz, 2H), 1.61 (s, 9H), 1.12 (s,6H), 1.04 (s, 6H), 0.70 (t, J = 7.8 Hz, 1H). ESI-MS m/z calc. 294.19434, found 295.0 (M+l)+; Retention time: 2.19 minutes Step B: 3-[(2,2,3,3-Tetramethylcyclopropyl)methoxy]-l H-pyrazole p-O NaOH p-£t To a solution of tert-butyl 3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazole-lcarboxylate (127 g, 431.4 mmol) in THE (317.5 mL) and ethyl alcohol (635.0 mL) was slowly added sodium hydroxide (approximately 431.4 mL of 2 M, 862.8 mmol) and stirred at room temperature overnight. Most of the solvent was removed under reduced pressure. The aqueous residue was d with water (400 mL) and extracted with methyl t-butyl ether (762.0 mL). The organic phase was washed twice with brine (2 x 300 mL) and the s phases were back extracted once with methyl t-butyl ether (250 mL). The combined organic phases were dried, filtered and evaporated to give 3- [(2,2,3,3-tetramethylcyclopropyl)methoxy]-lH-pyrazole (75 g, 89%) as a viscous oil. 'H NMR (400 MHz, DMSO-d6) 5 11.78 (s, 1H), 7.48 (t, J = 2.1 Hz, 1H), 5.65 (s, 1H), 4.05 (d, J = 7.7 Hz, 2H), 1.08 (s, 6H), 1.00 (s, 6H), 0.67 (t, J = 7.7 Hz, 1H). ESI-MS m/z calc. 194.1419, found 195.0 (M+l)+; Retention time: 1.43 minutes.
Step C: Ethyl 2-chloro [3- [(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylate ■ NH K2C03, DBU + % p-cyN. N Cl cr n ci SUBSTITUTE SHEET (RULE 26) To the ethyl 2,6-dichloropyridinecarboxylate (16.8 g, 76.35 mmol) and 3- [(2,2,3,3-tetramethylcyclopropyl)methoxy]-lH-pyrazole (approximately 14.83 g, 76.35 mmol) in DMF (201.6 mL) was added potassium carbonate (approximately 13.72 g, 99.26 mmol) followed by DABCO ximately 1.284 g. 11.45 mmol). The slurry was stirred at ambient temperature for 16 hours. The cream fine suspension was slowly diluted with water (201.6 mL), and the resulting thick slurry was d at ambient temperature for 30 minutes with an overhead stirrer. The precipitate was collected using an medium frit and washed 3 times with 25 mL of water. The solid was air dried for 30 s, and then dried in vacuo using an EtOAc azeotrope. Ethyl 2-chloro[3- [(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylate (28.8 g, 100%) was obtained as an off-white solid. ESI-MS m/z calc. 377.1506, found 378.37 (M+l)+; Retention time: 2.47 minutes, 'h NMR (400 MHz, DMSO-d6) 5 8.43 (dd, J = 2.9, 0.9 Hz, 1H), 8.39 (dd, J = 8.5, 0.9 Hz, 1H), 7.76 (dd, J = 8.5, 0.9 Hz, 1H), 6.24 (dd, J = 2.9, 0.9 Hz, 1H), 4.34 (td, J = 7.5, 6.6 Hz, 2H), 4.28 (d, J = 7.8 Hz, 2H), 1.34 (td, J = 7.1, 0.9 Hz, 3H), 1.11 (s, 6H), 1.05 (s, 6H), 0.75 (t, J = 7.8 Hz, 1H).
Step D: 2-Chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol- l-yl]pyridinecarboxylic acid O O N- N- ✓2 H P-QI N XI N Cl Ethyl 2-chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridine ylate (146 g, 386.4 mmol) in THE (730.0 mL) and EtOH (292.0 mL) was treated with NaOH ximately 772.8 mL of 1 M, 772.8 mmol) and the solution was stirred at room temperature for 5 hours. Most of the solvent was removed under reduced pressure, and the solution was acidified by addition of citric acid (approximately 148.5 g, 89.19 mL, 772.8 mmol) under ice cooling. The formed thick suspension (pH 2-3) was stirred in the ice bath for 1 hour, filtered, washed with plenty of water and dried in a drying cabinet under vacuum at 45 °C with a nitrogen bleed for two days to give 2- [3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridine carboxylic acid (128.2 g, 90%) as an off white solid. ESI-MS m/z calc. 349.11932, SUBSTITUTE SHEET (RULE 26) found 350.0 (M+l)+; Retention time: 2.11 minutes, 'll NMR (400 MHz, DMSO-d6) 5 13.64 (s, 1H), 8.69 - 8.22 (m, 2H), 7.73 (d, J = 8.4 Hz, 1H), 6.22 (d, J = 2.9 Hz, 1H), 4.28 (d, J = 7.8 Hz, 2H), 1.08 (d, J = 24.9 Hz, 12H), 0.75 (t, J = 7.8 Hz, 1H).
Step E: 2-Chloro-N-(o-tolylsulfonyl)[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide 0 9 9P "OH Qwp n:s is CPI, DBU H p^N N ■Cl + H2N P^N N XIN- 2-Chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg 0.429 mmol) and was dissolved/suspended in THF (2 mL), and carbonyl diimidazole (64.2 mg, 0.396 mmol) was added. The suspension was allowed to stir at room temperature for 1.5 hours. 2- benzenesulfonamide (73.4 mg, 0.429 mmol) was then added followed by DBU (59.2 pL, 0.396 mmol). The resulting on was then stirred for another 1.5 hours.
Volatiles were evaporated. The remaining residue was taken up in dichloromethane (2 mL) and washed with aqueous 1 M citric acid (1x2 mL). The organic layer was injected onto a silica gel column for chromatography: 12 gram silica gel column, 0-10% MeOH/DCM nt. 2-chloro-N-(o-tolylsulfonyl)[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (115 mg, 53%) was obtained. ESI-MS m/z calc. 502.14417, found 503.0 (M+l)+; Retention time: 2.25 minutes.
] Step F: N-(o-Tolylsulfonyl)[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrroli(lin-lyl ]pyridinecarboxamide ^1 HN'NS, K2CO3 vVvP P-^jN Cl p N SUBSTITUTE SHEET (RULE 26) 2-Chloro-N-methylsulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (115 mg, 0.229 mmol) and (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (106 mg, 0.935 mmol) were combined and dissolved in DMSO (1 mL). Finely ground potassium ate (258 mg, 1.87 mmol) was added. The reaction mixture was sealed and heated overnight at 130 °C. After g to room temperature, the reaction mixture was diluted with EtOAc (50 mL) and washed with aqueous citric acid (1 M, 2x 50 mL) and brine (1 x 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The product was isolated by silica gel column chromatography eluting with a 0-5% MeOH/DCM gradient on a 12 gram silica gel column. N-(o- Tolylsulfonyl)[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (57.2 mg, 42%) was obtained.
ESI-MS m/z calc. 579.2879, found 580.3 (M+l)+; Retention time: 2.52 minutes, fa NMR (400 MHz, DMSO-d6) 5 12.62 (s, 1H), 8.18 (d, J = 2.8 Hz, 1H), 8.04 (dd, J = 8.0, 1.4 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.59 (td, J = 7.5, 1.5 Hz, 1H), 7.50 - 7.40 (m, 2H), 6.93 (d, J = 8.2 Hz, 1H), 6.13 (d, J = 2.7 Hz, 1H), 4.24 (d, J = 7.8 Hz, 2H), 2.63 (s, 3H), 2.38 (d, J = 8.8 Hz, 2H), 2.16 (d, J = 10.3 Hz, 1H), 1.82 (dd, J = 11.9, 5.5 Hz, 1H), 1.52 (d, J = 1.6 Hz, 6H), 1.35 (t, J = 12.1 Hz, 1H), 1.10 (s, 6H), 1.04 (d, J= 1.1 Hz, 6H), 0.77 - 0.67 (m, 4H).
Synthetic e 16: Synthesis of Compound 16: N-(3- Fluorophenyl)sulfonyl[3-[(2,2,3>3-tetramethylcyclopropyl)methoxy]pyrazol-lyl 4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(3-fluorophenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide 0 o o 0 ‘OH u F GDI, DBU + h2n .voy©' p-XS N Cl 2-Chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0.429 mmol) was dissolved/suspended in THF (2 mL), and yl diimidazole (64.2 mg, 0.396 mmol) was added. The suspension SUBSTITUTE SHEET (RULE 26) was allowed to stir at room ature for 1.5 hours. 3-fluorobenzenesulfonamide (75.1 mg, 0.429 mmol) was then added followed by DBU (59.2 pL, 0.396 mmol). The resulting solution was then stirred for another 1.5 hours. Volatiles were evaporated.
The remaining residue was taken up in dichloromethane (2 mL) and washed with s 1 M citric acid (1x2 mL). The organic layer was injected onto a silica gel column to be purified by chromatography: 12 gram silica gel column, 0-10% MeOH/DCM gradient. 2-chloro-N-(3-fluorophenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (150 mg, 70%) was ed. ESI-MS m/z calc. 506.11908, found 507.0 (M+l)+; Retention time: 2.24 minutes Step B: N-(3-fluorophenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridmecarboxamide 9 oo o o 0 % Yxy. & K?CO, N' ■XCCp-' 2-Chloro-N-(3-fluorophenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (158 mg, 0.312 mmol), and ,2,4-trimethylpyrrolidine (Hydrochloride salt) (105.9 mg, 0.935 mmol) were ed and dissolved in DMSO (1 mL). Finely ground potassium carbonate (258 mg, 1.87 mmol) was added. The reaction mixture was sealed and heated overnight at 130 °C. After cooling to room temperature, the reaction mixture was diluted with EtOAc (50 mL) and washed with aqueous citric acid (1 M, 2x 50 mL) and brme (1 x 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced re. The product was isolated by column chromatography eluting with a 0-5% MeOH/DCM gradient on a 12 gram silica gel column. N-(3-fluorophenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide (28.4 mg, 16%) was obtained. ESI-MS m/z calc. 583.2629, found 584.6 (M+l)+; ion time: 2.46 minutes, 'H NMR (400 MHz, DMSO-d6) 8 12.61 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 7.87 - 7.81 (m, 2H), 7.79 - 7.71 (m, 2H), 7.63 SUBSTITUTE SHEET (RULE 26) (tdd, J = 8.6, 2.6, 1.1 Hz, 1H), 6.93 (d, J = 8.2 Hz, 1H), 6.13 (d, J = 2.8 Hz, 1H), 4.24 (d, J = 7.7 Hz, 2H), 2.44 (t, J = 10.4 Hz, 1H), 2.36 - 2.26 (m, 1H), 2.13 (td, J = 11.8, 6.0 Hz, 1H), 1.84 (dd, J= 11.8, 5.5 Hz, 1H), 1.54 (s, 3H), 1.52 (s, 3H), 1.39 (t, J= 12.1 Hz, 1H), 1.10 (s, 6H), 1.04 (s, 6H), 0.74 (d, J = 7.7 Hz, 1H), 0.70 (t, J = 6.6 Hz, 3H).
Synthetic Example 17: Synthesis of Compound 17: N-(Benzenesulfonyl)- 2- [(4S)-3,3-dideuterio-2,2-dimethyl(trideuteriomethyl)pyrrolidin-l-yl] [3- [2- [l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide VX AqsP o' "O. wCDs k2co3 p-^-N N F D D F F- F F F D D N-(Benzenesulfonyl)chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazoll-yl ]pyridinecarboxamide (2 g, 3.884 mmol) was dissolved inNMP (10.00 mL) and 1,2-diethoxy ethane (2.000 mL). Potassium carbonate (approximately 2.684 g, 19.42 mmol) and (4S)-3,3-dideuterio-2,2-dimethyl(trideuteriomethyl)pyrrolidine (Hydrochloride salt) ximately 1.502 g, 9.710 mmol) were added, and the resulting slurry heated was to 130 °C and stirred overnight. The reaction mixture was cooled and poured into rapidly stirred ice (60.00 mL) and acetic acid (approximately 3.499 g, 3.313 mL, 58.26 mmol). After stirring for 20 minutes to form a fairly uniform flowing solid, the solids were ed off and washed with water. The cake was dissolved in dichloromethane, and the resulting aqueous forced out was ted. The romethane layer was washed with water twice and brine and dried over sodium sulfate and concentrated. Ethanol (20 mL) was added, and the solution was concentrated to a few milliliters. Water was very slowly added dropwise. The suspension that formed was warmed to a thin suspension and allowed to cool over 30 minutes. Crystalline solids were filtered and washed with small amount of ethanol to give N-(benzenesulfonyl)[(4S)-3,3-dideuteno-2,2-dimethyl (trideuteriomethyl)pyrrolidin-l-yl][3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl]pyridinecarboxamide (600 mg, 26%).. ESI-MS m/z calc. 596.24, found 597.0 ; Retention time: 2.29 minutes.
SUBSTITUTE SHEET (RULE 26) Synthetic Example 18: Synthesis of Compound 18: N- (Benzenesulfonyl)[3-[(cis)(trifluoromethyl)cyclopropoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide n H °VN Ph3Rl DIAD 1) tBuOK.THF r w °^N. CF3CH2NH2HCI/NaN02/H2Q Br^0H NBoc °^N.r [[.NBoc nb°c 2) Boc20 Rh2(esp)2 N'NBoc N'NBoc N'NBoc N'NBoc f3c'-A,‘o^ + FsC^-'O^ and f3c'-A"o^ + F3C 'fp FjC^-'O^N'NBoc A N'NH A it /> ci N'N^N^CI C.0-U TFA DABCO, K2C03 TFA FjC''1 + ch2ci2 N'NBoc CHjCU i'o N'NH DMF f3cAA f3c 0-u jfi 0N'N^n^CI 0 X ?s"° f3c-<J 'OH F3C'"<J 5_^Sj^n ;| ^ 0 + h2n TO cdi.dbu + hn^A k2co3 f3c-<| OH A 'i"'0 °~ij.N-nAicI F'CA[°~v TO ^■QCwX %° F3C'"<] c.°-tJrJOTO *"0 AF ] Step A: te/7-Butyl romoethoxy)-li/-pyrazole-l-carboxylate D ^ Ph3P, DIAD /A/OH UVN^ Br r + NBoc Otj ] To the solution of 2-bromoethanol (1.69 g, 13.53 mmol), toV-butyl -2,3- dihydrooxopyrazole-l-carboxylate (2.08 g, 11.28 mmol) and triphenylphosphine SUBSTITUTE SHEET (RULE 26) (3.55 g, 13.53 mmol) in anhydrous tetrahydrofuran (45 mL) at 0 C diisopropyl azodicarboxylate (2.74 g, 13.53 mmol) was added dropwise. After the addition was te, the reaction solution was stirred at 0 °C for 1 hour, then warmed up to room temperature and stirred for additional 2 hours. Ether (400 mL) was added. The organic solution was washed with saturated sodium ate aqueous solution (80 mL), brine (50 mL), then dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue obtained was purified by silica gel chromatography using hexanes- ethyl acetate gradient method (0 to 15% ethyl acetate) to afford tert-butyl 3-(2- bromoethoxy^liZ-pyrazole-l-carboxylate (2.56 g, 78%) as white solid, 'll NMR (250MHz, CDCh) 6 (ppm): 7.85 (d, J= 3.0 Hz, 1H), 5.92 (d, 3.0 Hz, 1H), 4.63 (t,J = 6.0 Hz, 2H), 3.68 (t, J= 6.0 Hz, 2H), 1.64 (s, 9H). ESI-MS m/z calc. 292.0 found 292.9 /M+1)1, Retention time: 4.91 minutes.
Step B: fert-Butyl 3-(vinyloxy)-l//-pyiazole-l-cai boxylate 1) tBuOK.THF r O N, O. N v, 2) Boc20 NBoc To the solution of tert-butyl 3-(2-bromoethoxy)-lH-pyrazole-l-carboxylate (2.52 g, 8.66 mmol) in anhydrous tetrahydrofuran (90 mL) was added potassium tertbutoxide (1.46 g, 13.0 mmol). The resulting solution was stirred for 2 hours, then ditert-butyl dicarbonate (5.67 g, 26.0 mmol) and stirred for another 1 hour. Diethyl ether (400 mL) was added. c layers were washed with water (50 mL), brine (2 x 50mL), dried over dried over ium sulfate, filtered and concentrated under reduced pressure. The residue obtained was purified by silica gel chromatography using hexanes-ethyl acetate gradient method (0 to 10% ethyl acetate) to afford tert-butyl 3- (Yinylo\y)-l//-pyrazole-l-carbo\ylate (1.10 g, 60%) as colorless oil. 'h NMR (250MHz, CDC13) S (ppm): 7.89 (d, J= 3.0 Hz, 1H), 7.24 (dd, J= 6, 13.5 Hz, 1H), 5.95 (d, .7=3.0 Hz, 1H), 4.88 (dd, J= 1.8,13.5 Hz, 1H), 4.50 (dd,/= 1.8, 6.0 Hz, 1H), 1.62 (s, 9H). ESI-MS m/z calc. 210.1 found 211.0 (M+l)+ Retention time: 4.74 s.
Step C: tert-Butyl 3-(2-(trifluoromethyl)cyclopropoxy)-l//-pyrazole-l- ylate SUBSTITUTE SHEET (RULE 26) N'NBoc A N'NBoc CF3CH2NH2HCI/NaN02/H20 F3C' F3C 0n^N.
T NBoc N-NBoc and Rh2(esp)2 f3c^A'"o^ AA N'NBoc a} f3c"' "0'^/ utyl 3-(vinyloxy)-l//-pyrazole-l-carboxylate (1.10 g, 5.23 mmol) in pear-shape flask (100 mL) was added water (20 mL) and bubbled with argon for 5 minutes, then sodium acetate (85.8 mg, 1.05 mmol) was added followed by 2,2,2- oroethylamine hydrochloride (3.57 g, 26.17 mmol) and concentrated sulfunc acid (51.3 mg, 0.523 mmol). The solution was bubbled with argon for another 5 minutes before bis[rhodium(a,a,a',a'-tetramethyl-l,3-benzenedipropionic acid)] (397 mg, 0.523 mmol) was added. The reaction solution was kept under argon with balloon while s solution of sodium nitrite (2.17 g, 31.4 mmol) in water (12.8 mL) was added by syringe pump within 10 hours. After the addition was complete, the resulting solution was stirred for an additional 6 hours. Diethyl ether (300 mL) was added and the organic layer was separated. Then organic layer was washed with brine (30 mL), dried over magnesium sulfate, ed and concentrated under reduced re. The residue obtained was purified by silica gel chromatography using hexanes - dichloromethane gradient method (0 to 100% dichloromethane). The residue obtained was subjected to silica gel chromatography again (hexanes and ethyl acetate, 0 to 10% ethyl acetate gradient) to afford tert-butyl 3-(L2-trans(trifluoromethyl)cyclopropoxy)-li/- pyrazole-l-carboxylate and utyl 3-(l,2-cis(trifluoromethyl)cyclopropoxy) le-1 -carboxylate, /ert-buty 1 3-(1,2-trans(trifluoromethyl)cyclopropoxy)-\H- pyrazole-l-carboxylate: (366 mg, 24%); a white solid, 'fl NMR (250MHz, CDCfl) d (ppm): 7.84 (d, J = 2.8 Hz, 1H), 5.91 (d, 2.8 Hz, 1H), 4.49 (m, 1H), 1.75 (m, 1H), 1.62 (s, 9H), 1.56-1.25 (m, 2H). ESI-MS m/z calc. 292.1 found 293.1 (M+l)+.Retentiontime: 5,22 minutes. /677-butyl 3-(l.2-cis (trifluoromethyl)cyclopropoxy)pyrazole-l-carboxylate: (314mg, 21%); a white solid. ‘H NMR (250MHz, CDCfl) d (ppm): 7.90 (d, ./ = 2.8 Hz, 1H), 5.92 (d, ./ = 2.8 Hz, 1H), 4.49 (m, 1H), 1.94 (m, 1H), 1.62 (s, 9H), 1.30 (m, 2H). ESI-MS m/z calc. 292.1 found 293.1 (M+l)+ Retention time: 5 48 minutes.
Step D: 3-(L2-cis(Trifluoroinethyl)cyclopiopoxy)-l//-pyrazole SUBSTITUTE SHEET (RULE 26) N'NBoc A N'T f3c TFA f3c A N'T N-NBoc f3co‘ f3co- ] Trifluoroacetic acid (2.76 g, 24.3 mmol) was added to the solution of tertbutyl 3-(l,2-cis(trifluoromethyl)cyclopropoxy)-li7-pyrazole-l-carboxylate (708 mg, 2.43 mmol) in anhydrous dichloromethane (24 mL). The resulting solution was stirred at room temperature for 16 hours. 1,2-Dichloroethane (10 mL) was added to the reaction solution. All the solvents were removed under reduced re. The residue obtained was disolved in ethyl ether (150 mL), washed with satuated sodium bicarbonate aqueous on (30 mL). The organic solution was dried over magnesium sulfate, ed and concentrated under the reduced pressure to afford crude 3-(l,2-cis (trifluoromethyl)cyclopropoxy)-li7-pyrazole (461 mg, 99%) as yellow-brown oil. The crude product was used directly in next step without any further purification. ESI-MS m/z calc. 192.1 found 193.0 (M+l)+. Retention time: 3.26 minutes.
Step E: tert-Butyl 6-(3-(l,2-cis(trifluoromethyl)cyclopropoxy)-Lf7- pyrazol-l-yl)chloropyridinecarboxylate A N'NvH f3c 0 Cl -V DABCO, K2C03 + .0 O A N'NH Cl o f3c-<J '0 ^^^Hni^n^ci To the solution of crude -cis(trifluoromethyl)cyclopropoxy)-li7- pyrazole (461 mg, 2.43 mmol) in dimethylformarmde (8 mL) was added ferf-butyl 2,6- dichloropyridinecarboxylate (659 mg, 2.67 mmol), potassium carbonate (669 mg, 4.85 mmol) and 1,4-diazabicyclo [2.2.2]octane (55 mg, 0.49 mmol). The on was stirred at room temperature for 48 hours. The reaction solution was diluted with ether (200 mL), washed with water (4 x 20mL) and brine (20 mL). The organic layer was SUBSTITUTE SHEET (RULE 26) dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue obtained was purified by silica gel tography using hexanes - dichloromethane gradient method (0 to 100% dichloromethane) to afford tert-butyl 6- 2-cis(trifluoromethyl)cyclopropoxy)-l#-pyrazol-l-yl)chloropyridine carboxylate (731mg, 68%) as a white solid, ’tt NMR (250MHz, CDCh) S (ppm): 8.39 (d, J= 2.8 Hz, 1H), 8.22 (d, J= 8.5Hz, 1H), 7.74 (d, J= 8.5Hz, 1H), 6.01 (d, /= 2.8 Hz, 1H), 4.33 (m, 1H), 1.93(m, 1H), 1.62(s, 9H), 1.45-1.26(m, 2H). ESI-MS m/z calc. 403.1 found 404.1 . Retention time: 7.29 minutes.
Step F: 6-(3-(l,2-cis(Trifluoromethyl)cyclopropoxy)-li7-pyrazol-l-yl)- 2-chloropyridinecarboxylic acid O O 0 FX OH °-u °-u O O 0 f3c-<| OH F3C-0 ] Trifluoroacetic acid (2.03 g, 17.8 mmol) was added to the solution of tert- butyl 6-(3-(l,2-cis(trifluoromethyl)cydopropoxy)-l#-pyrazol-l-yl) chloropyridinecarboxylate (718 mg, 1.78 mmol) in anhydrous dichloromethane (18 mL). The resulting solution w as stirred at room temperature for 16 hours. 1,2- Dichloroethane (10 mL) was added to the reaction solution. All the solvents were removed under the reduced pressure. The crude solid obtained w as added 10% ethyl ether in hexanes (25 mL) and sonicated for 30 s, filtered, washed with 10% ethyl ether in hexanes (10 ml), hexances (10 mL) and dried under high vacumn to afford 6- (3-(l,2-cis(trifluoromethyl)cyclopropoxy)-li7-pyrazol-l-yl)chloropyridine carboxylic acid (517mg, 84%) as a white solid, 'h NMR (500MHz, DMSO) <5 (ppm): 13.6 (bs, 1H), 8.47 (d, J= 3.0 Hz, 1H), 8.42 (d, J= 8.8 Hz, 1H), 111 (d, J= 8.8 Hz, 1H), 6.27 (d, J= 3.0 Hz, 1H), 4.46 (m, 1H), 2.40 (m, 1H), 1.47 (m, 1H), 1.32 (m, 1H).
ESI-MS m/z calc. 347.0 found 347.9 (M+l)+. Retention time: 5.20 minutes.
SUBSTITUTE SHEET (RULE 26) Step G: N-(Benzenesulfonyl)chloro[3-[(cis) (trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyridinecarboxamide 9 qwp ‘OH p-Q* N Cl F> p-GN N Cl 9 Q.P Fv""<I QwP F F jr^OCPI, DBU TO P<J N p-Q N Cl pW pW F 6-(3-(l,2-Cis(trifluoromethyl)cyclopropoxy)-li/-pyrazol-l-yl) chloropyridinecarboxylic acid (125 mg, 0.360 mmol) was dissolved in THF (1 mL). l,r-Carbonyldiimidazole (75.6 mg, 0.431 mmol) was added. The reaction mixture was allowed to stir at room temperature for 1 hour, benzenesulfonamide (67.8 mg, 0.431 mmol) was added followed by DBU (64.5 pL, 0.431 mmol). The final reaction mixture was allowed to stir overnight at room temperature. Volatiles were removed by evaporation. It was taken up in EtOAc (50 mL) and washed with aqueous 1 M citric acid on (2x 50 mL) and brine (lx 50 mL). The organic layer was dried over sodium sulfate, ed and concentrated under reduced pressure. N-(benzenesulfonyl)- 2-chloro[3-[(cis)(trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyndine carboxarmde (201 mg) was ed. ESI-MS m/z calc. 486.03763, found 486.9 (M+l)+; Retention time: 0.67 minutes (1 minute run).
Step H; N-(Benzenesulfonyl)[3-[(cis) uoromethyl)cyclopropoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxainide SUBSTITUTE SHEET (RULE 26) WO 64632 CC'tJA^9 .nj p-0 N F HN"^ XXp9 Qp K?COs ^ 0 o 0 f4""<] F ..^'O P-U FW pW F N-(benzenesulfonyl)chloro[3-[(cis) (trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyridinecarboxamide (175 mg, 0.3595 mmol) was dissolved in DMSO (1 mL). (4S)-2,2,4-tnmethylpyrrolidine (Hydrochlonde salt) (161 mg, 1.08 mmol) was added followed by potassium carbonate (298 mg, 2.16 mmol). The reaction mixture was d to stir at 130 °C overnight. After cooling to room temperature, the reaction mixture was diluted with EtOAc (50 mL) and washed with s citric acid (1 M, 2x 50 mL) and brine (lx 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced re. The product was isolated by silica gel column chromatography on a 12 gram silica gel column eluting with a 0-10% EtOAc/hexane gradient. N-(benzenesulfonyl)[3-[(cis)- 2-(trifluoromethyl)cyclopropoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide (114.3 mg, 56%) was obtained. ESI-MS m/z calc. 563.1814, found 564.5 (M+l)+; Retention time: 2.08 minutes tic Example 19: Synthesis of Compound 19: N- (Benzenesulfonyl)[3-[(trans)(trifluoromethyl)cyclopropoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) F3C'"^Sy^N-NBoc A N'N\H F3C'"<3 fl° F3C"' c, TFA DABCO, K2C03 0-<^N N , U'bo CH2CI2 DMF CH2CI2 N-NBoc Cl 0 FaC^-tA^ FsC^-'O^A N-NH F3c-<q .N'N^N^-CIjTi 0 »£&A9sP F3C'"<^ jCtOH °-u^ ^naci 'O 0 H!N9s°/\' x> CPI, DBU hn'AS. k2co3 F3CK] J2&PAPsP A9sP >rv-XJ Anj IT h Step A: 3-(l,2-trans(Trifluoromethyl)cyclopropoxy)-l//-pyrazole N-NBoc ^\A N'xqjCj> f3c TFA f3c ^•.,a n-NBoc0aa ^•VA N'TqA/ f3c f3c Trifluoroacetic acid (3.15 g, 27.64 mmol) was added to the solution of lerl- butyl -trans(trifluoromethyl)cyclopropoxy)-li7-pyrazole-l-carboxylate (807 mg, 2.76 mmol) in anhydrous dichloromethane (28 mL). The resulting solution was stirred at room temperature for 16 hours. 1,2-Dichloroethane (15 mL) was added to the reaction solution. All the solvents were removed under the reduced pressure. The residue obtained was disolved in ethyl ether (200 mL), washed with satuated sodium onate aqueous solution (30 mL). The organic on was dried over magnesium e, filtered and concentrated under reduced pressure to afford crude 3-(l,2-trans SUBSTITUTE SHEET (RULE 26) (trifluoromethyl)cyclopropoxy)-l#-pyrazole (525mg, 99%) as yellow-brown oil. The crude product was used directly in next step without any further purification. ESI-MS mfz calc. 192.1 found 193.0 /M+l ) . Retention time: 2.97 minutes.
Step B: tert-Butyl 6-(3-(l,2-trans(trifluoromethyl)cyclopropoxy)-l//- l-l-yl)chloropyridinecarboxylate F3C''A>o^A N'NxH FoC" ?0 Cl -V °-u I DABCO, K2CO3 + .0 DMF 0 A N^H Cl 0 f3c-<| ‘0 b-AN^N^CI To the solution of crude 3-(l,2-trans(trifluoromethyl)cyclopropoxy) pyrazole (525 mg, 2.76 mmoL) in ylformamide (9.2 mL) was added fert-butyl 2,6-dichloropyridinecarboxylate (751 mg, 3.04 mmol), potassium carbonate (763 mg, .53 mmol) and 1,4-diazabicyclo [2.2.2]octane (62 mg, 0.55 mmol). The reaction was stirred at room temperature for 48 hours. The reaction solution was diluted with ether (250 mL), washed with water (4 x 20 mL) and brine (20 mL). The organic layer was dried over magnesium sulfate, filtered and concentrated under reduced pressure. The residue obtained was purified by silica gel chromatography using s - dichloromethane gradient method (0 to 100% dichloromethane) to afford tert-butyl 6-(3- rans(trifluoromethyl)cyclopropoxy)-li7-pyrazol-l-yl)chloropyridine ylate (314 mg, 21%) as a colorless oil. ESI-MS mfz calc. 403.1 found 404.1 (M+l)+. Retention time: 6.92 minutes, 'll NMR (250MHz, CDCf ) 5 (ppm): 8.38 (d, J = 3.0 Hz, 1H), 8.20 (d, T= 8.5 Hz, 1H), 7.73 (d, J= 8.5 Hz, 1H), 6.03 (d, 7 = 3.0 Hz, 1H), 4.39 (m, 1H), 1.77 (m, 1H), 1.62 (s, 9H), 1.44 (m, 1H), 1.31 (m, 1H).
Step C: 6-(3-(l,2-Trans(trilluoromethyl)cyclopropoxy)-l/f-pyrazol-l- yl)chloropyridinecarb oxylic acid SUBSTITUTE SHEET (RULE 26) 1 F3C"' <1 , OH F3C1" 0 b~fjNv N Cl f3c-<1 'OH F5C^0 '0 b-0NL N Cl N Cl Trifluoroacetic acid (2.39 g, 21.0 mmol) was added to the solution of tert- butyl l,2-trans(trifluoromethyl)cyclopropoxy)-li/-pyrazol-l-yl) chloropyridinecarboxylate (847 mg, 2.10 mmol) in anhydrous dichloromethane (21 mL). The resulting solution was stirred at room temperature for 20 hours. 1,2- Dichloroethane (15 mL) was added to the reaction mixture. All the solvents were removed under reduced pressure. Crude solid obtained was added 10% ethyl ether in hexanes (30 mL) and sonicated for 30 minutes, filtered, washed with 10% ethyl ether in hexanes (10 mL), hexances (10 mL) and dried under high vacumn to afford 6-(3-(l,2- trans(trifluoromethyl)cyclopropoxy)-li:/-pyrazol-l-yl)chloropyridinecarboxylic acid (600 mg, 82%) as a white solid. ESI-MS m/z calc. 347.0 found 347.9 (M+l)+.
Retention time: 4.91 minutes, 'll NMR z, DMSO) S (ppm): 8.46 (d, J= 2.8 Hz, 1H), 8.41 (d, J= 8.3 Hz, 1H), 7.74 (d, J= 8.3 Hz, 1H), 6.30 (d, 7 = 2.8 Hz, 1H), 4.46 (m, 1H), 2.15 (m, 1H), 1.40 (m, 1H), 1.34 (m, 1H).
Step D: zenesulfonyl)chloro[3-[(trans) (trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyridinecarboxainide O F ‘OH F V <Lo N'N iT'CI X H // ^ N' 'Cl CPI, DBU 0 F F ‘OH 9 %D P-^N N^CI <[ N 4'"<f b^QN N 'Cl 6-(3-(l,2-trans(Trifluoromethyl)cyclopropoxy)-l/f-pyrazol-l-yl) pyridinecarboxylic acid (125 mg, 0.360 mmol) was dissolved in THF (1 mL).
SUBSTITUTE SHEET (RULE 26) l,r-Carbonyldiimidazole (75.6 mg, 0.431 mmol) was added. The on e was allowed to stir at room temperature for 1 hour. Benzenesulfonamide (67.8 mg, 0.431 mmol) was added followed by DBU (64.5 pL, 0.431 mmol). The final on mixture was allowed to stir overnight at room temperature. Volatiles were removed by evaporation. It was taken up in EtOAc (50 mL) and washed with s 1 M citric acid solution (2x 50 mL) and brine (lx 50 mL). The c layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. N-(benzenesulfonyl)- 2-chloro[3-[(trans)(trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyridine amide (199 mg) was obtained. ESI-MS m/z calc. 486.0, found 486.9 (M+l)+; Retention time: 0.65 minutes (1 minute run) Step E: N-(Benzenesulfonyl)[3-[(trans) (trifluoromethyl)cyclopropoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide 9 q„p AQsP^ 's E F ,,H 'O R F N'b F F H ,,O^N N' "Jl HN-^S, K,CCK o o 0 9 Qwp R F ^*0 R F F^V F-V vao N-(Benzenesulfonyl)chloro [3 - [(trans) (trifluoromethyl)cyclopropoxy]pyrazol-l-yl]pyridinecarboxamide (175 mg, 0.3595 mmol) was dissolved in DMSO (1 mL). (4S)-2,2,4-Trimethylpyrrolidme (Hydrochloride salt) (161 mg, 1.08 mmol) was added followed by potassium carbonate (298 mg, 2.16 mmol). The reaction mixture was allowed to stir at 130 °C overnight.
After cooling to room temperature, the reaction mixture was diluted with EtOAc (50 mL) and washed with aqueous citric acid (1 M, 2x 50 mL) and brine (lx 50 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The product was ed by silica gel column chromatography on a 12 gram silica gel column eluting with a 0-10% EtOAc/hexane gradient. N-(benzenesulfonyl) [3-[(trans)(trifluoromethyl)cyclopropoxy]pyrazol-l-yl][(4S)-2,2,4- SUBSTITUTE SHEET (RULE 26) trimethylpyrrolidin-l-yl]pyridinecarboxamide (115.7 mg, 57%) was obtained. ESI- MS tn/z calc. 563.1814, found 564.5 (M+l)+; Retention time: 2.01 minutes Synthetic Example 20: Synthesis of Compound 20: N-(2- Hydroxyphenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl 4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(2-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide °vw,0;s 0 9 o,,o ?H 'OH N'S •% <^{2 N Cl GDI <,0^J N C, F3C f3c A solution of 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (181 mg, 0.5 mmol) and yldiirmdazole (approximately 97.3 mg, 0.60 mmol) in DMF (2.5 mL) was stirred for 30 minutes. A solution of 2-hydroxybenzenesulfonamide (approximately 113 mg, 0.65 mmol) and sodium hexamethyldisilazide (approximately 600 pL of 1 M, 0.60 mmol) in DMF (2.5 mL) was stirred for 30 minutes. The two solutions were combined and stirred for 15 h at room temperature. The reaction mixture was ied with lOmL 1 M aqueous citric acid, and extracted with 10 mL ethyl e. The combined extracts were dried over sodium sulfate and concentrated under reduced pressure. The crude material was purified by silica gel tography eluting with a 0-5% gradient of methanol in dichloromethane to give 2-chloro-N-(2- hy droxypheny onyl [3-[ [ 1 -(trifluoromethyl)cy clopropy 1] methoxy] py razol-1 - yl]pyridinecarboxamide (82 mg, 32%) ESI-MS mlz calc. 516.0, found 517.2 (M+l) +; ion time: 0.67 minutes.
Step B: N-(2-Hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrroli(lin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) 0 00 OH 0 qp OH K2C03 N'S N " Cl (S f3c HN 2-Chloro-N-(2-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (82 mg, 0.16 mmol), (4S)-2,2,4-tnmethylpyrrolidine (Hydrochloride salt) (approximately 71 mg, 0.48 mmol), and potassium carbonate (approximately 132 mg, 0.95 mmol) were combined in DMSO (793 pL) and heated at 130 °C for 15 h. The reaction was filtered and purified using a e phase S method using a Luna C18 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00CU0-AX), and a dual gradient run from 30-99% mobile phase B over 15.0 minutes [mobile phase A = H20 (5 mM HCI); mobile phase B = acetonitrile; flow rate = 50 mL/min, and column ature = 25 °C] to give N-(2-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-tnmethylpyrrolidin- I -yl |py ridinecarbo\amide (44 mg, 46%) ESI-MS m/z calc. 593.2, found 594.3 (M+l) +; Retention time: 2.07 s.
Synthetic Example 21: Synthesis of Compound 21: N-(3- Hydroxyphenyl)sulfonyl[3-[[l-(trifluoromethyl)cydopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(3-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cydopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide Qs ,p:s OH 9 o. ,o %• OH N'b' N. GDI N A H I <LP-ij N Cl N Cl DBU OH F3C f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (181 mg, 0.50 mmol) and carbonyl diimidazole (approximately 97 mg, 0.60 mmol) in DMF (2.5 mL) was d for 30 minutes. 3- SUBSTITUTE SHEET (RULE 26) WO 64632 hydroxybenzenesulfonamide (approximately 113 mg, 0.65 mmol) andNaH (approximately 24.0 mg of 60 %w/w, 0.60 mmol) in DMF (2.5 mL) was stirred for 30 minutes. The two solutions were combined and stirred for 4 h at room temperature. The reaction mixture was acidified with lOmL 1 M aqueous citric acid, and extracted with mL ethyl acetate. The combined extracts were dried over sodium sulfate and concentrated under d pressure. The crude material was purified by silica gel chromatography eluting with a 0-8% gradient of methanol in dichloromethane to give 2- chloro-N-(3-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (250 mg, 97%) E SI-MS m/z calc. 516.0482, found 517.2 (M+l) +; ion time: 0,67 minutes.
] Step B: N-(3-Hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 9P 0 00*!'/ N'S K2CO3 N'S 'jLP-iJ N OH OH f3c (S f3c 2-Chloro-N-(3-hydroxyphenyl)sulfonyl[3-[[l- (tnfluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyndinecarboxamide (290 mg, 0.56 mmol), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (approximately 252 mg, 1.68 mmol), and potassium carbonate (approximately 465 mg, 3.37 mmol) in DMSO (2.80 mL) was heated at 130 °C for 15 h. The reaction was filtered and purified using a reverse phase HPLC-MS method using a Luna Cl 8 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00CU0-AX), and a dual gradient run from 30-99% mobile phase B over 15.0 minutes [mobile phase A = HiO (5 mM HC1); mobile phase B = acetonitrile; flow rate = 50 mL/min, and column temperature = 25 °C] to give ydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide (37 mg, 11%) ESI-MS m/z calc. 593. 2, found 594.3 (M+l) +; Retention time: 1.98 minutes.
SUBSTITUTE SHEET (RULE 26) Synthetic Example 22: Synthesis of Compound 22: N-(4- Hydroxyphenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide 'OH II 0^°:s 'OH N' Ss GDI <a^0-O H Cl S? F3C f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (181 mg, 0.50 mmol) and carbonyl diimidazole (approximately 97 mg, 0.60 mmol) in DMF (2.5 mL) was stirred for 30 minutes. 4- hydroxybenzenesulfonamide (approximately 113 mg, 0.65 mmol) andNaH (approximately 24.0 mg of 60 %w/w, 0.60 mmol) in DMF (2.5 mL) was stirred for 30 minutes. The two ons were combined and stirred for 4 h at room temperature. The reaction mixture was acidified with lOmL 1 M aqueous citric acid, and extracted with mL ethyl acetate. The combined extracts were dried over sodium sulfate and concentrated under reduced pressure. The crude material was purified by silica gel chromatography eluting with a 0-8% gradient of methanol in dichloromethane to give 2- -N-(4-hydroxyphenyl)sulfonyl[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (210 mg, 81%) ESI-MS m/z calc. 516.0, found 517.2 (M+l) +; Retention time: 0.64 s.
Step B: N-(4-Hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]inethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 9P 0 00 S' K2C03 N'S H N-^ N Cl ‘OH WMJ N ‘OH f3c F3C SUBSTITUTE SHEET (RULE 26) 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (220 mg, 0.42 mmol), ,2,4-tnmethylpyrrolidine (Hydrochloride salt) (approximately 191 mg, 1.28 mmol), and potassium carbonate (approximately 353 mg, 2.56 mmol) in DMSO (2.13 mL) was heated at 130 °C for 15 h. The reaction was filtered and ed using a e phase HPLC-MS method using a Luna Cl 8 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00CU0-AX), and a dual gradient run from 30-99% mobile phase B over 15.0 minutes [mobile phase A = HiO (5 mM HC1); mobile phase B = acetonitrile; flow rate = 50 mL/min, and column temperature = 25 °C] to give N-(4-hydroxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazo l-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide (48 mg, 19%) ESI-MS m/z calc. 593.2, found 594.3 (M+l) +; Retention time: 1.98 minutes.
Synthetic Example 23: Synthesis of Compound 23: N-(o-Tolylsulfonyl)- 6- [3- [ [ l-(trifluoromethyl)cyclopropyl] methoxy] pyrazol- l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: ro-N-(o-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide SO2NH2 0 0 o, 0 N'S ■A- vs-O'w 'Cl^5 GDI 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.5529 mmol) and CDI ximately 107.6 mg, 0.6635 mmol) were combined in THE (960 uL) and stirred at room ature for 2 hours. 2-methylbenzenesulfonamide (approximately 123.1 mg, 0.7188 mmol) was added followed by DBU (approximately 101.0 mg, 99.21 pL. 0.6635 mmol) and the reaction was stirred for an additional 16 h at room temperature. AIM citric acid solution (1 mL) was added and the reaction was stirred for 20 min. The resulting solid was collected by vacuum filtration (washing with water) and dried under vacuum to SUBSTITUTE SHEET (RULE 26) give a white powder, which was used in the next step without further purification. 2- chloro-N-(o-tolylsulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (280 mg, 98%) ESI-MS m/z calc. 514.1, found 515.1 (M+l)+; Retention time: 0.73 minutes. lH NMR (400 MHz, DMSO) 5 d 13.56 - 12.55 (s, 1H), 8.42 (d, J = 2.8 Hz, 1H), 8.12 (d, J = 8.3 Hz, 1H), 8.04 (d, J = 7.9 Hz, 1H), 7.71 (d, J = 8.3 Hz, 1H), 7.63 (t, J = 7.6 Hz, 1H), 7.47 (t, J = 7.6 Hz, 2H), 6.23 (d, J = 2.9 Hz, 1H), 4.39 (s, 2H), 2.64 (s, 3H), 1.12-1.06 (m, 4H).
Step B: N-(o-Tolylsulfonyl) [3- [ [l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 q n 9 Q, .0 :s: HN HCI %■ N's' H (S) K2CO3 f3c f3c 2-Chloro-N-(o-tolylsulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (114.7 mg, 0.2227 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (100 mg, 0.6682) and, K2CO3 (184.6 mg, 1.336 mmol) were combined in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours.
After cooling to room temperature, the reaction mixture was diluted with 20 mL of ethyl acetate, and 10 mL water and transferred to a separatory funnel. AN aqueous 15 mL 1 M citric acid was added, and the organic layer was separated. The aqueous layer was extracted two onal times with 15 mL ethyl acetate, and the combined organics were washed with bnne, dried over sodium sulfate and concentrated. The resulting crude material was purified by flash chromatography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to give olylsulfonyl)[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrohdinl-yl inecarboxamide (101 mg, 77%). ESI-MS m/z calc. 591.21, found 592.3 (M+l)+; Retention time: 2.22 minutes, 'h NMR (400 MHz, DMSO) 5 12.74 (s, 1H), 8.38 (t, J = 1.7 Hz, 1H), 8.33 - 8.22 (m, 2H), 8.21 (d, J = 2.8 Hz, 1H), 7.90 (d, J = 7.9 Hz, 1H), 7.89 - 7.85 (m, 1H), 6.93 (d, J = 8.3 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.41 - 4.31 (m, 2H), 3.36 - 3.29 (m, 3H), 2.40 (t, J = 10.4 Hz, 1H), 2.27 (t, J = 8.6 Hz, 1H), SUBSTITUTE SHEET (RULE 26) WO 64632 2.11 (tt,J = 12.1, 6.3 Hz, 1H), .81 (m, 1H), 1.53 (d, J = 9.8Hz, 6H), 1.39(1, J = 12.1 Hz, 1H), 1.09 (dt, J = 6.7, 2.2 Hz, 4H), 0.68 (d, J = 6.2 Hz, 3H).
Synthetic Example 24: Synthesis of Compound 24: N-(p-Tolylsulfonyl)- 6- [3- [ [ l-(trifluoromethyl)cyclopropyl] methoxy] pyrazol-l-yl] [(4S)-2,2,4- trimethylpyrroMin-l-ylJpyridinecarboxamide Step A: 2-Chloro-N-(p-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide SO2NH2 o IhrCfrA V 'OH GDI XX 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.5529 mmol) and CDI (approximately 107.6 mg, 0.6635 mmol) were combined in THE (1.200 mL) and stirred at room temperature for 2 hours. 4-methylbenzenesulfonamide (approximately 123.1 mg, 0.7188 mmol) was added followed by DBU ximately 101.0 mg, 99.21 pL, 0.6635 mmol) and the reaction was stirred for an additional 16 h at room temperature. The reaction mixture was diluted with 1M aqueous citric acid and water, then extracted 3x 20 mL ethyl acetate. The combined organics were w ashed with 10 mL 1M citric acid, followed by brine, dried over sodium sulfate and concentrated, then purified by silica gel chromatography, eluting with 0-10% methanol/dichloromethane to give 2-chloro-N-(ptolylsulfonyl )[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridine carboxamide (262 mg, 92%) ESI-MS m/z calc. 514.0689, found 515.1 ; ion time: 0.74 minutes.
Step B: N-(p-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]inethoxy]pyrazol-l-yl][(4S)-2,2,4- hylpyrrolidin-l-yl]pyridinecarboxamide 9 V 9 Qp N'b' HN HCI N's yj-O N ‘Cl f3c k2co3 3C ' SUBSTITUTE SHEET (RULE 26) 2-chloro-N-(p-Tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (114.7 mg, 0.2227), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (100 mg, 0.6682) and, K2CO3 (184.6 mg, 1.336 mmol) were combined in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours. After cooling to room ature, the reaction mixture was diluted with 20 mL of ethyl acetate, and 10 mL water and erred to a separatory funnel. An aqueous 15 mL 1 M citric acid was added, and the organic layer was ted. The aqueous layer was extracted two additional times with 15 mL ethyl acetate, and the ed organics were washed with brine, dried over sodium sulfate and concentrated. The resulting crude al was purified by flash chromatography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to give N-(p- tolylsulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (65 mg, 49%). ESI-MS m/z calc. 591.21, found 592.3 (M+l)+; Retention time: 2.25 minutes, 'h NMR (400 MHz, DMSO) 5 12.74 (s, 1H), 8.38 (t, J = 1.7 Hz, 1H), 8.33 - 8.22 (m, 2H), 8.21 (d, J = 2.8 Hz, 1H), 7.90 (d, J = 7.9 Hz, 1H), 7.89 - 7.85 (m, 1H), 6.93 (d, J = 8.3 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.41 - 4.31 (m, 2H), 3.36 - 3.29 (m, 3H), 2.40 (t, J = 10.4 Hz, 1H), 2.27 (t, J = 8.6 Hz, 1H), 2.11 (tt, J = 12.1, 6.3 Hz, 1H), 1.88 - 1.81 (m, 1H), 1.53 (d, J = 9.8 Hz, 6H), 1.39 (t, J = 12.1 Hz, 1H), 1.09 (dt, J = 6.7, 2.2 Hz, 4H), 0.68 (d, J = 6.2 Hz, Synthetic Example 25: Synthesis of Compound 25: N-(3- Cyanophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(3-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide NCv^S02NH2U O 9 °,,o N''5' .CN o^Hr-N-'c, N Cl SUBSTITUTE SHEET (RULE 26) 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.5529 mmol) and CDI (approximately 107.6 mg, 0.6635 mmol) were combined in THF (1.200 mL) and d at room temperature for 2 hours. 3-cyanobenzenesulfonamide (approximately 131.0 mg, 0.7188 mmol) was added followed by DBU (approximately 101.0 mg, 99.21 pL. 0.6635 mmol) and the reaction was d for an additional 16 h at room temperature. The reaction mixture was diluted with 1M aqueous citric acid and water, and extracted 3x 20 mL ethyl acetate. The combined organics were washed with 10 mL 1M citric acid, followed by brine, then dried over sodium e and concentrated and used in the next step without further purification. 2-chloro-N-(3-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (228 mg, 78%) ESI-MS m/z calc. 525.0485, found 526.0 (M+l)+; Retention time: 0.7 minutes.
Step B: N-(3-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide HCI ifVV’Y'YCN F3c K2CO3 ro-N-(3-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (117.1 mg, 0.2227), (4S)-2,2,4-tnmethylpyrrolidine (hydrochloride salt) (100 mg, 0.6682) and, K2CO3 (184.6 mg, 1.336 mmol) were ed in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours. After cooling to room ature, the reaction mixture was diluted with 20 mL ethyl acetate, and 10 mL water and transferred to a separatory funnel. An aqueous 15 mL 1 M citric acid was added, and the organic layer was separated. The aqueous layer was extracted two additional times with 15 mL ethyl acetate, and the combined organics were washed with brine, dried over sodium sulfate and trated. The resulting crude material was purified by flash tography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to give N-(3- cyanophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide, (73 mg, 54%) ESI-MS m/z calc. 602.19, found 603.3 (M+l)+; Retention time: 2.04 minutes, 'll NMR (400 MHz, DMSO) 6 12.74 (s, 1H), 8.38 (t, J = 1.7 Hz, 1H), 8.30 (ddd, J = 8.1,1.9, 1.1 Hz, SUBSTITUTE SHEET (RULE 26) 1H), 8.24 (dt, J = 7.8, 1.3 Hz, 1H), 8.21 (d, J = 2.8 Hz, 1H), 7.92 - 7.84 (m, 2H), 6.93 (d, J = 8.3 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.42 - 4.31 (m, 2H), 2.40 (t, J = 10.4 Hz, 1H), 2.27 (t, J = 8.6 Hz, 1H), 2.11 (tt, J = 12.1, 6.3 Hz, 1H), 1.89 - 1.78 (m, 1H), 1.53 (d,J = 9.8 Hz, 6H), 1.39 (t, J = 12.1 Hz, 1H), 1.09 (dt, J = 6.7, 2.2 Hz, 4H), 0.68 (d, J = 6.2 Hz, Synthetic Example 26: Synthesis of Compound 26: N-(2- Cyanophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: ro-N-(2-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide S02NH2 o q q cn GDI I Vv°<JN N Cl 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyndinecarboxylic acid (200 mg, 0.5529 mmol) and CDI (approximately 107.6 mg, 0.6635 mmol) were combined in THE (1.200 mL) and stirred at room temperature for 2 hours. 2-Cyanobenzenesulfonamide (approximately 131.0 mg, 0.7188 mmol) was added followed by DBU (approximately 101.0 mg, 99.21 pL, 0.6635 mmol) and the reaction was d for an additional 16 h at room ature. AIM citric acid solution (1 mL) was added and the reaction was stirred for 20 minutes. The resulting solid precipitate was collected by vacuum filtration (washing with water) to give a white solid, which was dried on under vacuum and used in the next step without further cation, 2-chloro-N-(2-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (279 mg, 96%) ESI-MS m/z calc. 525.0485, found 526.1 (M+l)+; Retention time: 0.69 minutes. 1H NMR (400 MHz, DMSO) 8 12.23 (s, 1H), 8.49 (d, J = 2.9 Hz, 1H), 8.46 - 8.39 (m, 1H), 8.35 (d, J = 8.3 Hz, 1H), 8.21 - 8.13 (m, 1H), 7.96 - 7.90 (m, 2H), 7.82 (d, J = 8.3 Hz, 1H), 6.27 (d, J = 2.9 Hz, 1H), 4.42 (s, 2H), 1.11 (dt, J = 7.6, 2.2 Hz, 4H).
SUBSTITUTE SHEET (RULE 26) Step B: N-(2-Cyanophenyl)sulfonyl[3-[[l- (trifliioromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 0 q ,o CN 0 q o cn N:s' HN HCI s: tS) N' N Cl ys® N'^ f3c k2co3 2-Chloro-N-(2-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (117.1 mg, 0.2227), (4S)-2,2,4-tnmethylpyrrolidine (hydrochloride salt) (100 mg, 0.6682) and, K2CO3 (184.6 mg, 1.336 mmol) were combined in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours. After g to room temperature, the reaction mixture was diluted with 20 mL ethyl acetate, and 10 mL water and transferred to a separatory funnel. An s 15 mL 1 M citric acid solution was added, and the organic layer was separated. The aqueous layer was ted two additional times with mL ethyl acetate, and the combined cs were washed with brine, dried over sodium sulfate and concentrated. The ing crude material was purified by flash chromatography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to give. yanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide, (41 mg, 31%) ESI-MS m/z calc. 602.19, found 603.2 (M+l)+; Retention time: 2.12 minutes. 1HNMR (400 MHz, DMSO) 5 11.77 (s, 1H), 8.47 (s, 1H), 8.29 (d, J = 2.8 Hz, 1H), 8.17-8.11 (m, 1H), 8.06 (d, J = 8.3 Hz, 1H), 7.94 - 7.87 (m, 2H), 6.97 (d, J = 8.3 Hz, 1H), 6.20 (d, J = 2.8 Hz, 1H), 4.44 - 4.32 (m, 2H), 3.07 - 2.91 (m, 2H), 2.32 (d, J = 19 0 Hz, 1H), 1.98 (q, J = 5.9, 5.5 Hz, 1H), 1.67 (s, 3H), 1.63 (s, 3H), 1.57 (t, J = 10.4 Hz, 1H), 1.13 - 1.06 (m, 4H), 1.02 (d, J = 6.3 Hz, Synthetic Example 27: Synthesis of Compound 27: N-(4- Cyanophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) jaSO2NH2 0 NC x v 'OH GDI 0 /,N'N"k|/kCI Y-P^N'N iX-xxCN f3c f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0 4147 mmol) and CDI (81 mg, 0.4995 mmol) were combined in THF (900.0 pL) and stirred at room temperature for 2 hours. 4- cyanobenzenesulfonamide (98 mg, 0.5379 mmol) was added followed by DBU (75 pL, 0.5015 mmol) and the reaction was stirred at room temperature for 2 hours. Additional DBU (80 pL, 0.5350 mmol) was added, and the reaction was stirred for one additional hour at room temperature. The reaction mixture was diluted with 20 mL of a 1M citric acid solution and water and ted 3x 20 mL ethyl acetate. The combined organics were washed with 10 mL 1M citric acid, followed by brine, then dried over sodium sulfate and concentrated. The resulting material was further punfied by silica gel chromatography g with a 0-10% gradient of methanol in dichloromethane, to give a white solid; 2-chloro-N-(4-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyndinecarboxamide (192 mg, 88%) ESI-MS m/z calc. 525.0485, found 526.0 (M+l)+; Retention time: 0.71 s.
Step B: N-(4-Cyanophenyl)sulfonyl[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- hylpyrrolidin-l-yl]pyridinecarboxamide N CN f3c K2CO3 f3c ] 2-Chloro-N-(4-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (117.1 mg, 0.2227), (4S)-2,2,4-tnmethylpyrrolidine (hydrochloride salt) (100 mg, ) and, K2CO3 (184.6 mg, 1.336 mmol) were combined in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours. After cooling to room temperature, the reaction mixture was diluted with 20 mL ethyl acetate, and 10 mL water and transferred to a separatory funnel. An aqueous 15 mL 1 M citric acid solution was added, and the SUBSTITUTE SHEET (RULE 26) WO 64632 organic layer was separated. The aqueous layer was ted two additional times with mL ethyl acetate, and the ed organics were washed with brine, dried over sodium sulfate and concentrated. The resulting crude material was purified by flash chromatography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to giveN-(4-cyanophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide, (62 mg, 46%) ESI-MS m/z calc. 602.19, found 603.3 (M+l)+; Retention time: 2.04 minutes. 'H NMR (400 MHz, DMSO) 5 12.77 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 8.16 (s, 4H), 7.87 (d, J = 8.3 Hz, 1H), 6.93 (d, J = 8.3 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.50 - 4.17 (m, 2H), 2.33 (t, J = 10.3 Hz, 1H), 2.20 (dd, J = .2, 6.9 Hz, 1H), 2.11 (tt, J= 11.9, 6.4 Hz, 1H), 1.83 (dd, J = 11.8, 5.4 Hz, 1H), 1.52 (d, J = 5.6 Hz, 6H), 1.37 (t,J = 12.1 Hz, 1H), 1.13-1.05 (m, 4H), 0.66 (d, J = 6.2Hz, tic Example 28: Synthesis of Compound 28: N-(m-Tolylsulfonyl)- 6- [3- [ [ l-(trifluoromethyl)cydopropyl] methoxy] pyrazol-l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide ] Step A: 2-Chloro-N-(m-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide SO2NH2 0 9 <U>xc ’OH GDI O rTNC|H 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.5529 mmol) and CDI (approximately 107 6 mg, 0.6635 mmol) were combined in THE (964.9 pL) and stirred at room temperature for 2 hours. 3-methylbenzenesulfonamide (approximately 123.1 mg, 0.7188 mmol) was added followed by DBU (approximately 101.0 mg, 99.21 pL, 0.6635 mmol) and the reaction was stirred for an additional 16 h at room temperature. The reaction mixture was diluted with a 1M aqueous citric acid solution and water, and extracted 3x 20 mL ethyl acetate. The combined cs were washed with 10 mL 1M citric acid, followed by brine, dried over sodium sulfate, concentrated, and finally purified by silica gel chromatography eluting with 0-10% methanol/dichloromethane to give a white solid, 2- SUBSTITUTE SHEET (RULE 26) chloro-N-(m-tolylsulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl]rnethoxy]pyrazol-lyl ]pyridinecarboxamide (178 mg, 63%) ESI-MS m/z calc. 514.0689, found 515.1 (M+l)+; Retention time: 0.74 minutes Step B: N-(m-Tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 QwO 9 QwO:s: N'S HN- HCI N' -NJ-JviH (S) N Cl "XP f3c K2CO3 F3C ' 2-Chloro-N-(m-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (114.7 mg, 0.2227 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (100 mg, 0,6682) and, K2CO3 (184.6 mg, 1.336 mmol) were combined in DMSO (0.5 mL) in a screwcap tube and heated to 130 °C for 16 hours. After cooling to room temperature, the reaction mixture was diluted with 20 mL ethyl acetate, and 10 mL water and transferred to a separator}7 funnel. An s 15 mL 1 M citric acid on was added, and the organic layer was separated. The aqueous layer was extracted two additional times with 15 mL ethyl acetate, and the combined organics were washed with brme, dried over sodium sulfate and concentrated. The resulting crude material was ed by flash chromatography on silica gel, eluting with a 0-10% methanol in dichloromethane gradient to give N-(m-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide, (45 mg, 34%) ESI-MS m/z calc. 591.2, found 592.2 (M+l)+; Retention time: 2.24 minutes. 'H NMR (400 MHz, DMSO) 5 12.41 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 7.79 (tt, J = 6.0, 2.5 Hz, 3H), 7.57 - 7.50 (m, 2H), 6.91 (d, J = 8.2 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.48 - 4.24 (m, 2H), 2.46 (s, 1H), 2.42 (s, 3H), 2.29 (t, J = 8.8 Hz, 1H), 2.11 (dt, J = 13.2, 6.5 Hz, 1H), 1.83 (dd, J = 11.8, 5.5 Hz, 1H), 1.53 (d, J = 12.0 Hz, 6H), 1.38 (t, J = 12.1 Hz, 1H), 1.09 (dd, J = 4.5, 3.2 Hz, 4H), 0.66 (d, J = 6.2 Hz, 3H).
SUBSTITUTE SHEET (RULE 26) Synthetic Example 29: Synthesis of Compound 29: Synthesis ofN- (Benzenesulfonyl)[3-[(l-methylcyclopropoxy)methyl]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: (l-Methyl-l-(propyn-l-yloxy)cyclopropane ^-OH + 0 1-Methylcyclopropan-l-ol (1.0 g, 13.9 mmol) was dissolved in Et20 (50 mL) and cooled to 0°C. NaH (50% in oil, 0.67 g, 13.9 mmol) was added portion wise. The mixture was stirred for 10 min at 0 °C before propargyl bromide (80% in e, 3.1 g, .9 mmol) was added dropwise. The mixture was stirred for 1 hour at 0 °C. Since the reaction did not proceed, DMF (20 mL) was added. The mixture was stirred for an additional hour at 0° and quenched with sat. aq. NH4CI. The mixture was extracted with Et20 (2x50 mL). The combined c layers were washed with water twice and brine, dried over NajSCh and concentrated (at 40°C, 500 mbar) to afford crude (1-methyl-l- (propyn-l-yloxy)cyclopropane which was used as such in the next step, 'id NMR (CDCI3, 300 MHz): d 0.39 (m, 2H); 0.85 (m, 2H); 1.40 (s, 3H); 2.37 (s, 1H); 4.10 (s, ] Step B: 3-((l-methylcyclopropoxy)methyl)-lH-pyrazole TMS n2 Crude (l-methyl-l-(propyn-l-yloxy)cyclopropane from several batches (max 27.8 mmol, 3.0 g) was mixed with trimethylsilyl diazomethane (2.0 M in hexane, mL, 20 mmol) and stirred in a sealed tube at 115 °C for 18 hours. The mixture was cooled to 40 °C and ed with MeOH (20 mL) and concentrated. Column chromatography a; heptanes/EtOAc 2:1) gave 3-((l-methylcyclopropoxy)methyl)- IH-pyrazole as colorless oil (1.2 g, 28% over two steps). XHNMR (CDC13, 300 MHz): d 0.44 (m, 2H); 0.85 (m, 2H); 1.44 (s, 3H); 4.60 (s, 2H); 6.23 (s, 1H); 7.51 (s, 1H). 13C- NMR (75 MHz, CDC13): d 13.4, 20.3, 58.4, 61.9, 103.9,132.9 (one quatemaiy carbon not shown).
] Step C: N-(Benzenesulfonyl)[3-[(l- methylcyclopropoxy)methyl] pyrazol- l-yl] 2,2,4-trimethylpyrrolidin yl]pyridmecarboxamide SUBSTITUTE SHEET (RULE 26) 0 0 0 Sc(OTf)3 9 9wp % N'S NaH N'S ,N'NH + H or n H o N N'N^N^N^S) (S) 0 N-(Benzenesulfonyl)chloro[(4S)-2,2,4-trimethylpyrrolidin-l- yl]pyridinecarboxamide (83 mg, 0.2035 mmol), 3-[(l-methylcyclopropoxy)methyl]- IH-pyrazole (62 mg, 0.4074 mmol), and scandium triflate (10 mg, 0.02032 mmol) were combined in DMSO (1.660 mL). NaH (41 mg of 60 %w/w, 1.025 mmol) was added and the reaction was stirred for 15 minutes before it was sealed and heated to 160 °C for 16 h. The reaction was cooled and partitioned between ethyl acetate and a 1 M citric acid solution. The organics were separated, washed with brine, and dried over sodium sulfate. The organics were then evaporated under reduced re, and the crude material was purified by preparative HPLC (1-99 CH3CN in water with 5 mM HC1), over 30 minutes. Fractions ning product were diluted with water and extracted with ethyl acetate to give, upon concentration N-(benzenesulfonyl)[3-[(lmethylcyclopropoxy )methyl]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide (10 mg, 9%) ESI-MS m/z calc. 523.22534, found 524.2 (M+l)+; Retention time: 2.04 s.
Synthetic Example 30: Synthesis of Compound 30: N- (Benzenesulfonyl)[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide Step A: N-(Benzenesulfonyl)chloro[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide .so2nh2 9 CLP O N'S OH H GDI P^N 'Cl O^N NT XI✓5 2-Chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l- yl]pyridinecarboxylic 00 mg, 0.5717 mmol) and CDI (111 mg, 0.6846 mmol) were ed in THF (1.2 mL) and stirred at room temperature for 2 hours.
Benzenesulfonamide (117 mg, 0.7443 mmol) was added followed by DBU (102 pL SUBSTITUTE SHEET (RULE 26) 0.6821 mmol) and the reaction was stirred for an additional 6 h at room temperature.
The reaction mixture was diluted with a 1M citric acid on and water, and extracted 3 x 20 mL ethyl acetate. The combined organics were washed with brine, dried over sodium e and concentrated, then purified by silica gel chromatography using a gradient of 0-10% methanol in dichloromethane to give a white powder. N- (Benzenesulfonyl)chloro [3 - [(2,2,3,3-tetramethy Icy pyl)methoxy] pyrazol-1 - yl]pyridinecarboxamide (250 mg, 89%) ESI-MS m/z calc. 488.1285, found 489.2 ; Retention time: 0.81 minutes.
Step B: N-(Benzenesulfonyl)[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrroli(lin-lyl ]pyridinecarboxainide ? CLP II CLP N''3'- HM HCI N'S H I H p^N N^CI (S) p~0 n K2CO3 N-(Benzenesulfonyl)chloro[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (115 mg, 0.2352 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (approximately 105.9 mg, 0.7077 mmol), and potassium carbonate (approximately 195.6 mg, 1.415 mmol) were combined in DMSO (575.0 pL) and heated at 130 °C for 16 h. The reaction was cooled to room ature, diluted with 15 mL water, 15 mL 1M citric acid, and 30 mL ethyl acetate. The aqueous and the organic layers were separated, and the aqueous layer was extracted two additional times with 30 mL ethyl acetate, the organics were combined, washed with brine, dried over sodium sulfate and trated. The resulting solid was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane, and then additionally purified by silica chromatography using 0-100% ethyl e in dichloromethane, to give N-(benzenesulfonyl)[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidm-lyl ]pyridinecarboxamide (43 mg, 32%) ESI-MS m/z calc. 565.2723, found 566.3 (M+l)+; Retention time: 2.43 minutes. 'H NMR (400 MHz, DMSO) 5 12.47 (s, 1H), 8.18 (d, J = 2.8 Hz, 1H), 8.02 - 7.95 (m, 2H), 7.79 (d, J = 8.3 Hz, 1H), 7.76 - 7.69 (m, 1H), 7.68 - 7.62 (m, 2H), 6.92 (d, J = 8.3 Hz, 1H), 6.13 (d, J = 2.7 Hz, 1H), 4.24 (d, J = SUBSTITUTE SHEET (RULE 26) 7.7 Hz, 2H), 2.42 (t, J = 10.5 Hz, 1H), 2.28 (dd, J = 10.2, 7.1 Hz, 1H), 2.17 - 2.03 (m, 1H), 1.82 (dd, J = 11.8, 5.5 Hz, 1H), 1.52 (d, J = 9.4 Hz, 6H), 1.36 (t, J = 12.1 Hz, 1H), 1.10 (s, 6H), 1.04 (s, 6H), 0.73 (t, J = 7.7 Hz, 1H), 0.65 (d, J = 6.2 Hz, 3H).
Synthetic e 31: Synthesis of Compound 31: N N-(4- Hydroxyphenyl)sulfonyl[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide so2nh2 o 9 Qw,o 'OH iPd N*sT^s>l GDI ,N'N^r\rx:iH Cl p-O1 2-Chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (100 mg, 0.2661 mmol) and CDI (approximately 51.38 mg, 0.3169 mmol) were combined in THE (600.0 pL) and stirred at room temperature for 2 hours. 4-Hydroxybenzenesulfonamide (approximately 50.69 mg, 0.2927 mmol) was added followed by DBU (approximately 53.45 pL, 0.3574 mmol) and the reaction was d for an additional 16 h at room temperature. The reaction mixture was diluted with 10 mL 1M citric acid, and extracted 3 times with 10 mL of ethyl acetate.
The ed organics were washed with brine, dried over sodium sulfate, and concentrated to give a white solid, which was used in the next step without r purification. 2-chloro-N-(4-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (128 mg, 91%) ESI-MS m/z calc. 530.06384, found 531.0 (M+l)+; ion time: 0.69 minutes.
Step B: N-(4-Hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) HCI x V ivCtVO. '0hCj n OH K2CO3 cf3 cf3 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl]pyridinecarboxamide (134 mg, 0.2524 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (113 mg, 0.7550 mmol), and potassium carbonate (210 mg, 1.519 mmol) were combined in dimethyl sulfoxide (670.0 pL) and heated at 130 °C for 16 h. The reaction was cooled to room ature, and 1 mL of water was added. After 15 minutes stirring, the contents of the vial were allowed to settle, the liquid portion was removed by pipet and the remaining solids were dissolved with 20 mL ethyl acetate. The organics were washed with 15 mL 1M citric acid. The aqueous and the organic layers were ted, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were combined, washed with brine, dried over sodium e and concentrated. The resulting crude solid was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(4-hydroxyphenyl)sulfonyl- 6-[3-[2-[l-(tnfluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (43 mg, 28%) ESI-MS m/z calc. 607.20764, found 608.2 ; Retention time: 2.07 s, 'h NMR (400 MHz, DMSO) 5 12.25 (s, 1H), 10.58 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 7.87 - 7.79 (m, 2H), 7.75 (d, J = 8.2 Hz, 1H), 6.97 - 6.91 (m, 2H), 6.89 (d, J = 8.2 Hz, 1H), 6.10 (d, J = 2.7 Hz, 1H), 4.31 (t, J = 7.1 Hz, 2H), 2.44 (t, J = 10.4 Hz, 1H), 2.16 - 2.09 (m, 1H), 2.26 (t, J = 8.8 Hz, 1H), 2.07 (t, J = 7.0 Hz, 2H), 1.82 (dd, J = 11.9, 5.5 Hz, 1H), 1.54 (s, 3H), 1.51 (s, 3H), 1.38 (t, J = 12.1 Hz, 1H), 1.00 - 0.93 (m, 2H), 0.91 - 0.86 (m, 2H), 0.69 (d, J = 6.2 Hz, 3H).
Synthetic Example 32: Synthesis of Compound 32: Ar-(Benzenesulfonyl)- 6- [5-fluoro [2- [ l-(trifluoromethyl)cyclopropyl] ethoxy] pyrazol- 1-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-ylJpyridinecarboxamide SUBSTITUTE SHEET (RULE 26) 0 P,P NaH 9 QwP K2CO3 9 9wP 'OH + h2N :s N'S GDI H cr n ci CI^N^CI cr n N Sc(OTf)3 9 PP p^NH F3c —K Step A: nzenesulfonyl)-2,6-dichloro-pyridinecarboxamide 0. o P 0. 0 \y/ NaH \V/ OH + H2N GDI H cr n ci cr n ci A 5000 mL, 3 neck round bottom flask was fitted with a mechanical stirrer, a cooling bath, a J-Kem temperature probe/controller, a water cooled reflux condenser, an addition funnel and a nitrogen inlet/outlet. The vessel was charged under a nitrogen atmosphere with 60 wt% sodium e in mineral oil (26.04 g, 0.6510 mol). The vessel was then slowly charged with N,N-dimethylformamide (200 mL). Stirring was commenced and the pot temperature was recorded at 19 °C. The addition funnel was then charged with a solution of benzenesulfonamide (102.3 g, 0.6510 mol) in N,N- dimethylformarmde (868 ml, ~8.5 mL/g, 0.75M), requmng some gentle g to get a homogenous solution. The resulting clear pale yellow solution of benzenesulfonamide was subsequently added dropwise over 1 hour to the round bottom flask which resulted in some slight foaming and gas evolution. After the completed addition, the pot temperature was recorded at 28 °C. The vessel was then fitted with a heating mantle and the greyish mixture was warmed to 60 °C. Stirring of the mixture was continued at 60 °C for 1 hour at which point gas evolution appeared to have ceased. Stirring of the e was ued while the mixture was allowed to cool to room temperature.
Meanwhile, a 1000 mL, 3 neck round bottom flask was fitted with a mechanical stirrer, a heating mantle, a J-Kem ature probe/controller, a water cooled reflux condenser and a nitrogen inlet/outlet. The vessel was charged under a nitrogen atmosphere with SUBSTITUTE SHEET (RULE 26) 2,6-dichloropyridinecarboxylic acid (100 g, 0.5208 mol) andN,N- dimethylformamide (500 mL: 5 ml/g) which provided a clear light yellow on.
Stirring was commenced and the pot ature was recorded at 17 °C. The vessel was then charged with carbonyl diimidazole (84.45 g, 0.5208 mol) added as a solid in portions over 10 minutes which resulted in slight foaming and gas evolution, no exotherm was observed. Stirring of the resulting clear light amber solution was continued at room ature for 1 hour. The flask which contained the previously formed benzenesulfonamide sodium salt inN,N-dimethylformamide was treated dropwise over 45 minutes with the clear amber solution chloropyridinyl)(lH- imidazol-l-yl)methanone intermediate. After the completed addition, the vessel was fitted with a heating mantle and the mixture was warmed to 60 C'C and the condition was maintained for 1 hour when analysis by LC/MS indicated complete consumption of the intermediate. The reaction was allowed to cool to room temperature and then poured into ice cold 6M HC1 on (500 mL). The resulting mixture was further diluted with water (500 mL) and then transferred to a separatory funnel and partitioned with ethyl acetate (1000 mL). The organic layer was removed and the residual aqueous was extracted with ethyl acetate (2 x 500 mL). The combined organic layers were washed with ted sodium chloride on (3 x 500 mL), dried over sodium sulfate (300 g) and then ed through a glass frit r funnel. The clear pale yellow solution was trated under reduced pressure to a volume of about 200 mL. The clear residual oil was diluted with methyl tert-butyl ether (1000 mL) and then concentrated again under reduced pressure during which time a solid began to precipitate. The volume w as reduced to about 200 mL. The resulting slurry was allowed to stand at room temperature for 30 minutes and then filtered through a glass frit Buchner funnel. The filter cake was displacement washed methyl tert-butyl ether (2 x 150 mL) and then pulled in the Buchner funnel for 30 minutes. The al was further dried in a vacuum oven at 45°C for 2 hours to provide a white solid (101 g, 0.305 mol, 58% yield) as the desired product, N-(benzenesulfonyl)-2,6-dichloro-pyridinecarboxamide. ESI-MS m/z calc. 329.96326, found 330.9 (M+l)+; Retention time: 1.22 minutes.
Step B: A-(benzenesulfonyl)chloro[(4A)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) Q o. o k2co3 Q o. o \V/ \V/ N'S N'S H H cr n ci HN (S) cr N N (S) A 5000 mL 3 neck RB flask was fitted with a mechanical stirrer, a heating , a J-Kem temperature probe/controller, a water cooled reflux condenser and a en inlet/outlet. The vessel was charged under a nitrogen atmosphere with N- (benzenesulfonyl)-2,6-dichloro-pyridinecarboxamide (100 g, 0.3020 mol), (4S)- 2,2,4-trimethylpyrrolidine hydrochloride (54.24 g, 0.3624 mol) and dimethyl sulfoxide (500 ml, 5 mL/g) which provided a clear pale yellow solution. Stirnng was commenced and the pot temperature was recorded at 19 °C. The vessel was then charged with ium carbonate powder (167 g, 1.208 mol, 325 mesh) added as a solid in portions over 10 s which resulted in some minor gas evolution and foaming. The resulting off-white suspension was stirred at room temperature for 10 minutes and then heated to a pot ature of 115 °C and the condition was maintained for 24 hours. Analysis by LC/MS indicated reaction completion and the amber suspension was allowed to cool to room temperature. A 5000 mL 3 neck RB flask was fitted with a mechanical stirrer, a cooling bath and a J-Kem temperature probe. The vessel was charged with 2M HCI (1057 ml, 2.114 mol) and stirring was commenced at a us rate. The cooling bath was charged with crushed ter and the pot temperature was lowered to 0°C. The amber suspension reaction mixture was subsequently added slowly in portions over 30 minutes which resulted in the precipitation of a solid and an exotherm to 8 °C Note: Mild foaming upon addition. After the completed addition the resulting suspension was continued to stir at ~5 °C for 1 hour and then collected by vacuum filtration in a glass frit Buchner funnel. The filter cake was displacement washed with water (4 x 500 mL) and then pulled for 2 hours in the Buchner funnel to provide a white solid (150 g). A 5000 mL 3 neck RB flask was fitted with a mechanical stirrer, a g mantle, a JKem temperature controller, a water cooled reflux condenser and a en inlet/outlet. The vessel was charged under a nitrogen atmosphere with the isolated product (150g) and 2-propanol (1050 ml, 7 ml/g) which provided a pale yellow suspension. Stirring was commenced and the pot temperature was recorded at 19 °C.
The pot temperature was increased to reflux (~82 °C) and the condition was maintained SUBSTITUTE SHEET (RULE 26) for 10 minutes which resulted in a clear pale amber solution. Stirring of the solution was continued and the solution was allowed to slowly cool to room temperature during which time a solid began to form. ng of the suspension was continued and the vessel was fitted with a cooling bath which was charged with crushed ice/water. The pot temperature was d to 0 °C and stirring of the thick suspension continued at 0 °C for 1 hour. The material was collected by vacuum filtration in a glass frit Buchner funnel and the filter cake was displacement washed with ice cold 2-propanol (2 x 50 mL) and then pulled in the r for 30 minutes. The material was further dried in a vacuum oven at 45 °C for 15 hours to e a white solid (100 g, 0.245 mol, 81% yield) as the product, N-(benzenesulfonyl)chloro[(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide as 2-propanol solvate with 11 wt% anol. ESI-MS m/z calc. 703, found 408.1 (M+l)+; Retention time: 1.9 minutes.
Step C: 5-Fluoro[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]-lH- pyrazole N-mu ^'NH F3C f3c F A solution of 3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]-lH-pyrazole (0.68 g, 3.088 mmol) and l-(chloromethyl)fluoro-l,4- diazoniabicyclo[2.2.2]octane;ditetrafluoroborate (1.3 g, 3.7 mmol) in acetonitrile (15 mL) was stirred at 50 °C for 17 hours. The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were washed with water, dried over sodium sulfate, and evaporated. The residue was purified by silica gel column chromatography eluting with a 0-30% ethyl acetate in hexanes gradient to give the still impure t as a brown oil which was further punfied using a reverse phase HPLCMS method using a Luna Cl 8 (2) column (75 x 30 mm, 5 pm particle size) sold by Phenomenex (pn: 00CU0-AX), and a dual gradient run from 1-99% mobile phase B over 15.0 minutes (Mobile phase A = LEO (5 mM HC1). Mobile phase B = CEECN.
Flow rate = 50 mL/min, and column temperature = 25°C) giving 5-fluoro[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]-lH-pyrazole (90 mg) as a tan oil. ESI-MS m/z calc. 238.07292, found 239.1 (M+l)+; ion time: 0.56 minutes, 'hi NMR (400 SUBSTITUTE SHEET (RULE 26) MHz, DMSO-d6) 5 7.70 (d, J = 4.5 Hz, 1H), 4.30 - 4.16 (m, 2H), 2.04 (t, J = 7.1 Hz, 2H), 0.97 - 0.91 (m, 2H), 0.88 - 0.81 (m, 2H).
] Step D: 7V-(benzenesulfonyl) [5-fluoro [2- [1- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidiii-l-yl]pyridinecarboxamide XOCo o 0 AQsP g- Sc(OTf)3 -C&Q F3C- 0~<' A mixture of 5-fluoro[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]-lH- pyrazole (87 mg, 0.3653 mmol), A,-(benzenesulfonyl)chloro|(45')-2.2.4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (79 mg, 0.19 mmol), scandium triflate (10 mg, 0.020 mmol) and sodium hydride (38 mg of 60 %w/w, 0.95 mmol) in DMSO (0.92 mL) was stirred at 160 °C for 15 hours. The reaction was filtered and purified using a e phase HPLC-MS method using a Luna Cl 8 (2) column (75 x 30 mm, 5 pm le size) sold by Phenomenex (pn: 00CU0-AX), and a dual gradient run from 50-99% mobile phase B over 15.0 minutes (Mobile phase A = H20 (5 mM HC1).
Mobile phase B = CH3CN. Flow rate = 50 mL/min, and column temperature = 25 °C) giving /V-(benzenesuironyl)|5-fluoro|2-| 1- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (11 mg, 10%). ESI-MS m/z calc. 609.2033, found 610.3 (M+l)+; Retention time: 2.3 minutes. 'H NMR (400 MHz, DMSO-d6) 5 12.53 (s, 1H), 8.26 (d, J = 4.5 Hz, 1H), 8.00 (t, J = 1.3 Hz, 1H), 7.98 (d, J = 1.5 Hz, 1H), 7.82 (d, J = 8.3 Hz, 1H), 7.73 (d, J = 7.4 Hz, 1H), 7.66 (dd, J = 8.2, 6.7 Hz, 2H), 6.90 (d, J = 8.3 Hz, 1H), 4.41 (s, 2H), 2.38 (d, J= 10.5 Hz, 1H), 2.29 - 2.21 (m, 1H), 2.10 (q, J = 7.0 Hz, 3H), 1.82 (dd, J= 12.0, 5.5 Hz, 1H), 1.52 (s, 3H), 1.50 (s, 3H), 1.36 (s, 1H), 0.96 (dd, J = 3.7, 2.4 Hz, 2H), 0.89 (dt, J = 3.7, 1.9 Hz, 2H), 0.64 (d, J = 6.3 Hz, 3H).
Synthetic Example 33: sis of Compound 33: N-(4- Hydroxyphenyl)sulfonyl[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) Step A: 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide h2 0 OH HO GDI OH 0^N N" Cl 2-Chloro[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0,4288 mmol) and CDI (83 mg, 0.5119 mmol) were combined in THF (750 pL) and stirred at room temperature for 2 hours. 4- Hydroxybenzenesulfonamide (86 mg, 0.4966 mmol) was added followed by DBU (90 |iL. 0.6018 mmol) and the reaction was stirred for an additional 16 h at room temperature. The reaction mixture was diluted with 10 mL 1 M citric acid, and extracted 3x 10 mL ethyl acetate. The combined organics were washed with water, washed with brine, dried over sodium sulfate, and concentrated to give a white solid, which w as used in the next step without further cation. ro-N-(4- yphenyl)sulfonyl[3-[(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-lyl ]pyridinecarboxamide (235 mg, 94%) ESI-MS m/z calc. 504.1234, found 505.2 (M+l)+; Retention time: 0.75 minutes.
Step 2: N-(4-Hydroxyphenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxainide -■CClP.A°"s9 O-^N^N^CI ----------------- OH K2CO3 2-Chloro-N-(4-hydroxyphenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (235 mg, 0.4654 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (approximately 209.2 mg. 1.398 SUBSTITUTE SHEET (RULE 26) mmol), and potassium carbonate (approximately 387.4 mg, 2.803 mmol) were combined in DMSO (775.7 pL) and heated at 130 °C for 16 h. The reaction was cooled to room temperature, and 1 mL of water was added. After 15 s stirring, the contents of the vial were allowed to settle, the liquid portion was removed by pipet and the remaining solids were dissolved with 20 mL ethyl acetate, then washed with 15 mL 1M citric acid. The s and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were combined, washed with brine, dried over sodium sulfate and trated. The resulting solid was ed by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(4-hydroxyphenyl)sulfonyl[3-[(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide (30 mg, 11%) ESI-MS m/z calc. 581.2672, found 582.3 (M+l)+; Retention time: 2.26 minutes, 'll NMR (400 MHz, DMSO) 5 12.24 (s, 1H), .58 (s, 1H), 8.18 (d, J = 2.7 Hz, 1H), 7.86 - 7.78 (m, 2H), 7.74 (d, J = 8.2 Hz, 1H), 6.97 - 6.92 (m, 2H), 6.90 (d, J = 8.3 Hz, 1H), 6.13 (d, J = 2.6 Hz, 1H), 4.23 (d, J = 7.7 Hz, 2H), 2.43 (t, J = 10.4 Hz, 1H), 2.26 (t, J = 9.0 Hz, 1H), 2.10 (dt, J = 13.1, 6.8 Hz, 1H), 1.82 (dd, 3=11.9,5.4 Hz, 1H), 1.54 (s, 3H), 1.51 (s, 3H), 1.37 (t, J = 12.1 Hz, 1H), 1.10 (s, 6H), 1.04 (s, 6H), 0.73 (t, J = 7.7 Hz, 1H), 0.69 (d, J = 6.2 Hz, 3H).
Synthetic Example 34: Synthesis of Compound 34: N-(2- hydroxyphenyl)sulfonyl[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(2-hydroxyphenyl)sulfonyl[3-[2-[l- uoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide .S02NH2 0 o, 0 on N-'S' OH GDI I p^N n" Cl p-0 DBU AT' cf3 cf3 2-Chloro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (100 mg, 0.2661 mmol) and CDI (51 mg, 0.3145 mmol) were combined in THF (600.0 pL) and stirred at room temperature for 2 hours. 2- hydroxybenzenesulfonamide (51 mg, 0.2945 mmol) was added followed by DBU (55 SUBSTITUTE SHEET (RULE 26) |iL. 0.3678 mmol) and the reaction was stirred for an additional 16 h at room temperature. The reaction mixture was diluted with 10 mL 1 M citric acid, and extracted 3x 10 mL ethyl acetate. The combined organics were washed with water, brme, dried over sodium sulfate, and concentrated to give a white solid, which was used in the next step without further purification. 2-chloro-N-(2-hydroxyphenyl)sulfonyl [3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (132 mg, 93%) ESI-MS m/z calc. 530.06384, found 531.1 (M+l)+; Retention time: 0.7 minutes.
Step B: N-(2-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide IX OH HN HCI XOH 9-<J N Cl p-O N H N^(s) AC7 k2co3 aT cf3 cf3 2-Chloro-N-(2-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 ridinecarboxamide (132 mg, 0.2486 mmol), ,2,4-trimethylpyrrolidine (hydrochloride salt) (190 mg, 1.270 mmol), and ium carbonate (345 mg, 2.496 mmol) were combined in DMSO (660.0 pL) and heated at 130 °C for 16 h. The reaction was cooled to room temperature, and 1 mL of water was added. After 15 minutes stirring, the contents of the vial were allowed to settle, the liquid portion was removed by pipet and the remaining solids were ved with 20 mL ethyl acetate, then washed with 15 mL 1M citric acid. The aqueous and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were ed, washed with brme, dried over sodium e and concentrated. The resulting solid was further purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(2-hydroxyphenyl)sulfonyl[3-[2-[l- uoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (51 mg, 31%) ESI-MS m/z calc. 607.20764, found 608.3 SUBSTITUTE SHEET (RULE 26) (M+l)+; Retention time: 2.14 minutes 'H\MR (40() MHz. DMSO) 5 12.41 (s, 1H), .89 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 7.79 (dd, J = 8.1, 2.1 Hz, 2H), 7.56 - 7.43 (m, 1H), 7.07 - 6.95 (m, 2H), 6.88 (d, J = 8.2 Hz, 1H), 6.11 (d, J = 2.7 Hz, 1H), 4.31 (t, J = 7.1 Hz, 2H), 2.64 (d, J = 8.1 Hz, 1H), 2.58 (d, J = 10.6 Hz, 1H), 2.19 (d, J= 11.0 Hz, 1H), 2.08 (t, J = 7.0 Hz, 2H), 1.85 (dd, J = 11.9, 5.6 Hz, 1H), 1.54 (d, J = 8.1 Hz, 6H), 1.39 (t, J = 12.1 Hz, 1H), 1.00 - 0.92 (m, 2H), 0.90 (d, J = 10.8 Hz, 2H), 0.82 (d, J = 6.3 Hz, 3H).
Synthetic e 35: Synthesis of Compound 35: N-(3- Hydroxyphenyl)sulfonyl[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl 4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(3-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide HOXXso2nh2 o x %p OH GDI N-m p~U " 01 p^J ro[3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (100 mg, 0.2661 mmol) and CDI (51 mg, 0.3145 mmol) were combined in THF (600.0 pL) and stirred at room temperature for 2 hours. 3- Hydroxybenzenesulfonamide (51 mg, 0.2945 mmol) was added followed by DBU (55 |iL. 0.3678 mmol) and the reaction was stirred for an additional 16 h at room temperature. The reaction mixture was diluted with 10 mL 1 M citric acid, and extracted 3 x 10 mL ethyl acetate. The combined organics were washed with water, brine, dried over sodium sulfate, and trated to give a white solid, which was used in the next step without further purification. 2-chloro-N-(3-hydroxyphenyl)sulfonyl [3-[2-[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (135 mg, 96%) ESI-MS m/z calc. 530.06384, found 531.2 (M+l)+; Retention time: 0.69 minutes.
SUBSTITUTE SHEET (RULE 26) Step B: N-(3-Hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide nvo"0 0 o ■■-CCaV r* aPOH K2CO3 cf3 cf3 2-Chloro-N-(3-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide (135 mg, 0.2543 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (193 mg, 1.290 mmol), and potassium carbonate (352 mg, 2.547 mmol) were combined in DMSO (508.6 pL) and heated at 130 °C for 16 h. The reaction was cooled to room temperature, and 1 mL of water was added. After 15 minutes stirring, the ts of the vial were allowed to settle and the liquid portion was removed by pipet and discarded. The remaining solids were ved in 20 mL ethyl acetate then washed with 15 mL 1M citric acid. The aqueous and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were combined, washed with brine, dried over sodium sulfate and trated. The resulting solid was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(3-hydroxyphenyl)sulfonyl[3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (40 mg, 26%) ESI-MS m/z calc. 764, found 608.3 (M+l)+; Retention time: 2.05 minutes. 'H NMR (400 MHz, DMSO) 5 12.44 (s, 1H), .19 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.44 (t, J = 8.0 Hz, 1H), 7.40 - 7.37 (m, 2H), 7.06 (d, J = 7.9 Hz, 1H), 6.91 (d, J = 8.2 Hz, 1H), 6.11 (d, J = 2.7 Hz, 1H), 4.31 (t, J = 7.1 Hz, 2H), 2.47 (d, J = 10.0 Hz, 1H), 2.33 (s, 2H), 2.08 (m, J = 8.1, 7.0 Hz, 2H), 1.84 (dd, J = 11.8, 5.5 Hz, 1H), 1.54 (s, 3H), 1.52 (s, 3H), , J = 12.1 Hz, 1H), 0.96 (td, J = 5.0, 3.3 Hz, 2H), 0.90 (d, J = 11.1 Hz, 2H), 0.70 (d, J = 6.2 Hz, 3H).
Synthetic Example 36: Synthesis of nd 36: N- (Benzenesulfonyl) [3- [dideuterio-(2,2,3,3- SUBSTITUTE SHEET (RULE 26) WO 64632 2017/054611 tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxainide Step A: Dideuterio-(2,2,3,3-tetramethylcyclopropyl)methanol LiAI(2H)4 ■OH -OH O 2H2H 2,2.3,3-Tetramethylcyclopropanecarboxylic acid (1.077 g, 7.574 mmol) was dissolved in anhydrous diethyl ether in a nitrogen purged 100 mL round bottom flask.
The reaction mixture was cooled to 0 °C. Solid tetradeuterioalumanuide (lithium salt) (420 mg, 10.01 mmol) was added in 3 portions. The reaction mixture was allowed to gradually reach room ature and stirred for a total of 16 hours. The reaction mixture was then again cooled to 0 °C. HC1 (aq, 0.2 N, 5 mL) was added dropwise, followed by 20 mL water. The aqueous phase was extracted with diethyl ether (2 x 30 mL). The combined organic phases were washed with aqueous NaHCCf. followed by brme, then dried over sodium sulfate, filtered and evaporated to give dideuterio- (2,2,3,3-tetramethylcyclopropyl)methanol (920 mg, 93%). 'll NMR (400 MHz, DMSO) 5 4.11 (s, 1H), 1.04 (s, 6H), 0.93 (s, 6H), 0.37 (s, 1H).
Step B: tert-Butyl euterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazole-l-carboxylate n-n^o-^ HO. sJM. D DIAD .OH + N 2H2H PPh3 2-h ] DIAD (1.4 mL, 7.111 mmol) was added dropwise to a solution of triphenyl phosphine (1.815 g, 6.920 mmol) in 40 mL anhydrous toluene, at 0 °C. After 30 minutes at 0 0C, a solution of tert-butyl 3-hydroxypyrazole-l-carboxylate (1.155 g, 6.271 mmol) and dideuterio-(2,2,3,3-tetramethylcyclopropyl)methanol (980 mg, 7.525 mmol) in 30 mL toluene was slowly added by syringe. The reaction was warmed to room temperature for 45 minutes, and then was heated to 55 °C for 18 h. The mixture was evaporated and the resulting material was partitioned between ethyl acetate (30 mL) and IN sodium hydroxide (30 mL). The organics were separated, washed with brine (30 mL), dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-30% ethyl acetate in hexanes to give an oil that ally solidified to a slightly yellow solid: utyl 3-[dideuterio-(2,2,3,3- SUBSTITUTE SHEET (RULE 26) tetramethylcyclopropyl)methoxy]pyrazole-l-carboxylate (820 mg, 44%) ESI-MS m/z calc. 296.2069, found 297.2 (M+l)1: Retention time: 0.79 minutes.
Step C: 3-[Dideuterio-(2,2,3,3-tetramethylcyclopropyl)methoxy]-lH- nun^0J< N32CO3 f-oN-NH h2o *hh 'i 2h H To utyl 3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazole-l-carboxylate (800 mg, 2.699 mmol) in 1,2- dimethoxyethane (10 mL) was added sodium carbonate (460 mg, 4.340 mmol) in water (3 mL), and the reaction mixture was heated to 90 °C for 16 hours in a screwcap vial.
The reaction mixture was cooled to room ature and diluted with water (50 mL) and ethyl acetate (50 mL). The organics were separated, and the aqueous layer was ted 2 x 25 mL ethyl acetate. The combined organics were washed with bnne, and dried over sodium sulfate, then concentrated to give a colorless oil. euterio- (2,2,3,3-tetramethylcyclopropyl)methoxy]-lH-pyrazole (492 mg, 93%) ESI-MS m/z calc. 196.15446, found 197.1 (M+l)1; ion time: 0.57 minutes, 1HNMR(400 MHz, DMSO) 6 11.78 (s, 1H), 7.48 (t, J = 2.1 Hz, 1H), 5.65 (t, J = 2.3 Hz, 1H), 1.08 (s, 6H), 1.00 (s, 6H),0.66 (s, 1H).
Step D: Ethyl 2-chloro[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylate ,N'NH DABCO '0'"v N Cl 2h2h cr n ci K2C03 2h2h A round bottom flask was d under nitrogen with3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]-lH-pyrazole (485 mg, 2.471 mmol), ethyl 2,6- dichloropyridinecarboxylate (545 mg, 2.477 mmol), K2CO3 (513 mg, 3.712 mmol) (freshly ground in a mortar) and anhydrous DMF (4.128 mL). DABCO (50 mg, 0.4457 mmol) was added and the mixture was stirred at room temperature under nitrogen for 16 hours. The reaction mixture was diluted with ethyl acetate (50 mL) and water (50 mL) and the two phases were separated. The aqueous phase was further extracted with ethyl acetate (2 x 30 mL), and the combined extracts were washed with brine and dried over SUBSTITUTE SHEET (RULE 26) sodium sulfate, after which the solvent was removed under reduced pressure. The material was subjected to flash tography on silica gel using a gradient of 0-20% ethyl acetate in hexanes. The pure fractions were combined and the solvents removed under reduced pressure to provide a white solid; ethyl 2-chloro[3-[dideuterio- (2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylate (505 mg, 54%) ESI-MS m/z calc. 379.16318, found 380.1 (M+l)+; Retention time: 0.9 s lH NMR (400 MHz, DMSO) 5 8.42 (d, J = 2.9 Hz, 1H), 8.39 (d, J = 8.5 Hz, 1H), 7.76 (d, J = 8.4 Hz, 1H), 6.24 (d, J = 2.9 Hz, 1H), 4.34 (q, J = 7.1 Hz, 2H), 1.33 (t, J = 7.1 Hz, 3H), 1.11 (s, 6H), 1.04 (s, 6H), 0.74 (s, 1H).
Step E: 2-Chloro[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylic acid O O P^N N^CI o~Q N Cl A solution of sodium hydroxide (275 mg, 6.875 mmol) in water (2.500 mL) was added to a solution of ethyl 2-chloro[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxylate (500 mg, 1.316 mmol) in isopropanol (2.500 mL) and stirred at 90 °C for 45 minutes. The reaction was cooled to room temperature then diluted with 50 mL ethyl acetate, 20 mL 1M citric acid, and 10 mL water. The organics were ted, and the aqueous portion was extracted 2x 25 mL ethyl acetate. The combined organics were washed with brine, dried over sodium sulfate and concentrated. The ing solid was triturated in 40 mL water, briefly sonicated, and collected by tion then dried to give a white solid, 2- chloro[3-[dideuterio-(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridine- 3-carboxylic acid (404 mg, 87%) ESI-MS m/z calc. 351.13187, found 352.1 (M+l)+; Retention time: 0.77 minutes. ^NMR (400 MHz, DMSO) 5 8.41 (d, J = 2.9 Hz, 1H), SUBSTITUTE SHEET (RULE 26) 8.38 (d, J = 8.4 Hz, 1H), 7.74 (d, J = 8.4 Hz, 1H), 6.22 (d, J = 2.8 Hz, 1H), 1.11 (s, 6H), 1.04 (s, 6H), 0.74 (s, 1H).
Step F: N-(Benzenesulfonyl)chloro[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide SO2NH2 o O' X°"sP ‘OH ^C|H ^ f-u N Cl GDI p-^N « DBU =HH 2-Chloro[3-[dideuteno-(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol- l-yl]pyridinecarboxylic acid (100 mg, 0.2842 mmol) and CDI (55 mg, 0.3392 mmol) were combined in THF (600.0 pL) and stirred at room temperature for 2 hours, benzenesulfonamide (49 mg, 0.3117 mmol) was added followed by DBU (57 pL, 0.3812 mmol) and the reaction was stirred for an additional 16 h at room temperature.
The reaction mixture was diluted with 10 mL 1 M citric acid, and extracted 3x 10 mL ethyl acetate. The combined organics were washed with water, brine, dned over sodium sulfate, and concentrated to give approximately 135 mg of a white solid, which was used in the next step without further purification. N-(benzenesulfonyl)chloro[3- teno-(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridine armde ESTMS m/z calc. 490.14105, found 491.2 (M+l)+; Retention time: 0.81 minutes.
Step G: zenesulfonyl)[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxainide HCI x0*p •O-Cj n K2C03 2h 2h 2h zenesulfonyl)chloro[3-[dideuterio-(2,2,3,3- tetramethylcyclopropyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (135 mg, 0.2749 mmol), (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (128 mg, 0.8553 mmol), and potassium carbonate (237 mg, 1.715 mmol) were combined in DMSO (458.2 pL) and heated at 130 °C for 16 h. The reaction was cooled to room temperature, and 1 mL SUBSTITUTE SHEET (RULE 26) of water was added. After 15 minutes stirring, the contents of the vial were allowed to settle, the liquid portion was removed by pipet and the remaining solids were dissolved with 20 mL ethyl acetate then washed with 15 mL 1M citric acid. The s and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were combined, washed with brine, dried over sodium sulfate and concentrated. The resulting solid was further purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to giveN- (benzenesulfonyl)[3-[dideuterio-(2,2,3,3-tetramethylcyclopropyl)methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (112 mg, 72%) ESI- MS m/z calc. 567.28485, found 568.3 (M+l)+; Retention time: 2.42 minutes. 'H NMR (400 MHz, DMSO) 8 12.51 (s, 1H), 8.18 (d, J = 2.8 Hz, 1H), 7.99 (dt, J = 7.1, 1.4 Hz, 2H), 7.80 (d, J = 8.3 Hz, 1H), 7.76 - 7.70 (m, 1H), 7.70 - 7.62 (m, 2H), 6.92 (d, J = 8.3 Hz, 1H), 6.13 (d, J = 2.7 Hz, 1H), 2.40 (t, J = 10.4 Hz, 1H), 2.26 (t, J = 8.7 Hz, 1H), 2.09 (dq, J = 11.7, 6.0 Hz, 1H), 1.82 (dd, J = 11.8, 5.4 Hz, 1H), 1.52 (d, J = 9.5 Hz, 6H), 1.36 (t, J = 12.2 Hz, 1H), 1.10 (s, 6H), 1.04 (s, 6H), 0.72 (s, 1H), 0.64 (d, J = 6.2 Hz, ] tic Example 37: Synthesis of Compound 37: N- (Benzenesulfonyl)[3-[[l-(trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-lyl 4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: t-butoxymethyl)-lH-pyrazole TMS^Nj rU, NH 0 tert-Butylpropargyl alcohol (2.5 g, 22.2 mmol) was mixed with trimethylsilyl diazomethane (2.0 M in , 11.1 mL, 22.2 mmol) and stirred in a sealed tube at 115 °C for 18 hours. The mixture was cooled to 40 °C and quenched with methanol (5 mL) and concentrated. Column chromatography (silica; heptanes/EtOAc2:l to 1:1) afforded 3-(tert-butoxymethyl)-lH-pyrazole as colorless oil (1.5 g, 44%). JH NMR (CDC13, 300 MHz): 8 1.26 (s, 9H); 4.53 (s, 2H); 6.22 (s, 1H); 7.48 (s, 1H). R (75 MHz, CDC13): 8 27.3, 57.2, 73.9,103.5,134.0 (one quaternary carbon not shown).
Step B: tert-Butyl 6-[3-(tert-butoxymethyl)pyrazol-l-yl]chloro- pyridinecarboxylate SUBSTITUTE SHEET (RULE 26) DABCO *N'N ;,N'NH o cr n ci k2co3 A 100 mL round bottom flask was charged under nitrogen with 3-(tert- butoxymethyl)-lH-pyrazole (1.241 g, 8.047 mmol), tert-butyl 2,6-dichloropyridine carboxylate (2.0 g, 8.061 mmol), K2CO3 (1.448 g, 10.48 mmol) (freshly ground in a mortar) and anhydrous DMF (12.41 mL). DABCO (163 mg, 1.453 mmol) was added and the mixture was d at room temperature under nitrogen for 16 hours. The reaction e was diluted with ethyl acetate (50 mL) and water and brine (50 mL) and the two phases were separated. The aqueous phase was further extracted with ethyl acetate (2 x 30 mL). The combined extracts were washed with brine, dried over sodium sulfate and the t removed under reduced pressure. The material was subjected to flash chromatography on silica gel using a nt of ethyl acetate (0 to 10%) in s. The pure fractions were combined and the solvents removed under reduced pressure to provide tert-butyl 6-[3-(tert-butoxymethyl)pyrazol-l-yl]chloro-pyndine- 3-carboxylate (1.956 g, 66%) as a colorless oil, which solidified to a white solid overnight on high vac. ESI-MS m/z calc. 365.1506, found 366.2 (M+l)+; Retention time: 0.82 minutes Step C: 2-Chloro[3-(hydroxymethyl)pyrazol-l-yl]pyridine carboxylic acid O k O O HCI %■ OH °V//N'N' 'N' 'Cl HO *N'N' '1ST 'Cl✓5 tert-Butyl 6-[3-(tert-butoxymethyl)pyrazol-l-yl]chloro-pyridine carboxylate (538 mg, 1.471 mmol) was dissolved in HCI in dioxane (8.0 mL of 4 M, 32.00 mmol) and heated at 60 °C for 2 hours. The reaction e was then cooled to room temperature and concentrated to dryness, giving a white powder. 2-chloro[3- (hydroxymethyl)pyrazol-l-yl]pyridinecarboxylic acid (370 mg, 99%) ESI-MS m/z calc. 253.02542, found 254.1 (M+l)+; Retention time: 0.33 minutes SUBSTITUTE SHEET (RULE 26) Step D: 2-Chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl]pyridinecarboxylic A(CF3 o sA // OH °0 HO A rr°V//N'N^N^CI0OH '/N'N' 'N' 'ClxS KO*Bu [ 1 -(Tnfluoromethyl)cyclopropy 1] methyl 4-methyIbenzenesulfonate (1.3 g, 4.417 mmol), and 2-chloro[3-(hydroxymethyl)pyrazol-l-yl]pyridmecarboxylic acid (370 mg, 1.459 mmol), were combined in ous DMSO (9.250 mb), tertbutoxypotassium (660 mg, 5.882 mmol)was added and the reaction mixture was stirred at room temperature. After 30 minutes the reaction mixture was poured into 1 M citric acid (15 mL) and extracted 3 x 15 mL ethyl acetate. The combined organics were washed with brine, dried over sodium e and concentrated. The resulting material was purified by chromatography on silica gel using a 0-10% methanol in dichloromethane nt. The fractions containing product were collected and trated to give a white solid. 2-Chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl]pyridmecarboxylic acid (292 mg, 53%) ESI-MS m/z calc. 375.05975, found 376.1 (M+l)+; Retention time: 0.62 minutes.
Step E: N-(Benzenesulfonyl)chloro[3-[[l- uoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl]pyriclinecarboxamide SO2NH2 A rrM3 N-mM OH GDI c DBU 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l- yl]pyridinecarboxylic acid (60 mg, 0.1597 mmol) and GDI (31 mg, 0.1912 mmol) were combined in THF (360.0 pL) and stirred at room temperature for 2 hours.
Benzenesulfonamide (28 mg, 0.1781 mmol) was added followed by DBU (35 pL. 0.2340 mmol) and the reaction was stirred for an onal 16 h at room temperature.
SUBSTITUTE SHEET (RULE 26) The reaction e was diluted with 10 mL 1 M citric acid, and extracted 3 x 10 mL ethyl acetate. The combined organics were washed with water, brine, dried over sodium sulfate, and concentrated to give a white solid, which was used in the next step without further cation. N-(benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl]pyridmecarboxamide (approximately 78 mg) ESI-MS m/z calc. 514.0689, found 515.1 (M+l)+; Retention time: 0.69 minutes.
Step G: N-(Benzenesulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl] pyrazol- l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Yo J?!L %nXcVoC, ^o o O HN^vjjp1 r|I 9 qw0 \ N.kMi k2co3 N-(Benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl]pyridmecarboxamide (78 mg, 0.1515 mmol), (4S)-2,2,4-trimethylpyrrohdine (Hydrochlonde salt) (115 mg, 0.7684 mmol), and Potassium carbonate (210 mg, 1.519 mmol) were combined in DMSO (390.0 pL) and heated at 130 °C for 16 h. The reaction was cooled to room temperature, and 1 mL of water was added. After 15 minutes stirring, the contents of the vial were allowed to settle, the liquid portion was removed by pipet and the remaining solids were ved with 20 mL ethyl acetate, then washed with 15 mL 1M citric acid. The aqueous and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The cs were combined, washed with brine, dried over sodium sulfate and concentrated. The ing solid was further ed by silica gel tography eluting with 0-10% methanol in dichloromethane to give N-(benzenesulfonyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxymethyl]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (46 mg, 51%) ESI-MS m/z calc. 591.2127, found 592.3 (M+l)+; Retention time: 2.11 minutes. H NMR (400 MHz, DMSO) 5 12.56 (s, 1H), 8.33 (d, J = 2.6 Hz, 1H), 8.04 - 7.97 (m, 2H), 7,89 - 7.79 (m, 1H), 7.73 (t, J = 7.1 Hz, 1H), 7.70 - 7.60 (m, 2H), 7.07 (d, J = 8.2 Hz, 1H), 6.56 (d, J = 2.6 Hz, 1H), 4.55 (s, 2H), 3.59 (s, 2H), 2 42 (t, J = 10.4 Hz, 1H), 2.29 (d, J = 9.6 Hz, SUBSTITUTE SHEET (RULE 26) WO 64632 2017/054611 1H), 2.18 - 2.04 (m, 1H), 1.83 (dd, J = 11.7, 5.5 Hz, 1H), 1.54 (d, J = 9.4 Hz, 6H), 1.37 (t, J = 12.1 Hz, 1H), 1.03 - 0.94 (m, 2H), 0.91 - 0.80 (m, 2H), 0.65 (d, J = 6.2 Hz, 3H).
Synthetic Example 38: Synthesis of Compound 38: N-(Benzenesulfonyl)- 6- [3- [2-hydroxy [l-(trifluoromethyl)cyclopropyl]ethoxy] pyrazol- 1-yl] [(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Steps A-B: l-[l-(Trifluoromethyl)cyclopropyl]propen-l-ol 1.)PCC OH F3C ^\P°H f3c 2.) vinylmagnesium bromide ifluoromethyl)cyclopropyl]methanol (1 g, 7.138 mmol) was stirred in dry dichloromethane (30 mL) and cooled to 0 °C in an ice bath. Chloro-hydroxy- dioxo-chromium;pyridine (2.77 g, 12.85 mmol) was added in one portion, and the ice bath was removed and the reaction was stirred for 24 hours at room ature. The reaction mixture was poured into 100 mL diethyl ether and filtered through a pad of silica with a layer of celite on top, eluting with diethyl ether. The resulting filtrate was dried over sodium sulfate, then filtered through cotton and used in the next step without concentration due to the volatility of the aldehyde. The crude filtrate was cooled to 0 °C, and bromo(vinyl)magnesium (14.5 mL of 1 M, 14.50 mmol) (in THF) was slowly added. The reaction mixture was allowed to slowly warm to near room temperature over 2 hours. The reaction e was then cooled to 0 °C, quenched with 1M HC1, and diluted with water. The layers were separated, and the aqueous was extracted 4 additional times with diethyl ether. The combined cs were washed with brine, dried over sodium sulfate and partially trated. The resulting crude material was used in the next step without further purification. 1-[1- (trifluoromethyl)cyclopropyl]propen-l-ol (approximately 1.25g crude, with substantial THF remaining) Crude XH NMR (400 MHz, DMSO) 8 5.95 - 5.84 (m, 1H), .36 (d, J = 5.2 Hz, 1H), 5.32 (ddd, J = 17.1, 2.1, 1.4 Hz, 1H), 5.18 (ddd, J = 10.4, 2.0, 1.3 Hz, 1H), 4.17 (tt, J = 6.4, 1.4 Hz, 1H), 0.95 - 0.87 (m, 4H).
Step C: tert-Butyl-dimethyl-[l-[l- (trifluoromethyl)cyclopropyl]allyloxy]silane OH TBS-CI, imidazole f3c OTBS SUBSTITUTE SHEET (RULE 26) l-[l-(Trifluoromethyl)cyclopropyl]propen-l-ol (270 mg, 1.625 mmol) and imidazole (220 mg, 3.232 mmol) were dissolved in DMF (2 mL) and cooled to 0 °C in an ice bath. TBS-C1 (tert-butyldimethylsilyl chloride) (370 mg, 2.455 mmol) was then added in a single n, and after 15 s the ice bath was removed and the reaction mixture was allowed to stir 16 hours at room temperature. Saturated aqueous ammonium chloride (1 mL) was then added, and the on mixture was stirred for 20 minutes. The reaction mixture was then diluted with 30 mL diethyl ether and 20 mL of water. The organics were separated, and the aqueous layer was extracted 2x15 mL diethyl ether. The combined cs were washed with water, then brine and dried over sodium sulfate, and trated under vacuum. The crude material was then passed through a silica plug, eluting with hexanes, and concentrated to an oil containing substantial overlapping silyl impurities, but used in the next step without further purification, tert-butyl-dimethyl-[l-[l-(trifluoromethyl)cyclopropyl]allyloxy]silane (approximately 317 mg, crude).
] Steps D-F: tert-butyl 3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazole-l-carboxylate 1. ) 0s04, Nal04, tidine N'N^O^ OTBS 2. ) NaBH4 TBSO 3.) DIAD, PPh3 tert-Butyl-dimethyl-[l-[l-(trifluoromethyl)cyclopropyl]allyloxy]silane (220 mg, 0.7846 mmol) (crude) was combined in dioxane (6 mL)and water (2 mL) with 2,6- dimethylpyndine (185 |iL. 1.597 mmol) and sodium periodate (675 mg, 3.156 mmol).
Tetraoxoosmium (420 pL, 0.3139 mmol) was then added and the reaction mixture was stirred for 20 hours at room temperature. The reaction was then d with 30 mL dichloromethane and 30 mL water. The organics were separated, and the aqueous layer was extracted 2 x 25 mL dichloromethane. The combined organics were dried over sodium sulfate, and concentrated to about 8 mL and used in the next step, without isolation.
The crude mixture from the previous step was diluted with methanol (10 mL), and cooled to 0 °C. Sodium borohydride (90 mg, 2.379 mmol) was added and the SUBSTITUTE SHEET (RULE 26) on mixture stirred for 1 hour. The reaction e was then quenched with acetic acid and trated. The resulting material was partitioned between ethyl acetate and aqueous sodium bicarbonate and the layers were separated. The aqueous portion was extracted 2x ethyl acetate, and the combined organics were washed with brine, dried over sodium sulfate and concentrated. The resulting crude alcohol was used in the next step without purification. The crude t was ed with triphenylphosphine (300 mg, 1.144 mmol) and tert-butyl 3-hydroxypyrazole-l-carboxylate (145 mg, 0.7872 mmol) dissolved in THF (15 mL). The reaction mixture was cooled to 0 °C and DIAD (230 pL, 1.187 mmol) was slowly added. The ice bath was removed, and the reaction was stirred at room temperature for an hour then heated to 55 °C for an additional 16 hours. The reaction mixture was partially concentrated, dissolved in 100 mL ethyl acetate, washed with 25 mL IN NaOH, brine, dried over sodium sulfate and concentrated. The resulting material was purified by chromatography on silica gel eluting with 0-50% ethyl acetate in hexanes to give, with an overlapping UV active impurity utyl 3- [2- [tert-buty l(dimethy l] oxy[ 1 - (trifluoromethyl)cyclopropyl]ethoxy]pyrazole-l-carboxylate (63 mg, 18%) ESI-MS m/z calc. 450.21616, found 451.3 (M+l)+; Retention time: 0.95 minutes Step G: 3-(2-((tert-butyldimethylsilyl)oxy)(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyr azole TFA TBSO 0-/'NH tert-Butyl 3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazole-l-carboxylate (63 mg, 0.1398 mmol) (with a major impurity present) w as dissolved in dichloromethane (1.312 mL) with TFA (trifluroracetic acid) (approximately 194.3 mg, 131.3 pL, 1.704 mmol) and the reaction was stirred at room temperature for 30 minutes. s (1 mL) were added, and reaction was evaporated and the resulting oil was partitioned between ethyl acetate (10 mL) and a saturated sodium bicarbonate on. The organics were separated, washed with brine, dried over sodium sulfate and evaporated to give a colorless oil, 3-(2-((tertbutyldimethylsilyl 2-(l-(tnfluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole (37 SUBSTITUTE SHEET (RULE 26) mg, 76%) ESI-MS m/z calc. 350.16373, found 351.2 (M+l)+; Retention time: 0.81 minutes.
Step H: tert-Butyl 6-[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]chloro-pyridinecarboxylate DABCO TBSO ,0-y/N'NH TBSO 0_//N'NAN'>kCI K2CO3 cr N Cl cf3 cf3 A round bottom flask was charged under nitrogen with tert-butyl-dimethyl- [2-(lH-pyrazolyloxy)-l -[1 -(trifluoromethyl)cyclopropyl] ethoxy] silane (37 mg, 0.1056 mmol), tert-butyl 2,6-dichloropyridinecarboxylate (26 mg, 0.1048 mmol), K2CO3 (25 mg, 0.1809 mmol) (freshly ground in a mortar) and anhydrous DMF (250 pL). DABCO (2 mg, 0.01783 mmol) was added and the mixture was stirred at room temperature under nitrogen for 16 hours. The reaction mixture was diluted with ethyl acetate (15 mL) and water (15 mL) and the two phases were separated. The s phase was further extracted with ethyl e (2x15 mL). The combined extracts were washed with brine and dried over sodium sulfate and the solvent removed under reduced pressure. The material was subjected to flash chromatography on silica gel using a nt of ethyl e (0 to 20%) in hexanes. The pure fractions were ed and the ts removed under reduced pressure to provide a colorless oil, tert-buty l 6-[3- [2-[tert-butyl(dimethyl)silyl]oxy[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-lyl ]chloro-pyridinecarboxylate (20 mg, 34%). ESI-MS m/z calc. 561.20374, found 562.4 (M+l)+; Retention time: 0.84 minutes, 'H NMR (400 MHz, DMSO) 8 8.44 (d, J = 2.9 Hz, 1H), 8.34 (d, J = 8.4 Hz, 1H), 7.71 (d, J = 8.4 Hz, 1H), 6.17 (d, J = 2.9 Hz, 1H), 4.44 (dd, J = 10.7, 2.9 Hz, 1H), 0.13 - 0.03 (m, 6H), 4.32 - 4.25 (m, 1H), 3.89 (s, 1H), 1.56 (s, 9H), 0.98 (d, J = 35.8 Hz, 4H), 0.86 (s, 9H) Step I: 2-[tert-Butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]chloro-pyridinecarboxylic TBSO CW/N'N N Dl TBSO <W/N'N N' 'Cl cf3 CF3 SUBSTITUTE SHEET (RULE 26) WO 64632 tert-Butyl 6-[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cy pyl]ethoxy]pyrazol-1 -yl] chloro-pyridinecarboxylate (20 mg, 0.03558 mmol) and TFA (50 pL, 0.6490 mmol) were combined in dichloromethane (0.75 mL) and heated at 45 °C for 3 h. The reaction was evaporated. Hexanes were added and the mixture evaporated again to give a white solid 6-[3-[2-[tertbutyl (dimethyl)sily 1] oxy[ 1-(trifluoromethy l)cy clopropyl] ethoxy ] pyrazoly 1] chloro-pyridinecarboxylic acid (18 mg, 100%) ESI-MS m/z calc. 505.14114, found 506.3 (M+l)+; Retention time: 0.59 minutes.
Step J: N-(Benzenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy|l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]chloro-pyridine carboxamide fX* oso2nh2 0 QwP N'N^N^C|H ^ TBSO N'N^n^ci c: DBU cf3 6-[3-[2-[tert-Butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]chloro-pyndinecarboxylic acid (19 mg, 0.03755 mmol) and GDI (8 mg, 0.04934 mmol) were combined in THE (200 pL) and stirred at room temperature for 2 hours, benzenesulfonarmde (7 mg, 3 mmol) was added followed by DBU (10 pL, 0.06687 mmol) and the reaction was stirred for an additional 2 h at room temperature. The reaction mixture was then d with mL 1 M citric acid, and extracted 3 x 10 mL ethyl acetate. The combined cs were washed with water, brine, dried over sodium sulfate, and concentrated to give a white solid, which was used in the next step without further cation N- (benzenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]chloro-pyndinecarboxamide (approximately 22 mg) ESI-MS m/z calc. 644.1503, found 645.3 (M+l)+; Retention time: 0.8 minutes.
Step O: zenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) HCI £ <y> TBSO // 'N N TBSO K2CO3 cf3 CF3 zenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethy l)cy clopropy 1] ethoxy]pyrazol-1 -yl] chloro-pyndine-3 -carboxamide (22 mg, 0.03410 mmol), ,2,4-trimethylpyrrolidine (Hydrochloride salt) (approximately 40.83 mg, 0.2728 mmol), and potassium carbonate (approximately 75.40 mg, 0.5456 mmol) were combined in DMSO (180 pL) and heated at 130 °C for 24 hours. The reaction was cooled to room temperature and diluted with 15 mL 1M citric acid and 20 mL ethyl acetate. The aqueous and the organic layers were separated, and the aqueous layer was extracted two additional times with 15 mL ethyl acetate. The organics were combined, washed with brine, dried over sodium sulfate and concentrated. The resulting solid was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give the TBS protected material as a white solid, N-(benzenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-lyl ]pyridinecarboxamide (8 mg, 32%) ESTMS m/z calc. 721.2941, found 722.4 (M+l)+; ion time: 0.88 minutes Step P: N-(Benzenesulfonyl)[3-[2-hydroxy[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- hylpyrrolidin-l-yl]pyridinecarboxamide 0 0 0 IqsP TBSO HO ^-C&P cf3 CF3 zenesulfonyl)[3-[2-[tert-butyl(dimethyl)silyl]oxy[l- (trifluoromethyl)cy clopropy l]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (8mg, 0.0341 mmol) was then dissolved in THF (0.4 mL), cooled to 0 °C, and tetrabutylammomum fluoride (1M in THF, imately 0.17mL, 0.1705 mmol) was added by syringe. After 5 minutes the reaction was allowed to warm to room temperature. After 20 minutes at room temperature the reaction mixture was poured into 10 mL 1M citric acid, and extracted 3 x lOmL ethyl e. The combined SUBSTITUTE SHEET (RULE 26) organics were washed with brine, dned over sodium sulfate, and concentrated. The resulting crude material was then ed twice by silica gel chromatography eluting with a 0-10% methanol in dichloromethane to give zenesulfonyl)[3-[2- hydroxy[l-(trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (3 mg, 14%) ESI-MS m/z calc. 607.20764, found 608.3 (M+l)+; Retention time: 1.99 s, 'h NMR (400 MHz, DMSO) 5 12.51 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 7.97 (d, J = 7.5 Hz, 2H), 7.79 (d, J = 8.2 Hz, 1H), 7.70 (d, J = 9.6 Hz, 1H), 7.64 (d, J = 7.9 Hz, 2H), 6.89 (d, J = 8.2 Hz, 1H), 6.11 (d, J = 2.8 Hz, 1H), 5.57 (dd, J = 5.5, 2.6 Hz, 1H), 4.40 - 4.32 (m, 1H), 2.46 - 2.39 (m, 1H), 4.20 - 4.12 (m, 1H), 3.89 (s, 1H), 2.28 (s, 1H), 2.09 (s, 1H), 1.82 (dd, J = 12.1, .5 Hz, 1H), 1.52 (d, J = 9.7 Hz, 6H), 1.43 - 1.36 (m, 1H), 1.03 - 0.89 (m,4H), 0.65 (d, J = 6.2 Hz, 3H).
Synthetic Example 39: Synthesis of Compound 39: N-(benzenesulfonyl)- 6- [3- [(lR,2S,4S)-norbomanyl] oxypyrazol-l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: tert-Butyl 3-(((lR,2S,4S)-bicyclo[2.2.1]heptanyl)oxy)-lH- pyrazole-l-carboxylate HO. H n-n-V^ ^N. O DIAD N °~V O PPh3 tert-Butyl 3-hydroxypyrazole-l-carboxylate (1.632 g, 8.860 mmol), (+)-endo- 2-norbomeol (1 g, 8.915 mmol), and nyl phosphine (2.57 g, 9.798 mmol) were combined in THE (21.98 mL) and the on was cooled in an ice bath. To the mixture was added DIAD (2 mL, 10.16 mmol) dropwise and the reaction was allowed to warm to room temperature and stir for 16 h. The mixture was evaporated and the resulting material was partitioned between ethyl acetate (30 mL) and IN sodium hydroxide (30 mL). The organics were ted, washed with brine (30 mL), dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography SUBSTITUTE SHEET (RULE 26) eluting with 0-30% ethyl acetate in hexanes to give tert-butyl 3-(((lR,2S,4S)- bicyclo[2.2.1]heptanyl)oxy)-lH-pyrazole-l-carboxylate (2.08 g, 84%) ESI-MS m/z calc. 306, found 279.3 ; Retention time: 0.72 minutes. ^ NMR (400 MHz, DMSO) 6 8.05 (d, J = 3.0 Hz, 1H), 6.07 (d, J = 3.0 Hz, 1H), 4.47 (d, J = 6.8 Hz, 1H), 2.43 - 2.36 (m, 1H), 2.32 - 2.22 (m, 1H), 1.75 (td, J = 6.7, 2.4 Hz, 1H), 1.54 (s, 9H), 1.53 - 1.49 (m, 2H), 1.42 (ddt, J= 14.8, 7.8, 4.4 Hz, 2H), 1.18 - 1.07 (m, 3H).
Step B: 3-[(lR,2S,4S)-norbomanyl]oxy-lH-pyrazole H N-kAo-^ TEA ,N'NH H H tert-Butyl 3-[(lR,2S,4S)-norbomanyl]oxypyrazole-l-carboxylate (2.08 g, 7.473 mmol) was dissolved in CH2CI2 (20.80 mL) with trifluoroacetic acid (5.8 mL, 75.28 mmol) and the reaction was stirred at room temperature for 1 h. The reaction was evaporated under reduced re and the resulting oil was partitioned between ethyl acetate (50 mL) and a saturated sodium bicarbonate on (30 mL). The organics were separated, washed with brine, dried over sodium sulfate and concentrated under vacuum to give an oil, 3-[(lR,2S,4S)-norbomanyl]oxy-lH-pyrazole (1.29 g, 97%) ESI-MS m/z calc. 178.11061, found 179.2 (M+l)+; Retention time: 0.45 minutes.
Step C: tert-Butyl 2-chloro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol- 1-yl] pyridinecarboxylate DABCO H H N A A i0 A'Nn 4 N Cl cr n ci K2CO3 0 H H A 100 mL round bottom flask was charged under nitrogen with tert-butyl 2,6- dichloropyridinecarboxylate (1.796 g, 7.239 mmol), ,2S,4S)-norboman yl]oxy-lH-pyrazole (1.29 g, 7.238 mmol), and K2CO3 (1.310 g, 9.479 mmol) (freshly ground in a mortar) and anhydrous DMF (12 mL). DABCO (146 mg, 1.302 mmol) was added and the mixture was stirred at room ature under nitrogen for 8 hours. The reaction mixture was diluted with ethyl acetate (50 mL), water and brine (50 mL) and the two phases were separated. The aqueous phase was further ted with ethyl SUBSTITUTE SHEET (RULE 26) acetate (2 x 50 mL). The combined extracts were dried over sodium sulfate and the solvent removed under reduced pressure. The material was subjected to flash chromatography on silica gel using a gradient of ethyl acetate (0 to 20%) in hexanes.
The pure fractions were combined and the solvents removed under reduced pressure to provide tert-butyl 2-chloro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol-l-yl]pyridine- 3-carboxylate (1.814 g, 64%) ESI-MS m/z calc. 06, found 390.3 (M+l)+; Retention time: 0.92 minutes lH NMR (400 MHz, DMSO) 8 8.40 (d, J = 2.9 Hz, 1H), 8.32 (d, J = 8.4 Hz, 1H), 7.72 (d, J = 8.4 Hz, 1H), 6.18 (d, J = 2.9 Hz, 1H), 4.53 (d, J = 6.6 Hz, 1H), 1.88 - 1.78 (m, 1H), 2.45 (d, J = 4.6 Hz, 1H), 2.29 (t, J = 4.3 Hz, 1H), 1.56 (s, 9H), 1.55 - 1.39 (m, 4H), 1.22 - 1.08 (m, 3H).
Step D: ro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol-lyl inecarboxylic acid O 0 y N-MA A H Kl j. q__// IN N U ^ (W'T 'N' 'Cl H H tert-Butyl 2-chloro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol-lyl ]pyridinecarboxylate (1.814 g, 4.653 mmol) and TFA (5 mL, 64.90 mmol) were ed in dichloromethane (18.14 mL) and heated at 40 °C for 2 h. The reaction was evaporated. Hexanes were added and the mixture evaporated again to give a white solid, which was used in the next step without r purification. ro[3-[(lR,2S,4S)- norbomanyl]oxypyrazol-l-yl]pyndinecarboxylic acid (1.47 g, 79%) ESI-MS m/z calc. 333.088, found 334.2 (M+l)+; Retention time: 0.71 minutes.
Step E: N-(Benzenesulfonyl)chloro[3-[(lR,2S,4S)-norbornan yl]oxypyrazol-l-yl]pyridinecarboxainide so2nh2 0 9 QP 'OH %■ H GDI AnAn l/l H 7 N^rAciH H H 2-Chloro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol-l-yl]pyridine carboxylic acid (100 mg)) and GDI (52 mg, 0.323 mmol) were combined in THF and SUBSTITUTE SHEET (RULE 26) stirred for 2 hours at room temperature. Benzene sulfonamide (52 mg, 0.331 mmol) and DBU (0.048 mL, 0.323 mmol) were then added and the reaction was stirred an additional 2 hours at room temperature. The reaction mixture was then poured into 20 mL 1 M citric acid and extracted 3x 20 mL ethyl acetate. The combined organics were washed with water, then brine, dried over sodium sulfate, and concentrated to give approximately 122 mg N-(benzenesulfonyl)chloro[3-[(lR,2S,4S)-norboman yl]oxypyrazol-l-yl]pyridinecarboxamide which was used in the next step without further purification. ESI-MS m/z calc. 490.12, found 491.3 (M+l)+; Retention time: 0.75 s.
Step F: N-(Benzenesulfonyl)[3-[(lR,2S,4S)-norboman yl]oxypyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide flifc -,lC£po oo HN HCI k2co3 trv ] zenesulfonyl)chloro[3-[(lR,2S,4S)-norbomanyl]oxypyrazol- l-yl]pyridinecarboxamide (120 mg, 0.2537 mmol), (4S)-2,2,4-trimethylpyrrolidine (hydrochlonde salt) (113.9 mg, 0.7611 mmol), and potassium carbonate (210.3 mg, 1.522 mmol) were combined in 0.423 mL DMSO in a screwcap vial and heated to 130 °C for 16 hours. The reaction mixture was then cooled to room temperature, and 3 mL of water was added, resulting in the formation of a precipitate. After 30 minutes, the liquid portion was removed by syringe and discarded, and the remaining solids were dissolved in 15 mL ethyl e. The organics were washed with 15 mL 1M citric acid, and the aqueous layer was extracted an additional time with 15 mL ethyl acetate. The combined organics were washed with brine, dried over sodium sulfate and concentrated.
The crude material was ed by column chromatography on silica gel using a gradient of 0-10% methanol in dichloromethane. The pure fractions were combined and concentrated to give N-(benzenesulfonyl)[3-[(lR,2S,4S)-norbomanyl]oxypyrazoll-yl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (59 mg, 40%).
ESI-MS m/z calc. 549.24, found 550.4 ; Retention time: 2.37 minutes 'fl NMR (400 MHz, DMSO) 5 12.50 (s, 1H), 8.17 (d, J = 2.8 Hz, 1H), 8.02 - 7.97 (m, 2H), 7.80 (d, J = 8.2 Hz, 1H), 7.77 - 7.70 (m, 1H), 7.70 - 7.63 (m, 2H), 6.90 (d, J = 8.2 Hz, 1H), SUBSTITUTE SHEET (RULE 26) 6.08 (d, J = 2.7 Hz, 1H), 4.49 (d, J = 6.7 Hz, 1H), 2.46 - 2.37 (m, 2H), 2.27 (q, J = 10.0, 7.6 Hz, 2H), 2.09 (dq, J = 11.6, 5.9, 5.4 Hz, 1H), 1.86 - 1.77 (m, 1H), 1.56 (d, J = 6.9 Hz, 1H), 1.53 (s, 3H), 1.51 (s, 3H), 1.46 (dd, J = 13.8, 6.6 Hz, 3H), 1.36 (t, J = 12.1 Hz, 1H), 1.25 - 1.06 (m, 4H), 0.64 (d, J = 6.3 Hz, 3H).
Synthetic Example 40: Synthesis of Compound 40: N-(Benzenesulfonyl)- 6- [3- [ [(lR,4R)-norbomanyl] methoxyjpyrazol- l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: tert-butyl 3-[[(lS,4R)-norbornanyl]methoxy]pyrazole-l- carboxylate HOx^N OPm N-N OH + DIAD ^ o PPh3 tert-Butyl 3-hydroxypyrazole-l-carboxylate (1.327 g, 7.204 mmol),[(lS,4R)- norbomanyl]methanol (1 g, 7.924 mmol) (mixture of endo and exo), and triphenyl phosphine (2.09 g, 7.968 mmol) were combined in THF (17.87 mL) and the on was cooled in an ice bath. To the mixture was added DIAD (1.627 mL, 8.263 mmol) dropwise and the reaction was allowed to warm to room temperature and d for 72 h. The mixture was evaporated and the resulting al was partitioned between ethyl acetate (50 mL) and IN sodium hydroxide (50 mL). The organics were separated, washed with brine, dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-30% ethyl e in hexanes to give tert-butyl 3-[[(lS,4R)-norbomanyl]methoxy]pyrazole-l-carboxylate (1.698 g, 81%) ESI-MS m/z calc. 292.17868, found 293.3 (M+l)+; Retention time: 0.77 s. (2 diastereomers - mix of endo and exo substituted norbomane) 'H NMR (400 MHz, DMSO) 5 8.06 (d, J = 2.9 Hz, 1H), 6.10 (dd, J = 2.9, TO Hz, 1H), 4.23 - 3.81 (m, 2H), 2.29-2.15 (m, 2H), 1.69 (dq, J= 12.1, 4.2 Hz, 1H), 1.54 (d, J = 1.4 Hz, 9H), 1.51 -1.03 (m, 7H), 0.75 (dd, J = 5.0, 2.4 Hz, 1H).
Step B: 3-[[(lS,4R)-norbornanyl]methoxy]-lH-pyrazole SUBSTITUTE SHEET (RULE 26) hay~ H TFA H N-N N-NH H H tert-Butyl 3-[[(lS,4R)-norbomanyl]methoxy]pyrazole-l-carboxylate (1.698 g, 5.808 mmol) was dissolved in CH2CI2 (16.98 mL) with trifluoroacetic acid (approximately 6.622 g, 4.474 mL, 58.08 mmol) and the reaction was stirred at room temperature for 2 h. The reaction was evaporated and the resulting oil was partitioned between ethyl e (50 mL) and a saturated sodium bicarbonate solution (30 mL).
The organics were separated, washed with bnne, dried over sodium sulfate and concentrated under vacuum to give an oil, 3-[[(lS,4R)-norbomanyl]methoxy]-lH- pyrazole (1.11 g, 99%) ESI-MS m/z calc. 192.12627, found 193.2 (M+l)+; Retention time: 0.52 minutes.
Step C: tert-Butyl 2-chloro[3-[[(lS,4R)-norbornan yl]methoxy]pyrazol-l-yl]pyridinecarboxylate dho DABCO cr n ci H' K2CO3 A round bottom flask was charged under nitrogen with 3-[[(l S,4R)- norbomanyl]methoxy]-lH-pyrazole (1.11 g, 5.774 mmol) (mix of two diastereomers), tert-butyl 2,6-dichloropyndmecarboxylate (1.433 g, 5.776 mmol), K2CO3 (1.05 g, 7.597 mmol) (freshly ground in a mortar) and anhydrous DMF (10 mL).
DABCO (117 mg, 1.043 mmol) was added and the mixture was stirred at room temperature under nitrogen for 16 hours. The reaction mixture was diluted with ethyl acetate (50 mL) and water (50 mL) and the two phases were separated. The aqueous phase was further extracted with ethyl acetate (2 x 30 mL). The combined extracts were washed with brine, dried over sodium e and the solvent removed under reduced pressure. The material was subjected to flash chromatography on silica gel using a gradient of ethyl e (0 to 20% ) in hexanes. The pure fractions were combined and the solvents d under d pressure to provide tert-butyl 2-chloro[3- [[(lS,4R)-norbomanyl]methoxy]pyrazol-l-yl]pyridinecarboxylate (1.88 g, 81%) ESI-MS m/z calc. 403.16626, found 404.3 ; ion time: 0.94 minutes SUBSTITUTE SHEET (RULE 26) Step D: 2-Chloro[3-[[(lS,4R)-norbornanyl]methoxy]pyrazol-l- idinecarboxylic acid TFA *H .H P-J*'*} ] tert-Butyl 2-chloro[3-[[(lS,4R)-norbomanyl]methoxy]pyrazol-l- idinecarboxylate (1.88 g, 4.655 mmol) and TFA (5 mL. 64.90 mmol) were combined in dichloromethane (18.80 mL) and heated at 40 °C for 2 h. The reaction was ated. Hexanes were added and the mixture evaporated again to give a white solid 2-chloro[3-[[(lS.4R)-norbomanyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (1.58 g, 98%) ESI-MS m/z calc. 347.10367, found 348.2 (M+l)+; Retention time: 0.75 minutes.
Step E: N-(Benzenesulfonyl)chloro[3-[[(lR,4R)-norbornan yl] methoxy] py razol- 1-yl] pyridinecarb oxamidea so2nh2 OH vH xy-o9 q, P ci n H' DBU H' 2-Chloro[3-[[(lS,4R)-norbomanyl]methoxy]pyrazol-l-yl]pyndine carboxylic acid (100 mg, 0.2875 mmol) and GDI (60.59 mg, 0.3737 mmol) were stirred in THF (0.5 mL) at room temperature for 2 hours. Benzenesulfonamide (50 mg, 0.3181 mmol) was then added, followed by DBU (0.05588 mL, 0.3737) and the reaction was stirred an additional 4 hours at room temperature. The reaction mixture was then diluted with 25 mL ethyl acetate and poured 25 mL citric acid. The aqueous layer was extracted with an additional 25 mL ethyl acetate, and the combined organics were washed with water then brine, dried over sodium sulfate, and concentrated to give N- (benzenesulfonyl)chloro[3-[[(lR,4R)-norbomanyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (approximately 135 mg), which was used in the next step without r purification. ESI-MS m/z calc. 486.11, found 487.2 (M+l)+; ion time: 0.84 minutes.
SUBSTITUTE SHEET (RULE 26) ] Step F: N-(benzenesulfonyl)[3-[[(lR,4R)-norbornan yl] y] pyrazol- 1-yl] [(4S)-2,2,4-trimethylpy rrolidin- 1-yl] pyridine carboxamide HN HCI ivC#A °'s° (S) ?\0,P .H o k2co3 ] zenesulfonyl)chloro[3-[[(lR,4R)-norboman hoxy]pyrazol-l-yl]pyridinecarboxamide, (135 mg. 0.2772 mmol), (4S)-2,2,4- trimethylpyrrolidine (Hydrochloride salt) (124.5 mg, 0.8316 mmol), and potassium carbonate (229.8 mg, 1.663 mmol) were combined in DMSO in a screwcap vial and heated to 130 °C for 16 hours. The reaction mixture was then cooled to room temperature, and 3 mL of water was added, resulting in the formation of a precipitate.
After 30 minutes, the liquid portion was removed by syringe and discarded, and the remaining solids were dissolved in 15 mL ethyl acetate. The organics were washed with mL 1M citric acid, and the s layer was extracted an additional time with 15 mL ethyl acetate. The combined organics were washed with brine, dried over sodium sulfate and concentrated. The crude material was purified by column chromatography on silica gel using a gradient of 0-10% methanol in dichloromethane. The pure fractions were combined and concentrated to give N-(benzenesulfonyl)[3-[[(lR,4R)- anyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridine- 3-carboxamide, (mixture of endo and exo norbomane) ESI-MS m/z calc. 563.26, found 564.4 (M+l)+; Retention time: 2.45 minutes, 'h NMR (400 MHz, DMSO) 5 12.50 (s, 1H), 8.18 (dd, J = 2.8, 1.0 Hz, 1H), 7.99 (dd, J = 7.2, 1.7 Hz, 2H), 7.80 (dd, J = 8.3, 1.1 Hz, 1H), 7.79 - 7.70 (m, 1H), 7.66 (dd, J = 8.3, 6.7 Hz, 2H), 6.92 (dd, J = 8.3, 5.8 Hz, 1H), 6.12 (t, J = 2.9 Hz, 1H), 4.23 - 3.90 (m, 2H), 2.40 (t, J = 10.5 Hz, 1H), 2.35 - 2.16 (m, 4H), 2.09 (tt, J = 12.3, 6.2 Hz, 1H), 1.82 (dd, J = 11.9, 5.5 Hz, 1H), 1.73 (s, 1H), 1.52 (d, J = 9.7 Hz, 7H), 1.50 - 1.45 (m, 1H), 1.42 - 1.27 (m, 4H), 1.21 - 1.08 (m, 2H), 0.75 (ddd, J = 12.5, 5.0, 2.2 Hz, 1H), 0.64 (d, J = 6.2 Hz, 3H).
Synthetic Example 41: Synthesis of Compound 41: N- (Benzenesulfonyl)[3-(2,2-dicyclopropylethoxy) pyrazolyl][(4A)-2,2, 4- trimethylpyrrolidin-l-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) WO 64632 •OH 0 I fT 0 'N~NH ci^n^ci n-naJ< TFA 0=U DIAD, PPh3 P'C! K2CO3, DABCO O' TFA p^N' N XI PXJN N" Cl cqi, DBU svC CIH.HN'^IS *aP fT H 0 P~(j N' Cl p-^JNANiSA§i K2CO3, DMSO Step A: te/7-Butyl 3-(2,2-dicyclopropylethoxy) pyrazole-l-carboxylate OH M-nV^0 I N-nX,^ 0-tJ DIAD, PPh3 A solution of 2,2-dicyclopropylethanol (500 mg, 3.962 mmol), tert-butyl 3- hydroxypyrazole-l-carboxylate (730 mg, 3.963 mmol), and triphenylphosphane (1.1 g, 4.194 mmol) in dry THF (20.0 mL) was cooled in an ice bath, and DIAD (800.0 pL, 4.063 mmol) was slowly added under N2 atmosphere. The reaction was allowed to slowly warm to room temperature and was stirred for 16 h. The reaction e was diluted with ethyl acetate, washed with ted aqueous sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. The residue was purified by silica gel chromatography with 100% hexanes to 50% ethyl acetate in hexanes to afford tert-butyl 3-(2, 2-dicyclopropylethoxy)pyrazole-l-carboxylate (783 mg, 68%) as colorless oil.
ESI-MS m/z calc. 292.17868, found 293.3 (M+l)+; Retention time: 1.98 minutes. ^ NMR (400 MHz, Chloroform-d) 5 7.62 (d, J = 3.0 Hz, 1H), 5.67 (s, 1H), 4.13 (d, J = 5.3 Hz, 2H), 1.44 (s, 9H), 0.58 (qt, J = 8.2, 5.0 Hz, 2H), 0.36 (tt, J = 8.9, 5.6 Hz, 1H), 0.32 - 0.12 (m, 4H) 0.10 - 0.08 (m, 4H).
Step B: 3-(2,2-Dicyclopropylethoxy)-lH-pyrazole: SUBSTITUTE SHEET (RULE 26) TFA P^H A solution of tert-butyl 3-(2, 2-dicyclopropylethoxy) pyrazole-l-carboxylate (750 mg, 2.565 mmol) and trifluoroacetic acid (1.0 mL, 12.98 mmol) in dichloromethane (4 mL) was stirred for 2.5 hours. The volatiles were removed under reduced pressure and the residue was basified with saturated aqueous sodium bicarbonate and extracted with ethyl acetate. The combined extracts were dried over sodium sulfate and ated to give 3-(2.2-dicyclopropyletho\y)-1 /ole as colorless oil which was used as it is without further purification for next reaction. ESIMS m/z calc. 192.12627, found 193.3 (M+l)+; Retention time: 1.32 minutes.
Step C: fert-Butyl 2-chloro[3-(2,2-dicyclopropylethoxy) pyrazol-l-yl] pyridinecarboxylate V<9 | k2co3, dabco + A- — ^ Cl N^CI '°~U A mixture of 3-(2,2-dicyclopropylethoxy)-lH-pyrazole (493.0 mg, 2.564 mmol), fe/7-butyl 2,6-dichloropyridinecarboxylate (682.0 mg, 2.749 mmol), potassium carbonate (430.0 mg, 3.111 mmol), and azabicyclo[2.2.2]octane (60 mg, 0.5349 mmol) in DMSO (20.0 mL) was stirred at room temperature for 15 hours.
The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were washed with brine and dried over sodium e and ated. The residue was purified by silica gel column chromatography with 100% hexanes to 20% ethyl acetate in hexanes to afford utyl 2-chloro[3-(2,2- dicyclopropylethoxy)pyrazol-l-yl]pyridinecarboxylate (680 mg, 66%) as colorless oil. ESI-MS m/z calc. 403.16626, found 404.4 (M+l)+; ion time: 2.49 minutes. lH NMR (400 MHz, Chloroform-d) 5 8.35 (d, J = 2.8 Hz, 1H), 8.18 (d, J = 8.4 Hz, 1H), 7.70 (d, J = 8.5 Hz, 1H), 5.98 (d, J = 2.9 Hz, 1H), 4.32 (d, J = 5.6 Hz, 2H), 1.61 (s, 9H), 0.92 - 0.75 (m, 2H), 0.70 - 0.56 (m, 1H), 0.54 - 0.36 (m, 4H), 0.32 - 0.13 (m, 4H).
] Step D: 2-Chloro[3-(2,2-dicyclopropylethoxy) pyrazol-l-yl] pyridine- 3-carboxylic acid SUBSTITUTE SHEET (RULE 26) 0 0 TFA OH ,0'^NANA'CI A solution of fer/-butyl2-chloro[3-(2,2-dicyclopropylethoxy)pyrazol-l- yl]pyridinecarboxylate (675 mg, 1.671 mmol) in trifluoroacetic acid (15 mL, 19 47 mmol) and dichloromethane (4.5 mL) was stirred for 4 hours at room ature. The solvent was evaporated, and twice the residue was taken up in THF and concentrated under vacuum to afford 2-chloro[3-(2, 2-dicyclopropylethoxy) pyrazol-l-yl] pyridinecarboxylic acid (580 mg, 100%). ESI-MS m/z calc. 347.10367, found 348.3 (M+l) +; ion time: 1.95 minutes.
Step E: N-(Benzenesulfonyl)chloro[3-(2,2-dicyclopropylethoxy) pyrazol-l-yl] pyridinecarboxamide o 40 9 q OH NH2 %- is N “o H N. U P-^QN N Cl GDI, DBU O^N N 'Cl A solution of ro[3-(2, 2-dicyclopropylethoxy) pyrazol-l-yl] pyridinecarboxylic acid (100 mg, 0.2875 mmol) and carbonyl diimidazole (60.0 mg, 0.3700 mmol) in THF (2.0 mL) was stirred for 45 minutes. Then, benzenesulfonamide (50 mg, 0.3181 mmol) and DBU (60 pL, 0.4012 mmol) were added and the reaction mixture was stirred for additional 2 hours at room temperature. The reaction mixture was quenched with saturated ammonium chloride solution and extracted with ethyl acetate. The combined c extracts were washed with brine, dried over sodium sulfate and evaporated to afford N-(benzenesulfonyl)chloro[3-(2,2- dicyclopropylethoxy)pyrazol-l-yl]pyridinecarboxamide which was used as it is for next reaction. ESI-MS m/z calc. 486.11285, found 487.4 ; Retention time: 0.79 minutes.
Step F: N-(Benzenesulfonyl)[3-(2,2-<Hcyclopropylethoxy) pyrazol yl][(4A)-2,2,4-trimethylpyrrolidin-l-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) 9 Q HN-^S; 9 Q M-S .'s N 'N 'X H M 'X H N- ° ° P-^N N XI K2CO3, DMSO A mixture of N-(benzenesulfonyl)chloro[3-(2, 2-dicyclopropylethoxy) pyrazol-l-yl] pyridinecarboxamide (140.0 mg, 0.2875 mmol), (45)-2, 2, 4- trimethylpyrrolidine (Hydrochloride salt) (145.0 mg, 0.9689 mmol), and potassium carbonate (240.0 mg, 1.737 mmol) in DMSO (2 mL) was stirred at 130 °C for 15 hours.
The reaction mixture was filtered through Whatman filter disc (puradisc 25 IF) and filtrate was ed by a reverse phase HPLC-MS method using a dual gradient run from 50-99% mobile phase B over 15.0 minutes (Mobile phase A = H2O (5 mM HCI) and Mobile phase B = CH3CN) to affordN-(benzenesulfonyl)[3-(2,2- dicyclopropylethoxy)pyrazol-l-yl][(4<S)-2,2,4-tnmethylpyrrolidin-l-yl]pyridine carboxarmde (75.9 mg, 45%) as off white solid. ESI-MS m/z calc. 563.25665, found 564.5 (M+l) +; Retention time: 2.3 minutes. *H NMR (400 MHz, Chloroform-d) 8 8.35 (d, J = 8.6 Hz, 1H), 8.21 (d, J = 2.8 Hz, 1H), 8.18 - 8.08 (m, 2H), 7.66 - 7.47 (m, 5H), .96 (d, J = 2.8 Hz, 1H), 4.32 (d, J = 5.6 Hz, 2H), 3.48 (dd, J = 10.4, 8.4 Hz, 1H), 3.08 (dd, J = 10.4, 7.6 Hz, 1H), 2.61 (dt, J = 15.3, 7.8 Hz, 1H), 2.14 (dd, J = 12.4, 7.9 Hz, 1H), 1.73 (dd, J = 12.4, 9.5 Hz, 1H), 1.36 (s, 3H), 1.28 (s, 3H), 1.20 (d, J = 6.6 Hz, 3H), 0.81 (qt, J = 8.3, 5.0 Hz, 2H), 0.61 (tt, J = 8.8, 5.6 Hz, 1H), 0.55 - 0.38 (m, 4H), 0.23 (p, J = 4.8 Hz, 4H).
Synthetic Example 42: Synthesis of Compound 42: N- nesulfonyl)[3-(3, 3-dicyclopropylpropoxy) pyrazol-l-yl][(4A)-2, 2, 4- hylpyrrolidin-l-yl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) n-nh [\ T N-N CI^N^CI 'OH TFA 0=U DIAD, PPh3 K2CO3, DABCO 'OH ,'sX) TFA H2N '0 PXjN^N""CI N c GDI, DBU VP CIH.HN'^S; ^0Xj NCi p^N N k2co3, dmso ] 3,3-dicyclopropylpropan-l-ol 0 UAIH4 'OH OH To a on of 3, 3-dicyclopropylpropanoic acid (200 mg, 1.297 mmol) in dry THF (2.000 mL) was added lithium aluminum hydride (845.0 liL of 2 M, 1.690 mmol) in an ice/water bath under N2 atmosphere slowly drop wise. The mixture was d to gradually warm to room temperature and stirred for 16 hours. The flask was again cooled in an ice-bath and sequentially quenched with water (70.0 pL. 3.886 mmol) (slowly), followed by NaOH (70.0 liL of 6 M, 0.4200 mmol), then water (200 |iL. 11.10 mmol) affording a white granular solid in the mixture. To this mixture anhydrous MgSCh was added and stirred for 10 minutes. The resultant white heterogeneous mixture was filtered through celite and the precipitate was washed with ether. The filtrate was concentrated to afford 3, 3-dicyclopropylpropan-l-ol (140 mg, 77%). ESI-MS nvz calc. 140.12012, found 141.2 ; ion time: 0.5 minutes.
Step A: fert-butyl 3-(3,3-dicyclopropylpropoxy) pyrazole-l-carboxylate Wc 'OH O-u DIAD, PPh3 SUBSTITUTE SHEET (RULE 26) A solution of 3, 3-dicyclopropylpropan-l-ol (140.0 mg, 0.9984 mmol), tyl 3-hydroxypyrazole-l-carboxylate (185.0 mg, 1.004 mmol), and triphenylphosphane (278 mg, 1.060 mmol) in dry THF (7.0 mL) was cooled in an ice bath, and DIAD (200.0 pL, 1.016 mmol) was slowly added under aN2 atmosphere. The reaction was allowed to slowly warm to room temperature and was stirred for 16 hours.
The reaction mixture was diluted with ethyl acetate, washed with saturated aqueous sodium bicarbonate on, bnne, dried over sodium sulfate, and evaporated under . The residue was purified by silica gel chromatography using 100% hexanes to 50% ethyl acetate in hexanes to afford /m-butvl 3-(3,3-dicyclopropylpropoxy)pyrazole- 1-carboxylate (255 mg, 83%) as colorless oil. ESI-MS m/z calc. 306.19434, found 307.4 (M+l) +; Retention time: 0.81 minutes.
Step B: 3-(3,3-dicyclopropylpropoxy)-lH-pyrazole <Vnn TEA OX/NH A solution of /erf-butyl 3-(3, 3-dicyclopropylpropoxy) le-l- carboxylate (255 mg, 0.8322 mmol) and trifluoroacetic acid (325.0 pL, 4.218 mmol) in dichloromethane (1 mL) was d for 2.5 hours. The volatiles were removed under vacuum to afford 3-(3, 3-dicyclopropylpropoxy)-lH-pyrazole (Trifluoroacetate salt) as colorless oil which was used as it is t further purification for next reaction. ESIMS m/z calc. 206.1419, found 207.2 (M+l)+; Retention time: 0.59 minutes.
Step C: fert-Butyl 2-chloro[3-(3, 3-dicyclopropylpropoxy) pyrazol yl] ecarboxylate •O^N. K2CO3, DABCO XX'0' U™ N "Cl Cr N'ul A mixture of fer/-butyl 2,6-dichloropyridinecarboxylate (220.0 mg, 0.8867 mmol), 3-(3,3-dicyclopropylpropoxy)-lH-pyrazole (266.0 mg, 0.8305 mmol), potassium carbonate (230 mg, 1.664 mmol) and l,4-diazabicyclo[2.2.2]octane (20 mg, 0.1783 mmol) in DMSO (10 mL) was stirred at room temperature for 15 hours. The reaction was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate and evaporated. The residue SUBSTITUTE SHEET (RULE 26) was purified by silica gel column chromatography using 100% hexanes to 20% ethyl acetate in hexanes to afford rm-butyl 2-chloro[3-(3,3-dicyclopropylpropoxy)pyrazoll-yl ]pyridinecarboxylate (245 mg, 71%) as colorless oil. ESI-MS m/z calc. 417.18192, found 418.4 (M+l)+; Retention time: 1.28 minutes.
Step D: 2-Chloro[3-(3,3-dicyclopropylpropoxy) pyrazol-l-yl] pyridinecarboxylic acid O 0 •0^w 01 -°^y 'n''c, A solution of fert-butyl 2-chloro[3-(3, 3-dicyclopropylpropoxy) pyrazol yl]pyndinecarboxylate (245.0 mg, 0.5862 mmol) in tnfluoroacetic acid (500.0 pL, 6.490 mmol) and dichloromethane (1.5 mL) was stirred for 4 hours at room temperature. The solvent was ated, and twice the e was taken up in THF and concentrated under vacuum to afford 2-chloro[3-(3,3- dicyclopropylpropoxy)pyrazol-l-yl]pyndinecarboxylic acid (204 mg, 96%) as white solid which was used as it is for the next reaction. ESI-MS m/z calc. 361.11932, found 362.3 (M+l) +; ion time: 0.8 minutes. ’El NMR (400 MHz, ol-d4) 5 8.47 - 8.32 (m, 2H), 7.73 (d, J = 8.5 Hz, 1H), 6.03 (d, J = 2.9 Hz, 1H), 4.45 (t, J = 6.7 Hz, 2H), 1.98 (q, J = 7.0 Hz, 2H), 0.75 - 0.64 (m, 2H), 0.50 - 0.39 (m, 4H), 0.35 - 0.26 (m, 1H), 0.26 - 0.19 (m, 2H), 0.15 - 0.06 (m, 2H).
Step E: N-(Benzenesulfonyl)chloro[3-(3,3-dicyclopropylpropoxy) pyrazol- 1-yl] pyridinecarboxamide 0 q. ? vO 'OH :s . ,,'s; h2N'6 NIV V\H N. ° n Cl N Cl GDI, DBU ] A solution of 2-chloro[3-(3, 3-dicyclopropylpropoxy) pyrazol yl]pyridinecarboxylic acid (50.0 mg, 0.1382 mmol) and carbonyl diimidazole (30.0 SUBSTITUTE SHEET (RULE 26) mg, 0.1850 mmol) in THF (2.0 mL) was stirred for 45 minutes. Then, benzenesulfonamide (25.0 mg. 0.1590 mmol) and DBU (30 nL. 0.2006 mmol) were added. The reaction mixture was stirred for additional 2 hours at room temperature. The reaction mixture was ed with saturated ammonium chloride solution and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate and evaporated to afford zenesulfonyl)chloro[3- (3,3-dicyclopropylpropoxy)pyrazol-l-yl]pyridinecarboxarmde (84 mg, 121%) a s light brown viscous oil which was used (considering 100% conversion) as it is for next on. ESI-MS m/z calc. 500.1285, found 501.4 (M+l) +; Retention time: 0.83 minutes, ] Step F: N-(Benzenesulfonyl)[3-(3, 3-dicyclopropylpropoxy) pyrazol yl][(45)-2,2,4-trimethylpyrrolidin-l-yl] pyridmecarboxamide *0 HCI HN'NS, lQS-0 o-ry N ClII n 6 I I H,s° k2co3i dmso P-GN N A mixture ofN-(benzenesulfonyl)chloro[3-(3,3- opropylpropoxy)pyrazol-l-yl]pyridinecarboxamide (68.0 mg, 0.1357 mmol), (4,S')-2.2.4-tn methyl idine (Hydrochloride salt) (70.0 mg, 0.4677 mmol), and potassium carbonate (115.0 mg, 0.8321 mmol) in DMSO (1 mL) was stirred at 130 °C for 15 hours. The reaction mixture was filtered through Whatman filter disc (puradisc TF) and filtrate was purified by a reverse phase HPLC-MS method using dual gradient run from 50-99% mobile phase B over 15.0 minutes (Mobile phase A=H20 (5 mM HCI) and Mobile phase B=CH3CN to afford zenesulfonyl)[3-(3,3- dicyclopropylpropoxy)pyrazol-l-yl][(45)-2,2,4-tnmethylpyrrolidin-l-yl]pyndine carboxarmde (23.1 mg, 29%). ESI-MS m/z calc. 577.2723, found 578.5 (M+l) +; Retention time: 1.0 minutes, 'h NMR (400 MHz, Chloroform-d) 6 8.36 (d, J = 8.6 Hz, 1H), 8.21 (d, J = 2.8 Hz, 1H), 8.17 (d, J = 1.2 Hz, 1H), 8.15 (d, J = 1.6 Hz, 1H), 7.64 - 7.57 (m, 2H), 7.57 - 7.51 (m, 2H), 5.94 (d, J = 2.8 Hz, 1H), 4.43 (t, J = 6.8 Hz, 2H), 3.49 (dd, J = 10.3, 8.5 Hz, 1H), 3.09 (dd, J = 10.4, 7.6 Hz, 1H), 2.70 - 2.56 (m, 1H), 2.14 (dd, J = 12.4, 7.9 Hz, 1H), 1.97 (q, J = 6.8 Hz, 2H), 1.73 (dd, J = 12.4, 9.4 Hz, 1H), 1.36 (s, SUBSTITUTE SHEET (RULE 26) 3H), 1.28 (s, 3H), 1.21 (d, J = 6.7 Hz, 3H), 0.73 - 0.59 (m, 2H), 0.50 - 0.37 (m, 4H), 0.37 - 0.29 (m, 1H), 0.24 - 0.15 (m, 2H), 0.12 - 0.07 (m, 2H).
Synthetic Example 43: Synthesis of Compound 43: hlorophenyl) yl[3-[2-[l-(trifluoromethyl) cyclopropyl] ethoxy] pyrazol-l-yl][(45)-2, 2, 4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide Step A: 2-Chloro-N-(2-chlorophenyl) sulfonyl[3-[2-[l- (trifluoromethyl) cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxamide 0 vO 9 o, p 91 ‘OH Cl o H p^jN N^'CI N- F GDI, DBU F F- F- F F Step 1: 2-Chlorobenzenesulfonyl chloride (50 pL, 0.3667 mmol) was dissolved in ammonia in methanol (150 pL of 7 M, 1.050 mmol) and d at room temperature for 30 minutes. The mixture was evaporated to dryness and re-evaporated from dichloromethane. The solids were dissolved in THF (1 mL) and DBU (60 pi.. 0.4012 mmol) was added. The mixture was stirred at 70 °C for 30 minutes to liberate any remaining ammonia from the reaction.
Step 2: 2-Chloro[3-[2-[l-(trifluoromethyl) cyclopropyl]ethoxy]pyrazol-l- yl]pyridinecarboxylic acid (100 mg, 0.2661 mmol) and yl diimidazole (53 mg, 0.3269 mmol) were combined in THF (1.000 mL) and stirred for 2 hours. At this point, this mixture was added to the sulfonamide mixture (from step-1) and the reaction was stirred for overnight at room temperature. The reaction was diluted with ethyl acetate and washed with a 1 M citric acid solution, followed by brine. The organics were separated, dried over magnesium sulfate, and evaporated to afford 2-chloro-N-(2- chlorophenyl) sulfonyl[3-[2-[l-(trifluoromethyl) cyclopropyl]ethoxy]pyrazol-lyl ]pyridinecarboxamide along with starting material and primary amide. The mixture was used as it is for the next on. ESI-MS mlz calc. 548.02997, found 549.28 (M+l) +; ion time: 0.76 minutes.
Step B: hlorophenyl) sulfonyl[3-[2-[l-(trifluoromethyl) cyclopropyl] ethoxy] pyrazol-l-yl][(4A)-2,2,4-trimethylpyrrolidinyl] pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) HCI 9 ,o 91 ,^NXN^CI w jy jl h i H' K2C03l dmso A mixture of 2-chloro-N-(2-chlorophenyl) sulfonyl[3-[2-[l- (trifluoromethyl) cyclopropyl] ethoxy] pyrazol-l-yl]pyridinecarboxamide (50.0 mg, 0.09102 mmol) (mixture as it from Step A ), (45')-2.2.4-trmiethylpyrrolidine (Hydrochloride salt) (50.0 mg, 0.3341 mmol), and potassium carbonate (80.0 mg, 0.5788 mmol) in DMSO (2.0 mL) was stirred at 130 °C for 15 hours. The reaction mixture was filtered through Whatman filter disc (puradisc 25 TF) and filtrate was purified by a e phase S method using a dual gradient run from 50-99% mobile phase B over 15.0 minutes (Mobile phase A=H20 (5 mM HCI) and Mobile phase B=CH3CN) to afford N-(2-chlorophenyl) yl[3-[2-[l-(trifluoromethyl) cyclopropyl] ethoxy[pyra/ol-l-yl |(4,S)-2.2.4-trirnethylpyrrolidin-l-yl | pyridine carboxamide (18.7 mg, 31%). ESI-MS m/z calc. 625.17377, found 626.5 (M+l) +; ion time: 2.35 minutes, 'll NMR (400 MHz, Chloroform-d) 5 8.43 - 8.36 (m, 1H), 8.32 (d, J = 8.5 Hz, 1H), 8.21 (d, J = 2.8 Hz, 1H), 7.55 (d, J = 8.6 Hz, 1H), 7.53 - 7.44 (m, 3H), 5.94 (d, J = 2.7 Hz, 1H), 4.40 (t, J = 7.1 Hz, 2H), 3.51 (t, J = 9.5 Hz, 1H), 3.13 (dd, J = 10.6, 8.1 Hz, 1H), 2.73 - 2.55 (m, 1H), 2.16 (dd, J = 12.4, 7.7 Hz, 1H), 2.09 (t, J = 7.1 Hz, 2H), 1.77 (dd, J = 12 5, 9 6 Hz, 1H), 1 47 (s, 3H), 1.41 (s, 3H), 1.20 (d, J = 6.6 Hz, 3H), 1.09 - 0.97 (m, 2H), 0.81 - 0.64 (m, 2H).
Synthetic Example 44: Synthesis of Compound 44: ation of (S)- N-(phenylsulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-lyl )(2,2,4-trimethylpyrrolidin-l-yl)nicotinamide Step A: Methyl 6-(3-(2-(l-(trifhioromethyl)cyclopropyl)ethoxy)-lH- pyrazol-i-yl)chloropyridinecarboxylate N-m Cl Fsf Cl Cl •CL,N F NH SUBSTITUTE SHEET (RULE 26) WO 64632 2017/054611 To the solution of 3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazole (720 mg, 3.27 mmol) and methyl chloropyridinecarboxylate (742 mg, 3.60 mmol) in N,N-dimethylformamide (11 mL) were added potassium carbonate (9.36 g, 9.82 mmol) and 1,4-diazabicyclo [2.2.2]octane (110 mg, 0.98 mmol). The resulting solution was heated at 80 °C for 16 hours. The reaction on was cooled to room temperature and diluted with diethyl ether (400 mL). Then water (50 mL) was added, and the organic layers were separated. The organic layers were washed with IN s hydrogen chloride solution (15 mL), brine (3x15 mL), dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography using 0 - 20% hexanes - ethyl acetate to afford methyl 6-(3-(2-(l- (trifluoromethyl)cy clopropyl)ethoxy)- IH-pyrazol-1 -yl)chloropyridinecarboxylate (631 mg, 49%) as a white solid. ^NMR (250MHz, CDC13) 5 (ppm): 8.84 (s, 1H), 8.36 (d, J = 2.8Hz, 1H), 7.85 (s, 1H), 5.96 (d, J = 2.8Hz, 1H), 4.42 (t, J = 7.0Hz, 2H), 3.96 (s, 3H), 2.11 (t, J = 7.0Hz, 2H), 1.05 (m, 2H), 0.76 (m, 2H). ESI-MS m/z calc. 389.1 found 390.0 (Ml). Retention time: 7.08 minutes.
Step B: 6-(3-(2-(l-(Trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l- yl)chloropyridinecarb oxylic acid rr0O NT OH ,N'N 01 p^Jn-n 01 Methyl 6-(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl) chloropyndinecarboxylate(553 mg, 1.42 mmol) was dissolved in a mixture of tetrahydrofuran (3.5 mL) and methanol (3.5 mL), then 2N aqueous sodium hydroxide solution (1.4 mL, 2.84 mmol) was added. The resulting solution was d at room temperature for 3 hours. All solvents were removed under the reduced pressure. The residue was acidified with IN aqueous hydrogen chloride solution until pH value reached 2 and it was then extracted with ethyl e (3 x 80 mL). The organic layer was washed with brine (2 x 20 mL), dried over magnesium sulfate, filtered and SUBSTITUTE SHEET (RULE 26) concentrated to afford 2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-lyl )chloropyridinecarboxylic acid (534 mg, 97%) as a white solid, 'ft NMR (250MHz, DMSO) 5 (ppm): 8.85 (s, 1H), 8.51 (d, J = 3.0Hz, 1H), 7.76 (s, 1H), 6.21 (d, J = 3.0Hz, 1H), 4.36 (t, J = 7.0Hz, 2H), 2.11 (t, J = 7.0Hz, 2H), 0.95(m, 2H), 0.90 (m, 2H). ESI-MS m/z calc. 375.1 found 376.0 (M+l)+ Retention time: 5.80 minutes.
Step C: 4-Chloro-N-(phenylsulfonyl)(3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)nicotinamide -s;jD rf°H Qs^ 1X1 H2N Q n-n Cl 6-(3-(2-(l-(Trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl) chloropyridinecarboxylic acid (528 mg, 1.40mmol) and l,r-carbonyldiimidazole (341 mg, 2.11 mmol) in tetrahydrofuran (9 mL) was stirred for 2 hours at room temperature, then benzenesulfonamide (220 mg, 1.40 mmol) and 1,8- diazabicyclo[5.4.0]undecene (641 mg, 4.21 mmol) were added. The reaction solution was stirred for additional 16 hours and diluted with ethyl acetate (200 mL). The on was washed with saturated aqueous tartaric acid solution (25 mL), water (40 mL), brine (40 mL), then dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography using ethyl acetate to afford 4-chloro-N- (phenylsulfonyl)(3-(2-(l-(trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-lyl )nicotinamide (411 mg, 57%) as a white solid, 'h NMR z, DMSO) 5 (ppm): 8.56 (s, 1H), 8.47 (d, J = 2.5Hz, 1H), 7.94 (d, J = 6.8Hz, 2H), 7.67 (s, 1H), 7.59 (m, 3H), 6.16 (d, J = 2.5Hz, 1H), 4 34 (t, J = 7.0Hz, 2H), 2.09 (t, J = 7.0Hz, 2H), 0.94 (m, 2H), 0.89 (m, 2H). ESI-MS m/z calc. 514.1 found 515.0 (Ml). Retention time: 6.31 minutes.
Step D: (Phenylsulfonyl)(3-(2-(l- uoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)(2,2,4- trimethylpyrrolidin-l-yl)nicotinaiiiide SUBSTITUTE SHEET (RULE 26) kO 0 0 9 o. D n b H n '6 H H U H HN N'N’ 'Cl IS) To the on of 4-chloro-N-(phenylsulfonyl)(3-(2-(l- (trifluoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)nicotinamide (54.6 mg, 0.11 mmol) in dimethyl sulfoxide (0.5 mL) were added (S)-2,2,4-tnmethylpyrrolidine hydrochloride (96 mg, 0 64 mmol) and cesium fluoride (97 mg, 0.64 mmol). The resulting solution was heated at 120 °C for 48 hours. The mixture was purified by reverse phase HPLC using 5-100% water-acetomtnle (containing 0.1% tnfluoroacetic acid). Pure ons were ed and lyophilized to afford the product as trifluoroacetic acid salt, which was re-dissolved in 50% water-acetonitrile (0.1% hydrogen chloride) and lyophilized again to afford (S)-N-(phenylsulfonyl)(3-(2-(l- uoromethyl)cyclopropyl)ethoxy)-lH-pyrazol-l-yl)(2,2,4-trimethylpyrrolidin-lyl )nicotinamide hydrogen chloride salt (27.8 mg, 42%). 'h NMR (250MHz, DMSO) 5 (ppm): 12.65 (s, 1H), 8.40 (s, 1H), 8.08 (d, J = 1.3Hz, 1H), 8.02 (d, J = 8.0Hz, 2H), 7.70 (m, 3H), 7.24 (s, 1H), 6.05 (dd, J = 1.3, 2.5Hz, 1H), 4.34 (d, J = 7.0Hz, 2H), 2.50 (m, 3H), 2.09 (t, J = 7.0Hz, 2H), 2.10 (m, 1H), 1.91 (m, 1H), 1.53 (s, 6H), 0.87 (m, 2H), 0.84 (m, 2H), 0.64 (d, J = 6.0Hz, 3H). ESI-MS m/z calc. 591.2 found 592.6 (Ml).
Retention time: 2.88 s.
Synthetic Example 45: Synthesis of Compound 45,46, 47: N- (amino(oxo)(phenyl)-A,6-sulfaneylidene)(3-((l- (trifluoromethyl)cyclopropyl)methoxy)-lH-pyrazol-l-yl)((S)-2,2,4- trimethylpyrrolidin-l-yl)nicotinamide isomer 1 and isomer 2 Step A: tert-Butyl 2,6-difluoropyridinecarboxylate I O O | o 0 k DMAP, 2-methyl-THF '% OH 'o F N F F N F SUBSTITUTE SHEET (RULE 26) WO 64632 2,6-Difluoropyndinecarboxylic acid (1.0 g, 6.3 mmol) was dissolved in anhydrous 2-methyl tetrahydrofuran (12 mL). Di-rm-butyl dicarbonate (1.5 g, 6.9 mmol) was added in one n followed by 4-(dimethylamino)pyridine (462 mg, 3.78 mmol). The mixture became a slurry, with large amount of gas evolution. The heterogeneous mixture was stirred at room temperature over d and then diluted with methyl utvl ether (30 mL). The organic layer was washed successively with 1 M aqueous HC1 (10 mL), 5% w/v saturated aqueous sodium bicarbonate solution (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 0-30% ethyl e in es, to provide /m-butyl 2,6-difluoropyridinecarboxylate (360 mg, 26% yield) as a light yellow oil. 1HNMR(300 MHz, CDCl3)ppm 1.60 (s, 9 H), 6.88 (ddd, J= 8.4, 3.0, 0.5 Hz, 1 H), 8.40- 8.47 (m, 1 H). 19F NMR (282 MHz, CDC13) ppm -61.5 - -61.4 (m, IF), -60.3 (t, J= 8.6 Hz, 1 F).
Step B: tot-Butyl 2-fluoro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate CW/N'NH F F ^ "O P N N F K2CO3, DMSO F N F F /e/7-Butyl 2,6-difluoropyridinecarboxylate (1.8 g, 8.4 mmol), 3-[[l- (tnfluoromethvl)cvclopropvl xv |-1 //-pvrazole (1.8 g, 8.7 mmol) and freshly ground potassium carbonate (1.7 g, 12 mmol) were added to anhydrous dimethylsulfoxide (20 mL). The mixture was stirred at 20 °C under nitrogen for 16 hours and then diluted with ethyl acetate (100 mL). The organic layer was washed with water (3 x 30 mL), dned over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was ed by silica gel chromatography, eluting with 0- % ethyl acetate in heptanes, to afford fert-butyl 2-fluoro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate (1.9 g, 57% yield) as a white solid. 'H NMR (300 MHz, CDCI3) ppm 0.92 - 0.99 (m, 2 H), 1.13- 1.18 (m, 2 H), 1.60 (s, 9 H), 4.40 (s, 2 H), 6.00 (d, J= 2.8 Hz, 1 H), 7.61 (d, J= 8.4 Hz, SUBSTITUTE SHEET (RULE 26) 1 H), 8.30 (d, J= 2.8 Hz, 1 H), 8.37 (t, /= 8.4 Hz, 1 H). 19F NMR (282 MHz, CDCI3) ppm -69.7 (s, 3 F), -62.2 (d, 9.2 Hz, 1 F). LCMS: [M+H]+ = 402.1.
Step C: 2-Fluoro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol- l-yl]pyridinecarboxylic acid O k 0 o OH 0-^ n^n^f DCM qV'N^N^F F F- F F F F Trifluoroacetic acid (4 mL) was added to a solution of fert-butyl 2-fluoro [3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate (1.9 g, 4.7 mmol) in dichloromethane (16 mL). The mixture was stirred at 40 °C for 4 hours, after which TLC showed full conversion. The mixture was concentrated under reduced re and the e was triturated with heptanes, filtered and dried under high vacuum to provide 2-fluoro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxylic acid (1.6 g, 98% yield) as a white solid. 'H NMR (300 MHz, DMSO-ie) ppm 1.06- 1.11 (m,4H), 4.39 (s,2H), 6.24 (d,/=2.8Hz, 1 H), 7.66 (dd,J = 8.3, 1.0 Hz, 1 H), 8.43 (d,J= 8.3 Hz, 1 H), 8.47 (dd,J= 9.6, 8.4 Hz, 1 H). 19FNMR (282 MHz, DMSO-<4) ppm -67.9 (s, 3 F), -63.2 (d, J= 7.9 Hz, 1 F). LCMS: [M+H]+ = 346.1.
Step D: 2-Fluoro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol- 1-yl] pyridinecarboxamide O O •%- 'OH 1) oxalyl chloride NH2 OV N N F ^FM yk. 2) NH3 F F F F F F To a suspension of 2-fluoro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (1.6 g, 4.6 mmol) in dichloromethane (20 mL) was added one drop of At ,V-di methyl formamide ed by the dropwise addition of oxalyl chlonde (0.52 mL, 6.0 mmol). The reaction SUBSTITUTE SHEET (RULE 26) was stirred at room temperature for two hours until bubbling had stopped. The solvent was removed under reduced pressure. The resulting white solid was dissolved in anhydrous tetrahydrofuran (10 mL) and added to a mixture of 28% ammonium hydroxide (10 mL) and tetrahydrofuran (5 mL) cooled with an ice-water bath. The reaction was stirred at room temperature for 1 hour and then diluted with ethyl acetate (100 mL), washed with water (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under d pressure to afford 2-fluoro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (1.55 g, 98% yield) as a white solid. ^ NMR (300 MHz, CDC13) ppml.06-1.11 (m,4H),4.38 (s, 2 H), 6.21 (d, J= 2.7 Hz, 1 H), 7.61 (dd, J= 8.2, 1.5 Hz, 1 H), 7.76 (d, J= 8.2 Hz, 2 H), 8.33 (dd, T= 9.4, 8.4 Hz, 1 H), 8.4 (d, J= 2.7 Hz, 1 H). 19F NMR (282 MHz, CDCL,) .9 (s, 3 F), -66.3 (d, 8.9 Hz, 1 F). LCMS: [M+H]+ = 345.1.
StepE: 2-Fluoro-iY-phenylsulfanyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O Ph'S'S'Ph O NH2 N'S N A A H <1^-0 N F MeCN, Pyridine N F F3C f3c Bromine (0.14 mL, 2.7 mmol) was slowly added to a suspension of diphenyl disulfide (596 mg, 2.73 mmol) in anhydrous acetonitrile (4 mL) at 0 °C. The reaction was stirred at room temperature for 2 minutes to afford a solution. This solution was added to a on of 2-fluoro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazoll-yl inecarboxamide (940 mg, 2.73 mmol) in anhydrous acetonitrile (4 mL) and pyridine (4 mL) at 0 °C. The resulting dark mixture was stirred at room temperature ght, then concentrated under reduced pressure and porated with toluene (10 mL). The residual brown solid was purified by silica gel chromatography, eluting with 0-30% ethyl acetate in heptanes, to give ro-/V-phenylsulfanyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (380 mg, 31%yield) as a white solid. ^ NMR (300 MHz, CDCf) ppm0.93 - 1.00 , 1.14 - 1.19 (m, 2 H), 4.41 (s, 2 H), 6.03 (d, /= 3.0 Hz, 1 H), 7.20 - 7.26 (m, 1 H), 7.30 - 7.42 (m, 4 H), 7.73 (dd, J= 8.4, 1.8 Hz, 1 H), 7.84 (d, J= 15 Hz, 1 H), 8.28 (d, J= 3.0 Hz, 1 H), 8.63 (t, 9.0 Hz, 1 H). LCMS: [M+H]+ = 453.0.
SUBSTITUTE SHEET (RULE 26) WO 64632 Step F: rac-A,-(benzenesulfinyl)fluoio[3-[| l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide 0 0 0 ll ii N'S m-CPBA N'S N A A H N F □CM <Lp-tJn A -A H N F F3c f3c flieta-Chloroperoxybenzoic acid (469 mg of 77%, 2.1 mmol) was added to a solution of 2-fluoro-iV-phenylsulfanyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (860 mg, 1.90 mmol) in dichloromethane (30 mL) at 0 °C and the reaction was stirred at the same temperature for 1 hour. The reaction mixture was diluted with dichloromethane (70 mL), washed successively with 10% w/v sodium thiosulfate, 5% w/v sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and trated under reduced pressure. The residue was purified by silica gel tography, eluting with 0- 40% ethyl acetate in heptanes, to afford racemic /V-(ben/enesulfinyl)fluoro|3-|| I - (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (700 mg, 78% yield) as a white solid. ^ NMR (300 MHz, CDC13) ppm0.92- 1.00 (m,2H), 1.13 -1.20 (m, 2 H), 4.38 (d, J= 12.1, 1 H), 4.43 (d, J= 12.1, 1 H), 6.03 (d, J= 3.0 Hz, 1 H), 7.56 - 7.65 (m, 3 H), 7.70 - 7.78 (m, 1 H), 7.80 - 7.88 (m, 2 H), 8.24 (d, J= 3.0 Hz, 1 H), 8.40 (d, J= 12 Hz, 1 H), 9.61 (t, 8.5 Hz, 1 H). 19F NMR (282 MHz, CDC13) ppm -69.7 (s, 3 F), -63.2 (t, J= 10.5 Hz, 1 F). LCMS: [M+H]+ = 469.0.
Step G: .V-(Benzenesulfinyl)[3-[[l- |ti’ifluoromcthyl)cyclopi’opyl|mcthoxy|pyiazol-l-yl|[(4lS)-2,2,4- hylpyrrolidin-l-yl]pyridinecarboxamide MCI ,—\(S) O 0 Y'OII II NaH, DMF F3C f3c 2,2,4-Trimethylpyrrohdine hydrochloride (400 mg, 2.67 mmol) was dissolved in anhydrous /V.A'-dimethylformamide (10 mL), the mixture was cooled with SUBSTITUTE SHEET (RULE 26) WO 64632 ice-water and sodium hydride (287 mg of 60% dispersion in mineral oil, 7.2 mmol) was added. The reaction was stirred at room temperature for 10 minutes and cooled back to 0 °C. A solution of racemic ZV-(benzenesulfinyl)fluoro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (960 mg, 2.05 mmol) in AOV-dimethylformamide (10 mL) was added. After stirring at room temperature for 10 s the reaction e was stirred at 50 °C for 3 hours (LCMS showed 60% sion; there was a lot of unconsumed sodium hydride). Anhydrous tetrahydrofuran (0.5 mL) was added and the reaction was stirred at room temperature ght. The mixture was quenched with water (10 mL) at 0 °C then extracted with ethyl acetate (80 mL). The organic layer was washed with water (3 x 20 mL), brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The e was purified by silica gel chromatography, eluting with 0-50% ethyl acetate in heptanes. The fractions containing product and starting material were combined and concentrated under reduced pressure. The e was d in a mixture of heptanes (10 mL) and dichloromethane (10 mL) for 30 minutes and then filtered. The filtrate was concentrated under reduced pressure to afford Ar-(benzenesulfinyl)[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide (500 mg, 88% purity by LCMS, 38% yield) as ayellow solid that was used in the following step without further purification. LCMS: [M+H]+ = 562.2.
Step H: Synthesis of A-(phenylsulfonimidoyl)[3-[[l- !trifluoromethyl)cyclopi’opyl]methoxy]pyrazol-l-yl][(4A)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide O 0 9 o4jnh NCS, IMH3 in dioxane N'S- % MeCN H XX » f3c f3c Ammonia (7.8 mL of a 0.5 M solution in dioxane, 3.9 mmol) was added to a solution of A-(benzenesu]finyl)[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazoll-yl ][(45)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (500 mg, 88% , 0.78 mmol) in anhydrous acetonitrile (20 mL) at 0 °C. A-Chlorosuccinimide (120 mg, 0.90 mmol) was added in one portion (the mixture turned orange) and the reaction was stirred at 0 °C for one hour. Additional Ar-chlorosuccinimide (12 mg, 0.090 SUBSTITUTE SHEET (RULE 26) mmol) was added and the reaction was stirred at the same temperature for 30 minutes and then quenched with 10% w/v aqueous sodium thiosulfate solution and extracted with ethyl acetate (50 mL). The organic layer was washed sively with 5% w/v aqueous sodium bicarbonate solution, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 5-45% ethyl acetate in heptanes, to afford a white solid (300 mg) which was triturated with acetonitrile (3 mL) to provide a diastereomeric mixture ofN-(amino(oxo)(phenyl)-X6-sulfaneylidene)(3-((l- (trifluoromethyl)cyclopropyl)methoxy)-lH-pyrazol-l-yl)((S)-2,2,4- trimethylpyrrolidin-l-yl)nicotinamide (Compound 45) (180 mg, 97% purity by LCMS, 38%yield) as a white solid. ^ NMR (300 MHz, CDC13) ppm0.89 - 1.06 (m, 5 H), 1.08 -1.18 (m, 2 H), 1.59 - 1.72 (m, 7 H), 1.81 - 1.95 (m, 1 H), 2.07 - 2.40 (m, 1 H), 2.59 - 2.71 (m, 0.4 H), 2.82 - 2.96 (m, 0.6 H), 3.18 (t, J= 10.6 Hz, 0.4 H), 3.29 (t, J= 10.7 Hz, 0.6 H), 4.32 - 4.43 (m, 2 H), 5.90 (d, /= 2.6 Hz, 1 H), 6.25 (br. s., 2 H), 6.90 - 6.97 (m, 1 H), 7.48 - 7.57 (m, 2 H), 7.58 - 7.66 (m, 1 H), 7.97 - 8.09 (m, 2.6 H), 8.16 (d, /= 8.2 Hz, 0.4 H), 8.20 - 8.23 (m, 1 H). 19F NMR (282 MHz, CDC13) ppm -69.7 (s, 3 F).
LCMS: = 577.2.
The isomers were separated by chiral supercritical fluid chromatography utilizing a Phenomenex Lux-1 (250 x 21.2 mm), 5pm column and g with 20 % MeOH, 80 % C02 with a flow rate of 70 ute.
] Diastereoisomer 1 (Compound 46): >98 % de ESI-MS mJz calc. 576.2131, found 577.4 (M+l) + ; Retention time: 1.82 minutes; 'll NMR (400 MHz, DMSO-d6) 5 8.19 (d, J = 2.7 Hz, 1H), 7.95 (dd, J = 8.1, 1.6 Hz, 3H), 7.75 (s, 2H), 7.68 - 7.49 (m, 3H), 6.85 (d, J = 8.1 Hz, 1H), 6.12 (d, J = 2.7 Hz, 1H), 4.35 (s, 2H), 3.08 (t, J = 10.6 Hz, 1H), 2.76 (dd, J = 10.6, 7.1 Hz, 1H), 2.21 (dq, J = 12 1, 6 2 Hz, 1H), 1 97 - 1.79 (m, 1H), 1.55 (d, J = 1.7 Hz, 6H), 1.44 (t, J = 12.0 Hz, 1H), 1.09 (dd, J = 4.5, 3.1 Hz, 4H), 0.92 (d, J = 6.2 Hz, 3H).
Diastereoisomer 2 (Compound 47): >98% de. ESI-MS m!z calc. 576.2131, found 577.3 (M+l) + ; Retention time: 1.81 minutes; 1HNMR (400 MHz, DMSO-d6) 5 8.19 (d, J = 2.7 Hz, 1H), 7.99 - 7.88 (m, 3H), 7.79 (s, 2H), 7.69 - 7.55 (m, 3H), 6.87 (d, J = 8.1 Hz, 1H), 6.11 (d, J = 2.7 Hz, 1H), 4.44 - 4.28 (m, 2H), 2.63 (t, J = 10.8 Hz, 1H), SUBSTITUTE SHEET (RULE 26) WO 64632 2.21 - 2.03 (m, 1H), 1.80 (dd, J = 11.8, 5.4 Hz, 1H), 1.52 (d, J = 1.7 Hz, 6H), 1.33 (t, J = 12.2 Hz, 2H), 1.09 (dt, J = 5.6, 2.1 Hz, 4H), 0.71 (d, J = 6.2 Hz, 3H).
Synthetic Example 46: Synthesis of Compound 48: N-(Benzenesulfonyl)[3-(cyclopropoxy)pyrazol-l-yl][(4S)-2,2,4- hylpyrrolidin-l-ylJpyridinecarboxamide Step A: fert-Butyl 3-cyclopropoxy-LH-pyrazole-l-carboxylate n H °VN- □BAD, toluene Y XjNBoc °n^N. 110°C T NBoc ] To a solution of cyclopropanol (30.8 mg, 0.531 mmol), fert-butyl 2,3- dihydrooxopyrazole-l-carboxylate (97.7 mg, 0.531 mmol) and triphenylphosphine (139.3 mg, 0.531 mmol) in anhydrous toluene (2 mL) was added di-/e/7-butyl azodicarboxylate (122.2 mg, 0.531 mmol). The solution was purged with argon for 1 minute, and stirred at ambient temperature for 30 minutes. Then the reaction solution was heated at 110 °C for additional 5 hours before it cooled to ambient temperature. The soluhon was diluted with ether (50 mL), washed with NaOH aqueous solution, brine, dried over sodium e, filtered and concentrated under the reduced pressure. Residue obtained was purified by silica gel chromatography (hexane and ethyl acetate, 0 to 10% ethyl acetate gradient) to afford fert-butyl opropoxy-li7-pyrazole-l-carboxylate (52mg, 46%) as a white solid. ESI-MS m/z calc. 224.116, found 225.0 (M+l)+; Retention time: 4.38 minutes, 'h NMR (250 MHz, CDCT) 6 (ppm) 7.86 (d, J = 2.8Hz, 1H), 5.93 (d, J = 2.8Hz, 1H), .15 (m, 1H), 1.61 (s, 9H), 0.85-0.72 (m, 4H).
Step B: 3-Cyclopropoxy-l//-pyrazole Y TFA Y 0>*N ------- - °n^N.
T NBoc CH2CI2 To a solution of utyl 3-cyclopropoxypyrazole-l-carboxylate (131 mg, 0.584 mmol) in dichloromethane (6 mL) was added TFA (667 mg, 0.38 mL, 5.84 mmol). The resulting solution was stirred at ambient temperature for 3 hours. All solvents were removed under the reduced pressure. The residue ed was dissolved in ether (100 mL), washed with saturated sodium bicarbonate aqueous solution, dried over magnesium sulfate, filtered and concentrated under the reduced pressure to afford SUBSTITUTE SHEET (RULE 26) 3-cyclopropoxy-lff-pyrazole as a pale yellow oil. Crude product obtained was directly used in next step.
Step 3: fetf-Butyl 2-chloro(3-cyclopropoxy-l//-pyrazole-l-yl)pyridirie- 3-carboxylate Y Cl N Cl 7 DABCO, K2C03 o.
LNH DMF N=< 0 Crude 3-cyclopropoxy-lif-pyrazole (73mg, 0.584 mmol), tert-butyl 2,6- dichloro pyridinecarboxylate (159 mg, 0.643 mmol), K2CO3 (162mg, 1.17 mmol) and DABCO (13 mg, 0.117 mmol) were dissolved in anhydrous DMF (1.5 mL). The reaction solution was stirred at ambient temperature for 16 hours. The on solution was diluted with ether (100 mL), washed with water (3 x 25 mL) and brine (25 mL).
Organic layers were separated, dried over magnesium sulfate, filtered and concentrated under the reduced pressure. Residue ed was purified by silica gel chromatography (hexane and dichloromethane, 0 to 100% dichloromethane gradient) to afford /erf-butyl 2-chloro(3-cyclopropoxy-li/-pyrazole-l-yl)pyridinecarboxylate (153 mg, 78%) as a sticky oil. ESI-MS m/z calc. 335.104, found 336.1 (M+l)+; Retention time: 6.84 minutes.
Step D: 2-Chloro(3-cyclopropoxy-l//-pyrazole-l-yl)pyridine carboxylic acid Y Y VN- /TA Pf N-f VA TFA 0. ch2ci2 rV/vf N=< 0 N=< OH Cl Cl To a solution of /e/V-butyl ro('3-cyclopropox\ -1 a/ole-1 - yl)pyridinecarboxylate (153 mg, 0.456 mmol) in dichloromethane (2.2 mL) was added TFA (519 mg, 0.35 mL, 4.56 mmol). The resulting solution was stirred at t temperature for 48 hours. Then 1,2-dichloroethane (2 mL) was added, and all solvents were removed under the d pressure. White solid obtained was suspended in the mixture of hexane and ether (10 mL, hexane/ether, 19/1), sonicated, filtered.
SUBSTITUTE SHEET (RULE 26) washed with hexane (10 mL) and dried to afford 2-chloro(3-cyclopropo\y-l//- pyrazole-l-yl)pyndinecarboxylic acid (122 mg, 97%) as a white solid. ESI-MS m/z calc. 279.041, found 279.9 (M+l)+; Retention time: 4.43 minutes. ]H NMR (500 MHz, DMSO-d6) 5 (ppm) 13.6 (s, 1H), 8.43 (d, J = 3.0Hz, 1H), 8.39 (d, J = 8.5Hz, 1H), 7.72 (d, J = 8.5Hz, 1H), 6.28 (d,J = 3.0Hz, 1H), 4.16-4.13 (m, 1H), 0.79-0.71 (m, 4H).
Step E: N-(Benzenesulfonyl)chloro[3-(cyclopropoxy)pyrazol-l- yl]pyridinecarboxainide O q, 9 q, <{ N H2N'Sb 'S; N XI b-0 N Cl ro[3-(cyclopropoxy)pyrazol-l-yl]pyridinecarboxylic acid (30 mg, 0.1073 mmol) in DMF (600.0 |iL). HATU (l-[bis(dimethylamino)methylene]-lH- 1.2.3- lo[4,5-b]pyridinium 3-oxide uorophosphate) (85 mg, 0.2235 mmol), and DIEA (diisopropylethylamine) (38 uL. 0.2182 mmol) were combined and stirred at room temperature for 16 h. The reaction mixture was filtered and purified on reverse phase HPLC utilizing a gradient of 25-75% acetonitrile in water containing 5mM HC1 to giveN-(benzenesulfonyl)chloro[3-(cyclopropoxy)pyrazol-l-yl]pyridine carboxarmde !H NMR (400 MHz, DMSO-d6) 5 12.97 (s, 1H), 8.43 (d, J = 2.8 Hz, 1H), 8.12 (d, J = 8.3 Hz, 1H), 8.01 (d, J = 9.4 Hz, 2H), 7.77 (s, 1H), 7.69 (d, J = 6.0 Hz, 3H), 6.31 - 6.26 (m, 1H), 4.16 (s, 1H), 0.76 (s, 4H). ESI-MS m/z calc. 418.05026, found 419.0 (M+l)+; Retention time: 1.63 minutes (3 min run).
Step F: N-(Benzenesulfonyl)[3-(cyclopropoxy)pyrazol-l-yl][(4S)- 2.2.4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 1 N . rS' <1 N N '} b^QN N XI H O °<j N JiX A mixture of N-(benzenesulfonyl)chloro[3-(cyclopropoxy)pyrazol-l- yl]pyndinecarboxamide, (4S)-2,2,4-tnmethylpyrrolidme (Hydrochloride salt) (approximately 24.01 mg, 0.1604 mmol), CsF ximately 36.00 mg, 0.2370 mmol).
SUBSTITUTE SHEET (RULE 26) K2CO3 (approximately 72.01 mg, 0.5210 mmol) in DMSO (0.5 mL) was stirred at 140 °C for 16 h. The reaction was ed and purified on ed on reverse phase HPLC utilizing a gradient of 25-75% acetonitrile in water containing 5mM HC1 to give N- (benzenesulfonyl)[3-(cyclopropoxy)pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl inecarboxamide (5.6 mg, 11% over 2 steps) ESI-MS m/z calc. 495.19403, found 496.0 (M+l)+; Retention time: 1.98 minutes] (3 min run) Synthetic Example 47: Synthesis of Compound 49: 7V-(3- methoxyphenyl)sulfonyl [3- [ [l-(trifluoromethyl)cyclopropyl] methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: (l-(Trifluoromethyl)cyclopropyl)methanol OH orOH l-(Trifluoromethyl)cyclopropane-l-carboxylic acid (858 mg, 5.57 mmol, 1.00 eq.) was dissolved in diethyl ether (15 mL). The reaction mixture was cooled to 0 °C. Lithium aluminum hydride (274 mg, 7.24 mmol, 1.30 eq.) was added portionwise.
The reaction mixture was stirred ght and allowed to reach room temperature. The reaction mixture was cooled to 0 °C. HC1 (aq., 1 N, 25 mL) was added dropwise. The aqueous phase was extracted with diethyl ether (2 x 25 mL). The combined organic phases were washed with brine (25 mL), dried over sodium sulfate, filtered and evaporated in vacuo (Tbath < 30 °C) to give (l-(trifluoromethyl)cyclopropyl)methanol (547 mg, 3.90 mmol, 70% yield) as a colorless oil. ^ NMR (CDC13): d 3.73 (s, 2H), 1.58 (br, 1H), 1.07-1.01 (m, 2H), 0.82-0.75 (m, 2H).
Step B: l-(3-hydroxypyrazol-l-yl)ethanone HO-^JN'NH n-iA ho—<rJi A 100 mL round bottom flask equipped with a stir bar and a condenser was charged with lH-pyrazolol (4.97 g, 59.11 mmol) and ne (25 mL, 309.1 mmol).
The e was d at 95 °C. A solution of acetic anhydride (5.6 mL, 59.35 mmol) in pyridine (10 mL, 123.6 mmol) was added dropwise over a period of 3 minutes. The mixture was then stirred at 95 °C for an onal three hours. The solvents were SUBSTITUTE SHEET (RULE 26) removed under reduced pressure. The solid residue was triturated in 40 mL of diethyl ether, filtered, washed with diethyl ether and dried to give l-(3-hydroxypyrazol-l- yl)ethanone (6.96 g, 93%). ^ NMR (400 MHz, DMSO-d6) 5 10.96 (s, 1H), 8.13 (d, J = 3.0 Hz, 1H), 6.01 (d, J = 3.0 Hz, 1H), 2.48 (s, 3H).
Step C: (l-(trifluoromethyl)cyclopropyl)methoxy)-lH-pyrazol-l- yl)ethan-l-one o O nn'J' N'lA f-vOH + HO-^J ] 1 -(3-Hydrox} -1 //-pyrazol-1 -\ l)ethan-1 -one (443 mg, 3.51 mmol, 1.00 eq.) was ved in THF (8 mL). (l-(Trifluoromethyl)cyclopropyl)methanol (547 mg, 3.90 mmol, 1.11 eq.) and triphenyl phosphine (1.10 g, 4.21 mmol, 1.20 eq.) were added. The reaction mixture was cooled to 0 °C. Diisopropyl azodicarboxylate (829 mL, 851 mg, 4.21 mmol, 1.20 eq.) was added se (maintaining temperature < 5 °C). The reaction mixture was stirred at room ature over the weekend. Evaporation of the volatiles in vacuo gave a slightly yellow oil (2.88 g). The crude material was purified by silica gel chromatography eluting with 0-25% ethyl acetate in heptanes to give l-(3-((litnnuoromethyl )cyclopropYl)metho\y)-l//-pyrazol-1 -yl)ethanone (701 mg, 2.82 mmol, 80% yield) as a white solid. ^ NMR (CDC13): 5 8.06 (d, 1H), 5.99 (d, 1H), 4.36 (d, 2H), 2.57 (s, 3H), 1.18-1.12 (m, 2H), 0.98-0.90 (m, 2H). 19F NMR (CDC13): 5 - 69.77.
Step D: 3-((l-(Trifluoromethyl)cyclopropyl)methoxy)-l//-pyrazole <^°~vN'lA <^°~v•N'NH f3c F3c l-(3-((l-(Trifluoromethyl)cyclopropyl)methoxy)-l//-pyrazol-l-yl)ethan-l- one (695 mg, 2.80 mmol, 1.00 eq.) was dissolved in MeOH (30 mL). NaOH (aq., 30%, 421 mL, 560 mg, 4.20 mmol, 1.50 eq.) was added. The reaction mixture was stirred at room temperature overnight. Evaporation of the volatiles in vacuo gave a white solid (940 mg). The residue was partitioned between ethyl acetate (25 mL) and water (25 mL). The s phase was extracted with ethyl e (2 x 25 mL). The combined SUBSTITUTE SHEET (RULE 26) organic phases were washed with brine (25 mL), dried over sodium sulfate, filtered and evaporated in vacuo to give 3-((l-(trifluoromethyl)cyclopropyl)methoxy)-lif-pyrazole (548 mg, 2.66 mmol, 95% yield) as a slightly yellow oil. JH NMR (CDC13): 5 9.10 (hr, 1H), 7.36 (d, 1H), 5.77 (d, 1H), 4.29 (s, 2H), 1.14-1.08 (m, 2H), 0.96-0.89 (m, 2H). 19F NMR (CDC13): 5 .
Step E: fert-Butyl 2-chloro[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate <U°-C!N-NH cr n ci f3c f3o /erz-Butyl 2,6-dichloropyridinecarboxylate (approximately 451.3 mg, 1.819 mmol), 3-| 11 -(tri niioromethyljcyclopropyl [melhoxy|-1 //-pyra/ole (375 mg, 1.819 mmol), and potassium carbonate ximately 301.7 mg, 2.183 mmol) (freshly ground) were combined in anhydrous DMSO (9.026 mL). l,4-diazabicyclo[2.2.2]octane (approximately 40.81 mg, 0.3638 mmol) was added and the mixture was stirred at room temperature under nitrogen for 16 hours. The on mixture was diluted with ethyl acetate (10 mL) and water (2x5 mL) and the two phases were separated. The organics were washed with brine, dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-30% ethyl e in hexanes to give tert-butyl 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylate (620 mg, 82%) ESI-MS in z calc. 417.1067, found 418.1 (M+l) +; Retention time: 0.85 s.
Step F: 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol- l-yl]pyridinecarboxylic acid <LP-G N' Cl NN f3c F3C /e/7-But}'I 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol- l-yl]pyndinecarboxylate (620 mg, 1.484 mmol) and TFA (approximately 1.692 g.
SUBSTITUTE SHEET (RULE 26) 1.143 mL, 14.84 mmol) were combined in DCM (5 mL) and heated at 40 °C f or 16 h.
The reaction was evaporated to a white solid. Hexanes was added and the mixture was evaporated again to give 2-chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (500 mg, 93%) ESI-MS m/z calc. 361.0441, found 362.1 (M+l)+; Retention time: 0.66 minutes.
Step G: 2-Chloro-N-(3-methoxyphenyl)sulfonyl[3-[|l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide n^n^ciH ^ N- N- F3C f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- idinecarboxylic acid (200 mg, 0.55 mmol) and carbonyldiimidazole (110 mg, 0.66 mmol) were ed in THF (2 mL) and stirred at room ature for 2 hours. 3-methoxybenzenesulfonamide (104 mg, 0.55 mmol) was added, followed by DBU (0.25 mL, 1.66 mmol) and the reaction mixture was stirred for 2 h at room temperature.
The reaction mixture was diluted with 10 mL ethyl acetate, and washed with 10 mL 1M aqueous citric acid. The aqueous layer was extracted with ethyl e (2x10 mL), and the combined organics were washed with brine, dried over sodium sulfate, and concentrated under reduced re. The crude material was purified by silica gel chromatography eluting with a 0-10% gradient of methanol in dichloromethane to give 2-chloro-N-(3-methoxyphenyl)sulfonyl[3-[[l- uoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (217 mg, 74%) ESI-MS m/z calc. 530.06384, found 531.1 (M+l)+; Retention time: 0.72 minutes.
Step H: ;V-(3-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide wV- A 9 9,P l f3c f3c SUBSTITUTE SHEET (RULE 26) 2-Chloro-N-(3-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (120 mg, 0.23 mmol), (4S')-2.2.4-trimethylpyrrolidine (hydrochloride salt) (107 mg, 0.71 mmol), and potassium carbonate (173 mg, 1.25 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction was partitioned between ethyl acetate and water.
The organics were separated, washed with a 1M citric acid solution, brine, dried over sodium sulfate and evaporated. The crude material was ed by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give ,Y-(3- methoxyphenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]- 2-[(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (92 mg, 67%) ESI-MS m/'z calc. 764, found 608.3 (M+l)+; Retention time: 2.17 minutes. 'H NMR (400 MHz, 6) 5 12.43 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 7.82 (d, J = 8.2 Hz, 1H), 7.61 - 7.53 (m, 2H), 7.48 - 7.45 (m, 1H), 7.32 - 7.27 (m, 1H), 6.92 (d, J = 8.2 Hz, 1H), 6.15 (d, J = 2.8 Hz, 1H), 4.42 - 4.31 (m, 2H), 3.84 (s, 3H), 2.45 (d, J = 10.5 Hz, 1H), 2.35 -2.28 (m, 1H), 2.20-2.03 (m, 1H), 1.84 (dd, J = 11.9, 5.6 Hz, 1H), 1.53 (d,J = .9 Hz, 6H), 1.38 (t, J= 12.1 Hz, 1H), 1.12 -1.05 (m, 4H), 0.67 (d, J = 6.2Hz, 3H).
Synthetic Example 48: Synthesis of Compound 50: 7V-(2- phenyl)sulfonyl [3-[ [ l-(trifhioromethyl)cyclopropyl] methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-iV-(2-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide o 9 a ,o f 'OH N'S' 14 H <U0-iJ N Cl <LP-iJ N ClN- f3c f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- idinecarboxylic acid (181 mg, 0.5 mmol) and carbonyldiimidazole (97 mg, 0.6 mmol) were combined in THF (2.5 mL) and stirred at room temperature for 30 minutes. robenzenesulfonamide (114 mg, 0.65 mmol) was added, followed by DBU (0.09 mL, 0.6 mmol) and the reaction mixture was stirred for 3 h at room temperature. The reaction mixture was diluted with 10 mL ethyl acetate, and washed with 10 mL 1M aqueous citric acid. The organics were dried over sodium sulfate, and concentrated SUBSTITUTE SHEET (RULE 26) under d pressure. The crude material was purified by silica gel chromatography eluting with a 0-8% gradient of methanol in dichloromethane to give 2-chloro-N-(2- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (180 mg, 69%) ESI-MS m/z calc. 518.0439, found 519.1 (M+l)+; Retention time: 0.70 minutes.
Step B: fluoiophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide O oo Fw O Oo F n ci HCly "ft* f3c f3c 2-Chloro-N-(2-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (78 mg, 0.15 mmol), (4,S')-2.2.4-tri methyl idine (hydrochloride salt) (67 mg, 0.45 mmol), and potassium carbonate (124 mg, 0.9 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction mixture was ed and punfied by LC/MS ing a gradient of 30-99% acetonitrile in 5 mM aq HC1 to give N-(2- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (28 mg, 31%) ESI-MS m/z calc. 595.1876, found 596.3 (M+l)+; Retention time: 2.08 minutes.
Synthetic Example 49: Synthesis of Compound 51: N-(3- Fluorophenyl)sulfonyl [3- [ [ l-(trifluoromethyl)cydopropyl]methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-.Y-(3-fluorophenyl)sulfonyl[3-[[ l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O 9 o, ,oW " ci <}J^JN- " f3c f3c SUBSTITUTE SHEET (RULE 26) 2-chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (181 mg, 0.5 mmol) and carbonyldiimidazole (97 mg, 0.6 mmol) were ed in THF (2.5 mL) and stirred at room temperature for 30 minutes. 3-fluorobenzenesulfonamide (114 mg, 0.65 mmol) was added, followed by DBU (0.09 mL, 0.6 mmol) and the reaction mixture was stirred for 3 h at room ature. The reaction mixture was diluted with 10 mL ethyl e, and washed with 10 mL 1M aqueous citric acid. The organics were dried over sodium sulfate, and concentrated under reduced pressure. The crude material was purified by silica gel chromatography eluting with a 0-8% gradient of methanol in dichloromethane to give 2-chloro-N-(3- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (190 mg, 73%) ESI-MS m/z calc. 518.0439, found 519.1 (M+l)+; Retention time: 0.72 minutes.
Step B: N-(3-Fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- hylpyrrolidin-l-yl]pyridinecarboxamide 9 ist 0 9P .N jTXh TJ+ 4N~N/%'XI ^ HCl7\ f3c f3c ro-N-(3-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (78 mg, 0.15 mmol), (45')-2.2.4-tri methyl pyrrolidine (hydrochloride salt) (67 mg, 0.45 mmol), and potassium carbonate (124 mg, 0.9 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction mixture was filtered and punfied by LC-MS to give A-(3-nuorophenyl)sul fonyl| 3-||l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidml-yl ]pyridinecarboxamide (47 mg, 52%) ESI-MS m/z calc. 595.1876, found 596.3 (M+l)+; Retention time: 2.14 minutes.lH NMR (400 MHz, DMSO-d6) 8 8.21 (d, J = 2.8 Hz, 1H), 7.90 - 7.82 (m, 2H), 7.80 - 7.69 (m, 2H), 7.68 - 7.59 (m, 1H), 6.93 (d, J = 8.2 Hz, 1H), 6.15 (d, J = 2.8 Hz, 1H), 4.43 - 4.27 (m, 2H), 2.44 (t, J = 10.4 Hz, 1H), 2.30 (dd, J = 10.2, 7.0 Hz, 1H), 2.23 - 2.08 (m, 1H), 1.84 (dd, J = 11.9, 5.5 Hz, 1H), SUBSTITUTE SHEET (RULE 26) 1.53 (d, J = 9.7 Hz, 6H), 1.39 (t, J = 12.2 Hz, 1H), 1.17 -1.02 (m, 4H), 0.69 (d, J = 6.3 Hz, 3H).
] Synthetic Example 50: sis of Compound 52: N-(4- phenyl)sulfonyl [3-[ [ l-(trifhioromethyl)cyclopropyl] methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-Ar-(4-fluorophenyl)sulfonyl[3-[[ l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide XxaV 'OH Cl N-N^N^CI " XX F3C f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (181 mg, 0.5 mmol) and carbonyldiimidazole (97 mg, 0.6 mmol) were combined in THF (2.5 mL) and stirred at room temperature for 30 minutes. 4-fluorobenzenesulfonamide (114 mg, 0.65 mmol) was added, followed by DBU (0.09 mL, 0.6 mmol) and the reaction mixture was stirred for 3 h at room temperature. The reaction e was diluted with 10 mL ethyl acetate, and washed with 10 mL 1M aqueous citric acid. The organics were dried over sodium sulfate, and trated under reduced pressure. The crude al was purified by silica gel chromatography eluting with a 0-8% gradient of methanol in dichloromethane to give 2-chloro-N-(4- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (160 mg, 62%) ESI-MS m/z calc. 518.0439, found 519.1 (M+l)+; Retention time: 0.72 minutes.
Step B: N-(4-Fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]inethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxaniide n m xx+11 SP (§/ ys-v [\U1.1..J-l /e> ■n'nan'x'ci hcVn f3c f3c SUBSTITUTE SHEET (RULE 26) ro-N-(4-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (78 mg, 0.15 mmol). (4S')-2.2.4-trimethylpyrrolidine (hydrochloride salt) (67 mg, 0.45 mmol), and potassium carbonate (124 mg, 0.9 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction e was filtered and punfied by LC/MS utilizing a gradient of 30-99% acetonitrile in 5 mM aq HC1 to give /V-(4- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (34 mg, 38%) ESI-MS m/z calc. 595.1876, found 596.3 (M+l)+; Retention time: 2.16 minutes. 1H NMR (400 MHz, DMSO-d6) 6 8.20 (d, J = 2.8 Hz, 1H), 8.12 - 8.02 (m, 2H), 7.82 (d, J = 8.3 Hz, 1H), 7.59 - 7.45 (m, 2H), 6.92 (d, J = 8.2 Hz, 1H), 6.15 (d, J = 2.7 Hz, 1H), 4.43 - 4.30 (m, 2H), 2.37 (t, J= 10.4 Hz, 1H), 2.22 (dd, J = 10.1, 7.0 Hz, 1H), 2.18 - 2.05 (m, 1H), 1.83 (dd, J = 11.9, 5.5 Hz, 1H), 1.52 (d, J = 8.8 Hz, 6H), 1.37 (t, J= 12.1 Hz, 1H), 1.15 - 1.00 (m, 4H), 0.67 (d, J = 6.3 Hz, 3H). tic Example 51: Synthesis of Compound 53: 7V-(2- methoxyphenyl)sulfonyl [3- [ [l-(trifluoromethyl)cyclopropyl] methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-chloro-N-(2-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O 9 o. .o 9 "OH 4. N'S' ■4 14 14 H <^J " Cl N Cl f3c f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.55 mmol) and carbonyldiimidazole (110 mg, 0.66 mmol) were combined in THE (2 mL) and stirred at room temperature for 2 hours. 2-methoxybenzenesulfonamide (104 mg, 0.55 mmol) was added, followed by DBU (0.25 mL, 1.66 mmol) and the on mixture was stirred for 2 h at room temperature.
The reaction mixture was diluted with 10 mL ethyl acetate, and washed with 10 mL 1M aqueous citric acid. The s layer was ted with ethyl acetate (2x10 mL), and the combined organics were washed with brine, dried over sodium sulfate, and SUBSTITUTE SHEET (RULE 26) concentrated under d pressure. The crude material was used without further purification. 2-Chloro-N-(2-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (286 mg, 97%) ESTMS m/z calc. 530.06384, found 531.1 (M+l)+; Retention time: 0.70 minutes.
Step B: \-(2-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 9P ? O 00 o IN'S w* Cl HCI/ f3c f3c 2-Chloro-N-(2-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (120 mg, 0.23 mmol), (4^)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (107 mg, 0.71 mmol), and ium carbonate (173 mg, 1.25 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction was partitioned between ethyl acetate and water.
The organics were separated, washed with a 1M citric acid solution, brine, dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography g with 0-10% methanol in dichloromethane to give N-(2- yphenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]- )-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (82 mg, 60%) ESI-MS m/z calc. 607.20764, found 608.3 (M+l)+; Retention time: 2.15 minutes.
Synthetic Example 52: Synthesis of Compound 54: 7V-(4- methoxyphenyl)sulfonyl [3- [ ifluoromethyl)cyclopropyl] methoxy] pyrazol yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-A'-(4-methoxyphenyl)sulfonyl[3-|[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O 9 o, ,o N- N- ciHU <lp~On n u°u N f3c f3c SUBSTITUTE SHEET (RULE 26) 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (300 mg, 0.83 mmol) and carbonyldiimidazole (162 mg, 1.0 mmol) were combined in THF (4 mL) and stirred at room temperature for 30 minutes. 4-Methoxybenzenesulfonamide (203 mg, 1.08 mmol) was added, followed by DBU (0.15 mL, 1.0 mmol) and the reaction mixture was stirred for 2 h at room temperature. The reaction mixture was diluted with 10 mL ethyl e, and washed with 10 mL 1M aqueous citric acid. The organics were dried over sodium sulfate and trated under reduced pressure. The crude material was purified by silica gel chromatography eluting with 0-8% methanol in romethane to give 2-chloro-AL(4- methoxyphenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (430 mg, 97%) ESI-MS m/z calc. 530.06384, found 531.1 (M+l)+; Retention time: 0.71 minutes.
Step B: /V-(4-Methoxyphenyl)sulfonyl[3-[[ l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 (s/ 0 QP N jfX" + HnQ irV^Vi )LP-0,N'NXNA'CI HCl/x I I f3c F3c 2-Chloro-N-(4-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (210 mg, 0.39 mmol), 2.2.4-tri methyl pyrrol i di nc (hydrochlonde salt) (180 mg, 1.2 mmol), and potassium carbonate (330 mg, 2.39 mmol) were combined in DMSO (2 mL) and heated at 130 °C for 15 h. The reaction was partitioned n ethyl acetate and water.
The organics were separated, washed with a 1M citric acid solution, brine, dried over sodium e and evaporated. The crude al was purified by silica gel chromatography eluting with 0-5% methanol in dichloromethane. The material was further ed by LC/MS utilizing a gradient of 30-99% acetonitrile in 5 mM aq HC1 to yield /V-(4-methoxyphenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyndinecarboxamide (25 mg, 10%) ESI-MS m/z calc. 607.20764, found 608.3 (M+l)+; Retention time: 2.16 minutes. 1H NMR (400 MHz, DMSO-d6) 6 8.19 (d, J = 2.8 Hz, 1H), 7.99 - 7.87 (m, 2H), 7.77 (d, J = 8.2 Hz, 1H), 7.26 - 7.10 (m, 2H), 6.91 (d, J SUBSTITUTE SHEET (RULE 26) = 8.2 Hz, 1H), 6.14 (d, J = 2.8 Hz, 1H), 4.44 - 4.28 (m, 2H), 3.84 (s, 3H), 2.40 (t, J = .5 Hz, 1H), 2.29-2.21 (m, 1H), 2.16 - 2.00 (m, 1H), 1.82 (dd. J = 11.9, 5.6 Hz, 1H), 1.52 (d, J = 10.7 Hz, 6H), 1.37 (t, J= 12.1 Hz, 1H), 1.16 -1.02 (m, 4H), 0.64 (d, J = 6.3 Hz, 3H).
Synthetic Example 53: Synthesis of Compound 55: N-(Benzenesulfonyl)- 6- [3- [ [ l-(difluoromethyl)cyclopropyl] methoxy]pyrazol-l-yl] [(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: /er/-Butyl 3-((l-(difluoromethyl)cyclopropyl)methoxy)-li/-pyrazole- 1-carboxylate HF2C~£ ^CnbocrOHo t! DIAD, Ph3P, THF 0°C to it, then 50°C °Xj Boc ] To the solution of (l-(difluoromethyl)cyclopropyl)methanol (867 mg, 7.11 mmol), fe/7-butyl 2,3-dihydrooxopyrazole-l-carboxylate (1.19 g, 6.46 mmol), and triphenylphosphine (1.86g, 7.11 mmol) in tetrahydrofuran (22 mL) at 0 °C was added diisopropyl azodicarboxylate (1.44 g, 7.11 mmol) se. After the addition was complete, the reaction was allowed to warm to room temperature, then heated at 50 °C for 1 hour. The reaction solution was cooled to room temperature and ethyl acetate (300 mL) was added. The solution was then washed with aqueous sodium hydroxide (20 mL, 1M), water, brine and dried over magnesium sulfate, filtered and concentrated. The residue obtained was purified by silica gel chromatography (hexane and dichloromethane, 0 to 100% dichloromethane nt) to afford fert-butyl (difluoromethyl)cyclopropyl) y)-1//-pyra/ole-1 - carboxylate as a white solid (1.50 g, 80% yield). ESI-MS m/z calc. 288.1, found 289.2 (M+l)+. Retention time: 3.08 minutes, 'h NMR z, CDCh) d(ppm): 7.84(d, J= 3.0Hz, 1H), 5.97(t, J= 57.8Hz, 1H), 5.89(d, /= 3.0 Hz, 1H), 4.32(s, 2H), 1.61(s, 9H), 0.97(m, 2H), 0.75(m, 2H).
SUBSTITUTE SHEET (RULE 26) Step B: 3-((l-(Difluoromethyl)cyclopropyl)methoxy)-lF-pyrazole CF2H cf2h HCI in dioxane °xy VS, M Boc o^Qih Cold hydrogen e solution (30 mL, 4.0M in 1,4-dioxane) was added to /e/7-butyl 3-((l-(difluoromethyl)cyclopropyl)methoxy)-li7-pyrazole-l-carboxylate (1.69 g, 5.88 mmol) in a round-bottom flask, and the reaction solution was warmed to room temperature and stirred for 3 hours. After removal of all the solvents under reduced pressure, the residue so obtained was partitioned n water (50 mL) and diethyl ether (80 mL). The organic layer was separated and the s layer extracted with diethyl ether (2 x 80 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over sodium sulfate, filtered and trated under reduced pressure. The residue so obtained was ed by silica gel chromatography (hexanes and ethyl acetate, 0 to 40% ethyl acetate gradient) to afford 3-((l- (difluoromethyl)cyclopropyl)methoxy)-li/-pyrazole as a white solid (997 mg, 90% yield). ESI-MS »i/z calc. 188.1, found 189.1 (M+l)+. Retention time: 1.94 minutes.
^ NMR (250MHz, DMSO) <5(ppm): 11.87(s, 1H), 7.51(m, 1H), 5.98(t. ./ = 57.0Hz. 1H), 5.66(m, 1H), 4.10(s, 2H), 0.80(m, 4H).
Step C: tert-butyl 2-chloro [3- [ [1- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylate 0^//N'NH + o-//N'NA'isrXi cr n ci f- F F F ] tert-Butyl 2,6-dichloropyridinecarboxylate (approximately 659.2 mg, 2.657 mmol), 3-[[l-(difluoromethyl)cyclopropyl]methoxy]-lH-pyrazole (500 mg, 2.657 mmol), and potassium carbonate (approximately 440.6 mg, 3.188 mmol) (freshly ) were combined in anhydrous DMSO (13.18 mL). l,4-diazabicyclo[2.2.2]octane (approximately 59.61 mg, 0.5314 mmol) was added and the mixture was stirred at room temperature under nitrogen for 16 hours. The reaction mixture was diluted with water SUBSTITUTE SHEET (RULE 26) (20 mL) and stirred for 15 min. The resulting solid was collected and washed with water. The solid was ved in dichloromethane and the small amount of aqueous layer removed. The cs were dried over sodium sulfate and evaporated to give tertbutyl 2-chloro[3-[[l-(difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridine carboxylate (842 mg, 79%). ESI-MS m/z calc. 399.11612, found 400.1 (M+l)+; Retention time: 0.82 s.
Step D: 2-chloro[3-[[l-(difluoromethyl)cyclopropyl]methoxy]pyrazol- yridinecarboxylic acid N O^'N^N rDI 0-('• N N Cl F F F F tert-Butyl 2-chloro[3-[[ 1 -(difluoromethyl)cyclopropyl]methoxy]pyrazol-1 - yl]pyridinecarboxylate (842 mg, 2.106 mmol) and TFA (approximately 2.401 g, 1.622 mL, 21.06 mmol) were dissolved in dichloromethane (8.420 mL) and heated at 40 0C for 3 h. The reaction was evaporated and the ing solid was triturated with hexanes and re-evaporated to give 2-chloro[3-[[l- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxylic acid (710 mg, 98%). ESI-MS m/z calc. 343.05353, found 344.1 (M+l)+ ; Retention time: 0.62 minutes. 1H NMR (400 MHz, DMSO-d6) 5 13.59 (s, 1H), 8.43 (d, J = 2.9 Hz, 1H), 8.39 (d, J = 8.4 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 6.22 (d, J = 2.9 Hz, 1H), 5.98 (t, J = 56.4 Hz, 1H), 4.32 (s, 2H), 0.93 - 0.84 (m, 4H).
Step E: N-(Benzenesulfonyl)chloro[3-[[l- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O Sv O^'N^N^CI F- F F F 2-Chloro[3-[[l-(difluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (200 mg, 0.5819 mmol) and carbonyl diimidazole SUBSTITUTE SHEET (RULE 26) WO 64632 (approximately 113.2 mg, 0.6983 mmol) were combined in THF (2.5 mL) and stirred for 2 h. At this point, benzenesulfonamide (approximately 91.47 mg, 0.5819 mmol) was added followed by DBU (approximately 265.8 mg, 261.1 |iL. 1.746 mmol) and the reaction was stirred for an additional 2 h at room temperature. A 1M citric acid solution (5 mL) was added and the reaction was stirred for 20 min. The solution was extracted with ethyl acetate. The organics were washed with brine, dried over sodium e and evaporated. The crude matenal was d by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(benzenesulfonyl)chloro[3-[[l- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (153.6 mg, 55%) ESI-MS m/z calc. 482.0627, found 483.1 (M+l)+; Retention time: 0.68 minutes.
Step F: N-(benzenesulfonyl)[3-[[l- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide V-O oo 9 (sjf 0-/N XX"0 +l~N'A'N''XI ^ HCI/% F F N-(Benzenesulfonyl)chloro[3-[[l- (difluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (116 mg, 0.24 mmol), (45)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (71 mg, 0.63 mmol), and potassium carbonate (145 mg, 1.05 mmol) were combined in DMSO (600 pL) and heated at 130 °C for 16 h. The reaction was partitioned between ethyl acetate and water.
The organics were separated, washed with a 1M citric acid solution, brine, dried over sodium e and evaporated. The crude al was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to giveN- nesulfony l)[3- [ [1 -(difluoromethy l)cy clopropy 1] methoxy ] py razol-1 -y 1] [(4S)-2,2,4-trimethylpyrrolidm-l-yl]pyridinecarboxamide (87 mg, 65%) ESI-MS m/z calc. 559.2065, found 560.3 ; Retention time: 2.06 minutes. 1H NMR (400 MHz, DMSO-d6) 8 12.47 (s, 1H), 8.19 (d, J = 2.8 Hz, 1H), 8.00 (t, J = 1.3 Hz, 1H), 7.98 (d, J = 1.6 Hz, 1H), 7.80 (d, J = 8.2 Hz, 1H), 7.76 - 7.70 (m, 1H), 7.65 (tt, J = 6.8, 1.6 Hz, 2H), 6.91 (d, J = 8.3 Hz, 1H), 6.13 (d, J = 2.8 Hz, 2H), 4.32 - 4.23 (m, 2H), 2.47 - 2.37 (m, 1H), 2.28 (dd, J = 10.3, 7.0 Hz, 1H), 2.09 (dq, J 11.9, 6.2 Hz, 1H), 1.82 (dd, J SUBSTITUTE SHEET (RULE 26) = 11.9, 5.6 Hz, 1H), 1.52 (d, J = 9.5 Hz, 6H), 1.36 (t, J = 12.1 Hz, 1H), 0.87 (dt, J = 5.1, 2.0 Hz, 4H), 0.65 (d, J = 6.2 Hz, 3H).
Synthetic Example 54: Synthesis of Compound 56: N-(Benzenesulfonyl)- 2-(2,2,4,4-tetramethylpyrrolidin- l-yl) [3- [ [1- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide Step A: ,4-Trimethylpentanyl)picolinamide o 0 h2n■KM ‘OH NH HATU, DIEA, DMF At 5 °C, to picolinic acid (20.5 g, 167 mmol) in DMF (200 mL) was added HATU (65 g, 171 mmol, -1.0 eq), followed by 2,4,4-trimethylpentanamine (27.0 mL, ~1.0 eq), and then DIEA (65 mL, ~2.5 eq). The reaction was stirred at RT for 1.0 h. The reaction mixture was poured to ice-water (350 mL) and extracted with EtOAc (2x600 mL). The combined extract was washed with water (2x300 mL) and brine (150 mL), dried over , and concentrated to give crude product, which was purified by plug filtration through a pad of silica gel, eluted with 25% EtOAc in hexanes, giving a lightyellow oil. N-(2,4,4-tnmethylpentanyl)picolinamide (36 g, 92%).: MS [M+l]: Step B: Pyridinyl(2,2,4,4-tetramethylpyrrolidin-l-yl)methanone O O Phl(OAc)2, Pd(OAc)2 'NH vs* PhMe, 80 °C 1 d N-(2,4,4-trimethylpentanyl)picolinamide (36.0 g, 153 mmol), Pd(OAc)2 (1.72 g, 5%), PhI(OAc)2 (99.2 g, 308 mmol) were mixed in toluene (600 mL) and heated at 80 °C overnight (~18 h). The reaction was concentrated to remove most of the e and the residue loaded to a silica gel column, eluted with 50% EtOAc in hexanes, resulting in a lightyellow solid. pyndinyl(2,2,4,4-tetramethylpyrrolidin-lyl none (32 g, 90%).; MS [M+l]: 233. 1HNMR (250 MHz, CDC13) 8.56 (d, J = 4.8 Hz, 1H), 7.76 (td, J = 7.7, 1.7 Hz, 1H), 7.61 (d, J = 7.8 Hz, 1H), 7.35 - 7.25 (m, 1H), 3.36 (s, 2H), 1.79 (s, 2H), 1.67 (s, 6H), 1.08 (s, 6H) Step C: 2,2,4,4-Tetramethylpyrrolidine (HC1 salt) SUBSTITUTE SHEET (RULE 26) V\| NaOH HN' 150 °C .HCI Pyridinyl(2,2,4,4-tetramethylpyrrolidin-l-yl)methanone (6 g, 25.9 mmol) was dissolved in a mixture of NaOH (9.0 g, 225 mmol) in water (6 mL) and EtOH (18 mL) in a small pressure reactor (~45 mL) and heated at 140 °C for 48 h. The reaction was completed. The mixture was dissolved with 80 mL of water and extracted with Et20 (3x200 mL). The extract was washed with water (2x100 mL) and dried over Mg2S04. After filtenng, HCI gas was bubbled through the filtrate for 5 min. An oil formed on the bottom of the flask. The top ether layer was ed carefully, the remaining oil was washed with ether (2x30 mL) and the washing was decanted. The final oil was evaporated to give a white olid, which was dned in a vacuum oven at 50°C for 1 day, to give an off-white solid. 2,2,4,4-tetramethylpyrrolidine (HCI salt) (4.0 g, 95%). MS [M+l]: 128. 1HNMR (250 MHz, DMSO-d6) 9.39 (s, 2H), 3.01 (t, J = .5 Hz, 2H), 1.69 (s, 2H), 1.40 (s, 6H), 1.16 (s, 6H).
Step D: zenesulfonyl)(2,2,4,4-tetramethylpyiroli(lin-l-yl)[3- [ [ l-(trifluoromethyl)cyclop ropyl] y] py razol- 1-yl] pyridinecarboxamide 9 V 0 00 N JTI h' 10 + HnO N'M^N^CI ^ HCI/x OS' OS'■XOvP F- FF N-(Benzenesulfonyl)chloro[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (100 mg, 0.2 mmol), 2,2,4,4-tetramethylpyrrolidine (hydrochloride salt) (98 mg, 0.6 mmol), and potassium carbonate (138 mg, 1.0 mmol) were combined in DMSO (500 pL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was ed. The solid was dissolved in ethyl acetate and washed with a 1M citric acid solution, then brine. The organics were dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-10% ol in dichloromethane to giveN- (benzenesulfonyl)(2,2,4,4-tetramethylpyrrolidin-l-yl)[3-[[l- SUBSTITUTE SHEET (RULE 26) (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (57 mg, 48%) ESI-MS m/z calc. 591.2127, found 592.2 (M+l)+; Retention time: 2.20 minutes. 1H NMR (400 MHz, 6) 5 12.52 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 8.04 - 7.97 (m, 2H), 7.82 - 7.72 (m, 2H), 7.72 - 7.63 (m, 2H), 6.94 (d, J = 8.0 Hz, 1H), 6.14 (d, J = 2.8 Hz, 1H), 4.36 (s, 2H), 2.38 (s, 2H), 1.72 (s, 2H), 1.58 (s, 6H), 1.14 - 1.04 (m, 4H), 0.81 (s, 6H).
Synthetic Example 55: Synthesis of Compound 57: ethoxy methyl-phenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-lyl ][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(4-methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide ii o, p OH N:'S N- H N Cl N Cl O'' f3c f3c 2-Chloro[3-[[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0.4023 mmol) and carbonyl diimidazole (82 mg, 0.5057 mmol) were combined in THF (1.299 mL) and stirred for 2 h. At this point, 4- methoxymethyl-benzenesulfonamide (85 mg, 0.4224 mmol) was added followed by DBU (200 pL, 1.337 mmol) and the reaction was stirred for an additional 2 h at room temperature. The reaction was diluted with ethyl acetate and washed with a 1M citric acid solution, ed by brine. The organics were ted, dried over sodium sulfate, and evaporated to give 2-chloro-N-(4-methoxymethyl-phenyl)sulfonyl[3- [[l-(trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (205 mg, 94%) ESI-MS m/z calc. 544.0795, found 545.0 (M+l) +; Retention time: 0.73 minutes.
Step B: N-(4-Methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide SUBSTITUTE SHEET (RULE 26) l9i 0 ooV- + HN- 5Zv0~O N Cl O HCI 0 I y-p-<J jy1 I ro-N-(4-methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (100 mg, 0.18 mmol), (4»S)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (90 mg, 0.6 mmol), and potassium ate (138 mg, 1.0 mmol) were combined in DMSO (500 pL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was decanted. The solid was dissolved in ethyl acetate and washed with a 1M citric acid solution, then brine. The organics were dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(4- methoxymethyl-phenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclopropyl]methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidinl-yl ]pyridinecarboxamide (75 mg, 67%) ESI-MS m/z calc. 327, found 622.3 (M+l)+; Retention time: 2.23 minutes.lHNMR(400 MHz, DMSO-d6) 5 12.46 (s, 1H), 8.20 (d, J = 2.8 Hz, 1H), 7.98 (d, J = 8.7 Hz, 1H), 7.78 (d, J = 8.3 Hz, 1H), 7.01 - 6.94 (m, 2H), 6.92 (d, J = 8.2 Hz, 1H), 6.14 (d, 1 = 2.1 Hz, 1H), 4.41 - 4.32 (m, 2H), 3.82 (s, 3H), 2.59 (s, 3H), 2.41 - 2.29 (m, 2H), 2.22 - 2.10 (m, 1H), 1.82 (dd, J = 11.9, 5.6 Hz, 1H), 1.52 (s, 6H), 1.36 (t, J = 12.1 Hz, 1H), 1.12 - 1.06 (m, 4H), 0.70 (d, J = 6.2 Hz, Synthetic Example 56: Synthesis of Compound 58: N-(o-Tolylsulfonyl)- 6-[3-[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: 2-Chloro-N-(o-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O 9 o, ,o 'OH N'S' N^CI H O^'N^N^CI F3c f3c SUBSTITUTE SHEET (RULE 26) WO 64632 ro[3-[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0.3992 mmol) and carbonyl diimidazole (approximately 81.69 mg, 0.5038 mmol) were combined in THF (1.339 mL) and stirred for 2 h. At this point, 2-methylbenzenesulfonamide (approximately 71.77 mg, 0.4192 mmol) was added, followed by DBU (approximately 202.6 mg, 199.0 pL, 1.331 mmol) and the reaction was stirred for an additional 2 h at room temperature. The reaction was diluted with ethyl acetate and washed with a 1 M citric acid solution, followed by brine.
The organics were separated, dried over sodium e, and evaporated to give 2- chloro-N-(o-tolylsulfonyl)[3-[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-lyl ]pyridinecarboxamide (208 mg, 99%) ESI-MS m/z calc. 528.0846, found 529.0 (M+l) +; Retention time: 0.77 minutes Step B: N-(o-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 0 00 O oo (s! % N-mA.A„.h + HN- AN, H n ci HCly N f3c f3c 2-Chloro-N-(o-tolylsulfonyl)[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridmecarboxamide (100 mg, 0.19 mmol), (45')-2.2.4-tri methyl pyrrolidine (hydrochloride salt) (90 mg, 0.6 mmol), and potassium carbonate (138 mg, 1.0 mmol) were combined in DMSO (500 pL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was decanted. The solid was dissolved in ethyl acetate and washed with a 1M citric acid solution, then brine. The organics were dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(otolylsulfonyl -[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)- trimethylpyrrolidin-l-yl]pyridinecarboxamide (69 mg, 60%) ESI-MS m/z calc. 605.22833, found 606.5 (M+l)+; Retention time: 2.33 minutes. 1H NMR (400 MHz, DMSO-d6) 5 12.63 (s, 1H), 8.22 (d, J = 2.8 Hz, 1H), 8.04 (d, J = 7.9 Hz, 1H), 7.82 (d, J = 8.2 Hz, 1H), 7.59 (td, J = 7.5, 1.5 Hz, 1H), 7.49 - 7.40 (m, 2H), 6.96 (d, J = 8.3 Hz, SUBSTITUTE SHEET (RULE 26) 1H), 6.18 (d, J = 2.7 Hz, 1H), 4.48 (s, 2H), 2.63 (s, 3H), 2.39 (d, J = 8.8 Hz, 2H), 2.35 - 2.23 (m, 2H), 2.21 - 2.04 (m, 4H), 2.02 -1.91 (m, 1H), 1.83 (dd, J= 11.9, 5.6 Hz, 1H), 1.53 (s, 6H), 1.35 (t, J = 12.1 Hz, 1H), 0.69 (d, J = 6.2 Hz, 3H). tic Example 57: Synthesis of Compound 59: N-(3- Fluorophenyl)sulfonyl [3- [ [ l-(trifluoromethyl)cyclobutyl]methoxy] pyrazol- 1-yl]- 2- [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide Step A: 2-Chloro-N-(3-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridinecarboxamide O II O, ,p 0^-n'nxn^ci N^N^CI ^ F3c f3c 2-Chloro[3-[[l-(tri£luoromethyl)cyclobutyl]methoxy]pyrazol-l- yl]pyridinecarboxylic acid (150 mg, 0.3992 mmol) and yl diimidazole (approximately 81.69 mg, 0.5038 mmol) were combined in THF (1.339 mL) and stirred for 2 h. At this point, 3-fluorobenzenesulfonamide (approximately 69.93 mg, 0.3992 mmol) was added followed by DBU (approximately 202.6 mg, 199.0 pL, 1.331 mmol) and the reaction was d for an additional 2h at room temperature. The reaction was diluted with ethyl acetate and washed with a 1M citric acid solution, followed by brine.
The organics were separated, dried over sodium sulfate, and evaporated to give 2- chloro-N-(3-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridmecarboxamide (210 mg, 99%) ESI-MS m/z calc. 95, found 533.0 (M+l)+; Retention time: 0.77 minutes.
Step B: N-(3-Fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 9,9 (S)/ 0 9,p „ jfxVlY4 hiQ a,°-csN'N^N^CI Hci/\ f3c f3c SUBSTITUTE SHEET (RULE 26) ] ro-N-(3-fluorophenyl)sulfonyl[3-[[l- (trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl]pyridinecarboxamide (100 mg, 0.19 mmol), (4S')-2.2.4-trimethylpyrrolidine (hydrochloride salt) (90 mg, 0.6 mmol), and potassium carbonate (138 mg, 1.0 mmol) were ed in DMSO (500 pL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was decanted. The solid was dissolved in ethyl acetate and washed with a 1M citric acid solution, then brine. The organics were dried over sodium sulfate and evaporated. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(3- fluorophenyl)sulfonyl[3-[[l-(trifluoromethyl)cyclobutyl]methoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (73 mg, 63%) ESI-MS m/z calc. 609.2033, found 610.2 ; Retention time: 2.27 minutes. 1H NMR (400 MHz, DMSO-d6) 5 12.63 (s, 1H), 8.22 (d, J = 2.8 Hz, 1H), 7.86 (d, J = 2.3 Hz, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.78 - 7.70 (m, 2H), 7.66 - 7.59 (m, 1H), 6.96 (d, J = 8.2 Hz, 1H), 6.18 (d, J = 2.7 Hz, 1H), 4.48 (s, 2H), 2.43 (d, J = 10.4 Hz, 1H), 2.35 - 2.25 (m, 3H), 2.19 - 2.05 (m, 4H), 1.96 (td, J = 10.0, 5.3 Hz, 1H), 1.84 (dd, J = 11.8, 5.6 Hz, 1H), 1.55 (s, 3H), 1.52 (s, 3H), 1.40 (t, J= 12.2 Hz, 1H), 0.69 (d, J = 6.2 Hz, 3H). tic Example 58: Synthesis of Compound 60: N-(Benzenesulfonyl)- 6-[3-(spiro[2.2]pentanylmethoxy)pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxamide Step A: spiro[2.2]Pentyl-methanol ‘OH ‘OH To a suspension of lithium aluminum e (888 mg, 23.4 mmol) in tetrahydrofuran (30 mL) was added spiro[2.2]pentane-l-carboxylic acid (1.75g, 15.6 mmol) in tetrahydrofuran (5 mL) dropwise over 5 minutes. The reaction was heated to 50 °C for 16 hours. The reaction was diluted with diethyl ether (20 mL) and quenched with solid sodium sulfate decahydrate. The mixture was diluted with diethyl ether (100 mL), filtered through celite pad and concentrated to give spiro [2.2] pentyl-methanol (793 mg, 52%) as an oil. ESI-MS m/z calc. 98.15 found 98.8 (M+l)+ . Retention time: SUBSTITUTE SHEET (RULE 26) 2.54 minutes. 1H NMR (250 MHz, CDC13) ppm 0.58 - 0.89 (m, 4 H) 0.91 -1.09 (m, 1 H) 1.20 - 1.37 (m, 1 H) 1.43 (m, 1 H) 3.60 (dd, J = 11.98, 6.37 Hz, 2 H) ] Step B: 3-(spiro[2.2]Pent-l-ylmethoxy)-pyrazole-l-carboxylic acid tertbutyl ester N'NA0^0 I 0 I 'OH XLP-XJ To a on of crude spiro[2.2]pent-l-yl-methanol (966 mg, 9.8 mmol) in tetrahydrofuran (40 mL) was added triphenyl phosphine (2.58 g, 9.8 mmol), 3-hydroxypyrazolecarboxylic acid utyl ester (1.64 g, 8.9 mmol). The reaction mixture was cooled in an ice bath followed by the addition of diisopropyl azodicarboxylate (1.9 mL, 9.8 mmol). The ice bath was removed and the reaction was stirred for 2 hours. The solvent was removed in vacuum and the crude mixture was purified by silica gel column chromatography using 10-20% hexanes-diethyl ether to give 3-(spiro[2.2]pent-lylmethoxy )-pyrazole-l-carboxylic acid tert-butyl ester (1.20 g, 44%) as a clear oil. ESI- MS m/z calc. 264.33 found 265.1 (M+l)+ . Retention time: 3.36 minutes Step C: 3-(spiro[2.2]Pent-l-ylmethoxy)-lH-pyrazole To 3-(spiro[2.2]pent-l-ylmethoxy)-pyrazole-l-carboxylic acid tert-butyl ester (1.2 g, 4.54 mmol) was added dichloromethane (30 mL) and trifluoroacetic acid (3.4 mL, 45 mmol). The on mixture was stirred for 2 hours at room temperature and trated to dryness in vacuum. The residue was azeotroped twice with 1,2- dichloroethane (15 mL) to give cmde 3-(spiro[2.2]pent-l-ylmethoxy)-lH-pyrazole (1.87 g, 51%) as ayellow oil. ESI-MS m/z calc. 164.09 found 164.6 (M+l)+ . Retention time: 2.11 minutes Step D: 2-Chloro[3-(spiro[2.2]pentylmethoxy) l-l-yl]- nicotinic acid methyl ester SUBSTITUTE SHEET (RULE 26) 0 'o CXLv0'UNH + O'' N^CI cr n 'ci>£ To crude ro[2.2]pent-l-ylmethoxy)-lH-pyrazole (1.87 g, assumed 4.54 mmol) was added methyl 2,6-dichloromcotinate (935 mg, 4.54 mmol), 1,4- diazabicyclo[2.2 2]octane (102 mg, 0 91 mmol), ylformamide (8 mL) and potassium carbonate (1.9 g, 13.6 mmol). The reaction was stirred for 48 hours at room temperature, diluted with diethyl ether (75 mL) and washed with water ning a small amount of brine (3 x 50 mL) and brine (50 mL). This organic layer was dried over sodium sulfate and concentrated in vacuum. The crude reaction mixture was purified by silica gel column chromatography using 0-15% s:diethyl ether to afford 2-chloro- 6-[3-(spiro[2.2]pent-l-ylmethoxy) pyrazol-l-yl]-nicotinic acid methyl ester (1.02 g, 67%) as an off-white solid. ESI-MS m/z calc. 333.09 found 333.9 (M+l)+ . Retention time: 3.85 minutes.
Step E: ro[3-(spiro[2.2]pent-l-ylmethoxy)-pyrazol-l-yl]- nicotinic acid O O O 'OH N- NU ^5 \X\__p-<J N Clyi XLP-Cj To 2-Chloro[3-(spiro[2.2]pent-l-ylmethoxy) l-l-yl]-nicotinic acid methyl ester (990 mg, 2.97 mmol) was added water (6 mL), methanol (6 mL) and tetrahydrofuran (6 mL) followed by lithium hydroxide (285 mg, 11.88 mmol). The reaction was stirred for 1 hour and 1M hydrochloric acid (12 mL) was added. Formed white solid was filtered off, washed with water and hexanes to give 2-chloro[3- (spiro[2.2]pent-l-ylmethoxy)-pyrazol-l-yl]-nicotinic acid (927 mg, 98%) as a white solid. ESI-MS m/z calc. 319.07 found 320.0 (M+l)+ . Retention time: 3.25 minutes 1H NMR (250 MHz, CDC13) ppm: 0.76 - 0.88 (m, 5 H), 1.11-1.13 (m, 1 H), 1.60-1.75 (m, 1H), 4.22 (dd, J=7.0, 3.3, Hz, 2H) 6.00 (d, J=2 5 Hz, 1H), 7.76 (d, J=8.5 Hz, 1H), 8.38 (d, J=2.5 Hz, 1H), 8.43 (d, J=8.5 Hz, 1H).
Step F: N-(benzenesulfonyl)chloro[3-(spiro[2.2]pentan ylmethoxy)pyrazol-l-yl]pyridinecarboxainide SUBSTITUTE SHEET (RULE 26) 0 QvP \XLP~iJ N ClN- N'CI ro[3-(spiro[2.2]pentanylmethoxy)pyrazol-l-yl]pyridine carboxylic acid (50 mg, 0.16 mmol) and carbonyl diimidazole (38 mg, 0.23 mmol) were combined in THF (1.5 mL) and stirred for 2 h. At this point, benzenesulfonamide (25 mg, 0.16 mmol) was added followed by DBU (70 |iL. 0.47 mmol) and the on was stirred for an additional 2 h at room temperature. The reaction was d with ethyl acetate and washed with a 1M citric acid solution, ed by brine. The organics were separated, dried over sodium sulfate, and evaporated to give N-(benzenesulfonyl) [3-(spiro[2.2]pentanylmethoxy)pyrazol-l-yl]pyridinecarboxamide (72 mg, 98%) ESI-MS m/z calc. 458.08154, found 459.2 (M+l)+; Retention time: 0.75 minutes.
Step G: N-(benzenesulfonyl)[3-(spiro[2.2]pentanylmethoxy)pyrazol- l-yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 %9 (s/ O 00 XX H TQ + HnQ 0-^N'NAN^CI ^ HciS N-(benzenesulfonyl)chloro[3-(spiro[2.2]pentanylmethoxy)pyrazol-l- yl]pyndinecarboxamide (72 mg, 0.16 mmol), (4<S,)-2,2,4-trimethylpyrrohdme (hydrochlonde salt) (63 mg, 0.42 mmol), and potassium carbonate (97 mg, 0.7 mmol) were combined in DMSO (1 mL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was decanted. The solid was dissolved in ethyl acetate and washed with a 1M citric acid solution, then brine. The organics were dried over sodium sulfate and ated. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in dichloromethane to give N-(benzenesulfonyl)[3-(spiro[2.2]pentan ylmethoxy)pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide (38 mg, 45%) ESI-MS m/z calc. 535.22534, found 536.1 (M+l)+; Retention time: 2.22 minutes.
SUBSTITUTE SHEET (RULE 26) Synthetic Example 59: sis of Compound 61: (5S) (benzenesulfonyl)-3,3,5-trimethyl(3-{2-[l- (trifluoromethyl)cyclopropyl]ethoxy}- IH-pyrazol- l-yl)-2,7,13- tricyclo[7.4.0.02,6]trideca-l(9),10,12-trienone 9 9PYn O oo N- p^QN NN- nA(S)^ F^ F Fn F F F N-(Benzenesulfonyl)[3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (52 mg, 0.08789 mmol), NaOAc (14 mg, 0.1707 mmol), water (16 pL, 0.89 mmol), and [Ir{dF(CF3)ppy}2(dtbpy)]PF6 (5 mg, 0.004 mmol) were combined in DMA (dimethylacetamide) (0.9 pL) and the reaction mixture was placed next to a 23 W CFL light source for 1.5 h. The reaction was injected directly onto a silica gel column without any workup. The crude mixture was purified by silica gel chromatography eluting with 0-100% ethyl acetate in hexanes to give (5S) (benzenesulfonyl)-3,3,5-trimethyl(3-{2-[l-(trifluoromethyl)cyclopropyl]ethoxy}- lH-pyrazol-l-yl)-2,7,13-triazatricyclo[7.4.0.02,6]trideca-l(9),10,12-trienone (10 mg, 19%) ESI-MS m/z calc. 71, found 590.3 (M+l)+; Retention time: 2.51 minutes.
^ NMR (400 MHz, DMSO-d6) 5 8.26 (d, J = 2.8 Hz, 1H), 8.06 (d, J = 7.7 Hz, 2H), 7.81 (d, J = 8.2 Hz, 1H), 7.73 (t, J = 7.4 Hz, 1H), 7.66 (t, J = 7.6 Hz, 2H), 6.97 (d, J = 8.2 Hz, 1H), 6.07 (d, J = 2.7 Hz, 1H), 6.02 (d, J = 3.9 Hz, 1H), 4.36 (t, J = 7.0 Hz, 2H), 3.04 (dt J = 12.2, 5.7 Hz, 1H), 2.17 (dd, J = 13.4, 7.3 Hz, 1H), 2.11 (m, 2H), 1.86 (d, J = 13.2 Hz, 1H), 1.71 (s, 3H), 1.61 (s, 3H), 1.21 (d, J = 6.3 Hz, 3H), 0.97 (m, 2H), 0.88 (m, 2H) Synthetic Example 60: Synthesis of Compound 62: (5S)-3,3,5-trimethyl- 12-(3-{2-[l-(trifluoromethyl)cyclopropyl]ethoxy}-lH-pyrazol-l-yl)oxa-2,13- diazatricyclo[7.4.0.02,6]trideca-l(13),9,ll-trienone Step A: 2-[l-(Trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl] [(4S)-2,2,4-trimethylpyrrolidin- 1-yl] pyridinecarboxylic acid SUBSTITUTE SHEET (RULE 26) 0 0 'OH ‘OH ,O^N N' Cl N- y-i Fv F Fv F p-tj Njy F F To a mixture of 2-chloro[3-[2-[l- (trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl]pyridinecarboxylic acid (1 g, 2.661 mmol) and (4S)-2,2,4-trimethylpyrrolidine (Hydrochloride salt) (620 mg, 4.143 mmol) in N-methylpyrrolidinone (5 mL) and 1,2-diethoxy ethane (1 mL) was added potassium carbonate (1.8 g, 13.02 mmol). The slurry was heated at 125 °C for 68 h. LC/MS showed 40% conversion. More (4S)-2,2,4-trimethylpyrrolidine (400 mg) was added and the on was continued for 18 h at 135 °C. Cooled reaction suspension to ambient temperature and added slowly to a rapidly stirred solution of HC1 (2 mL of 6 M, 12.00 mmol) in ice (foams!) affording a brown slurry. The slurry was extracted with ethyl e. The organic was washed with water, brine, dried over sodium sulfate and concentrated. The crude material was chromatographed on silica using a gradient of hexane/ethyl acetate. Product came out -30% ethyl acetate. 6-[3-[2-[l- (trifluoromethyl)cy clopropyl]ethoxy]pyrazol-1 -yl] [(4S)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxylic acid (667.8 mg, 55%). ESI-MS m/z calc. 352, found 453.0 (M+l) +; Retention time: 1.87 minutes. 1H NMR (400 MHz, DMSO-d6) 5 12.68 (s, 1H), 8.24 (s, 1H), 7.92 (d, J = 8.2 Hz, 1H), 6.86 (dd, J = 32.9, 7.9 Hz, 1H), 6.29 - 6.01 (m, 1H), 4.31 (s, 2H), 3.54 (s, 1H), 2.89 (s, 1H), 2.33 (s, 1H), 2.08 (s, 2H), 1.95 (s, 1H), 1.74 - 1.46 (m, 7H), 1.12 - 1.01 (m, 3H), 0.92 (d, J = 29.3 Hz, 4H).
Step B: (5S)-3,3^-Trimethyl(3-{2-[l- (trifluoromethyl)cyclopropyl]ethoxy}-lH-pyrazol-l-yl)oxa-2,13- diazatricyclo[7.4.0.02,6]trideca-l(13),9,ll-trienone O O •OH 'Y o p^N ; p-uN. nT'nAis) F F F. F F F 6-[3-[2-[l-(Trifluoromethyl)cyclopropyl]ethoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxylic acid (50 mg, 0.1105 mmol), water (20 SUBSTITUTE SHEET (RULE 26) HL, 1.110 mmol),NaOAc (18 mg, 0.22 mmol), and [Ir{dF(CF3)ppy}2(dtbpy)]PF6 (4 mg, 0.003565 mmol) were dissolved in DMA (0.9 mL) and the reaction mixture was placed next to a 23 W CFL light source for 1.5 h. The reaction was injected directly onto a silica gel column without any workup. The crude mixture was purified by silica gel tography g with 0-100% ethyl acetate in hexanes to give (5S)-3,3,5- trimethyl- 12-(3 - {2- [1 -(trifluoromethy l)cy clopropy 1] ethoxy} -1 H-pyrazol-1 -yl)oxa- 2,13-diazatricyclo[7.4.0.02,6]trideca-l(13),9,ll-trienone (30.8 mg, 62%) ESI-MS m/z calc. 450.18787, found 451.3 (M+l)+; Retention time: 2.35 minutes.
Synthetic Example 61: Synthesis of Compound 63: N-(Benzenesulfonyl)- 6- [3- [(3-fluoro- l-bicyclo[l. 1.1] pentanyl)methoxy] pyrazol- l-yl|[(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: tert-Butyl 3-[(3-fluoro bicyclo [ 1.1.1] yl)methoxy] py razole- oxylate ^3% N- F OH + HO— OW A on of (3-fluoro-l-bicyclo[l.l.l]pentanyl)methanol (0.27 g, 2.3 mmol), tert-butyl 3-hydroxypyrazole-l-carboxylate (0.46 g, 2.5 mmol), and triphenyl phosphine (0.67 g, 2.6 mmol) in THF (12 mL) was cooled in an ice bath, and isopropyl N-isopropoxycarbonylimmocarbamate (0.50 mL, 2.6 mmol) was slowly added. The reaction was allowed to slowly warm to room temperature and was stirred for three days. It was diluted with ethyl acetate, washed with saturated s sodium bicarbonate, dried over sodium sulfate, and evaporated under vacuum. The residue was ed by silica gel tography with 0-40% ethyl acetate in hexanes to give tertbutyl 3-[(3-fluoro-l-bicyclo[l.l.l]pentanyl)methoxy]pyrazole-l-carboxylate (0.43 g, 66%) ESI-MS m/i calc. 282.13797, found 283.3 (M+l)1. Retention time: 0.65 minutes.
Step B: 3-[(3-Fluoro-l-bicyclo|l.l.l]pentanyl)methoxy]-lH-pyrazole F- N.
F >N'NH OW O W A solution of utyl 3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]pyrazole-l-carboxylate(0.43 g, 1.523 mmol) and SUBSTITUTE SHEET (RULE 26) trifluoroacetic acid (587 (xL, 7.62 mmol) in dichloromethane (4 mL) was stirred for 5 hours. The volatiles were removed under vacuum, and the residue was basified with saturated aqueous sodium bicarbonate and ted with ethyl e. The combined extracts were dried over sodium sulfate and evaporated to give 3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]-lH-pyrazole (0.28 g, 100%) ESI-MS m/z calc. 182.08554, found 183.1 (M+l)+ ; Retention time: 0.39 minutes.
Step C: tert-Butyl 2-chloro[3-[(3-fluoro-l- o[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxylate -N- F NH + oW F An N Cl Cl N Cl oW A mixture of 3-[(3-fluoro-l-bicyclo[l.l.l]pentanyl)methoxy]-lH-pyrazole (0.28 g, 1.5 mmol), tert-butyl chloropyridinecarboxylate (0.38 g, 1.5 mmol), potassium carbonate (0.26 g, 1.9 mmol), and l,4-diazabicyclo[2.2.2]octane (34 mg, 0.30 mmol) in DMSO (7.5 mL) was d at room temperature for 16 h. The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were washed with brine and water, dried over sodium sulfate, and evaporated. The residue was ed by silica gel chromatography with 0-5% methanol in dichloromethane to give tert-butyl 2-chloro[3-[(3-fluoro-l-bicyclo[l.l.l]pentanyl)methoxy]pyrazol-lyl ]pyridinecarboxylate (0.50 g, 85%) ESI-MS m/z calc. 393.12555, found 394.2 (M+l) +; Retention time: 0.86 minutes.
Step D: 2-Chloro[3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyri(linecarboxylic acid v<0 I o F- An N Cl✓5 F An N Cl OW 0 W A solution of tert-butyl 2-chloro[3-[(3-fluoro-l- o[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxylate (0.50 g, 1.270 mmol) and trifluoroacetic acid (978 pL, 12.7 mmol) in dichloromethane (6 mL) was stirred for 15 hours. The solvent was evaporated, and the residue was taken up in SUBSTITUTE SHEET (RULE 26) acetonitrile. The solvent was evaporated to give 2-chloro[3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxylicacid (0.43 g, 100%) ESI-MS m/z calc. 296, found 338.1 (M+l)+ ; Retention time: 0.63 minutes 1H NMR (400 MHz, Chloroform-d) 5 8.43 (d, J = 8.5 Hz, 1H), 8.39 (d, J = 2.9 Hz, 1H), 7.73 (d, J = 8.5 Hz, 1H), 6.00 (d, J = 2.8 Hz, 1H), 4.51 (s, 2H), 2.13 (d, J = 2.6 Hz, 6H).
Step E: N-(benzenesulfonyl)chloro[3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxamide O 9 Q„P N''S' H % F- N Cl F- ,n'n N XI OW oW 2-Chloro[3-[(3-fluoro-l-bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l- idinecarboxylic acid (100 mg, 0.3 mmol) and carbonyl diimidazole (58 mg, 0.36 mmol) were combined in THF (1.5 mL) and stirred for 2 h. At this point, benzenesulfonamide (61 mg, 0.39 mmol) was added followed by DBU (54 |iL 0.36 mmol) and the on was stirred for an additional 16 h at room temperature. The reaction was dduted with ethyl acetate and washed with a 1M citric acid solution, ed by brine. The organics were separated, dried over sodium sulfate, and evaporated to give N-(benzenesulfonyl)chloro[3-[(3-fluoro-lbicyclo [l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (190 mg, 135%) ESI-MS m/z calc. 476.07214, found 477.2 (M+l)+; Retention time: 0.69 minutes.
Step F: N-(Benzenesulfonyl)[3-[(3-fluoro-l- bicyclo|l.l.l]pentanyl)methoxy]pyrazol-l-yl][(4S)-2,2,4-trimethylpyrrolidin-l- yl] pyridinecarb oxamide N /X H T) + HnQ F- n-n^n^ci W hciA N-(Benzenesulfonyl)chloro[3-[(3-fluoro-l- bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l-yl]pyridinecarboxamide (140 mg, 0.29 mmol), (45)-2,2,4-trimethylpyrrolidine (hydrochloride salt) (131 mg, 0.88 mmol), and potassium carbonate (243 mg, 1.76 mmol) were combined in DMSO (1.5 mL) and SUBSTITUTE SHEET (RULE 26) heated at 130 °C for 16 h. The reaction mixture was filtered and punfied by LC/MS utilizing a gradient of 30-99% acetonitrile in 5 mM aq HC1 to give N-(benzenesulfonyl)- 6-[3-[(3-fluoro-l-bicyclo[l.l.l]pentanyl)methoxy]pyrazol-l-yl][(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide (89 mg, 54%) ESI-MS m/z calc. 553.2159, found 554.4 (M+l)+; Retention time: 2.16 minutes. 1HNMR (400 MHz, DMSO-d6) 5 8.19 (d, J = 2.8 Hz, 1H), 8.05 - 7.95 (m, 2H), 7.81 (d, J = 8.3 Hz, 1H), 7.77 - 7.70 (m, 1H), 7.70 - 7.61 (m, 2H), 6.91 (d, J = 8.2 Hz, 1H), 6.14 (d, J = 2.8 Hz, 1H), 4.47 (s, 2H), 2.45 - 2.36 (m, 1H), 2.31 - 2.22 (m, 1H), 2.15 - 2.08 (m, 7H), 1.82 (dd, J = 11.9, 5.5 Hz, 1H), 1.52 (d, J = 9.2 Hz, 6H), 1.36 (t, J = 12.1 Hz, 1H), 0.64 (d, J = 6.2 Hz, 3H) ] Synthetic Example 62: Synthesis of Compound 64: N- (Benzenesulfonyl)[3-(dispiro[2.0.2.1]heptanylmethoxy)pyrazol-l-yl][(4S)- 2,2,4-trimethylpyrrolidin-l-yl]pyridinecarboxamide Step A: totf-Butyl 3-(dispiro[2.0.2.1]heptanyl methoxy)-l//-pyrazole-l- carboxylate NUrBoc OH p-V A solution of dispiro[2.0.2.1]heptanyl methanol (1.36 g, 11.0 mmol) (Meijere, et al., Eur. J. Org. Chem. 2002,485-492), fert-butyl 3-hydroxypyrazole-l-carboxylate (2.3 g, 12 mmol), and triphenyl phosphine (3.2 g, 12 mmol) in THE (28 mL) was cooled in an ice bath, and diisopropyl azodicarboxylate (DIAD) (2.4 mL, 12 mmol) was slowly added. The cooling bath was removed, and the reaction was stirred for 15 hours. The on was diluted with ethyl acetate, washed with ted aqueous sodium onate, dried over sodium sulfate, and evaporated under vacuum. The residue was ed by silica gel chromatography eluting with 0-20% ethyl acetate in hexanes to give /m-bulyl 3-(dispiro[2.0.2.1]heptanyl methoxy)-1 a/ole- 1-carboxylate (1.57 g, 49% yield) as a colorless oil. ESI-MS m/z calc. 290.16306, found 291.3 (M+l)+; Retention time: 0.76 minutes.
] Step B: 3-(Dispiro[2.0.2.1 |heptanylmethoxy)-l//-pyrazole SUBSTITUTE SHEET (RULE 26) WO 64632 N.m.Boc ,0-^N - 'NH A solution of /cvv-butyl piro[2.0.2.1]heptanyl melhoxx )-1//- pyrazole-l-carboxylate (1.57 g, 5.41 mmol) and trifluoroacetic acid (2.2 mL, 29 mmol) in dichloromethane (20 mL) was d for three hours. The volatiles were removed under vacuum, and the residue was basified with saturated aqueous sodium bicarbonate and extracted with ethyl acetate. The combined extracts were dried over sodium sulfate and evaporated to give 3-(dispiro[2.0.2.1]heptanylmethoxy)pyrazole (0.94 g, 91%yield) as pale yellow oil. ESI-MS m/z calc. 190.11061, found 191.1 (M+l)+; Retention time: 0.52 minutes Step C: Ethyl 2-chloro(3-(dispiro[2.0.2.1]heptanylmethoxy)-l//- pyrazol-l-yl)nicotinate - NH P_An N' -Cl✓i A mixture of 3-(dispiro[2.0.2.l]heptanylmethoxy)pyrazole (0.94 g, 4.9 mmol), ethyl chloropyridinecarboxylate (1.15 g, 5.23 mmol), potassium carbonate (0.83 g, 6.0 mmol), and azabicyclo[2.2.2]octane (0.12 g, 1.1 mmol) in DMSO (16 mL) was stirred for 24 hours. The reaction was diluted with water and extracted with ethyl acetate. The combined ts were washed with brine and water, dried over sodium sulfate, and evaporated under vacuum. The residue was purified by silica gel column chromatography eluting with 0-20% ethyl acetate in hexanes to give ethyl 2-chloro(3-(dispiro[2.0.2.1]heptanylmethoxy)-li7-pyrazol-l-yl)nicotinate (1.39 g, 75% yield) as a colorless solid. ESI-MS m/z calc. 373.11932, found 374.2 ; Retention time: 0.87 minutes. 'H NMR (400 MHz, Chloroform-d) 5 8.36 (d, J = 2.8 Hz, 1H), 8.27 (d, J = 8.5 Hz, 1H), 7.72 (d, J = 8.5 Hz, 1H), 5.96 (d, J = 2.9 Hz, 1H), 4.41 (q, J = 7.1 Hz, 2H), 4.30 (d, J = 7.0 Hz, 2H), 1.94 (t, J = 7.0 Hz, 1H), 1.42 (t, J = 7.1 Hz, 3H), 1.02-0.89 (m, 4H), 0.75-0.65 (m, 2H), 0.65-0.53 (m, 2H) Step D: 2-Chloro[3-(dispiro[2.0.2.1]heptanylmethoxy)pyrazol-l- yl]pyridinecarboxylic acid SUBSTITUTE SHEET (RULE 26) 0 0 O^QN N-'CIxi P-A N''GIxl A solution of ethyl ro(3-(dispiro[2.0.2.1]heptanylmethoxy)-li/- pyrazol-l-yl)nicotinate (1.39 g, 3.72 mmol) and sodium hydroxide (7.5 mL of 1 M solution, 7.5 mmol) in THF (6 mL) and ethanol (3 mL) was stirred for 90 s. The volatiles were removed under vacuum, and water was added. The reaction was cooled in an ice bath, and hydrochloric acid (7.5 mL of 1 M solution, 7.5 mmol) was slowlyadded.
The reaction was diluted with water and extracted with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, and evaporated to give 2-chloro[3-(dispiro[2.0.2. l]heptanylmethoxy)pyrazol-l-yl]pyridine carboxylic acid (1.16 g, 82% yield) as a colorless solid. ESI-MS m/z calc. 345.088, found 346.1 (M+l)+; Retention time: 0.73 minutes. 'H NMR (400 MHz, DMSO-d6) 8 8.41 (d, J = 2.9 Hz, 1H), 8.38 (d, J = 8.4 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 6.19 (d, J = 2.8 Hz, 1H), 4.27 (d, J = 7.0 Hz, 2H), 1.93 (t, J = 7.0 Hz, 1H), 0.97 - 0 79 (m, 4H), 0 76 - 0.66 (m, 2H), 0.65 - 0.56 (m, 2H) Step E: A/-(Benzenesulfonyl)chloro[3-(dispiro[2.0.2.l]heptan ylmethoxy)pyrazol-l-yl]pyridinecarboxamide O 9 QwP N:s'' p^N N^CIH ] A solution of 2-chloro[3-(dispiro[2.0.2.1]heptanylmethoxy)pyrazol-l- yl]pyridinecarboxylic acid (0.10 g, 0.29 mmol) and carbonyl diimidazole (0.06 g, 0.4 mmol) in THF (1.4 mL) was stirred for 45 minutes, and esulfonamide (55 mg, 0.35 mmol) and l,8-diazabicyclo(5.4.0)undecene (DBU) (130 pL, 0.87 mmol) were added. After 15 hours the reaction was diluted with 1 M aqueous citric acid and ted with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, and evaporated to give crude A-(benzenesulfonyl)chloro[3- (dispiro [2.0.2.1 ]heptan-7 -y xy)pyrazol-1 -y 1] pyridine-3 -carboxamide (0.16 g) which was used in the next step as-is. ESI-MS m/z calc. 484.0972, found 485.2 ; Retention time: 0.81 minutes.
SUBSTITUTE SHEET (RULE 26) ] Step F: 7V-(Benzenesulfonyl)[3-(dispiro[2.0.2.1]heptan ylmethoxy)pyrazol-l-yl][(4V)-2,2,4-trimethylpyrrolidin-l-yl]pyridine carboxamide 9 qpVo o o 0 •N'N-CC%P A mixture of jV-(benzenesulfonyl)chloro[3-(dispiro[2.0.2.1]heptan ylmethoxy)pyrazol-l-yl]pyridinecarboxamide (0.14 g, 0.29 mmol), (4S)-2,2,4- trimethylpyrrolidine hydrochloride (0.14 g, 0.94 mmol), and potassium carbonate (0.24 g, 1.7 mmol) inNMP (1.3 mL) was stirred at 130 °C for 15 hours. The reaction was filtered and purified using a reverse phase HPLC-MS method using a dual gradient run from 30-99% acetonitrile in 5 mM aqueous HC1. .V-iBenzenesulfonvl )|3- (dispiro[2.0.2.1 ]heptan-7 -ylmethoxy)pyrazol-l -yl] [(45)-2,2,4-trimethylpyrrolidin-1 - yl]pyridinecarboxamide (65 mg, 40% yield) was ed. ESI-MS m/z calc. 561.24097, found 562.3 (M+l)+; Retention time: 2.35 minutes.
Synthetic Example 63: Synthesis of Compound 65: zenesulfonyl)- 6-(3-hydroxypyrazol-l-yl)[(4S)-2,2,4-trimethylpyrrolidin-l-yl]pyridine carboxamide Step A: fezt-Butyl 3-((l-ethylcyclopropyl)methoxy)-lf7-pyrazole-l- carboxylate ^ X)°v-N DIAD, Ph3R, THF 0°C to rt, then 50°C T NBoc To the solution of (l-ethylcyclopropyl)methanol (1.68 g, 16.73 mmol), tertbutyl 2,3-dihydrooxopyrazole-l-carboxylate (2.80 g, 15.21 mmol) and triphenylphosphine (4 39 g, 16.73 mmol) in tetrahydrofuran (40 mL) at 0 °C was added diisopropyl arboxylate (3.38 g, 16.73 mmol) se. The reaction was warmed up to room temperature, then heated at 50 °C for 21 hours. The reaction solution was cooled to room temperature and ethyl e (500 mL) was added. The organic layer was washed with 0.3M aqueous sodium hydroxide solution (100 mL), brine (2 x 50 mL) and dried over magnesium sulfate, filtered and concentrated. The residue was purified SUBSTITUTE SHEET (RULE 26) by silica gel chromatography using 0 to 80% hexanes/dichloromethane gradient method to afford re/Y-butyl 3-(( I -ethylcyclopropyl)methoxy)-1 //-pyrazole-1 -carboxylate (1.73 g, 43%) as a yellow oil. ESI-MS m/z calc. 266.2, found 267.3 (M+l)+. ion time: 3.47 s. 1HNMR (250MHz: CDCh) 8 (ppm): 7.82 (d, J= 2.5Hz, 1H), 5.88 (d, / = 2.5Hz, 1H), 4.09 (s, 2H), 1.60 (s, 9H), 1.48 (q, J= 7.5Hz, 2H), 0.94 (t, J= 7.5Hz, 3H), 0.49 (m, 2H), 0.42 (m, 2H).
Step B: 3-((l-Ethylcyclopropyl)methoxy)-l//-pyrazole HCI in e 0V^N. tVN.X/NH T NBoc A solution of 4M hydrogen chloride in 1,4-dioxane (65 mL) was added to /e/V-butyl 3-(( l-cthylcyclopropyi)mctho\))-1//-pyrazole-1 -carboxylate (1.73 g, 6.49 mmol) and the reaction solution was stirred at room temperature for 16 hours and concentrated to dryness to obtain crude 3-((l-ethylcyclopropyl)methoxy)-l//-pyrazole as an oil which was used ly in next step. ESI-MS m/z calc. 166.1, found 167.3 (1VM) . Retention time: 0.60 minutes.
Step C: Methyl 2-chloro(3-((l-ethylcyclopropyl)methoxy)-lH-pyrazol- l-yl)nicotinate / K2C03/DABCO Ox^N.
WNH x: f O' cr n ci DMF .Cl 3-((l-Ethylcyclopropyl)methoxy)-lH-pyrazole (6.49 mmol) and methyl 2,6- dichloronicotinate (1.81 g, 8 mmol) were added into dimethylformamide (20 mL).
Potassium carbonate (2.8 g, 20 mmol) and l,4-diazabicyclo[2.2.2]octane (0.167 g, 1.5 mmol) were added into reaction e which was allowed to stir at room temperture for 16 hours. The reaction mixture was diluted with water (60 mL) and ted with diethyl ether (3 x 60 mL). Combined organic layer was dried over sodium e and evaporated under reduced pressure. The residue was subjected to flash chromatography on silica gel using 0 to 100% hexanes/dichloromethane gradient method to give methyl 2-chloro(3-((l-ethylcyclopropyl)methoxy)-lH-pyrazol-l-yl)nicotinate (1.7 g, 77%) as a white solid. ESI-MS m/z calc. 335.10, found 336.5 (M+l)+. Retention time: 4.29 minutes, fa NMR (250 MHz, DMSO) S (ppm): 8.43 (d, J = 2.8Hz, 1H), 8.41 (d, / = SUBSTITUTE SHEET (RULE 26) 8.4Hz, 1 H), 7.74 (d, J = 8.4Hz, 1 H), 6.23 (d, 2.8Hz, 1 H), 4.08 (s, 2H), 3.88 (s, 3H), 1.44 (q, J= 7.4Hz, 2H), 0.93 (t. ./= 7.4Hz, 3H): 0.53 (m, 2H), 0.42 (m: 2H).
Step D: 2-Chloro(3-((l-ethylcyclopropyl)methoxy)-lH-pyrazol-l- yl)nicotinic acid 0 NaOH 0 f '0- // OH -c, THF/MeOH Methyl 2-chloro(3-((l-ethylcyclopropyl)methoxy)-lH-pyrazol-l- yl)nicotinate (1.7 g, 50 mmol) was added in a ice-cooled round bottom flask with tetrahydrofuran (5 mL) and methanol (5 mL). Sodium hydroxide (0.4 g, 10 mmol) in water (5 mL) was added slowly and reaction solution was stirred at room temperature for 5 hours. The resulting reaction mixture was concentrated under d pressure to remove tetrahydrofuran and acidified by IN aqueous hydrogen chloride solution to pH = 1 in ice bath. Formed white solid was filtered off to give 2-chloro(3-((lethylcyclopropyl )methoxy)-lH-pyrazol-l-yl)-nicotinic acid (1.54 g, . ESI-MS m/z calc. 321.09, found 322.2 . Retention time: 3.59 minutes. 'H NMR (250 MHz, DMSO) S (ppm): 8.43 (d, J= 2.8Hz, 1H), 8.42 (d, J= 8.4Hz, 1 H), 7.72 (d, J= 8.4Hz, 1 H), 6.24 (d, J= 2.8Hz, 1 H), 4.09 (s, 2H), 1.45 (q, J= 7.4Hz, 2H), 0.92 (t, J 7.4Hz, 3H), 0.54 (m, 2H), 0.44 (m, 2H).
Step E: N-(Benzenesulfonyl)chloro[3-[(l- ethylcyclopropyl)methoxy] l-l-yl] pyriilinecarboxamide O Q o..o OH In ' N‘N N-m H .0^ N Cl✓5 0—& N N Cl 2-Chloro[3-[(l-ethylcyclopropyl)methoxy]pyrazol-l-yl]pyridine carboxylic acid (157 mg, 0.4879 mmol) and carbonyl diimidazole (100 mg, 0.6167 mmol) were combined in THE (2 mL) and d for 2 h. At this point, benzenesulfonamide (77 mg, 0.4899 mmol) was added followed by DBU (243 pL. 1.625 mmol) and the reaction was stirred an additional 30 min at room temperature. The reaction was diluted with ethyl e and washed with a 1M citric acid solution, followed by brine. The organics were separated, dried over sodium sulfate, and SUBSTITUTE SHEET (RULE 26) evaporated to give crude N-(benzenesulfonyl)chloro[3-[(lethylcyclopropyl )methoxy]pyrazol-l-yl]pyridinecarboxamide (224 mg, 100%) ESI- MS mxz calc. 460.0972, found 461.1 (M+l) +; Retention time: 0.75 minutes.
Step F: N-(Benzenesulfonyl)chloro(3-hydroxypyrazol-l-yl)pyridine- oxamide Q 0 0 \'0 Q oo N-S \V/ H H o-^n'n N Cl ho-^n N Cl zenesulfonyl)chloro[3-[(l-ethylcyclopropyl)methoxy]pyrazol-l- yl]pyridinecarboxamide (224 mg, 100%) was dissolved in dichloromethane (2 mL) with TFA (1 mL, 12.98 mmol) and the reaction was stirred for 4 h. The reaction was evaporated to dryness to give N-(benzenesulfonyl)chloro(3-hydroxypyrazol-lyl )pyridinecarboxamide (136 mg, 74%) ESI-MS m/z calc. 378.01895, found 379.1 (M+l) +; Retention time: 0.54 minutes.
Step G: N-(benzenesulfonyl)(3-hydroxypyrazol-l-yl)[(4S)-2,2,4- trimethylpyrrolidin-l-yl]pyridinecarboxamide 9 9P .(s) + HN N- ho^n^n^ci joifio N-(benzenesulfonyl)chloro(3-hydroxypyrazol-l-yl)pyridine carboxarmde (100 mg, 0.26 mmol), ('4T)-2.2.4-tri methyl pyrrolidine (hydrochloride salt) (110 mg, 0.76 mmol), and potassium carbonate (180 mg, 1.3 mmol) were combined in DMSO (0.5 mL) and heated at 130 °C for 16 h. The reaction was diluted with water (3mL) and stirred for 20 min. A solid formed and the aqueous liquid was decanted. The solid was ved in ethyl acetate and washed with a 1M citric acid solution, then brme. The organics were dried over sodium sulfate and ated. The crude material was purified by silica gel chromatography eluting with 0-10% methanol in romethane to give N-(benzenesulfonyl)(3-hydroxypyrazol-l-yl)[(4S)-2,2,4- SUBSTITUTE SHEET (RULE 26) trimethylpyrrolidin-l-yl]pyridinecarboxamide (4.6 mg, 4%) ESI-MS m/z calc. 272, found 456.3 (M+l)+; Retention time: 1.50 minutes.
Examples: ASSAYS & DATA 5A. Assays for Detecting and Measuring F508del-CFTR modulator Properties of Compounds Membrane potential optical methods for assaying properties of F508del-CFTR The assay utilizes fluorescent voltage sensing dyes to measure changes in membrane potential using a scent plate reader (e.g., FLIPR III, Molecular Devices, Inc.) as a readout for increase in functional F508del in NIH 3T3 cells. The driving force for the se is the creation of a chlonde ion gradient in conjunction with channel activation and concurrent with compound ent by a single liquid addition step after the cells have usly been loaded with a voltage sensing dye. 5A-A1. Identification of F508del-CFTR modulators To identify modulators of F508del, a fluorescence based 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 F508del NIH 3T3 cells. The driving force for the se is the creation of a chloride ion gradient in conjunction with channel activation and rent with compound treatment by a single liquid on step after the cells have previously been loaded with a voltage sensing dye. Data for nds 1-65 that were obtained using the assay described here are summarized in Table 6 below. For example, using this method, Compound 1 had an EC50 of less than 3 pM and a % Efficacy of > 100% relative to Compound II.
Solutions Bath on#!: (inmM)NaCl 160, KC14.5, CaCl22, MgCl21,HEPES 10, pH 7.4 with NaOH, Glucose 10.
Chloride-free bath solution: Chloride salts in Bath Solution #1 (above) are substituted with gluconate salts.
SUBSTITUTE SHEET (RULE 26) Cell Culture NIH3T3 mouse fibroblasts stably expressing F508del are used for optical measurements of membrane ial. The cells are maintained at 37 °C in 5% CCh and 90 % humidity in Dulbecco’s ed Eagle’s medium supplemented with 2 mM glutamine, 10 % fetal bovine serum, 1 XNEAA, [3-ME, 1 X pen/strep, and 25 mM HEPES in 175 cm2 culture flasks. For all optical , the cells were seeded at 12,000 cells/well in 384-well matrigel-coated plates and cultured for 18-24 hrs at 37 °C for the potentiator assay. For the correction assays, the cells are cultured at 37 °C with and without compounds for 18 - 24 hours.
Electrophysiological Assays for assaying F508del modulation properties of compounds.
Ussing Chamber Assay Ussing chamber experiments were performed on polarized airway epithelial cells expressing F508del to further characterize the F508del modulators fied in the optical assays. Non-CF and CF airway epitheha were isolated from bronchial , cultured as previously described (Gahetta, L.J.V., Lantero, S., Gazzolo, A., Sacco, O., Romano, L., Rossi, G.A., & Zegarra-Moran, O. /« Vitro Cell. Dev. Biol. 34, 478- 481), and plated onto Costar® 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 ed from non-smokers that did not have any known lung disease. E were isolated from patients homozygous for F508del or compound heterozygous for F508del with an different e causing on on the other .
HBE grown on Costar® Snapwell™ cell culture inserts were mounted in an Ussing chamber (Physiologic Instruments, Inc., San Diego, CA), and the transepithelial ance and short-circuit current in the presence of a basolateral to apical CF gradient (Isc) were measured using a e-clamp system (Department of Bioengineering, University of Iowa, IA). Briefly, HBE were examined under voltage-clamp recording conditions (Vhoid = 0 mV) at 37 °C. The basolateral solution contained (in mM) 145 SUBSTITUTE SHEET (RULE 26) NaCl, 0.83 K2HPO4, 3.3 KH2PO4,12 MgCl2,1.2 CaCl2,10 Glucose, 10 HEPES (pH adjusted to 7.35 with NaOH) and the apical solution contained (in mM) 145 NaGluconate, 1.2 MgCl2,1.2 CaCE, 10 glucose, 10 HEPES (pH adjusted to 7.35 with NaOH).
SA-A2. fication of F508del-CFTR modulators Typical protocol utilized a teral to apical membrane Cl' concentration nt. To set up this gradient, normal ringers was used on the basolateral membrane, whereas apical NaCl was replaced by equimolar sodium gluconate ted to pH 7.4 with NaOH) to give a large CE concentration gradient across the epithelium.
Modulators were added either to the basolateral side 18-24 prior to assay or to the apical side during the assay. lin (10 pM) was added to the apical side during the assay to stimulate CFTR-mediated Cl' transport.
Patch-clanw Recordings Total Cl" current in F508del-NIH3T3 cells was monitored using the perforated-patch recording configuration as previously described (Rae, J., Cooper, K., Gates, P., & Watsky. M. (1991) J. Neurosci. s 37, . Yoltage-clamp recordings were performed at 22 °C using an Axopatch 200B patch-clamp amplifier (Axon Instruments Inc., Foster City, CA). The e solution contained (in mM) 150 A-methy 1-D-glucamine (NMDG)-Cl, 2 MgCl2, 2 CaCl2, 10 EGTA, 10 HEPES, and 240 pg/mL amphotericm-B (pH adjusted to 7.35 with HC1). The extracellular medium contained (in mM) 150 NMDG-C1, 2 MgCf, 2 CaCE, 10 HEPES (pH adjusted to 7.35 with HC1). Pulse generation, data acquisition, and analysis were performed using a PC equipped with a Digidata 1320 A D ace in conjunction with Clampex 8 (Axon Instruments Inc.). To activate F508del, 10 pM forskolin and 20 pM genistein were added to the bath and the current-voltage relation was monitored every 30 sec. 5A-A3. Identification of -CFTR modulators The ability of F508del-CFTR modulators to increase the macroscopic F508del CF current (Esnsdci) in NIH3T3 cells stably expressing F508del was also investigated using perforated-patch-recording ques. Modulators identified from the optical assays SUBSTITUTE SHEET (RULE 26) evoked a dose-dependent increase in IAfsos with similar potency and efficacy ed in the l assays.
Cell e NIH3T3 mouse fibroblasts stably expressing F508del are used for whole-cell ings. The cells are maintained at 37 °C in 5% CO2 and 90 % ty in Dulbecco’s modified Eagle’s medium supplemented with 2 mM glutamine, 10 % fetal bovine serum, 1 X NEAA, [1-ME. 1 X pen/strep, and 25 mM HEPES in 175 cm2 culture flasks. For whole-cell recordings, 2,500 - 5,000 cells were seeded on -lysinecoated glass coverslips and cultured for 18 - 24 hrs in the presence or absence of tors 37 °C.
Single-channel recordings Gating activity of F508del-CFTR expressed in NIH3T3 cells following modulator treatment was observed using excised inside-out membrane patch recordings as previously described (Dalemans, W., Barbry , P., Champigny, G., Jallat, S., Dott, 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 CaCl2, 2 MgCl2, and HEPES (pH adjusted to 7 35 with Tris base). The bath contained (in mM): 150 NMDG-C1, 2 MgCl2, 5 EGTA, 10 TES, and 14 Tris base (pH adjusted to 7.35 with HC1). After excision, both wt- and l were activated by adding 1 mM Mg-ATP, 75 nM of the catalytic subunit of ependent protein kinase (PKA; Promega Corp. Madison, WI), and 10 mMNaF 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 ed from 120 sec of F508del activity was filtered “off-line” at 100 Hz and then used to construct mt amplitude histograms that were fitted with multigaussian functions using Bio-Patch Analysis software (Bio-Logic Comp. France). The total microscopic current and open probability (P0) were ined from 120 sec of channel activity. The P0 was determined using the Bio-Patch software or from the relationship SUBSTITUTE SHEET (RULE 26) P0 = I/i(N), where I = mean current, i = single-channel current amplitude, and N = number of active channels in patch.
Cell Culture NIH3T3 mouse fibroblasts stably expressing F508del are used for excised- membrane patch-clamp recordings. The cells are maintained at 37 °C in 5% CO2 and 90 % humidity in Dulbecco’s modified Eagle’s medium supplemented with 2 mM ine, 10 % fetal bovine serum, 1 XNEAA, [3-ME, 1 X pen/strep, and 25 mM HEPES in 175 cm2 culture flasks. For single channel recordings, 2,500 - 5,000 cells were seeded on poly-L-lysine-coated glass lips and cultured for 18 - 24 hrs in the ce or absence of modulators at 37 °C. 5B. Chromatographic determination of Human Serum Albumin (HSA) Assay Chromatographic determination of Human Serum Albumin (HSA) values was performed on aUPLC-MS system using a Pak® HSA column (p/n: 58469AST) from Sigma Aldrich. Mobile phase A consisted of 50 mM ammonium acetate buffer in water adjusted to pH=7.4, and mobile phase B was 2-propanol. The column compartment was kept at constant temperature of 30°C. ination of retention time on the HSA column was performed by injecting 3 mL of 0.5 mM of compound (in DMSO) using a linear gradient from 0% - 30% B in 2.5 minutes, followed by a hold at 30 %B for 2 minutes, and the final equilibration step from 30% - 0% B in 1.5 minutes, for a total run time of 6 minutes. Flow rate was kept constant throughout the gradient and set to 1.8 mL/min. Compound retention time on the HSA column was converted to %HS A values according to a previously published protocol (Valko, et. al, 2003) correlating column retention times to standard plasma protein binding (PPB) values obtained from is experiments. HSA data for certain compounds are summarized below in Table 8 below.
Valko, K., Nunhuck, S., Bevan, C., Abraham, M. H., Reynolds, D. P. Fast Gradient HPLC Method to Determine Compounds Binding to Human Semm Albumin. onships with Octanol Water and Immobilized Artificial Membrane Lipophilicity. ./. m. Sci. 2, 2236-2248. ] 5C. Experimental Protocol for Rat IV and PO PK studies SUBSTITUTE SHEET (RULE 26) The tested compound was administered to male Sprague-Dawley rats as a single nominal intravenous dose of 3.0 mg/kg as a solution in 10% NMP, 10% solutol, % EtOH, 35% PEG400 and 30% D5W. The tested nd was also administered to male Sprague-Dawley rats at single nominal oral dose of 3 mg/kg as a solution in % NMP, 30% PEG400, 10% TPGS, 5% PVP-K30 at 5 mL/kg dose volume. es of plasma and dose preparations were performed using LC/MS/MS.
Plasma concentration-time profiles of the tested compound in Sprague- Dawley rats at scheduled (nominal) sampling times were analyzed by noncompartmental pharmacokinetic methods using PK function within Watson LIMS re, Version 7.4.2 (Thermo Scientific Inc, m, MA). AUC values were calculated using the linear trapezoidal rule. 5D. Experimental Protocol for PXR assay The propensity for PXR mediated CYP3A4 induction is assessed using the DPX-2 cell line in vitro. This cell line, which has been licensed from Puracyp Inc. was derived from HepG2 cells and has been stably transfected with genes ng human PXR as well as a modified luciferase reporter linked to the CYP3A4 promoter region and related distal and al enhancers.
The assay is run in 384 well format and each test article is administered in 11 doses ranging from 0.1 to 60 gM. On day 1, DPX-2 cells which have previously been expanded in-house and cryopreserved are thawed and seeded in tissue culture plates.
The following day, media is changed and cells are cultured in media containing test e, vehicle control or the positive control compound, the clinically ted CYP3A4 inducer rifampicin. Cells are cultured in the presence of test article for 48 hours and then cell viability is assessed using fluorescence based assay (Cell Titer- Fluor, Promega) with an EnVision Plate Reader (PerkinElmer). Subsequently, CYP3A4 transactivation, which is proportional to luciferase activity, is measured by g luminescense using the Promega One-Glo reagent system using the same plate reader.
Data processing within the Genedata software package allows reporting of max fold induction ed to vehicle control, an EC5o value for CYP3 A4 rs and an 11 point-dose response curve. Wells with cell viability less than 70% are not used for the analysis and plates where the rifampicin positive control se falls outside of the expected range, either in y or max fold induction, are not reported.
SUBSTITUTE SHEET (RULE 26) ] 5E. CFTR Data of Compounds 1-65 ] The compounds of formula (I) are useful as modulators of CFTR activity.
The Table 6 below illustrates the EC50 of the compounds of Table 6 using procedures described above (assay described above in Example 5A-A1). In Table 6 below, the following meanings apply. EC50: “+++” means < 0.1 uM; “++” means between 0.1 uM and 1 uM; “+” means greater than 1 uM.
Table 6. CFTR Activity Comp. No. CFTRdF508 EC50 ]uM) 1 +++ 2 ++ 3 +++ 4 +++ +++ 6 NAa 7 NAa 8 +++ 9 ++ +++ 11 ++ 12 +++ 13 ++ 14 +++ +++ 16 +++ 17 +++ 18 ++ 19 ++ ++ 21 +++ 22 ++ 23 +++ 24 +++ +++ 26 + 27 ++ 28 +++ 29 ++ +++ 31 +++ SUBSTITUTE SHEET (RULE 26) Comp. No. CFTRdF508 EC50 (uM) 32 +++ 33 +++ 34 +++ +++ 36 +++ 37 +++ 38 ++ 39 +++ 40 +++ 41 +++ 42 +++ 43 +++ 44 ++ 45 ++ 46 ++ 47 +++ 48 + 49 +++ 50 +++ 51 +++ 52 ++ 53 +++ 54 +++ 55 +++ 56 ++ 57 +++ 58 +++ 59 +++ 60 ++ 61 NAa 62 + 63 ++ 64 +++ 65 + a.: notmeasurec ] 5F. Metabolites It has been determined that Compound 1 is metabolized both in vitro and in vivo, mostly by oxidative metabolism. nd 1 and the metabolites shown in the following table were prepared and tested.
Table?. Data for Metabolites Metabolite CFTRdF508 Dculernled nnnlo« of EC50 (llM) metabolite SUBSTITUTE SHEET (RULE 26) Mef.nholifo CFTRdF508 Deulci'nlcd nl EC50 (uM) metabolite Compound 1 (Parent 0.03 Compound 17 Compound) V-Y;Xub._J XiH (jP+h” Compound 38 0.24 Compound 7 9 o. o A N'-3' v 9 9 •hrHO o fjyy. a. n,° ‘V M Compound 62 >30 Compound 31 vW°° o N-\ ^ OH Compound 34 L H I •k' °iJJJ.. Vn^Vl 5G. lfoxamides. Compounds comprising an acylsulfoxamide moiety (i.e., sulfonimidoylamide moiety - wherein X in Formulae I or II is chosen from SUBSTITUTE SHEET (RULE 26) substituted or unsubstitued amines) were determined to result in decreased human serum albumin binding and enhanced free fraction as compared to compounds comprising an acylsulfonamide group (i.e., wherein X in Formulae I or II is chosen from 0). The HAS data were measured as described above in Example 5B. Decreased human serum albumin g may result in a higher amount of free (unbound) drug which can affect biological activity.
Tables. HSAData Ae\lsiiironamide MSA Siilfbviiiiine HSA ng binding 99.1% VY 98.1% Compound 15 '~N _>.jCcCq0 o 0 0 /Ss, fyk'-aH.ll r o'-y) 0 F'A ereomeric mixture: Compound 45))_______ 97.9% o y /•=», fVs--N 'o"Yl W o F-/ (diastereoisomer 1: Compound 46) XX 98.4% o A*.
A'S':N W o Ft( (diastereoisomer 2: nd 47) Table 8 below summarizes CFTR activity (CFTR dF508 EC50), PXR Max induction. Rat IV clearance, Rat PO AUC, and Rat PO data for certain nds described above.
Table 9. Comparative Data SUBSTITUTE SHEET (RULE 26) Cnmp nds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li riliimpicinl min.' hi ml. in ku) 3 mu dose1 Comp. 0.06 2 17.8 1.2a 28% A 0 o 0 NHi Comp. 0.12 9 29.2 B 0 00 Comp. 0.26 3 C 0 00 0 I I II H /) Comp. 0.4 20 D 0q p w. !l V^'A1 Comp. 0.21 10 E Jo.,o oJ-MV/) SUBSTITUTE SHEET (RULE 26) Cnmp Compounds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li picinl min.' hi ml. in ku) 3 mu dose1 Comp. o 0.02 19 49 vO FX/'° Comp. 0.02 4 4 0 0.
(Y'wr IX/■—-/ '^4 f A-1 Comp. 0.02 0 0 00 vi o7'N « N' /? Comp. 0.02 2 24.8 0.25 12% 16 0 00 Yf “TV Comp. 0.03 3 1.6 21 63% 1 0 00 F LX FI rv F I H ks Comp. 0.03 32 50 0 00 SUBSTITUTE SHEET (RULE 26) WO 64632 Cnmp Compounds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li riliimpicinl min.' hi ml. in ku) 3 mu dose1 Comp. 0.03 22 2.7 12.5 69% F c® Comp. 0.03 17 Comp. 0.03 7 31 o 0.0 F •aOXO.
Comp. o 0 0 0.04 19 2.0 14.7 58% v-erft-jctfo Comp. n 0.04 39 2.3 13.7 98% (rtCQ,o a 53 /yVV'eu yn —j'-j Comp. 0.04 6 11.2 2.2 41% ?0. .0 Comp. 0.04 4 32 H1J:s y Fktfp SUBSTITUTE SHEET (RULE 26) WO 64632 Cnmp Compounds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li riliimpicinl min.' hi ml. in ku) 3 mu dose1 Comp. 0.05 17 5.2 10.5 100 51 -krtvo ooi! •«" % Comp. 0.05 22 55 ? 'i.JJ tfc&rI li H ° Comp. 0.05 0 9.6 2.7 50% A.liWF 0-// H /"J W Comp. 0.05 4 58 ? o. .0 Comp. 0.05 4 3.4 9b 79% nY // Comp. 0.05 12 43 w0 o 0 Cl )Hr H Comp. vs 0.05 3 5.4 4.4b 51% 42 0 00 ^Arr^ SUBSTITUTE SHEET (RULE 26) Cnmp Compounds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li riliimpicinl min.' hi ml. in ku) 3 mu dose1 Comp. 0.07 6 1.9 6.4 41% 41 ooo Comp. 0.07 9 2.6 20 98% 24 »°-P Comp. 0.07 8 37 ?0..0 F o^W Comp. 0.07 13 39 ?0.,0 Comp. 0.09 0 6.1 2.3 26% 9o..o . o. n is ^:s: Comp. 0.08 2 /y % 0. /> 1 'O’ F [-1 SUBSTITUTE SHEET (RULE 26) Cnmp Compounds Cl IK P\K Max Kill i\ Knt po Rat (Hind dl'508 I ml n d ion Cl. \l ( po No. l'. ('50 I'1.) of (mL IMS?- %F (ii\li riliimpicinl min.' hi ml. in ku) 3 mu dose1 i 0.11 2 Comp. ?0 0 Comp. 0.08 0 33 JLs:»°- 0 Comp. 0.08 2 40 °0..° r JL:s: Comp. 0.27 8 3.6 20.3 100 27 % Comp. ft 0.62 18 5.0 8.5 81% 44 vM V ^■N ft Comp. 0.12 4 6.7 3.6 31% 22 000 a: 10 mg/kg for Compound A, and for the other compounds at 3 mg/kg.
Example 6: Chloride Transport ments SUBSTITUTE SHEET (RULE 26) In one Ussing Chamber experiment with F508del/F508del-HBE cells. nd 1 enhanced chloride transport. The effect of Compound 1 on chloride transport was additive to the effect of Compound IF In addition, F508del-CFTR delivered to the cell surface by either Compound 1 alone or in combination with Compound II was potentiated by Compound III. The triple combination of Compound 1 / Compound II / Compound III provided a superior (approximately 3-fold) increase in chloride transport compared to the 3 dual ns under most conditions tested.
Example 7: F508del-CFTR Processing and Trafficking In Vitro Experiments In vitro, Compound 1 improved the processing and trafficking of F508del-CFTR, thereby increasing the quantity of functional F508del-CFTR protein at the cell surface. The CFTR protein delivered to the cell surface by Compound 1 alone or in combination with Compound II (Compound 1 / Compound II) was iated by Compound III. In human bronchial epithelial (HBE) cells studied in vitro, the triple combination of Compound 1, nd II, and Compound III (Compound 1 / Compound II / Compound III) increased CFTR de transport more than any of the dual combinations und 1 / nd II, Compound 1 / Compound III, and Compound II / Compound III) or individual components (Compound 1, Compound II, and nd III) under most conditions studied.
Processing and cking of F508del-CFTR was directly monitored by the appearance of a 170 to 180 kDa band Such monitoring established that Compound 1 is a CFTR corrector, as it facilitates the processing and trafficking of F508del-CFTR to increase the amount of onal F508del-CFTR at the cell surface.
Incubation of F508del/F508del-HBE cells for 16 to 24 hours with 1 pIYI Compound 1 alone or in combination with 3 pM Compound II resulted in an increase in -state levels, reaching 6.5-fold and 18.7-fold of ted levels, respectively.
Other Embodiments The foregoing discussion discloses and describes merely exemplary embodiments of this disclosure. One skilled in the art will readily recognize from such discussion and from the accompanying drawings and claims, that s changes, modifications and variations can be made therein without departing from the spirit and SUBSTITUTE SHEET (RULE 26) scope of this disclosure as defined in the following claims.
SUBSTITUTE SHEET (RULE 26)

Claims (11)

1. A compound of Formula I: 0 o x vi^V^N^ H X(R2)p / N Y N R1 -r (R3), (R4), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein: - one of Y1 and Y2 is N and the other is CH; - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 lkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, wherein each R is ndently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, ns, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8. SUBSTITUTE SHEET (RULE 26)
2. A compound of Formula II: 0 O X H X(R2)p '/' N N N (R3)q (II), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein: - X is chosen from 0, NH, and N(Ci-C4 alkyl) groups; - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is ndently chosen from C3-C10 cycloalky] groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl , and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally tuted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R3 is independently chosen from C1-C2 alkyl groups optionally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8. SUBSTITUTE SHEET (RULE 26)
3. A compound of Formula III: 0 o. o V(r2)p / N N N / \R3)q (R4)r (ni), a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the ing, wherein: - R1 is chosen from -(CR2)k(CR2)m(CR)n(Ring A)n+i groups, wherein each Ring A is independently chosen from C3-C10 cycloalky l groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl , halogenated C1-C2 alkyl groups, and halogens, wherein each R is independently chosen from H, OH, and C1-C2 alkyl groups optionally substituted with one or more halogens; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, halogens, and cyano; - each R is independently chosen from C1-C2 alkyl groups ally substituted with one or more OH groups; - each R4 is independently chosen from halogens; - k is 0 or 1; - ris 0 or 1; - mis 0,1, 2, or 3; - n is 0 or 1; - p is 0, 1, 2, 3, 4, or 5; and - qis 0, 1, 2, 3, 4, 5, 6, 7, or 8. SUBSTITUTE SHEET (RULE 26)
4. A compound according to any of claims 1-3, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the ing, wherein if R2 is cyano, then said R2 is meta or para relative to the sulfur atom.
5. A compound according to any of claims 1-3, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein: - each Ring A is independently chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, and - each R is independently chosen from H and OH; - each R2 is independently chosen from C1-C2 alkyl groups, OH, C1-C2 alkoxy groups, and halogens; - R4 is F; - kis 0; - p is 0, 1, or 2; - qis 0, 1, 2, 3, or 4; - r is 0; and n m and n are not 0 at the same time.
6. A compound according to claim 5, a pharmaceutically acceptable salt thereof, or a ated tive of any of the foregoing, wherein: - R1 is chosen from 2)m-Ring A groups, wherein Ring A is chosen from C3-C10 cycloalkyl groups groups optionally substituted with one or more substituents each independently chosen from C1-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens, - m is 1 or 2.
7. A compound ing to claim 6, a pharmaceutically acceptable salt thereof, or a deuterated derivative of any of the foregoing, wherein each R3 is a methyl group and q is 3 or 4. SUBSTITUTE SHEET (RULE 26)
8. A compound according to claim 7 having Formula FV: 0 °n P ¥ H X(R2)p Ring AJrk ' N N N (IV), a pharmaceutically able salt thereof, or a deuterated derivative of any of the - Ring A is chosen from C3-C10 cycloalkyl groups optionally substituted with one or more substituents each independently chosen from Ci-C2 alkyl groups, halogenated C1-C2 alkyl groups, and halogens; and - each R2 is independently chosen from C1-C2 alkyl groups, OH, F, Cl, and Ci- C2 alkoxy groups; - m is 1 or 2; and - p is 0, 1, or 2.
9. A compound according to claim 8, wherein p is 0 or 1.
10. A compound according to claim 8, wherein p is 0.
11. A compound according to claim 8 having Formula V: O O. pY ' N N l
NZ792410A 2016-09-30 2017-09-29 Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator NZ792410A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US62/402,838 2016-09-30
US62/410,353 2016-10-19
US62/415,409 2016-10-31
US62/419,935 2016-11-09

Publications (1)

Publication Number Publication Date
NZ792410A true NZ792410A (en) 2022-09-30

Family

ID=

Similar Documents

Publication Publication Date Title
US20220306606A1 (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
AU2021211993B2 (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
AU2019381750A1 (en) Methods of treatment for cystic fibrosis
NZ792410A (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
OA19370A (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator.
NZ795112A (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator
EA040454B1 (en) MODULATOR OF CYSIC FIBROUS TRANSMEMBRANE CONDUCTIVITY REGULATOR, PHARMACEUTICAL COMPOSITIONS, METHODS OF TREATMENT AND METHODS OF OBTAINING THE MODULATOR
OA19688A (en) Modulator of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulator.