OA17460A - Pyrrolo [2, 3 - D]pyrimidine derivatives as inhibitors of Janus-Related Kinases (JAK). - Google Patents

Pyrrolo [2, 3 - D]pyrimidine derivatives as inhibitors of Janus-Related Kinases (JAK). Download PDF

Info

Publication number
OA17460A
OA17460A OA1201500328 OA17460A OA 17460 A OA17460 A OA 17460A OA 1201500328 OA1201500328 OA 1201500328 OA 17460 A OA17460 A OA 17460A
Authority
OA
OAPI
Prior art keywords
linear
branched chain
chain alkyl
cycloalkyl
methyl
Prior art date
Application number
OA1201500328
Inventor
Matthew Frank Brown
Ashley Edward Fenwick
Mark Edward Flanagan
Andrea GONZALES
Timothy Allan JOHNSON
Neelu Kaila
Mark J. Mitton-Fry
Joseph Walter Strohbach
Ruth E. Tenbrink
John David TRZUPEK
Rayomand Jal Unwalla
Michael L. Vazquez
D. Parikh Mihir
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Publication of OA17460A publication Critical patent/OA17460A/en

Links

Abstract

Described herein are pyrrolo{2,3-d} pyrimidine derivatives, their use as Janus Kinase (JAK) inhibitors, and pharmaceutical compositions containing them.

Description

Described herein are pyrrolo{2,3-d} pyrimidine dérivatives, their use as Janus Kinase (JAK) inhibitors, and pharmaceutical compositions containing them.
O.A.P.I. - B.P. 887, YAOUNDE (Cameroun) - Tel. (237) 222 20 57 00- Fax: (237) 222 20 57 27- Site web: http:/www.oapi.int - Email: oapi@oapi.int
DEMANDE DE BREVET D’INVENTION
PCT/IB2014/058889 DU 11/02/2014
Nom du Déposant(s): PFIZER INC.
Titre d’invention:
PYRROLO [2, 3 -DJPYRIMIDINE DERIVATIVES AS INHIBITORS OF JANUS- RELATED
KINASES (JAK)
Noms d’inventeur(s):
1. BROWN,. MatthewFrank
2. FENWÏCK, Aëhley Edward
3. FLANAGAN, Mark Edward
4. GONZALES, Andrea
5. JOHNSON, Timothy Allan
6. KAILA, Neelu
7. MITTON-FRY, Mark J.
8. STROHBACH, Joseph Walter
9. TENBRINK, Ruth E.
10. TRZUPEK, John David
11. UNWALLA.' Rayomand Jal
12. VAZQUEZ, Michael L.
13. MIHIR, D. Parikh
Mandataire : SCP AKKUM, AKKUM & Associâtes
B.P 4966 Yaoundé - Cameroun.
l
PYRROLO[2,3-DJPYRIMIDlNE DERIVATIVES
FIELD OF THE INVENTION
The présent invention provides pharmaceutically active pyrrolo[2,3-d]pyrimidine compounds and analogues. Such compounds are useful for inhibiting Janus Kinase (JAK). This invention also is directed to compositions comprising methods for making such compounds, and methods for treating and preventing conditions mediated by JAK.
BACKGROUND OF THE INVENTION
Protein kinases are families of enzymes that catalyze the phosphorylation of spécifie residues in proteins, broadly classified into tyrosine and serine/threonine kinases, inappropriate kinase activity, arising from mutation, over-expression, or inappropriate régulation, dys-regulation or de-regulation, as well as over- or under-production of growth factors or cytokines has been implicated in many diseases, including but not limited to cancer, cardiovascular diseases, allergies, asthma and other respiratory diseases, autoimmune diseases, inflammatory diseases, bone diseases, metabolic disorders, and neurological and neurodegenerative disorders such as Alzheimer's disease. Inappropriate kinase activity triggers a variety of biological cellular responses relating to cell growth, cell différentiation, survival, apoptosis, mitogenesis, cell cycle control, and cell mobility implicated in the aforementioned and related diseases.
Thus, protein kinases hâve emerged as an important class of enzymes as targets for therapeutic intervention. In particular, the JAK family of cellular protein tyrosine kinases (JAK1, JAK2, JAK3, and Tyk2) play a central rôle in cytokine signaling (Kisseleva et al., Gene, 2002, 285,1; Yamaoka et al. Genome Biology 2004, 5, 253)). Upon binding to their receptors, cytokines activate JAK which then phosphorylate the cytokine receptor, thereby creating docking sites for signaling molécules, notably, members
of the signal transducer and activator of transcription (STAT) family that ultimateiy lead to gene expression. Numerous cytokines are known to activate the JAK family. These cytokines include, the IFN family (IFN-alpha, IFN-beta, IFN-omega, Limitin, IFN-gamma, IL-10, IL-19, IL-20, IL-22), the gp130 family (IL-6, IL-11, OSM, LIF, CNTF, NNT-1/BSF-3, G-CSF, CT-1, Leptin, IL-12, IL-23). gamma C family (IL-2, IL-7, TSLP, IL-9, tL-15, IL-21, IL-4, tL-13), IL-3 family (IL-3, IL-5, GM-CSF), single chain family (EPO, GH, PRL, TPO), receptor tyrosine kinases (EGF, PDGF, CSF-1, HGF), and G-protein coupled receptors (AT1).
There remains a need for new compounds that effectively and selectively inhibit spécifie JAK enzymes, and JAK1 in particuiar, vs, JAK2. JAK1 is a member of the Janus family of protein kinases composed of JAK1, JAK2, JAK3 and TYK2. JAK1 is expressed to various levels in ali tissues. Many cytokine receptors signal through pairs of JAK kinases in the following combinations: JAK1 /JAK2, JAK1 /JAK3, JAK1/TYK2, JAK2/TYK2 or JAK2/JAK2. JAK1 is the most broadly paired JAK kinase in this context and ts required for signaling by γ-common (IL-2Ry) cytokine receptors, IL-6 receptor family, Type I, II and III receptor families and IL-10 receptor family. Animal studies hâve shown that JAK1 is required for the development, function and homeostasis of the immune system. Modulation of immune activity through inhibition of JAK1 kinase activity can prove useful in the treatment of various immune disorders (Murray, P.J. J, Immunol., 178, 2623-2629 (2007); Kisseleva, T„ et al., Gene, 285,1-24 (2002); O'Shea, J. J., et al., Cell, 109, (suppl.) S121-S 131 (2002)) while avoiding JAK2 dépendent erythropoietin (EPO) and thrombopoietin (TPO) signaling (Neubauer H,, et al., Cell, 93(3), 397-409 (1998); Parganas E,, et al„ Cell, 93(3), 385-95 (1998)),
SUMMARY OF THE INVENTION
The présent invention provides a compound of formula I having the structure:
or a pharmaceutically acceptable sait thereof, wherein R1 is hydrogen or C1-C4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, hydroxy, methoxy, amino, CF3, and Ca-Ce cycloalkyl; R2 and R3 are each independently hydrogen, deuter5 ium, Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, Ci-Cs linear or branched chain perfluoroalkyl, Ci-Ce linear or branched chain alkoxy, Ci-Ce linear or branched chain perfluoroalkoxy, halogen, cyano, hydroxyl, amino, carboxy, aminocarbonyl, aryl, heteroaryl, (aryl)Ci-Ce linear or branched chain alkyl, (heteroaryl)Ci-Ce linear or branched chain alkyl, (heterocyclic)Ci-Ce linear or branched chain alkyl, (Ci-Ce linear or branched chain alkyl)aryl, (Ci-Ce linear or branched chain alkyljheteroaryl, (Ci-Ce linear or branched chain alkyl)heterocyclic, (Ci-Ce linear or branched chain alkoxyl)carbonyl, (Ci-Ce linear or branched chain alkyl)amino-carbonylamino, or (Ci-Ce linear or branched chain alkyl)aminocarbonyl; R4 is selected from hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, Ci-Ce linear or branched chain perfluoroalkyl, aryl, and alkylaryl; X is selected from -NH-- and —CRaRb-, where (a) Ra and Rb are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (aryl)Ci-Ce linear or branched chain alkyl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, (heteroaryl)CiCe linear or branched chain alkyl. (heterocyclic)Ci-C6 linear or branched chain alkyl, or (b) Ra and Rb together form a chain comprising —(CRcRd)/-, where Rc and Rd are independently hydrogen, deuterium, C1Cë linear or branched chain alkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryi, heteroaryl, (Ci-Ce linear
or branched chain alkylheteroaryl, haio, CN, CFa, hydroxyl, CONH2, or SO2CH3; Y is -A-R5, where A is a bond, --(CH2)k-- or -(CD2)k- and Rs is Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, or -NRa'Rb·, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxy!, CF3, -ORa, --NReRf, S(0)PRe and C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and SO2CH3, where (a) Ra- and Rb- are independently hydrogen, deuterium, Ci-Cb linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (Ct-Ce linear or branched chain alkyl )aryl, heteroaryl, or (Ci-Ce linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc·, or (b) Ra- and Rb- together form a chain comprising —(CRcRd')r-. where Rc- and Rp are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryi, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, (Ci-Ce linear or branched chain alkylheteroaryl, halo, CN, hydroxyl, CF3, CONH2, --ORe, NRaRf, or -S(O)PRe; where Re and Rt are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2;y is 2, 3, 4 or 5; /ris 1, 2; 3, or 4; p is 0, 1 or 2; and, n is 1 or 2.
In other aspects, the présent invention also provides:
pharmaceutical compositions which comprise a pharmaceutically acceptable carrier and a compound of formula I;
methods for treating conditions or disorders including myositis, vasculitis, pemphigus, Crohn’s disease, lupus, nephritis, psoriasis, multiple sclerosis, major dépréssion disorder, allergy, asthma, Sjogren's disease, dry eye syndrome, transplant rejection, cancer, inflammatory bowel disease, septic shock, cardiopulmonary dysfonction, acute respiratory disease, or cachexia by administering to a subject in need a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable sait thereof;
methods for treating conditions or disorders including atopie dermatitis, eczema, psoriasis, scleroderma, lupus, pruritus, other pruritic conditions, allergie réactions including allergie dermatitis in mammal, horse allergie diseases including bite hypersensitîvity, summer eczema, sweet itch in horses, heaves, inflammatory airway disease, récurrent airway obstruction, airway hyper-responsiveness, and chronic obstruction pulmonary disease by administering to a mammal in need a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable sait thereof; and, methods for the préparation of compounds of the présent invention. The présent invention will be further understood from the following description given by way of exampte only. The présent invention is directed to a class of pyrrolo[2,3-d]pyrimidine dérivatives. In particuiar, the présent invention is directed to pyrrolo[2,3-d]pyrimidine compounds useful as inhibitors of JAK, and particularly JAK1. While the présent invention is not so limited, an appréciation of various aspects of the invention will be gained through the following discussion and the examples.
The term alkyl, alone or in combination, means an acyclic, saturated hydrocarbon group of the formula CnH2n+i which may be linear or branched. Examples of such groups include methyl, ethyl, npropyl, isopropyl, π-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl and hexyl. Unless otherwise specified, an alkyl group comprises from 1 to 6 carbon atoms. The carbon atom content of alkyl and various other hydrocarbon-containîng moieties is indicated by a prefix designating a lower and upper number of carbon atoms in the moiety, that is, the prefix Ci-C, indicates a moiety of the integer i to the integer j carbon atoms, inclusive, Thus, for example, Ci-Ce alkyi refers to alkyi of one to six carbon atoms, inclusive.
The term “hydroxy, as used herein, means an OH radical. The term “heterocyclic refers to a saturated or partially saturated (i.e. non aromatic) heterocycle which may be attached via a ring nitrogen atom (when the heterocycle is attached to a carbon atom) or a ring carbon atom (in ail cases). Equally, when substituted, the substituent may be located on a ring nitrogen atom (if the substituent is joined through a carbon atom) or a ring carbon atom (in ail cases). Spécifie examples include oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, 1,4-dioxanyl, morpholinyl, piperazinyl, azepanyl, oxepanyl, oxazepanyl and diazepinyl.
The term “aryl” refers to an aromatic monocyclic or bicyclic hydrocarbon which may be attached via a ring carbon atom. Equally, when substituted, the substituent may be located on a ring carbon atom. Spécifie examples include phenyl, toluyl, xylyl. trimethylphenyl, and naphthyl. Examples of aryl substituents include alkyi, hydroxyl, halo, nrtrile, alkoxy, trifluoromethyl, carboxamido, SOîMe, benzyl, and substi15 tuted benzyl.
The term heteroaryl refers to an aromatic heterocycle which may be attached via a ring carbon atom (in ali cases) or a ring nitrogen atom with an appropriate valency (when the heterocycle is attached to a carbon atom). Equally. when substituted, the substituent may be located on a ring carbon atom (in ail cases) or a ring nitrogen atom with an appropriate valency (if the substituent is joined through a carbon 20 atom). Spécifie examples include thienyl, furanyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, îsothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrîmidinyl and pyrazinyl. The term cycloalkyl means a monocyclic, saturated hydrocarbon group of the formula CnHin17460
i. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cydohexyi, and cyclohaptyl. Unless otherwise specified, a cycloalkyl group comprises from 3 to 8 carbon atoms.
The terms “halo and “halogen refer to fluoride (F), chloride (Cl), bromide (Br) or iodide (I).
The term marnmal refers to human, livestock or companion animais.
The term companion animal or companion animais refers to animais kept as pets or household animal. Examples of companion animais include dogs, cats, and rodents Încluding hamsters, guinea pigs, gerbils and the like, rabbits, ferrets and birds.
The term livestock refers to animais reared or raised in an agricultural setting to make products such as food or fiber, or for its iabor. In some embodiments, livestock are suitable for consumption by mammals, for example humans. Examples of livestock animais include cattle, goats, horses, pigs, sheep, Încluding lambs, and rabbits, as well as birds, such as chickens, ducks and turkeys.
The term treating or “treatment means an alleviation of symptoms associated with a disease, disorder or condition, or hait of further progression or worsening of those symptoms. Dépending on the disease and condition of the patient, the term “treatment as used herein may include one or more of curative, palliative and prophyiactic treatment Treatment can also include administering a pharmaceutical formulation of the présent invention in combination with other thérapies.
The term therapeutically-effective indicates the capability of an agent to prevent, or improve the severity of, the disorder, while avoiding adverse side effects typically associated with alternative thérapies. The phrase therapeutically-effective is to be understood to be équivalent to the phrase effective for the treatment, prévention, or amelioration, and both are intended to qualify the amount of each agent for use in the combination therapy which will achieve the goal of improvement in the severity of cancer, cardiovascular disease, or pain and inflammation and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative thérapies.
Pharmaceutically acceptable means suitable for use in mammals, companion animais or livestock animais.
If substituents are described as being independently selected from a group, each substituent is selected independent of the other, Each substituent therefore may be identical to or different from the other substituent s).
DETAILED DESCRIPTION OF THE INVENTION
The présent invention is related to novel oompounds which are seiectrve JAK1 modulators useful for the treatment of diseases and conditions associated with dysrégulation of the JAK1. The présent invention further provides pharmaceutical compositions comprising such JAK1 modulators as well as methods of treating and/or preventing such diseases and conditions. Accordingly, the présent invention provides a compound of formula I having the structure:
or a pharmaceutically acceptable sait thereof, wherein R1 is hydrogen or C1-C4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consîsting of halo, hydroxy, methoxy, amino, CF3, and C3-C6 cycloalkyl; R2 and R3 are each independently hydrogen, deuterium, C-i-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, Ci-Ce linear or branched chain perfluoroalkyl, Ci-Ce linear or branched chain alkoxy, Ci-Ce linear or branched chain perfltioroalkoxy, halogen, cyano, hydroxyl, amino, carboxy, aminocarbonyl, aryl, heteroaryl, (aryl)Ci-Ce linear or branched chain alkyl, (heteroaryl)Ci-C6 linear or branched chain alkyl, (heterocycircJCi-Ce linear or branched chain alkyl, (Ci-Ce linear or branched chain alkyl)aryl, (Ci-Ce linear or branched chain alkyljheteroaryl, (Ci-Ce linear or branched chain alkyl)heterocyclic, (Ci-Ce linear or branched chain alkoxy l)carbonyl, (Ci-Cg linear or branched chain alkyl)amino-carbony1amino, or (Ci-Ce linear or branched chain alkyl)aminocarbonyl; R4 is selected from hydrogen, deuterium, Ch-Ce linear or branched chain alkyl, Ci-Cs linear or branched chain perfluoroalkyl, aryl, and alkylaryl; X is selected from --NH-- and —CRaRb-, where (a) Ra and Rb are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (aryl)CiCë linear or branched chain alkyl, heteroaryl, (Ci-Ce linear or branched chain aikyt)heteroaryl, (heteroarylJCi-Cs linear or branched chain alkyl, (heterocyclicJCi-Ce linear or branched chain alkyl, or (b) Ra and Rb together form a chain comprising — (CRcRdY. where Rc and Rd are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryl, (Ci-Cs linear or branched chain alkyl)aryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, CF3, hydroxyl, CONH2, or SO2CH3; Y is -A-R5, where A is a bond, -(CHz)*-- or -(Œtejk-- and R5 is Ci-Ce linear or branched chain alkyl, C3Ce cycloalkyl, aryl, or ~NRaRb·, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, Ca-Cs cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Cs linear or branched chain alkyl, CN, hydroxyl, CF3, -ORe, -NReRf, -S(O)pRe and Ca-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl.
CONHî, and SO2CH3, where (a) Ra· and Rb· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl. C3-C6 cycloalkyl, aryl, (Ci-Ce linear or branched chain alkyljaryl, heteroaryl, or (C1Cs linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc , or (b) Ra· and R&· together form a chain comprising —(CRc-Rd·)/-, where Rc and Rd· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyljaryl, heteroaryl, (C1-C5 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl,
CFs, CONH2, “0Re, “NReRf, or --S(O)PRB: where Re and Rr are independently hydrogen, deuterium, C1Ce linear or branched chain alkyl, or C3-C5 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; /is 2, 3, 4 or 5; k is 1, 2; 3, or 4; p is 0, 1 or 2; and, n ts 1 or 2.ln one embodiment, the inven tion provides a compound of formula IA having the structure:
or a pharmaceutically acceptable sait thereof, wherein Y is -A-Rs, where A is a bond, -(CH2)k- or (CD2)k-- and Rs is Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, or --NRa'Rb·, or is an un15 saturated, saturated or partially saturated monocyclîc or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-Ce cycloalkyl, aryl, or monocyclîc or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deu-
terium, halo, Ci-Cê linear or branched chain alkyl, CN, hydroxyI, CF3, -ORe, --NReRf, --S(O)PRa and Cs-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and SO2CH3, where (a) Ra· and Rb· are independently hydrogen, deuterium, Ci-Ca linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (Ci-Ce 5 linear or branched chain alkyljaryl, heteroaryl, or (Ci-Cs linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc·, or (b) Ra· and Rb· together form a chain comprising —(CRcRd·)/-, where Rc- and R<j· are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, aryl, (Ci-Cs linear or branched chain alkyljaryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CFs, CONH2, -ORe, -NReRt, or --S(O)PRe; where Re 10 and Rf are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or Ca-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CFa, and CONH2;/is 2, 3, 4 or 5; fris 1.2; 3, or 4; and, p is 0, 1 or 2.
In one embodiment, the invention provides a compound of formula IA wherein A is a bond and R5 15 is a Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl or aryl. In another embodiment, the invention provides a compound of formula IA wherein A is a bond or —(CHîJk-, and R5 is C3-C6 cycloalkyl wherein said Ca-Ce cycloalkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, Ci-Ce linear or branched chain alkyl, and CN where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, 20 CN, hydroxyl, CONH2, and SO2CH3; where fris 1,2, or 3. In yet another embodiment, the invention provides a compound of formula IA wherein A îs a bond or --(CH2X-, and R5 is an unsaturated, saturated or partially saturated monocycltc or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sul-
fur, wherein said alkyl, C3-C6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyI, CF3, -NRaRt>·, --0Re, -S(O)PRe and C3-C6 cycloalkyl; where fris 1, 2, or 3.
tn another embodiment, the invention provides the compound of formula IB having the structure:
Ra
or a pharmaceutically acceptable sait thereof, wherein (a) Ra· and Rb are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, Cî-Ce cycloalkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, or (Ci-Cs linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc, or (b) Ra- and Rb· together form a chain comprising --(CRc Rd )/-, where Rc and R<r are independently hydrogen, deuterium, Ci-Cs linear or branched chain alkyl, aryl, (Ci-Cb linear or branched chain alkyljaryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2, -ORa, --NReRt, or-S(O)pRe: where Re and Rf are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; or, (c) Ra· and Rt>· together form an unsaturated, saturated or partially
I3 saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, 5 hydroxyl, CF3, -NRa'Rb·, -0Re, --S(O)PRe and Ca-Ce cycloalkyl; /is 2, 3, 4 or 5; and, p is 0, 1 or 2.
In another embodiment, the invention provides the compound of formula IC having the structure:
IC or a pharmaceutically acceptable sait thereof, wherein (a) Ra- and Rb1 are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, 10 heteroaryl, or (Ci-Cs linear or branched chain alkylheteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc, or (b) Ra· and Rb· together form a chain comprising —(CRc Rd )/-, where Rc· and Rd· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryl, (Ci-Cs linear or branched chain alkyl)aryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CFa, CONH2, -OR0, --NReRt, or --S(O)PRa; where Re and Rt are independently hydro15 gen, deuterium, Ci-Ce linear or branched chain aikyi, or C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo,
CN, hydroxyl, CF3, and CONH2; or, (c) Ra- and Rb- together form an unsaturated, saturated or partialiy saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consîsting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure îs further optionally substituted with one or more sub5 stituents selected from the group consîsting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyl, CFa, --NRa'Rb’, --0Ra, --S(O)PRa and Ca-Ce cycloalkyl; jis 2, 3, 4 or 5; and, p is 0, 1 or 2.
In another embodiment, the invention provides the compound of formula ID having the structure:
or a pharmaceutically acceptable sait thereof, wherein
Y is -AR5, where A is a bond or ~(CH2)k-, and R5 is Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consîsting of oxygen, nitrogen, and sulfur, wherein said alkyl, Cs-Ce cycloalkyl, aryl, or monocy15 clic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consîsting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyl, CF3, --NRaRb·, -ORa, -S(O)pRb and C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consîsting of halo, CN, hydroxyl, CONH2, and SO2CH3,
I5 where (a) Ra· and Rb' are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyi, Ca-Ce cycloalkyl, aryl, (aryl)Ci*C6 linear or branched chain alkyi, heteroaryl, (Ci-Cs linear or branched chain alkyljheteroaryl, (heteroaryl)Ci-Ce linear or branched chain alkyi, (heterocyclic)Ci-Cs linear or branched chain alkyi, where said alkyi and cycloalkyl may be optionally substituted with one or more Rc·, or (b) Ra5 and Rb· together form a chain comprising —(CRc'Rd·)/-, where Rc and Rd are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyi, aryl, (Ci-Cs linear or branched chain alkyljaryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CFa, CONH2, -ORa, -NReRr, or -S(0)PRB; where Re and Rr where are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyi, or C3-C6 cycloalkyl, where said alkyi and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; /is 2, 3, 4 or 5; fris 1, 2, or 3; and, p is 0, 1 or 2. In one embodiment, the invention provides the compound of formula ID wherein R5 is a Ci-Ce linear or branched chain alkyi or C3-Ca cycloalkyl.
In another embodiment, the invention provides the compound of formula of formula ID wherein A îs a bond or -(CHîJk-, and R5 is an unsaturated, saturated or partially saturated monocyclic or bicyclic 15 ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyi, C3-C5 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyi, CN, hydroxyl, CF3, --NRaRb·, —ORe, —S(O)pRe and C3-C6 cycloalkyl; where Re and Rr are independently hydrogen, deu20 terium, Ci-Ce linear or branched chain alkyi, or Cs-Ce cycloalkyl, where said alkyi and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; fris 1, 2, or 3; and, p is 0, 1 or 2, In another embodiment, the invention provides the compound of formula I wherein R5 is an unsaturated ring structure containing a total of five to eleven
atoms having one or two heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur. In other embodiments, the invention provides the compound of formula I wherein R5 is furyl, thiofuryl, pyrrolyl, pyrazolyl, oxazolyl, azetidinyl, piperidinyl or thiazolyl, optionally substituted by one or two methyl.
In another embodiment, the invention provides the compound selected from the group consisting of:
4-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}pyridine-2-sulfonamide; 2,2,2-trifluoro-N-{cis-3-[methyf(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}ethanesulfonamide;
2-methyJ-N-{cis-3-[methyl(7H-pyrro!o[2,3-d]pyrimîdin-4-yl)amino]cyclobutyl}-propane-1sutfonamide;
N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}propane-1-sulfonamide; 1-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methanesulfonamide;
N-{cis-3-[(butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyrrofo[2,3-d]pyrimidin-4-amine;
1-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yi)amino]cyclobutyl}-azetidîne-3sulfonamide;
3-cyano-N-{cis-3-[methy I ( 7 H-pyrrolo[2,3 - d ] py rimidin-4-y Ijami no]cyciobuty l}-azetid ine-1 sulfonamide;
(1R,5S)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yi)amino]cyclobutyl}-6-oxa-3azabicyclo[3.1.1]heptane-3-sulfonamide;
(3R)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2r3-d]pyrimidin-4-yl)amino]cycfobutyl}-pyrroiidine-1sulfonamrde;
(3S)-3-cyano-N-(cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidinZ-yi)amino]cyclobutyl}-pyrrolidine-1sulfonamîde;
N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-1-(oxetan-3'yl)methanesulfonamîde;
1-(3,3-difluorocyc!obutyl)-N-{cis-3-[methyl(7H-pyrToio[2,3-d]pyrimidin-4yl)amino]cyclobuty1}methane-sulfonamide;
trans-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrirnidin-4-yi)arnino]-cyclobutyl}cyclobutanesulfonamîde;
cîs-3-(cyanomethyl)-N-{cis-3-[methyl(7H'pyrrolo[2,3-d]pyrimidin-4-yl)amino]10 cyclobutyljcyclobutane-sulfonamide;
N-[cis-3-({[(3,3-difluorocyclobutyi)methyl]sulfonyl}methyl)cyclobutyl]-N-methyl-7H-pyrrolo[2,3dJpyrimidin-4-amine;
(1S,5S)-1-cyano-N-{cfS-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}-3azabicyclo[3.1.0]hexane-3-sulfonamide;
(1R,5R)-1-cyano-N-{cis-3-[methyl(7H-pynOlo[2,3-d]pyrïmidin-4-yl)amino]cyclobutyi}-3azabicyclo[3.1.0]hexane-3-sulfonamide:
(3R)-1-[({cis-3-[methyl(7H-pyrTolo[2,3-d]pyrïnntdin-4-yt)amÎno]cyclobutyi}methyl)sulfonyl]pyrrolidine-3-carbonitrile;
1-[({cis-3-[methyî(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]-420 (trifluoromethyl)piperidin-4-ol;
N-(cis-3-{[(4,4-dίfluoropiperidinΊ-yl)sulfonyl]methyl}cyclobutyl)-N-methyl·7H-pyπΌJo[2,3d]pyrimîdin-4-amine;
(3S)-1-[({ds-3-[methyl(7H-pyirolo[2,3-d]pyrimidin-4-yl)arnino]cyclobutyl}methyl)sulfonyl]pyaolidine-3-carbonitrile;
N-(ds-3'{[(3-chloro-4-fluorûphenyl)sulfonyl]methyl}cyclobutyl)-N-rnethyl-7H-pynOlo[2,3d]pyrimidin-4-amine;
N-(cis-3-{[(2-cyc!opropylethyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4amine;
N-methyl-N-[ds-3-({[1-(propan-2-yl)pyrroiidin-3-yl]sulfonyl}methyl)cydobutyl]-7H-pyrrolo[2,3d]pyrimidin-4-amine;
3,3-difluoro-N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}-cydobutane10 sulfonamide;
1-[3-(cyanomethyi)oxetan-3-yl]-N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}methanesulfonamide;
cis-S-lcyanomethyO-S-methyl-N-Îcis-S-IrnethyKZH-pyrrolop.S-dlpyrimidin^-ylJaminoJcyclobutyt}cydobutanesulfonamide;
trans-3-(cyanomethyl)-3-methyl-N-{ds-3-[methyl(7H-pyrTolo[2,3-d]pynmidin-4yl)amino]cydobutyl}cyclobutanesulfonamide;
N-(2-cyanoethy!)-N-methy!-N'-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amîno]cydobutyl}sulfuric diamide;
N-{( 1 S, 3R)-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydopentyl}propane-1 -sulfonamide;
3-(2-hydroxypropan-2-yl)-N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}benzene-sulfonamide;
N-(cydopropylmethy l)-N '-{ds-3-[methy I (7H-py rrolo[2,3-d]pyrimidin-4-y l)am ino]-cydobuty l}su Ifuric diamide;
N-{cÎs-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-4'(1 H-pyrazol-3-yl)piperidine-1 sulfonamide;
Z-methyl-N-icis-S-Imethyl^H-pyrroîofZ.S-dJpyrimidirM-ylJaminoJcyclobutylJYSdihydropyrrolo(3,4-c]pyrazole-5(4H)-su!fonamide;
2-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)-sulfonyl]pyridine-4carbonitrile;
(1SI3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]cyclopentanecarbonitrile;
(1R,3R)-3-[({cis-3-[methyl(7H-pyrroto[2,3-d]pyrimidin-4-yi)amino]cyclobutyl}-methyl)sulfonyi] cyclopentanecarbonitrile;
1- cyclopropyl-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-methane sulfonamide;
3-cyano-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-y!)amino]cyclobutyl}-pyrrolidine-1sulfonamide;
N-methyl-N-{trans-3-Kpropyisulfonyi)methyl]cyclobutyl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine; and,
2- methyl-N-{ciS’3-[methy[(7H-pyrrolo[2,3-d]pyrimÎdin-4-yl)amino]cyclobutyl}-1,3-thiazole-5sulfonamide; or, a pharmaceutically acceptable sait thereof.
In another embodiment, the invention provides the compound selected from the group consisting of;
1-(3,3-difluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyljmethanesu Ifonamid e;
trans-3-(cyanomethyl)-N-{ciS-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyl}cyclobutanesulfonamide;
N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}propane-1-sulfonamide;
3,3-difluoro-N-{ds-3-[methyl(7H-pyrrdo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}-cyclobutanesulfonamide; and,
N4(1 S, 3R)-3-[methyl(7H-pyrroIo[2,3'd]pyrimidin-4-yl)amino]cyclopentyl}propane-1-sulfonamide;
or, a pharmaceutically acceptable sait thereof.
In other embodiments, the invention provides the compound selected from the group consisting of:
(3R)-3-cyano-N-{cis-3-[methyî(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobuty1}pyrrolidine-1sulfonamide;
('lR,5S)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-y1)amino]cyclobutyl}-6-oxa-3azabicyclo[3.1.1]heptane-3-sulfonamide;
(1S,5S)-1-cyano-N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}-3azabicyclo[3.1.0]hexane-3-sulfonamide;
N-(2-cyanoethyl)-N-methyl-N‘-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-415 yl)amino]cyclobutyl}sulfuric diamide; and,
2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-2,6dîhydropyrrolo[3,4-c]pyrazole-5(4H)-sulfonamide; or, a pharmaceutically acceptable sait thereof.
In another embodiment, the invention provides the compound selected from the group consisting of:
(3R)-1-[({ds-3-[methyt(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyljpyrrolidine-3-carbonitrile;
1-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}methyl)sulfonyl]-4(trifluoromethyl)piperidin-4-ol;
2l
N-(cis-3-{[(4,4-difluoropiperidin-1-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrTolo[2,3d]pyrimidin-4-amine;
(3S)-1-[({cis-3-[rnethy](7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyiIpyrrolidine-3-carbonitriie; and, or, a pharmaceutically acceptable sait thereof.
In yet another embodiment, the invention provides the compound selected from the group consisting of:
(1R,3R)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyljcyclo-pentanecarbonitrile;
( 1 S, 3S)-3-[({cis-3-[methyl(7H-pyrrol o[2,3-d]pyrimidin-4-yl)amino]cyc!obuty l}-methyl JsulfonylJcyclopentanecarbonitrile;
2-[({ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)-sulfony!]pyridine-4carbonitrile;
N-[cis-3-({[(3,3-difluorocyclobutyl)methyl]sulfonyl}methyl)cyclobutyl]-N-methyl-7H-pyrrolo[2,315 dJpyrimidin-4-amine; and,
N-{cis-3-[(butylsulfonyl)methyî]cyclobutyl}-N-methyl-7H-pyrroÎo[2,3-d]pyrimidin-4-amine; or, a pharmaceutically acceptable sait thereof.
Particularly preferred embodiments include 2-methyl-N-{cis-3-[methyl(7H-pyrTOlo[2,3-d]pyrimidin4-yl)amino]cyclobutyl}-1,3-thiazole-5-sulfonamide, N-{ds-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-420 yl)amino]cyclobutyl}-propane-1-sulfonamide; N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}-1-oxetan-3-ylmethanesulfonamide; 1-(3,3-difluorocyclobutyl)-N-{cis-3-[methyl(7Hpyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-methanesulfonamide; 3,3-difluoro-N-{cis-3-[methyl(7Hpyrrolo[2,3-d]pyrimrdin-4-yl)amino]cyclobutyl}cydobutanesulfonamide ; trans-3-(cyanomethyl)-N-(cis-3-
[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-cyclobutanesulfonamide; (1S,5S)-1-cyano-N{cis-3-[meÜiyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-3-azabi-cyclo[3.1.0]hexane-3sulfonamide; and, (3S)-1-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolidine-3-carbonitrile; or, a pharmaceutically acceptable sait thereof.
The présent invention also provides a pharmaceutical or a veterinary composition comprising a compound of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier.
The présent invention also provides a method of treating a disorder or condition related to dysrégulation of JAK, and particulariy of JAK1, in a subject, comprising administering to the subject a therapeutically effective amount of the compound having the structure of formula I, IA, JB, IC or ID, or a pharmaceutically acceptable sait thereof. In certain embodiments, the disorder or condition treated by the method is selected from among rheumatoid arthritis, myositis, vasculitis, pemphigus, Crohn's disease,, ulcerative colîtis, Alzheimer’s disease, lupus, nephritis, psoriasis, atopie dermatitis, autoimmune thyroid disorders, multiple sclerosis, major dépréssion disorder, allergy, asthma, Sjogren’s disease, dry eye syndrome, organ transplant rejection, xeno transplantation, Type I diabètes and complications from diabètes, cancer, leukemia, T cell acute lymphoblastic leukemia, adult T cell leukemia activated B-cell like, diffuse large B cell lymphoma, înflammatory bowel disease, septîc shock, cardiopulmonary dysfunction, chronic pulmonary obstructive disorder, acute respiratory disease, and cachexia comprising the step of adminis20 tering to a subject an effective amount of a composition comprising a compound of formula I, IA, IB, IC or ID. In certain embodiments, the therapeutically effective amount used in accord with the method is from 0.01 mg/kg of body weight/day to 100 mg/kg of body weight/day. In certain other embodiments, the therapeutically effective amount used in accord with the method is the therapeutically effective amount is
from 0.1 mg/kg of body weight/day to 10 mg/kg of body weight/day. In the practice of the method, the compound of formula I is preferably selected from N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}-propane-1-sulfonaniide, N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}-1-oxetan-3-ylmethanesulfonamrde; 1-(3,3-difluorocyclobutyl)-N-{cis-3-[methyl(7H5 pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-methanesulfonamide; 3,3-difluoro-N-{cis-3-[methyl(7Hpyrrolo[2,3-d]pyrimidin-4-yl)amino]cyctobutyl}cyclobutanesulfonamide ; trans-3-(cyanomethyl)-N-{cis-3[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclQbutyl}-cyctobutanesulfonamide; (1S,5S)-1-cyano-N(cis-3-[rnethyl(7H-pyrrolo[2,3-d]pyrinnidin-4-yl)amino]cyctobutyl)-3-azabi-cyclo[3.1.0]hexane-3sulfonamide; and, (3S)-1-[({cis-3-[methyl(7H-pynolo[2,3-d]pyrimidin-410 yl)amino]cycÎobutyl)methyi)sulfonyl]pyrrolidine-3-carbonitrile:, or a pharmaceutically acceptable sait thereof.
The présent invention further provides a method for treating or preventing a disorder or condition selected from atopie dermatitis, eczema, scleroderma, pruritus, other pruritic conditions, allergie reactions including allergie dermatitis in mammal, horse allergie diseases including bite hypersensitivity, summer eczema, sweet itch in horses, heaves, inflammatory airway disease, récurrent airway obstruction, and airway hyper-responsiveness by administering to a mammal in need a therapeutically effective amount of a compound of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable sait thereof.
In certain embodiments, the therapeutically effective amount used in accord with the method is from 0.01 mg/kg of body weight/day to 100 mg/kg of body weight/day. In certain other embodiments, 20 the therapeutically effective amount used în accord with the method is wherein the therapeutically effective amount is from 0.1 mg/kg of body weight/day to 10 mg/kg of body weight/day. In accord with the method, the mammal treated with the compound of the invention is selected from companion animais, dogs, and livestock. In certain embodiments, the compound of formula I, IA, IB, IC or ID, or a pharma-
ceutically acceptable sait thereof, may be administered in accord with the method orally, parenterally, or topically.
In the practice ofthe method, the compound of formula I is preferably selected from N-{cîs-3-[methyl(7Hpyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-propane-1-sulfonamide; N-{cis-3-[methyl(7H-pyrrolo[2,35 d]pyrimidin-4-yl)amino]cyclobutyl}-1-oxetan-3-ylmethanesulfonamide; 1-(3,3-difluorocyclobutyl)-N-{cis-3[methyl(7H-pynOlo[2,3’d]pyrimidin-4-yl)amino]cyclobutyl}methanesulfonamide; 3,3-difluoro-N-{cîs-3[methyl(7H-pyrTolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyi}cyclobutanesulfonamide ; trans-3-(cyanomethyl)N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-cyclobutanesulfonamide; (1 S,5S)-1 cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-3-azabi-cyclo[3.1.0]hexane-310 suifonamide; and, (3S)-1-[({cîs-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}methyl)suifonyl]pyrrolidine-3-carbonîtrile; or, a pharmaceutically acceptable sait thereof.
Compounds that hâve the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed ''isomers. Isomers that differ in the 15 arrangement of their atoms in space are termed stereoisomers”. It will be appreciated by those skilled in the art that the compound of formula 1, IA, IB, IC or ID can exist as cis- and trans- achiral diastereomers.
Included within the scope of the described compounds are ail isomers (e.g. cis-, trans-, or diastereomers) of the compounds described herein alone as well as any mixtures. Ail of these forms, including enantiomers, diastereomers, cis, trans, syn, anti, solvatés (including hydrates), tautomers, and 20 mixtures thereof, are included in the described compounds. Stereoisomeric mixtures, e.g. mixtures of diastereomers, can be separated into their correspondîng tsomers in a known manner by means of suitable séparation methods. Diastereomeric mixtures for example may be separated into their individual diastereomers by means of fractionated crystallization, chromatography, solvent distribution, and similar
procedures. This séparation may take place either at the level of one of the starting compounds or in a compound of formula I, IA, IB, IC or ID rtself. Enantiomers may be separated through the formation of diastereomeric salts, for example by sait formation with an enantiomer-pure chiral acid, or by means of chromatography, for example by HPLC, using chromatographie substrates with chiral ligands.
in therapeutic use for treating disorders in a mammal, a compound of the présent invention or its pharmaceutical compositions can be administered orally, parenterally, topically, rectally, transmucosally, or intestinally. Parentéral administrations include indirect injections to generate a systemic effect or direct injections to the afflicted area. Topical administrations include the treatment of skin or organs readily accessible by local application, for exampîe, eyes or ears. It also includes transdermal delivery to generate a systemic effect. The rectal administration includes the form of suppositories. The preferred routes of administration are oral and parentéral.
Pharmaceutically acceptable salts of the compounds of formula I, IA, IB, IC or ID include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besyiate, bicarbonate/carbonate, bi15 sufphate/sulfate, borate, camsylate, citrate, cyclamate, edisytate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsuifate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stéarate, succinate, tannate, tartrate, tosylate, 20 trifluoroacetate and xinofoate salts.
Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnésium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. For a revîew on suitable salts, see Handbook of Pharmaceutical Salts: Properties, Sélection, and Useby Stahl and Wermuth (Wiley-VCH, 2002).
Pharmaceutically acceptable salts of compounds of formula I, IA, IB, IC or ID may be prepared, respectively, by one or more of three methods: (i) by reacting the compound of formula I, IA, IB, IC or ID with the desired acid or base; (ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of formula I, IA, IB, IC or ID or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or (iii) by converting one sait of the compound of formula I, IA, IB, IC or ID to another by reaction with an appropriate acid or base or by means of 10 a suitable ion exchange column. Ail three reactions are typically carried out in solution. The resulting sait may precipitate out and be collected by filtration or may be recovered by évaporation of the solvent. The degree of ionization in the resulting sait may vary from completely ionized to almost non-ionized.
Pharmaceutical compositions of the présent invention may be manufactured by methods well known in the art, e.g., by means of conventional mixing, dissoiving, granulation, dragee-making,
I5 levigating, emulsifying, encapsulating, entrapping, lyophilizing processes or spray drying.
Pharmaceutical compositions for use in accordance with the présent invention may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compound into préparations, which can be used pharmaceutically. Proper formulation is dépendent upon the route of administration chosen. Pharmaceu20 tically acceptable excipients and carriers are generally known to those skilled in the art and are thus included in the instant invention. Such excipients and carriers are described, for example, în Remington's Pharmaceutical Sciences Mack Pub. Co., New Jersey (1991). The formulations ofthe invention can be
designed to be short-acting, fast-releasing, long-acting, and sustained-releasing. Thus, the pharmaceutical formulations can also be formulated for controlled release or for slow release.
Pharmaceutical compositions suitable for use in the présent invention include compositions wherein the active ingrédients are contained in an amount sufficient to achieve the intended purpose, i.e., 5 control or the treatment of disorders or diseases. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms/signs of disease or prolong the survival of the subject being treated.
The quantity of active component, which is the compound of this invention, in the pharmaceutical composition and unit dosage form thereof, may be varied or adjusted widely depending upon the manner 10 of administration, the potency of the particuiar compound and the desired concentration. Détermination of a therapeutically effective amount is well within the capability of those skilled in the art. Generally, the quantity of active component will range between 0,01 % to 99% by weight of the composition.
Generally, a therapeutically effective amount of dosage of active component will be in the range of about 0.01 to about 100 mg/kg of body weight/day, preferably about 0.1 to about 10 mg/kg of body 15 weight/day, more preferably about 0.3 to 3 mg/kg of body weight/day, even more preferably about 0,3 to 1.5 mg/kg of body weight/day It is to be understood that the dosages may vary depending upon the requirements of each subject and the severity of the disorders or diseases being treated.
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day, The sub-dose 20 itself may be further divided, e.g,, into a number of discrète loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
Also, it is to be understood that the initial dosage administered may be increased beyond the above upper level in order to rapidly achieve the desired plasma concentration. On the other hand, the
initial dosage may be smaller than the optimum and the daily dosage may be progressive^ increased during the course of treatment depending on the particular situation. If desired, the daily dose may also be divided into multiple doses for administration, e.g., two to four times per day.
Compounds ofthe présent invention are directed to pyrrolo[2,3-d]pyrimidine compounds useful as
Janus Kinase inhibitors (JAK-î). They are useful as therapeutic agents in connection with the treating or preventing a disorder or condition selected from rheumatoid arthritis, myositîs, vasculitis, pemphigus, Crohn's disease, ulcerative colitis, Alzheimer's disease, lupus, nephritis, psoriasis, atopie dermatitis, autoimmune thyroid disorders, multiple sclerosis, major dépréssion disorder, allergy, asthma, Sjogren's disease, dry eye syndrome, organ transplant rejection, xeno transplantation, Type I diabètes and complica10 lions from diabètes, cancer, leukemia, T cell acute lymphoblastic leukemia, adult T cell leukemia activated B-cell like, diffuse large B cell lymphoma, inflammatory bowei disease, septic shock, cardiopulmonary dysfunction, chronic pulmonary obstructive disorder, acute respiratory disease, cachexia, and other indications where immunosuppression/immunomodulation would be désirable, comprising the step of administering to a subject an effective amount of a compound of the invention.
There are substantial needs for safe and efficadous agents to control disorders related to JAK, such as atopie dermatitis, both in human and animais. The market for treating atopie dermatitis in animais is currently dominated by corticosteroids, which cause distressing and undesirable side effects in animais, specifically in companion animais such as dogs. Antihistamines are also used, but are poorly effective. A canine formulation of cyclosporine (ATOPICA™) is currently being marketed for atopie der20 matitis, but is expensive and has a slow onset of effîcacy. In addition, there are Gl toleration issues with ATOPICA™. Compounds of the présent invention are JAK inhibitors with sélective effîcacy against JAK1. These compounds are expected to provide an alternative to steroid usage and provide resolution of
chronic pruritus and inflammation that would either persist in atopie dermatitis or slowly regress following removal of allergen or causative agent, such as fleas in flea-allergic dermatitis.
Compounds of the présent invention may be administered in a pharmaceutically acceptable form either alone or in combination with one or more additional agents which modulate a mammalian immune
System or with anti-inflammatory agents. These agents may include but are not limited to cyclosporin A (e.g., Sandimmune™ or Neoral™, rapamycin, FK-506 (tacrolimus), leflunomlde, deoxyspergualin, mycophenolate (e.g., Cellcept™, azathioprine (e.g., Imuran™), daclizumab (e.g., Zenapax™), OKT3 (e.g., Orthocolone™), AtGam, aspirin, acetaminophen, ibuprofen, naproxen, piroxicam, and antiinflammatory steroids (e.g., prednisolone or dexamethasone). These agents may be administered as part of the same or separate dosage forms, via the same or different routes of administration, and on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.
Accordingly, the invention provides methods of treating or preventing a disease, condition or disorder associated with JAK in a subject, such as a human or non-human mammal, comprising administer15 ing an effective amount of one or more compounds described herein to the subject. Suitable subjects that can be treated include domestic or wild animais, companion animais, such as dogs, cats, horses and the like; tivestock including, cows and other ruminants, pigs, poultry, rabbits and the like; primates, for example monkeys, such as rhésus monkeys and cynomolgus (also known as crab-eating or long-tailed) monkeys, marmosets, tamarins, chimpanzees, macaques and the like; and rodents, such as rats, mice, gerbils, guinea pigs and the like. In one embodiment, the compound is administered in a pharmaceutically acceptable form, optionally in a pharmaceutically acceptable carrier.
Conditions in which sélective targeting of the JAK pathway or modulation of the JAK kinases, particularly JAK1, are contemplated to be therapeutically useful include, arthritis, asthma, autoimmune dis-
eases, cancers or tumors, diabètes, certain eye diseases, disorders or conditions, inflammation, intestinal inflammations, allergies or conditions, neurodegenerative diseases, psoriasis, and transplant rejection. Conditions which can benefit from sélective inhibition of JAK1 are discussed in greater detail below.
Accordingly, the compound of formula I, IA, IB, IC or ID, or its pharmaceutically acceptable salts, and pharmaceutical compositions thereof can be used to treat a variety of conditions or diseases such as the following;
Arthritis, including rheumatoid arthritis, juvénile arthritis, and psoriatic arthritis;
Autoimmune diseases or disorders, including those designated as single organ or single cell-type autoimmune disorders, for example Hashimoto's thyroiditis, autoimmune hemolytic anémia, autoimmune atrophie gastritis of pernîcious anémia, autoimmune encephalomyelitis, autoimmune orchitis, Goodpasture's disease, autoimmune thrombocytopenia, sympathetic ophthalmia, myasthenia gravis, Graves' disease, primary biliary cirrhosis, chronic aggressive hepatitis, ulcerative colitis and membranous glomerulopathy, those designated as invotving systemic autoimmune disorder, for example systemic lupus erythematosis, rheumatoid arthritis, Sjogren's syndrome, Reiter’s syndrome, polymyositis15 dermatomyositis, systemic sclerosis, polyarteritîs nodosa, multiple sclerosis and bullous pemphigoid, and additional autoimmune diseases, which can be O-cell (humoral) based or T-cell based, including Cogan’s syndrome, ankylosing spondylitis, Wegener's granulomatosis, autoimmune alopecia, Type I or juvénile onset diabètes, or thyroiditis;
Cancers or tumors, including alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, including acute myelogenous leukemia and chronic myelogenous leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi's sarcoma, myelomas including mul-
tiple myeloma, myeloproliferative disorders, proliférative diabetic retinopathy, or angiogenic-assocrated disorders including solid tumors;
Diabètes, including Type I diabètes or complications from diabètes;
Eye diseases, disorders or conditions including autoimmune diseases of the eye, keratoconjunctivitis, vernal conjunctivitis, uveitis including uveitis associated with Behcet's disease and lens-induced uveitis, keratitis, herpetic keratitis, contcal keratitis, corneai épithélial dystrophy, keratoleukoma, ocular premphigus, Mooren’s ulcer, scleritis, Grave's ophthalmopathy, Vogt-KoyanagiHarada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, îridocyclitis, sarcoidosis, endocrine ophthaimopathy, sympathetic ophthalmitis, allergie conjunctivitis, or ocular neovascularization;
Intestinal inflammations, allergies or conditions including Crohn’s disease and/or ulcerative colitis, inflammatory bowel disease, celiac diseases, proctitis, éosinophilie gastroenteritis, or mastocytosis;
Neurodegenerative diseases including motor neuron disease, Alzheimeris disease, Parkinson’s disease, amyotrophie latéral sclerosis, Huntington's disease. cérébral tschemia, or neurodegenerative disease caused by traumatic injury, strike, glutamate neurotoxicity or hypoxia; ischemic/reperfusion injury in stroke, myocardial ischemica, rénal ischemia, heart attacks, cardiac hypertrophy, atherosclerosis and arteriosclerosis, organ hypoxia, or platelet aggregation;
Skin diseases, conditions or disorders including atopie dermatitis, eczema, psoriasis, scleroderma, pruritus or other pruritic conditions;
Allergie reactions including allergie dermatitis in mammal (including horse allergie diseases such as bite hypersensitivity), summer eczema, sweet itch in horses, heaves, inflammatory airway disease, récurrent airway obstruction, airway hyper-responsiveness, or chronic obstruction pulmonary disease;
Asthma and other obstructive airways diseases, including chronic or inveterate asthma, late asthma, bronchitis, bronchial asthma, allergie asthma, intrinsrc asthma, extrinsic asthma, or dust asthma;
Transplant rejection, including pancréas islet transplant rejection, bone marrow transplant rejection, graft-versus-host disease, organ and cell transplant rejection such as bone marrow, cartilage, cornea, heart, intervertébral dise, islet, kidney, limb, liver, Jung, muscle, myoblast, nerve, pancréas, skin, small intestine, or trachea, or xeno transplantation; and
Another embodiment provides a method of selectively inhibiting a JAK1 enzyme, which includes contacting the JAK enzyme with either a non-therapeutic amount or a therapeutically effective amount of one or more of the presently taught compounds. Such methods can occur in vivo or in vitro, in vitro contact can involve a screening assay to détermine the efficacy of the one or more compounds against a selected enzyme at various amounts or concentrations, in vivo contact with a therapeutically effective amount of the one or more compounds can involve treatment of a described disease, disorder or condition or prophylaxie of organ transplant rejection in the animal in which the contact occurs, The effect of the one or more compounds on the JAK enzyme and/or host animal can also be determined or measured. Methods for determining JAK activity include those described in the Exemples as well as those disclosed in WO99/65908, WO 99/65909, WO01/42246, W002/00661, W002/096909, W02004/046112 and W02007/012953.
Chemical Synthesis
The following schemes and written descriptions provide general details regarding the préparation of the compounds of the invention.
SULFONAMIDES
Compounds of formula I, wherein p is 2, X is NH, Y is AR5,and A is a bond, may be prepared according to Scheme 1.
Step 1
VII
Step 5
I (p = 2, X = NH, Y = AR5; A = bond)
It will be apparent to those skilled in the art that sensitive functional groups (PG) may need to be protected and deprotected during the synthesis of a compound of the invention. Protection and deprotection may be achieved by conventional methods, as described, for example, in “Protective Groups in Organic Synthesis by T. W. Greene and P. G. M. Wuts, John Wiley & Sons Inc. (1999), and référencés therein. Thus, in Scheme 1, Step 1, a compound of formula II, wherein Q1 is halogen, is treated with a protecting agent to provide a compound of formula III, wherein PG' is an arylsulfonyl protecting group such as benzenesulfonyl, or preferably pa/a-toluenesulfonyl (“tosyl). The protecting group may be installed by reaction of the compound of formula II with an arylsulfonyl chloride, preferably tosyl chloride, 10 in the presence of a base such as aqueous sodium hydroxide solution and an organic solvent such as acetone. The reaction is typically run at 0°C to about 50°C, preferably at about 23°C (room température).
Altematively, bases such as sodium hydride and potassium tert-butoxide may be used, employing a suit-
able solvent such as N,N-dimethylfomnamide or tetrahydrofuran. Several compounds of formula II are known in the literature and hâve been prepared by the methods above. For example, the synthesis of compound of formula ll, wherein Q1 is Cl and R2 and R3 are hydrogen has been reported previously, for example in WO 2007 012953.
in Scheme 1, Step 2, the protected compound of formula III is combined with 1-2 équivalents of an amine of formula IV in the presence of a 1-3 équivalents of a base and a protic solvent to afford a compound of formula V. Suitable bases include triethylamine, diisopropylethylamine, and potassium carbonate while suitable solvents include methanol, éthanol, diisopropyl alcohol and water or mixtures thereof. The réaction is typically run at about 23°C to about 15Û°C, preferably about 75°C. It will be noted that the amine of formula IV contains a second amino group that îs protected with a protecting group PG2 that can be removed under conditions that do not lead to loss of PG1. Suitable protecting groups PG2 include t-butoxycarbonyl (“Boc’j and f'Cbz), preferably benzyloxycarbonyl.
In Scheme 1, Step 3, the protecting group PG2 is removed from the compound of formula V under conditions that do not lead to loss of PG1 to give a primary amine (or a sait thereof) of formula VI. When
PG2 is benzyloxycarbonyl, the benzyloxycarbonyl protecting group may be removed by hydrogenolysis wherein the compound of formula V is exposed to hydrogen or a hydrogen transfer reagent such as cy~ clohexene in the presence of a hydrogénation catalyst such as palladium hydroxide using a solvent such as methanol, acetic acid or, preferably, éthanol. Altematively, when PG2 is benzyloxycarbonyl, the benzyloxycarbonyl protecting group may be removed by treatment of the compound of formula V with a solution of hydrogen bromide (about 6 équivalents) in acetic acid optionally in the presence of a suitable solvent such as ethyl acetate at a température from about minus 20DC to about 40°C, preferably less than 25°C. This latter deprotectîon method is preferred wherein n is 1, R2, R3 and R4 are hydrogen, R’ is methyl, PG1 is tosyl and PG2 is benzyloxycarbonyl and provides the amine of formula VI as the
dihydrobromide sait. When PG2 is t-butoxycarbonyl, the t-butoxycarbonyl protecting group may be removed by treatment with an excess of an acid such as hydrochlorrc acid or trifluoroacetic acid in a solvent such as dichloromethane or 1,4-dioxane.
In Scheme 1, Step 4, the primary amine of formula VI (or sait thereof) is converted to a sulfona5 mide dérivative of formula VII by treatment with an activated sulfonic acid dérivative of formula VIII, wherein Q2 is halogen, O-alkyl or O-aryl in the presence of a base. Most commonly, VIII is a sulfonyl chloride dérivative wherein Q2 is CI. Many sulfonyl chlorides may be obtained from commercial sources. Also, several methods exist for the préparation of sulfonyl chlorides, which are well known to those skilled in the art and hâve been described in texts such as Advanced Organic Chemistry by J. March, John
Wiley & Sons (1985). Typically, the amine of formula VI is treated with a sulfonyl chloride dérivative of formula VIII wherein Q2 is Cl in the presence of at least one équivalent of a base such as triethylamine or diisopropylamine in a suitable solvent such as dichloromethane, tetrahydrofuran or acetonitrile. When a sait form of the amine is used, an additional équivalent of base is used for each équivalent of acid forming the sait. For example, using a dihydrobromide sait, two extra équivalents of base are used. The reaction may be run from about minus 20°C to about 50°C, preferably starting the reaction at about 0°C and then allowing it to warm to about 23DC (room température).
Finally, in Scheme 1, Step 5, the sulfonamide dérivative of formula Vil is deprotected to afford a compound of formula 1, wherein p is 2. X is NH, Y is AR5and A is a bond. Two methods are typically employed, the choice of which is determined by the compatibility of the conditions with other functional groups on the molécule. The first method involves exposure of the compound of formula VII to an excess (about 4 équivalents) of a base such as lithium hydroxide or sodium hydroxide. The reaction is run in a solvent mixture containing water and an alcohol such as methanol or éthanol. It may also be run în a mixture of water and tetrahydrofuran, and, optionally an alcohol such as methanol or éthanol. The reaction
may be run at a température of about 23 “C to about 100°C, typically about 60’C. The second method, which is preferred in instances where there is hydroxide-sensitive functionality such as nitrile présent in the molécule, involves reaction of the compound of formula VII with an excess of tetrabutylammonium fluoride (4-25 équivalents) in a solvent such as 1,2-dimethoxyethane or, preferably tetrahydrofuran, The deprotection is conducted at a température of about 0°C to about 60°C, preferably about 23°C.
Compounds of formula II, wherein Q’ is halogen, are commercialiy available or are known in the chemical literature. For exampie, 4-chloro-7W-pyirolo[2,3-a]pyrimidine, wherein Q1 is Cl and R2 and R3 are both hydrogen, is a readily available commercial compound.
Compounds of formula IV are known in the chemical literature or may be prepared by standard chemical reactions well known to one skilled in the art.
An alternative method of preparing compounds of the invention wherein p is 2, X is NH, Y is AR5, A is a bond is shown in Scheme 2.
Scheme 2
= bond)
In Scheme 2, Step 1, a compound of formula IX is combined with a benzyloxycarbamate dériva tive of formula X in the presence of a base (1-5 équivalents) to provide a benzyloxycarbamate dérivative
of formula XI. The reaction is carried out in a solvent such as water or an alcohol such as éthanol, optionally with addition of a miscible co-solvent such as tetrahydrofuran. Suitable bases include potassium carbonate, césium carbonate, triethylamine and diisopropylethylamine. The reaction is run at about 23°C to about 100°C. When n is 1, R2, R3 and R4 are hydrogen, and R1 is methyl, the preferred conditions are to run the reaction in water, using potassium carbonate (3 équivalents) as base, starting the reaction at about 23°C and then heating to about 95°C.
In Scheme 2, Step 2, the benzyloxycarbamate dérivative of formula XI is deprotected by exposure to hydrogen or a hydrogen transfer reagent such as cyclohexene in the presence of a hydrogénation catalyst such as palladium hydroxide. At the same time, under the conditions of the deprotection, the chlorine atom at the 2-position of 7H-pyrrolo[2,3-d]pyrimidine ring is replaced with hydrogen to provide an amine hydrochloride sait of formula XII. The reaction is run in a solvent such as methanol or éthanol at a température of about 50°C to about 80°C. When R2, R3 and R4 are hydrogen, and R’ is methyl, the preferred conditions are to run the réaction in éthanol at about 78°C using palladium hydroxide as catalyst, and cyclohexene (about 20 équivalents) as a hydrogen transfer reagent.
Finaily, in Scheme 2, Step 3, the amine hydrochloride of formula XII is converted to a sulfonamide of formula I, wherein p is 2, X is NH, Y is AR5, A is a bond by reaction with an activated sulfonic acid dérivative of formula VIII, wherein Q2 is halogen, O-alkyl or O-aryl in the presence of at least two équivalents of a base. Most commonly, VIII is a sulfonyl chloride dérivative wherein Q2 is Cl. Suitable bases include triethylamine, diisopropylethylamine and potassium carbonate. Suitable solvents include N,N20 drmethylformamide, and a mixture of tetrahydrofuran and water. The reaction may be run at a température of about minus 20“C to about 50°C preferably at about 23°C. Altematively, the amine hydrochloride of formula XII is first treated with about 2 équivalents of trimethylchlorosilane in the presence of about 2-3 équivalents of a base such as lithium bis(dimethylsilyl)amide or sodium bis(dimethylsilyl)amide in an suit-
able aprotrc solvent such as tetrahydrofuran. Then, after about 1 hour, about 1.2 équivalents of the sulfonyl chloride of formula VIII, Q2 is Cl is added to provide, after workup, the sulfonamide of formula I, wherein p is 2, X is NH, Y is AR5, A is a bond The reaction may be run at a température of about minus 20°C to about 50°C, preferably at about 23°C.
Compounds of formula IX, are commercially available or are known in the chemical literature. For example, 2,4-dichloro-7Z/-pyrrolo[2,3-a]pyrimidine1 wherein R2 and R3 are both hydrogen, is commercially available. Its synthesis is described in PCT International Publication No. W02007/012953.
SULFAMIDES
Compounds of formula I, wherein p is 2, X is NH, and Y is NRaRb, may be prepared according to
Scheme 3.
l(p = 2,X = NH, Y = NRaRb)
In Scheme 3, Step 1, an amine of formula VI (or sait thereof), wherein PG1 is an arylsulfonyl protecting group such benzenesulfonyl, or preferably tosyi, is converted to oxazolidinone dérivative of formu la XIII. First, a solution of N-chlorosulfonylisocyanate (1 équivalent) is slowly added to a solution of 2bromoethanol (1 équivalent) at a température of about -40°C to about 10°C, preferably about 0°C. Subsequently, after 0.5 to 2 hours, a solution of the amine of formula VI (1 équivalent) and a base such as triethylamine or diisopropylethylamine (about 3 équivalents, plus one équivalent for each mole of acid
forming a sait) is added slowly and frie reaction is allowed to warm to about 23°C over a period of about to 24 hours. Suitable solvents for the reaction include chloroform or preferably dichloromethane.
In Scheme 3, Step 2, the oxazolidinone dérivative of formula Xlll is reacted with 1-3 équivalents of an amine of the formula HNRaRb, in the presence of a base (2-5 équivalents), to afford a sulfamide de5 rivative of formula XIV. Suitable bases include triethylamine and diisopropylethylamine. The reaction is preferably carried out by heating to about 90°C to about 150°C in a pressure vessel using a suitable solvent such a N.N-dimethylformamide or acetonitrile.
In Scheme 3, Step 3, the compound of formula XIV is deprotected, removing the arylsulfonyl protecting group PG1 to provide a sulfamide dérivative of formula 1, wherein p is 2, X is NH, and Y is NRaRt>.
The reaction may be carried out by one of the two general methods described for Scheme 1, Step 5.
Again, the choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molécule. Altematively, the sulfamides of the formula XIV may be obtained directly from an amine of formula VI (or sait thereof). Thus, in Scheme 3, Step 4, the amine of formula VI (or sait thereof) is treated with a sulfamoyl chloride of the formula CI-SOïNRaRb and a base such as tri15 ethylamine or diisopropylethylamine as described for Scheme 1, Step 4. Sulfamoyl chlorides of the formula CI-SOîNRaRb may be prepared, in turn, from amines of the formula HNRaRb according to the procedures reviewed by W. R. Bowman and R. J. Marmon in Comprehensive Organic Functional Group Transformations, Volume 2”, Pergamon (1995).
The compounds of formula I, wherein p is 2, X is NH, and Y is NRaRb may also be obtained di20 rectly from an amine of formula XII (or sait thereof). Thus, in Scheme 3, Step 5, the amine of formula Xll(or sait thereof) is treated with a sulfamoyl chloride of the formula CI-SOzNRaRb and a base such as triethylamine or diisopropylethylamine as described for Scheme 1, Step 4. Amines of formula XII are obtained as described for Scheme 2. Amines of formula XII (or salts thereof) may be obtained by the re-
moval of the arylsulfonyl protecting group PG1 from a compound of the formula VI (refer to Scheme 1).
The deprotection may be carried out by one of the two general deprotection methods described for Scheme 1, Step 5. The chotce of deprotection method is determined by the compatibility of the conditions with other functional groups on the molécule.
REVERSE SULFONAMIDES
Compounds of formula I, wherein p is 2, X is CH2, and Y is NRaRb, may be prepared according to Scheme 4.
Scheme 4
Jn Scheme 4, Step 1, a compound of formula IJI (refer to Scheme 1), is combined with an amino alcohol of formula XV in the presence of a base and a polar solvent to afford a compound of formula XVI. Suitable bases include triethylamine and diisopropylethylamine while suitable solvents include methanol,
diisopropyl alcohol and acetone. The reaction is typically run at about 23°C to about 70°C. Preferably, a catalytic amount (about 1 mole %) of potassium iodide is added to the reaction.
In Scheme 4, Step 2, the compound of formula XVI is converted to a compound of formula XVII, wherein LG is a leaving group such as bromo, iodo, methanesulfonate or, preferably, para5 toluenesulfonate. Methods for installing such leaving groups are well-known to those skilled in the art and hâve been described in texts such as “Advanced Organic Chemistry” by J. March, John Wiley & Sons (1985). In the case where LG is pa/a-toluenesulfonate, the compound of formula XVI is treated with para· toluenesulfonyl chloride in the presence of a base such as triethylamine, diisopropylethylamine or N,Ndimethylaminopyridine in an aprotic solvent such as dichloromethane or tetrahydrofuran. The reaction is run at a température of about -10°C to about 40°C, preferably beginning at around 0°C and allowing the reaction to warm to about 23°C.
In Scheme4, Step 3, the compound of formula XVII is combined with a sait of thioacetic acid, preferably potassium thioacetate to yield a thioester dérivative of formula XVIII. The reaction is carried out in a polar solvent such as Ν,Ν-dimethylformamide or N-methytpyrrolidine, at a température of about 23°C 15 to about 80°C, preferably at about 55°C.
In Scheme 4, Step 4, the thioester dérivative of formula XVIII is converted to a sulfonic acid dérivative of formula XIX by reaction with an aqueous solution of hydrogen peroxide, typically 30% by weight. The reaction is carried out in an acidic solvent such as formic or acetic acid at a température from about 0°C to about 40°C, preferably at about 23°C.
In Scheme 4, Step 5, the sulfonic acid dérivative of formula XIX is converted to a sulfonyl chloride dérivative of formula XX. Several methods for carrying out this functional group transformation are known in the literature. The preferred method is to treat the compound of formula XIX with an excess (3-15 équivalents) of thionyl chloride in the presence of a catalytic amount of Ν,Ν-dimethylforrnamide in an
aprotic solvent such as dichloromethane or chloroform. The reaction may be run from about minus 20°C to about 100°C, preferably beginning the réaction at about 0°C, and then warming to about 75°C.
Altematively, in Scheme 4, Step 6, the thîoester dérivative of formula XVIII may be directly converted to the sulfonyl chloride dérivative of formula XX by treatment with a chlorinating agent. Several methods for canrying out this functional group transformation are known in the literature. Chlorinating agents include chlorine gas and N-chlorosuccinimide, and the reaction is commonly run in the presence of an acid such as hydrochloric acid or acetic acid. Mixed aqueous solvents Systems are often used, such as water and dichloromethane and water and acetonitrile.
In Scheme 4, Step 7, the sulfonyl chloride dérivative of formula XX is combined with 1-3 équivalents of an amine ofthe formula HNRaRb to form a sulfonamide dérivative of formula XXI. The reaction is run in the presence of at least one équivalent of a base such as trrethylamine or diiosopropylethylamine at a température from about minus 20°C to about 50°C, preferably starting the reaction at about 0°C and allowing the reaction to warm to about 23°C. The reaction is run in an aprotic solvent such as tetrahydrofuran or dichloromethane.
Finally in Scheme 4, Step 8, the arylsulfonyl protecting group PG1 is removed to provide a compound ofthe formula I, wherein p is2, X isCI-b, and Y is NRaRb. The reaction may be carried out by one ofthe two general deprotection methods described for Scheme 1, Step 5. The choice of deprotection method îs determined by the compatibility ofthe conditions with other functional groups on the molécule. Amino alcohols of formula XV are known in the chemical literature or may be prepared by methods well known to one skilled in the art.
SULFONES, Sulfoxides and Thioethers
Compounds of formula I, wherein p is 0, 1, or 2, X is CHs, Y is AR5 and A is a bond, may be prepared according to Scheme 5.
Scheme 5
In Scheme 5, Step 1, a compound of formula XVII (refer to Scheme 4) is treated with 1-2 équivalents of a thiol of the formula R5SH in the presence of 1-2 équivalents of a base to give a sulfide of the formula XXIII. Suitable bases include sodium hydride, sodium bis(trimethylsilyl)amide, 1,55 diazabicyclo[4.3.0]non-5-ene (DBN) and preferably, 1,8-diazabicyc!o[5.4.0]undec-7-ene (DBU). The reaction is carried out in a solvent such as N.N-dimethylforrriamide or N-methylpyrrolidinone at a température from about 0°C to about 50°C, preferably at about 23°C.
In Scheme 5, Step 2, the compound of formula XXIII is deprotected, removing the aryfsuifonyi protecting group PG1 to provide a compound of formula XXIV. The reaction may be carried out by one of
the two general deprotection methods described for Scheme 1, Step 5. The choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molécule.
In Scheme 5, Step 3, a sulfide of formula XXIV is oxidized to yield a sulfone of formula I, wherein p is 2, X is CH2, Y Is AR5and A is a bond. Several methods are known in the literature and ail involve the use of an oxidant such as meta-chloroperbenzoic acid, hydrogen peroxide, or potassium peroxymonosulfate (Oxone®). A preferred method is to treat the compound of formula XXIV with 2 équivalents of potassium peroxymonosulfate (Oxone®) in a solvent mixture of tetrahydrofuran, éthanol and water at a température of about 23°C. The sulfide of formula XXIV may also be oxidized under milder conditions, for exampie using 1 équivalent of meta-chlorobenzoic acid in a solvent such as dichloromethane at 10 about Û°C to produce a sulfoxide of formula I, wherein p is 1, X is CH2, Y rs AR5 and A is a bond
It is noted that the order of Steps 2 and 3 in Scheme 5, may optionally be reversed such that the oxidation step is carried out prior to the deprotection step.
Compounds of the formula I, wherein p is 0, X is CH2, Y is AR5and A is a bond are prepared in
Scheme 5, Step 4 by removing the arylsulfonyl protecting group PG1 from a compound of formula XXIII.
The reaction may be carried out by one of the two general deprotection methods described for Scheme 1,
Step 5. Again, the choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molécule.
In Scheme 5, Step 5, compounds of the formula XXIII are altematively prepared from a thîoacetate dérivative of formula XVIII. First, the thioacetate of formula XVIII is dissolved in a solvent such 20 as éthanol, methanol, or water (or a mixture thereof). A suitable base such as potassium carbonate or césium carbonate (about 2 équivalents) is added and nitrogen is bubbled through the solution to remove oxygen. An aikylating agent of the formula R5-LG is then added, wherein LG is a leaving group such as bromo, iodo, methanesulfonate or, para-toluene-sulfonate. The reaction is conducted at a température
from about minus 20°C to about 30°C. Preferably, the reaction is started at about 0°C and then allowed to warm to about 23°C.
Many thiols of the formula R5SH and alkylating agents of the formula R5-LG may be obtained from commercial sources. Also, several methods exist for the préparation of such compounds, which are well known to those skilled in the art and hâve been described in texts such as Advanced Organic Chemistry” by J. March, John Wiley & Sons (1985).
It is noted that certain compounds of the invention can be obtained by functional group transformations at a late stage of the synthesis, for example, by chemical modification of the groups R4 or R5 after carrying out Steps 4 or 5 in Scheme 1, Step 3 în Scheme 2, Steps 2, 3 or 4 in Scheme 3, Steps 7 or 8 10 Scheme 4 and Steps 2, 3, 4 or 5 in Scheme 5. Such functional group transformations may include one step or multiple steps, for example, réduction of an ester to an alcohol, reoxidation to an aldéhyde, addition of an organomagesium reagent to form a secondary alcohol, reoxidation to a ketone and, finally, addition of an organomagesium reagent to yield a tertiary alcohol.
In executing the synthesis of the compounds of the invention, one skilled in the art will recognize the need to sample and assay reaction mixtures prior to work up in order to monitor the progress of reactions and décidé whether the reaction should be continued or whether it is ready to be worked up to obtain the desired product. Common methods for assaying reaction mixtures include thin-layer chromatography (TLC), liquid chromatography/mass spectroscopy (LCMS), and nuclear magnetic résonance (NMR).
One skilled in the art will also recognize that the compounds of the invention may be prepared as mixtures of diastereomers or géométrie isomers (e.g., cis and trans substitution on a cycloalkane ring). These isomers can be separated by standard chromatographie techniques, such as normal phase chromatography on silica gel, reverse phase préparative high pressure liquid chromatography or supercritical
fluid chromatography. One skilled in the art will also recognize that some compounds of the invention are chiral and thus may be prepared as racemic or scalemic mixtures of enantiomers. Several methods are available and are well known to those skilled in the art for the séparation of enantiomers. A preferred method for the routine séparation enantiomers is supercritical fluid chromatography empioying a chiral stationary phase.
EXPERIMENTAL SECTION
Except where otherwise noted, reactions were run under an atmosphère of nitrogen. Chromatography on silica gel was carried out using 250-400 mesh silica gel using pressurized nitrogen (-10-15 psi) to drive solvent through the column (flash chromatography). Where indicated, solutions and reaction mixtures were concentrated by rotary évaporation under vacuum.
Example 1: 2,2,2- Trifluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}ethanesuifonamide
Step 11 Benzyl[cis-3-(methylamino)cyclobutyl]carbamate and benzyl[trans-315 (methylamino)cyclobutyijcarbamate
A 33% solution of methylamine (1000 mL, 9.13 mol) in absolute éthanol was added to a mixture of benzyl (3-oxocyclobutyl)carbamate (WO2012/75381 A1 and WO2012/09678 A1) (200 g. 0.913 mol) and acetic acid (88 mL) stirring in éthanol (1000 mL) at 0°C. The réaction mixture stirred for at 0°C for 1.5 hours and then stirred at room température for 2 hours. Lithium borohydrîde (41g, 2.05 mol) was added in portions to the reaction mixture at -70°C. After addition was complété, the reaction mixture was stirred at -70°C for 1 hour and then allowed to warm to room température over 12 hours. The reaction mixture was quenched with water (400 mL), and concentrated under vacuum to remove éthanol. The aqueous layer was acidîfïed with concentrated hydrochloric acid to pH 2, washed with ethyl acetate (2 x 1000 mL),
basified with 10 % sodium hydroxide to pH 9-10 and then extracted with dichloromethane (3 x 1000 mL). The combined organic layers were washed with brine (1000 mL), dried over sodium sulfate, and concentrated to obtain the crude product as a pale brown liquid. This was dissolved in dichloromethane (400 mL) and cooled to 0°C. To the resulting solution was added a solution of 4M HCI in dioxane (300 mL). The mixture was stirred at 0°C for 30 minutes, and then at room température for 12 hours. The reaction mixture was filtered and the remaining solid was recrystallized from a mixture of methanol and methyl tertbutyl ether to afford the cis-isomer as a white solid (111.09 g, 52%). 1H NMR: (400 MHz, D2O): δ 7.337.38 (m, 5H); 5.02 (s, 2H), 3.83-3.87 (m, 1H), 3.89-3.41 (m, 1H), 2.66-2.70 (m, 2H), 2.56 (s, 3H), 2.032.05 (m, 2H). LC/MS (exact mass) calculated for C13H18N2O2; 234.137, found (M + H+); 235,1.
The trans isomer was isolated from the mother liquor using supercritical fluid chromatography.
Step 2: Benzyl (cis-3-[(2-chloro-7H-pyrrolo[2,3-d]pyrimidin~4~yl)(rnethyl)amino]-cyclobutyl}carbamate To a solution of potassium carbonate (20.47 g, 148 mmol) in water (180 mL) was added benzyl [cis-3-(methylamino)cyc1obutyl]carbamate (13.57 g, 50.2 mmol), followed by 2,4-dichloro-7H-pyrrolo(2,3djpyrimidine (9.0 g, 47.9 mmol) at room température. After addition was complété, the reaction mixture was stirred at 95°C ovemight. The mixture was filtered to collect the solid. The filter cake was washed with water and dried under vacuum to afford the title compound (16.5 g, 89.7%) as a yellow solid. 1H NMR (400 MHz, DMSO-ûfe): δ 11.81 (sm 1 H), 7.65 (d, 1 H), 7.38 (m, 5 H). 7.16 (m, 1 H), 6.67 (d, 1 H), 5.02 (s, 2 H), 4.81 (m, 1 H), 3.85 (m, 1 H), 3.25 (s, 3 H), 2.53 (m, 2 H), 2.25 (m, 2 H). LC/MS (exact mass) calculated for C19H20CIN5O2; 385.131, found (M + H*); 386.1.
Step 3: cis-N-Methyl-N-7H-pyrroio[2,3-d]pyrimidîn-4-ylcyclobutane-1,3-diamîne hydrochioride
A mixture of {cis-3-[(2-chloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)(methyl)-amino]cyclobutyl}carbamate (13.0 g, 34,0 mmol), Pd(OH)2 (40.3 g, 40.8 mmol) and cyclohexene (72.5 mL, 0.71 mol) in éthanol (300 mL) was stirred at reflux for 3 hours. The reaction mixture was filtered through a pad of Celite® and the
pad was washed with methanol. The filtrate was concentrated under vacuum to afford the title compound (4.8 g, 66%) as a white solid. 1H NMR (400 MHz, DMSO-de): δ 11.68 (br, 1H), 8.11 (s, 1H), 7.67 (br, 2H), 7.17 (d, 1H), 6.65 (d, 1H), 5.08 (m, 1 H), 3.45 (m, 1H), 3.26 (s, 3H), 2.31 (m. 4H). LC/MS (exact mass) calculated for CnHisNsl 217.133, found (M + H*); 218.1.
Step 4: 2,2,2-Trifluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-dJpyrimidin-4-yl)3mino]cyclobuty!}ethanesulfonam/de
To a solution of cis-N-methyl-N-7H-pyrrolo[2.3-d]pyrimidin-4-ylcyclobutane-1,3-diamine hydrochioride (100 mg, 0.39 mmol) în tetrahydrofuran (0.8 mL) was added lithium bis(trimethylsilyl)amide (1M solution in tetrahydrofuran) (0.9 mL, 0.9 mmol) and chlorotrimethylsilane (94 mg, 0.88 mmol) at room température. The reaction mixture was stirred for 45 minutes and then 2,2,2-trifluoroethanesulfonyl chtoride (86 mg, 0.47 mmol) was added slowly. The mixture was stirred at room température for 18 hours and then partitioned between dichloromethane and water. The aqueous layer was and extracted twice with dichloromethane and the combined organic layers were concentrated to afford the crude product as a tan solid. The crude material was purified by chromatography on silica gel eluting with a mixture of dîchloro15 methane and methanol (93:7) to afford the title compound as a white solid (93 mg, 65%). 1H NMR (400 MHz, DMSO-de): δ 11.61 (br. s„ 1 H), 8.20 (d. 1 H), 8.08 (s. 1 H), 7.13 (d, 1 H), 6.60 (d, 1 H), 4.80-4.94 (m, 1 H). 4.34 (q. 2 H), 3.58-3.71 (m, 1 H), 3.23 (s, 3 H), 2.55-2.67 (m, 2 H), 2.17-2.30 (m, 2 H). LC/MS (exact mass) calculated for CiaHieFaNsOïS; 363.098, found (M + H+); 363.9.
The following compounds, Examples 2-7, were prepared from cis-N-methyl-N-7H-pyrrolo[2,3-d]pyrimidin4-ylcyctobutane-1,3-diamine hydrochioride (Example 1, Step 3) in a similar manner to that described in Example 1, Step 4, substituting the indicated sulfonyl chloride for 2,2,2-trifluoroethanesulfonyl chloride.
Example 2: N-{às-3-[Methyi(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cydobutyl}-propane- 1-sulfonamide
This compound was prepared using 1-propanesulfonyl chloride. The crude compound was purified by chromatography on silica gel eluting with a mixture of dichloromethane and methanol (93:7) to afford the title compound as a tan solid (78% yield). 1H NMR (400 MHz, DMSO-de): δ 11.60 (br s, 1 H),
8.08 (s, 1 H), 7.46 (d, 1 H), 7.12 (d, 1 H), 6.61 (d, 1 H), 4.81-4.94 (m, 1 H), 3.47-3.62 (m, 1 H), 3.23 (s, 3
H), 2.87-2.96 (m. 2 H), 2.52-2.63 (m, 2 H), 2.14-2.27 (m, 2 H) 1.60-1.73 (m, 2 H) 0.96 (t, 3 H). LC/MS (exact mass) calcutated for C14H21N5O2S; 323.142, found (M + H+); 324.1.
Example 3: 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyc!obutyl}propane-110 sulfonamide
This compound was prepared using 2-methyl-1propanesulfonyl chloride, The crude compound was purified by chromatography on silica gel eluting with a mixture of dichloromethane and methanol (93:7) to afford the title compound as a white solid (52%). 1H NMR (400 MHz, DMSO-de): δ 11.64 (br s, 1 H). 8.12 (s, 1 H), 7.51 (d, 1 H). 7.03-7.26 (m, 1 H), 6.65 (d, 1 H), 4.82-5.02 (m, 1 H). 3.52-3.70 (m. 1 H),
3.26 (s, 3 H), 2.87 (d, 2 H), 2.55-2.67 (m, 2 H), 2.18-2.30 (m, 2 H), 2.11 (dt, 1 H), 1.04 (d, 6 H). LC/MS (exact mass) calculated for C15H23N5O2S; 337.157, found (M + H+); 338.0.
Example 4A and Example 4B: c/s- andÎrans-3-(Cyanomethyl)-N-{cis-3-[methyl(7H-pyrmlc>[2,3d]pyrimidin-4-yl)amino]cydobutyÎ}cydobutanesulfonamîde
These compounds were prepared using a mixture (-1:1) of cis- and trans-3(cyanomethyl)cyclobutanesulfonyl chloride. The crude mixture of cis and trans isomers was purified by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 10:1) to
afford a mixture (420 mg) of the title compounds as a white solid (67%). The ois and trans isomers were separated by supercritical fluid chromatography.
cis-isomer 4A: 160 mg (21%). ’H NMR (400 MHz, methanol-d4): δ 8.12 (s, 1H), 7.13-7.12 (d. 1H), 6.696.69 (d, 1 H), 4.92-4.89 (m, 1 H), 3.84-3.78 (m, 1 H), 3.76-3.67 (m, 1 H), 3.36 (s, 3 H), 2.79-2.73 (m, 2 H), 2.65-2.64 (m, 3H), 2.58-2.52 (m, 2 H), 2.32-2.19 (m, 4 H). LC/MS (exact mass) calculated for C17H22N6O2S; 374.152, found (M + H+); 375.3.
trans-isomer 4B: 155 mg (20%). 1H NMR (400 MHz, methanol-d4): 6 8.13 (s, 1H). 7.13 (d. 1H). 6.70 (d, 1 H), 4.94-4.89 (m, 1H), 3.89-3.85 (m, 1H), 3.72-3.69 (m, 1H), 3.36 (s, 3H), 2.85-2.62 (m, 7H), 2.31-2.23 (m, 4H). LC/MS (exact mass) calculated for C17H22N6O2S; 374.152, found (M + H*); 374.9.
The mixture of cis- and trans-3-(cyanomethyl)cyclobutanesu1fonyl chlorides was prepared as follows:
Step 1 : [3-(Benzyloxy)cyclobutylidene]acetonitrile
To a cold suspension of sodium hydride (125 mg, 3.12 mmol) in tetrahydrofuran (12 mL) at 0°C was added diethyl cyanomethylphosphonate (1.21 g, 3.40 mmol). The mixture was stirred at room température for 1 hour before adding a solution of 3-(benzyloxy)cyclobutanone (500 mg, 2.84 mmol) in tetrahydrofuran (8 mL). The mixture was stirred at room température overnight, and was then quenched with water. The mixture was extracted with ethyl acetate (3 x 25m L) and the combined organic layers were dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 85:15) to afford the title compound (450 mg, 80%) as a yellow oil.
Step 2: [3-(Benzyloxy)cyclobutyl]acetoniïrile
A mixture of [3-(benzyloxy)cyclobutylidene]acetonitrile (10.2 g, 51 mmol,) and 10% Pd/C (2.0 g) in dry tetrahydrofuran was pressurized to 50 psi with hydrogen and stirred at room température for 3 days.
The mixture was then filtered and concentrated under vacuum. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 80:20) to give the title compound (7 g, 70%) as a colorless oil. 1H NMR (400 MHz, CDCb): δ 7.36-7.28 (m, 5 H), 4.44-4.43 (m, 2 H), 4.30-4.09 (m, 1 H). 3.98-3.95 (m, 1 H), 2.64-2.45 (m, 4 H), 1.81-1.759 (m, 2 H).
Step 3: (3-Hydroxycyclobutyl)acetoniïrile
To a solution of [3-(benzyloxy)cyclobutyl]acetonitrile (1 g, 5.00 mmol) in acetonitrile (15 mL) was added dropwise iodotrimethylsilane (1.5 g, 7.50 mmol) at 0°C. The mixture was stirred at room température overnight. The mixture was quenched with triethylamine, concentrated and then purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (1:0 to 1:1) to afford the title compound (340 mg, 62%) as yellow oil. 1H NMR (400 MHz, CDCIa): 64.55-4.15 (m, 1H), 2.49-2.46 (m, 2H), 2.25-2.21 (m, 2H), 2.14-2.08 (m, 1H), 1.79-1.72 (m, 2H).
Step 4: 3-(Cyanomethyl)cyclobutyl-4-methylbenzenesulfonate
To a solution of (3-hydroxycyclobutyl)acetonitrile (333 mg, 3.0 mmol) in dry dichloromethane (25 mL) was added 4-dimethylaminopyridine (732 mg, 6.0 mmol). The mixture stirred ai room température for
5 minutes and then p-toluenesulfonyl chloride (859 mg, 4.5 mmol) was added. The resulting mixture was stirred at room température overnight. The mixture was washed with water (2 x 15mL). The organic layer was dried over sodium sulfate and concentrated. The residue was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10:0 to 7:3) to afford the title compound (520 mg, 65% yield) as colorless oil.
Step 5: S-[3-(Cyanomethyl)cycfobutyl] ethanethioate
The mixture of 3-(cyanomethyl)cyclobutyl 4-methylbenzenesulfonate (1.5 g, 5.7 mmol) and potassium thioacetate (1.29 g, 3.00 mmol) in N,N-dimethylformamide (8 mL) was heated at 80°C overnight. The mixture was diluted with ethyl acetate (15 mL), washed with water (30 mL) and brine (2 x 30 mL),
dried over sodium sulfate and concentrated. The residue was purified by préparative thin layer chromatography eluting with a mixture of petroleum ether and ethyl acetate (3:1) to afford the title compound (750 mg, 78%) as colorless oil. 1H NMR (400 MHz. CDCb): δ 4.12-3.92 (m, 1 H), 2.86-2.77 (m, 2 H), 2.712.47 (m, 2 H), 2.42-2.37 (m, 2 H), 2.30-2.29 (m,3 H), 1.97-1.90 (m, 1 H).
Step 6: 3-(Cyanomethyl)cyclobutanesulfonyl chloride
A mixture of N-chlorosuccinimide (1.6 g, 12.0 mmol) in concentrated HCl (3 mL) and acetonitrile (12 mL) was stirred at room température for 10 minutes. S-[3-(cyanomethyî)cyclobutyl] ethanethioate (507 mg, 3.0 mmol) in acetonitrile (3 mL) was added at 0°C and stirred for 10 minutes. The mixture was diluted with aqueous sodium bicarbonate (50 mL), and extracted with methyl tert-butyl ether (3 x 50 mL). The combined dried organic layers were dried over anhydrous sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a mixture of petroleum ether and ethyl acetate (100:0 to 50:50) to afford the title compound (400 mg, 69%) as a yellow oil. Ή NMR (400 MHz, CDCb): δ 4.45-4.40 (m, 1H), 3.06-2.71 (m, 3H), 2.61-2.49 (m, 4H).
Example 5: 1-[3-(Cyanomethyl)oxetan-3-yl]-N-(cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimîdîn-4yl)amino]cyclobutyl}methanesulfonamîde
This compound was prepared from [3-(cyanomethyl)oxetan-3-yl]methanesulfonyl chloride. The crude compound was purified using préparative thin layer chromatography eluting with ethyl acetate to afford the title compound as a white solid (32%). Ή NMR (400 MHz, methanol-d4): δ 8.13 (s, 1 H), 7.14-
7.13 (m, 1 H), 6.71-6.70 (m, 1 H), 5.06-5.05 (m, 1 H), 4.85-4.81 (m, 2 H), 4.52-4.50 (m, 2 H), 3.77-3.75 (m, 1 H), 3.63 (m, 2 H), 3.39 (s, 3 H). 3.29-3.26 (m, 2 H), 2.85-2.78 (m, 2 H), 2.38-2.30 (m, 2 H). LC/MS (exact mass) calculated for C17H22N6O3S; 390.147, found (M + H+); 391.0. [3-(Cyanomethyl)oxetan-3-yl]methanesulfonyl chloride
Step 1: [3-(Cyanomethyl)oxetan-3-yl]methyl 4-methylbenzenesulfonate
This compound was prepared following the procedure of Example 4 Step 4, substituting [3(Hydroxymethyl)-3-oxetanyl]acetonitnle for (3-hydroxycyclobutyl)acetonitrile. The crude compound was purified by chromatography on silica gel eluting with a mixture of petroieum ether and ethyl acetate (1:0 to
1:1) to afford the title compound as a white solid (10%). 1H NMR (400 MHz, CDCIa): S 7.82-7.80 (m, 2 H),
7.41-7.39 (m. 2 H), 4.54-4.35 (m, 4 H), 4.31 (s, 2 H), 2.79 (s, 2 H), 2.45 (s, 3H).
Step 2: [3-(Cyanomethyl)oxetan-3-yl]methyl thiocyanate
A solution of [3-(cyanomethyi)oxetan-3-yl]methyl 4-methylbenzenesulfonate (150 mg, 0.53 mmol) and potassium thiocyanate (104 mg, 1.07 mmol) was stirred in éthanol (10 mL). The reaction was heated
I0 to 85°C and stirred for 16 hours. The solvent was evaporated to afford the crude title compound as a white solid.
Step 3; [3-(Cyanomethyl)oxetan-3-yl]methanesulfonyl chloride
Chlorine gas was bubbled through a solution of [3-(cyanomethyl)oxetan-3-yl]methyl thiocyanate (0.53 mmol, crude) in water (10 mL) at 0°C for 30 minutes. The reaction mixture was extracted with me15 thyl tert-butyl ether (2 x 20 mL). The combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (20 mg, 18%).
Example 6: N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- 1-oxetan-3ylmethanesulfonamide
This compound was prepared using oxetan-3-ylmethanesulfonyl chloride. The crude compound was purified by chromatography on silica gel eluting with a mixture of dichloromethane and methanol (85:15) to afford the title compound as a white solid (23%). 1H NMR (400 MHz, methanol-di): δ 8.13 (m, 1 H), 7.13 (d, J = 4Hz, 1 H), 6.70-6.69 (m, J = 4Hz, 1 H). 4.93-4.91 (m,1 H), 4.84-4.83 (m, 2 H). 4.63-4.59
(m, 2 H), 3.74-3.68 (m, 1 H), 3.58-3.56 (m, 1 H), 3.47-3.45 (m, 2 H), 3.37 (s, 3 H), 2.79-2.77 (m, 2 H), 2.32-2.29 (m, 2 H). LC/MS (exact mass) calculated for C15H21N5O3S; 351.136, found (M + H*); 352.1.
Oxetan-3-ylmethanesulfonyl chloride
Step 1: Oxetan-3-ylmethyl thiocyanate
This compound was prepared according to the procedure of Example 5, Step 2, substituting oxetan-3-ylmethyl 4-methylbenzenesulfonate (WO2012/117000A1) for [3-(cyanomethyl)oxetan-3yljmethyl 4-methylbenzenesulfonate to afford the crude title compound as a white solid. (100%).
Step 2: Oxetan-3-ylmethanesulfonyI chloride
This compound was prepared in crude form (25% yield) following the procedure of Example 5 Step 3, substituting oxetan-3-ylmethyl thiocyanate for [3-(cyanomethyl)oxetan-3-yl]methyl thiocyanate.
Example 7A and 7B: cis- and trans-3-(Cyanomethyl)-3-methyl-N-{cis-3-[methyl(7/-l-pyrrolo[2,315 d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide
These compounds were prepared using a mixture (-1:1) of cis- and trans-3-(cyanomethyl)-3methylcyclobutanesulfonyl chloride. The crude mixture of cis- and trans isomers was purified by chromatography on silica gel eluting with a gradient of petroleum ether:ethyl acetate (10:1 to 1:15) to afford a mixture (70 mg) of the title compounds as a light brown solid (28%). The cis and trans isomers were then 20 separated by supercritical fluid chromatography (SFC).
cis-isomer (7A): 26 mg (10%); SFC rétention time = 7.11 minutes; Ή NMR (400 MHz, methanol-d4): δ
8.13 (s, 1 H), 7.13-7,13 (d, 1 H), 6.69 (d, 1 H), 4.93-4.86 (m. 1 H), 3.91-3.87 (m, 1 H), 3.71-3.65 (m, 1 H),
3.37-3.33 (m, 3 H), 2.77-2.75 (m, 2 H), 2.68 (s, 2 H), 2.41-2.36 (m, 2 H), 2.26-2.21 (m, 2 H), 1.34 (m. 3 H). LC/MS (exact mass) calculated for C18H24N6O2S; 388.168, found (M + H+); 389.1.
trans-isomer (7B) 24 mg (10%); SFC rétention time = 11.35 minutes; 1H NMR (400 MHz, methanol-do): δ
8.13 (s, 1 H), 7.14 (d, 1 H). 6.69 (d, 1 H), 4.93-4.86 (m, 1 H), 3.96-3.86 (m, 1 H), 3.72-3.65 (m, 1 H), 3.36-
3.31 (m, 3 H), 2.77-2.75 (m, 2 H), 2.71 (s. 2 H), 2.34-2.26 (m, 6 H), 1.33 (m, 3 H). LC/MS (exact mass) calculated for C18H24N6O2S; 388.168, found (M + H+); 389.0.
The mixture of cis- and trans-3-(cyanomethyl)-3-methylcyclobutanesulfonyl chlorides was prepared as follows:
Step 1: 1-Methyl-3-methyienecyciobutanecarbonitrile
To a solution of 3-methylenecyclobutanecarbonîtrile (35.0 g, 373.0 mmol) în tetrahydrofuran (200 mL) was added dropwise lithium bis(trimethylsilyl)amide (450 mL, 1M) at -78°C. The solution was stirred for 1 hour at -78C and iodomethane (30 mL, 448 mmol) was added to the reaction. After 1 hour, the mixture was warmed to room température and stirred overnight. The reaction mixture was quenched with aqueous ammonium chloride (380 mL) and extracted with methyl tert-butyl ether (3 x 400 mL). The com15 bined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by distillation under reduced pressure to afford the title compound (20 g, 50%) as ciear oil. ’H NMR (400 MHz, CDCb): δ 4.90-4.89 (m, 2 H), 3.24-3.20 (m, 2 H), 2.67-2.62 (m, 2 H), 1.50 (s, 3H).
Step 2; 1-Methyl-3-methylenecyclobutanecarboxylic acid
To a solution of 1-methyl-3-methylenecyclobutanecarbonitrile (10.0 g, 93.3 mmol) in water (50 mL) and éthanol (50 mL) was added potassium hydroxide (25.6 g, 466.6 mmol). The reaction mixture was heated to reflux and stirred overnight. The éthanol was removed under reduced pressure, and the solution was cooled to below 10‘C, acidified with concentrated hydrochloric acid to pH 1. The aqueous phase was extracted with ethyl acetate (3 x 150 mL). The combined organic layers were dried over sodi-
um sulfate and concentrated to afford the title compound (9 g, 77%). 1H NMR (400 MHz, CDCb): δ 11.90 (s, 1 H), 4.88-4.85 (m, 2 H), 3.23-3.17 (m, 2 H), 2.53-2.41 (m, 2 H), 1.45 (s, 3 H).
Step 3: Ethy! 1-methyl-3-methylenecyclobutanecarboxylate
To a solution of 1-methyl-3-methylenecyclobutanecarboxylic acid (6 g, 47.6 mmol) in dichloromethane (30 mL) at 0°C was added dropwise thionyl chloride (11.0 mL, 143 mmol). The solution was stirred at 0°C for 1 hour. Three drops of Ν,Ν-dimethylformamide were added to the solution. The solution was stirred at 0'C for 30 minutes. The solvent was evaporated and dichloromethane (20 mL) and éthanol (125 mL) were added to the residue. The resulting solution was stirred for 16 hours at room température. The solvent was evaporated and water (20 mL) was added to the residue. The aqueous layer was extracted with dichloromethane (4 x 20 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20:1 to 10:1) to afford the title compound (5 g, 68%). 1H NMR (400 MHz, CDCb): δ 4.85-4.83 (m, 2 H), 4.17-4.12 (m, 2 H), 3.18-3.12 (m, 2 H), 2.48-2.42 (m, 2H), 1.41 (s, 3 H), 1.27-1.23 (m, 3 H).
Step 4: (1-Methyl-3-methylenecyclobutyl)methanol
A mixture of ethyl 1-methyl-3-methyîenecyclobutanecarboxylate (4.55 g, 29.5 mmol) lithium aluminum hydride (2.8 g, 72 mmol) in tetrahydrofuran (50 mL) was stirred ovemight at room température. To the reaction mixture was added Na2SO4'10H2O (3.7 g, 11.5 mmol) and the resulting mixture was stirred for 1 hour at room température. The solrds were removed by filtration and the filtrate was concentrated under vacuum. The residue was extracted with dichloromethane (3 x 50 mL). The combined organic extracts were dried over sodium sulfate and concentrated to afford the title compound (2.6 g, 79%) as a colorless oil. Ή NMR (400 MHz, CDCb): δ 4.79-4.78 (m. 2 H), 3.48 (s, 2 H), 2.53-2.48 (m, 2 H), 2.36-2.27 (m, 2 H), 1.16 (s, 3 H).
Step 5: (1-Methyl-3-methylenecyclobutyl)methyl 4-methylbenzenesulfonate
This compound was prepared following Example 4, Step 4, substituting (1-methyl-3methylenecyclobutyl) methanol for (3-hydroxycyclobutyl)acetonitrile. The crude compound was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20:1 to 4:1) to afford the title compound (70%). 1H NMR (400 MHz, CDCh): δ 7.79 (d, 2 H), 7.34 (d, 2 H), 4.79-4.78 (m, 2 H), 3.90 (s, 2 H), 2.51-2.47 (m, 2 H), 2.44 (s, 3 H), 2.35-2.31 (m, 2 H). 1.15 (s, 3 H).
Step 6: (1-Methyl-3-methylenecyclobutyl)acetonitrile
A mixture of (1-methyl-3-methylenecyclobutyl)methyl 4-methylbenzenesulfonate (2.5 g, 9.4 mmol), potassium cyanide (1.3 g, 19 mmol) and Ν,Ν-dimethylformamide (8mL) was stirred ovemight at
70°C. Water (10 mL) and methyl tert-butyl ether (20 mL) were added to the mixture and the organic layer was separated. The aqueous phase was extracted with methyl tert-butyl ether (3 x 30 mL). The combined organic layers were washed with an aqueous saturated sodium bicarbonate solution (15 mL), dried over sodium sulfate, and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10:1 to 5:1) to afford the title compound (1.1 g, 97%) as light brown oil. 1H NMR (400 MHz, CDCh): 54.88-4,87 (m, 2 H), 2.62-2.54 (m, 2 H), 2.50 (s, 2 H), 1.33 (s, 3 H).
Step 7: (1-Methyl-3-oxocyciobutyl)acetonitrile
Ozone gas was bubbled through a solution of (1-methyl-3-methyîenecyc!o-butyi)acetonitrile (1.08 g, 8.91 mmol) in dichloromethane (30 mL) -78'C for 10 minutes. After purging the solution with nitrogen gas, dimethylsulfide (10 mL) was added dropwise to the solution at -78°C. The solution was stirred for 30 minutes at -78C and the solvent was removed under reduced pressure. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20:1 to 8:1)
to afford the title compound (920 mg, 84%) as a colorless oil. ’H NMR (400 MHz, CDCb): δ 3.11-3.06 (m, 2 H), 2.96-2.91 (m, 2 H), 2.69 (s, 2 H), 1.53 (s, 3 H).
Step 8: (3-Hydroxy-1-methylcyclobutyl)acetonitrile
To a solution of (1-methyl-3-oxocyclobutyl)acetonitrile (400 mg, 3.25 mmol) in tetrahydrofuran (15 mL) was added sodium borohydride (246 mg, 6.5 mmol). The mixture was stirred for 3 hours at room température. Acetone (2 mL) was added and then the solvent was evaporated. Water (10 mL) was added to the residue and the aqueous phase was extracted with dichloromethane (4x15 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10:1 to 1:1 ) to afford the title compound (300 mg, 74%) as a colorless oil. ’H NMR (400 MHz, CDCb): 64.38-4.34 (m, 2 H), 2.46-2.27 (m, 4 H), 1.94-1.86 (m, 2 H), 1.33-1.12 (m, 3 H).
Step 9: 3-(Cyanomethyl)-3-methylcyclobutyl 4-methylbenzenesulfonate
This compound was prepared following Example 7, Step 5, substituting (3-hydroxy-1methylcyclobutyl)acetonitrile for (1-methyl-3-methylenecyclo-butyl)methanol. The crude compound was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20:1 to 4:1) to afford the title compound (36%). ’H NMR (400 MHz. CDCb): δ 7.77 (d, 2 H), 7.35 (d, 2 H), 4.894.81 (m, 1 H), 2.45 (s, 3 H), 2.43-2.34 (m, 3 H), 2.26-2.21 (m, 1 H), 2.15-2.11 (m, 2 H), 1.33 (s, 3 H). Step 10: S-[3-(Cyanomethyl)-3-methylcyclobutyl]ethanethioate
This compound was prepared in 89% yield (crude) following the procedure of Example 4, Step 5, substituting 3-(cyanomethyl)-3-methylcyclobutyl 4-methylbenzenesulfonate for 3-(cyanomethy1)cyclobutyl 4-methylbenzene-sulfonate. ’H NMR (400 MHz, CDCb): δ 3.12 (s, 1 H), 2.46-2.30 (m, 4 H), 2.19 (s, 2 H), 1.29 (s, 1 H) 1.26-1.24 (m, 1 H), 1.18-1.14 (m, 1 H), 1.13 (s, 3H).
Step 11: 3-(Cyanomethyl)-3-methylcyclobutanesulfonyl chloride
6l
This compound was prepared following Example 4 Step 6, substituting S-[3-(cyanomethyl)-3methylcyclobutyl] ethanethioate for S-[3-(cyanomethyl)-cyclobutyl] ethanethioate. The crude compound was purified using chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (90:10 to 30:70) to afford the title compound as a yellow liquid (66%). 1H NMR (400 MHz, CDCh): δ
4.45-4,38 (m, 1 H), 2.67-2.55 (m, 4 H), 2.46-2.40 (m, 2 H), 1.42-1.40 (m, 3 H).
Example 8: 4-Cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyl}pyridine-2sulfonamide
Step 1:2-(Benzylthio)isonicotinonitrile
A 60% suspension of sodium hydride in minerai oil (8.36 g, 210.0 mmol) was suspended in tetrahydrofuran (100 mL). A solution of benzyl mercaptan (21.5 g, 173 mmol) in tetrahydrofuran (50 mL) was then added dropwise. A thick slurry formed during the addition. 4-Cyano-2-chloropyridine (12.5 g, 90.2 mmol) was added and the resulting mixture was stirred for 3 hours at room température. After carefully quenching with water, the mixture was partitioned between water and diethyl ether. The ether layer was washed with saturated aqueous sodium bicarbonate solution, dried over magnésium sulfate and concentrated under reduced pressure. Heptane was added to the residue with solids forming rapidly. The solids were collected by filtration, washed with heptane, and dried to give (33.02 g, 84%) of the title compound as an off-white solid. 1H NMR (400 MHz, CDCta): δ 8.61 (d, 1 H), 7.25-7.46 (m, 6 H), 7.16-7.22 (m, 1 H), 4.47 (s, 2 H). LC/MS (exact mass) calculated for C13H10N2S; 226.056, found (M + H+); 227.1.
Step 2: 4-cyanopyridine-2-sulfonyl chloride
To a mechanically-stirred mixture of 2-(benzyithio)isonicotinonitrile (8.92 g, 39.4 mmol) in dichloromethane (139 mL) and water (31 mL) was added dropwise sulfuryl chloride (22.5 mL, 278 mmol), keeping the température of the mixture below 3°C. After addition was complété, the mixture was stirred
for 30 minutes with continued cooling in an ice bath. A slurry of water (50 mL) and ice (20 g) was added. The aqueous phase was extracted twice with dichloromethane. The combined extracts were dried over magnésium sulfate and concentrated under reduced pressure to afford the crude title compound. Step 3: 4-Cyano-N-{cis-3-[methyl(7H-pyrro!o{2,3-d]pyrimidin-4-yl)amino]cydobutyl}pyr-idine-25 suiïonamide
A solution of 4-cyanopyridine-2-sulfonyl chloride (9.7 g, 47.9 mmol) in N,N-dimethylformamide (10 mL) was added to a solution of cis-N-methyl-N-7H-pyrrolo[2,3-d]pyrimidin-4-ylcyclobutane-1,3-diamine hydrochloride (8.0 g, 36.8 mmol) and 4-dimethylaminopyridine (150 mg, 0.03 mmol) in N,Ndimethylformamide (90 mL) at room température. Diisopropylethylamine (13 mL, 77 mmol) was added and the resulting mixture was stirred at room température for 2 hours. The mixture was diluted with ethyl acetate (200 mL) and aqueous saturated sodium bicarbonate solution was added. Water was added to dissolve the precipitated solids. The aqueous phase was extracted three times with ethyl acetate. The combined organic extracts were washed four times with brine, dried over anhydrous magnésium sulfate and concentrated under reduced pressure. A 1:1 mixture of ethyl acetate and hexanes was added to the residue. The solids were coliected by filtration and then dissolved in dichloromethane and a minimum amount of methanol. The resulting solution was passed through a silica gel plug eluting with a 5% solution of methanol in dichloromethane. The solvents were evaporated to afford a solid to which was added a solution of 10% methanol in dichloromethane. The mixture was briefly stirred and then let stand overnight. The solids were filtered, washed with dichloromethane and dried to afford the title compound (5.58 g, 39%) as an off-white solid. 1H NMR (400 MHz, DMSO-de): δ 11.62 (br. s., 1 H), 9.02 (d, 1 H), 8.52 (d, 1 H), 8.38 (s, 1 H), 8.17 (dd, 1 H), 8.07 (s, 1 H), 7.10-7.15 (m, 1 H), 6.59 (dd, 3.41 Hz. 1 H), 4.80-4.91 (m, 1 H), 3.58-3.71 (m, 1 H), 3.19 (s, 3 H), 2.25-2.36 (m, 2 H), 2.10 (m, 2 H). LC/MS (exact mass) calculated for C17H17N7O2S: 383.116, found (M + H+); 384.1.
Example 9: 3-(1-Hydroxy- 1-methy!ethyl)-N-{cis-3-[methy!(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyc!obutyl}benzenesulfonamide
Step 1 : Methyl 3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyl}amino)5 sutfonytjbenzoate
To a suspension of cis-N-methy!-N-7H-pynOlo[2,3-d]pyrimidin-4-ylcyclobutane-1,3-diamine hydrochloride (1.8 g, 8.29 mmol) in N.N-dimethylformarnide (100 mL) was added portionwise triethylamine (6.7 mL, 49 mmol) at 0°C. Methyl 3-{chlorosulfonyÎ)benzoate (2.3 g, 9.9 mmol) was added at 0°C. The resulting mixture was stirred at room température for 3 hours. The solvent was removed under vacuum.
The residue was chromatographed on silica gel eluting with a gradient of methanol in dichloromethane (3% to 10%) to afford the title compound (1.6 g, 47%) as a yellow solid.
Step 2; 3-(Hydroxymethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc!obutyl}benzenesutfonamide
To a solution of methyl 3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-415 yl)amino]cyclobutyl}amino)sulfonyl]benzoate (800 mg, 1.92 mmol) in tetrahydrofuran (120 mL) was added lithium aluminum hydride (0.25 g, 6.7 mmol) at 0°C. The reaction was warmed to 25°C and stirred for 3 hours. The reaction was quenched with water (2 mL) and stirred for 15 minutes. The reaction mixture was filtered. The filter cake was stirred in tetrahydrofuran (50 mL) and filtered again. The combined filtrate was concentrated to dryness to afford the title compound (430 mg, 58%) as a yellow solid.
Step 3: 3-Formyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobuty!}benzenesulfonamide To a solution of 3-(hydroxymethyl)-N-(cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}benzenesulfonamide (400 mg, 1.03 mmol) in chloroform (50 mL) and methanol (5 mL) was added manganèse dioxide (0.89 g, 10.0 mmol). The reaction mixture was stirred at 25°C overnight.
The réaction mixture was filtered and the filter cake was washed with chiorofomn (3 x 25 mL). The cornbined filtrâtes were concentrated. The residue was chromatographed on silica gel eluting with a gradient of methanol tn dichloromethane (2% to 8%) to afford the title compound (240 mg, 60%) as an oil.
Step 4: 3-(1-Hydroxyethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d}pyr!midin-4yl)amino]cyclobutyl}benzenesulfonamide
To a solution of 3-formyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}benzenesulfonamide (260 mg, 0.68 mmol) in tetrahydrofuran (20 mL) was added méthylmagnésium bromide (1.8 mL, 5.4 mmol) at 0°C under nitrogen. The reaction was stirred at 25°C ovemight and was then quenched with aqueous ammonium chloride (10 mL). The reaction mixture was extracted with ethyl acetate (3 x25 mL), The combined organic layers were dried over sodium sulfate and concentrated. The residue was purified by préparative high performance liquid chromatography to afford the title compound (60 mg. 22%) as a white solid. 1H NMR (400 MHz, DMSO-de): δ 11.6 (s, 1 H), 8.08 (s, 1 H), 8.03 (d, 1 H), 7.86 (s, 1 H), 7,70 (m, 1 H), 7.55 (m, 2 H), 7.15 (m, 1 H), 6.61 (m, 1 H), 5.44 (m, 1 H), 4.85 (m. 1 H), 3.56 (m. 1 H), 3.18 (s. 3 H), 2.18 (m, 2 H). 2.04 (m. 2 H), 1.32 (d, 3 H). LC/MS (exact mass) calculated for C19H23N5O3S; 401.152, found (M + H+); 402.2.
Step 5: 3-Acety!-N-{cis-3-[methyÎ(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyl}benzenesulfonamide To a solution of 3-(1-hydroxyethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl)benzenesulfonamide (60 mg, 0.15 mmol) in chloroform (30 mL) and methanol (5 mL) was added manganèse dioxide (190 mg, 2.2 mmol). The reaction mixture was stirred at 45°C ovemight. Then the reaction mixture was filtered and the filter cake was washed with chloroform (3 x 25 mL), The combined filtrâtes were concentrated. The residue was purified by préparative high performance liquid chromatography to afford the title compound (15 mg, 25%) as a white solid. 1H NMR (400 MHz, DMSOde): 8 11.58 (s, 1 H), 8.31 (s, 1 H), 8.21 (m, 2 H), 8.16 (m, 2 H), 7.76 (m, 1 H), 7.09 (d, 1 H), 6.56 (s. 1 H),
4.82 (m, 1 H), 3.54 (m, 1 H), 3.14 (s, 3 H), 2.81 (m, 3 H), 2.26 (m, 2 H), 1.98 (m, 2 H). LC/MS (exact mass) calculated for C19H21N5O3S; 399.136, found (M + H+); 400.1.
Step 6: 3-(1-Hydroxy- 1-methylethyl)-N-{cis-3-[methyi(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyi}benzenesulfonamide
To a solution of 3-acetyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyciobutyl}benzenesulfonamide (240 mg, 0.58 mmol) in tetrahydrofuran (20 mL) was added méthylmagnésium bromide (2.4 mL, 7.2 mmol) at 0°C under nitrogen. The reaction was stirred at 25°C for 2 hours and was quenched with aqueous ammonium chloride solution (10 mL). The reaction mixture was extracted with ethyl acetate (3 x 25 mL). The combined organic layers were dried over sodium sulfate and concentrated. The residue was purified by préparative high performance liquid chromatography to afford the title compound (101 mg, 42%) as a white solid. 1H NMR (400 MHz, DMSO-de): δ 11.6 (s, 1 H), 8.05 (s, 1 H), 7.97 (m, 2 H), 7.67 (m, 2 H). 7.52 (m, 1 H), 7.12 (m, 1 H), 6.57 (m, 1 H), 5.29 (s, 1 H), 4.85 (m, 1 H), 3.53 (m. 1 H), 3.15 (s, 3 H), 2.24 (m, 2 H), 1.98 (m, 2 H), 1.44 (s, 6 H). LC/MS (exact mass) calculated for CaoHïsNsOaS; 415.168, found (M + H+); 416.0.
Example 10: 1-Cyclopropyl-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidîn-4yl)amîno]cyclobutyl}methanesulfonamide
This compound was synthesized starting from benzyl (trans-3-(methylamino)cyclobutyljcarbamate (Example 1, Step 1), following procedures similar to those described for Example 1, Steps 2 and 3 to obtain ΐΓθη5-Ν-ιιπ6ΐΐΊνΙ-Ν-7Η-ργΓΓθΙο[2,3<1]ρνΓΪΓηίάίη-4-νΙογο^υΐ3ηβ-1,3-άί3Γηίηβ hydrochloride. To the resulting hydrochloride (60 mg, 0.28 mmol) in THF (10 mL) was added potassium carbonate (76 mg, 0,55 mmol), H2O (5 mL) and cycfopropylmethanesulfonyl chloride (52 mg, 0.33 mmol). The mixture stined for two hours, was diluted with dichloromethane, washed with brine, dried over sodium
sulfate, filtered, and concentrated. The crude product was purified by préparative high performance liquid chromatography to afford the title compound as a white solid (7 mg; 8%). 1H NMR (400 MHz, methanold+): δ 8.14 (s, 1 H), 7.16 (d, 1 H,), 6.72 (d, 1 H), 5.44-5.40 (m, 1 H), 4.07-4.06 (m, 1 H), 3.41 (s. 3 H), 3.01-2.99 (m, 2 H), 2.81-2.74 (m, 2 H), 2.54-2.49 (m, 2 H), 1.15-1.13 (m, 1 H), 0.720-0.69 (m, 2 H), 0.425 0.41 (m, 2 H). LC/MS (exact mass) calculated for C15H21N5O2S; 335.142, found (M + H+); 336.1.
Example 11: N-{(1S,3R)-3-[Methyl(7H-pyrrolo[2,3-d]pyrimîdin-4-yl)amino]cyclopentyl}propane-1sulfonamide
This compound was prepared following Example 10 substituting (1 S,3/î)-N-benzyl-N'10 methylcyclopentane-1,3-diamine for benzyl [trans-3-(methylamino)-cyclobutyl]carbamate, (1R,3S)-Nmethyl-N-7H-pynolo[2,3-d]pyrimidin-4-ylcyclopentane-1,3-diamine hydrochioride for trans-N-methyl-N-7Hpyrrolo[2,3-d]pyrimîdin-4-ylcyclobutane-1,3-diamine hydrochioride, and propane-1-sulfonyl chloride for cyclopropylmethanesuifonyi chloride to afford the title compound as an off-white solid (11%). The crude compound was purified using préparative high performance liquid chromatography. 1H NMR (400 MHz, methanol-d4): δ 8.08 (s, 1 H), 7.08 (s, 1 H,), 6.65 (s, 1 H), 5.27-5.23 (m, 1 H), 3.81-3.76 (m, 1 H), 3.31 (s,
H), 2.33-2.29 (m, 1 H), 2.13-2.04 (m, 1 H), 1.98-1.92 (m. 2 H), 1.82-1.75 (m. 4 H). 1.06 (t. 3 H), 0.420.41(m, 2 H). LC/MS (exact mass) calculated for C15H23N5O2S; 337.157, found (M + H+); 337.8. (1S,3R)-N-benzyl-N'-methylcyclopentane-1,3-diamine was prepared as follows:
Step 1: Senzy! [(1R,3S)-3-aminocydopenty!]carbamate
Trifluoroacetic acid (15 mL, 190 mmol) was added to a solution of benzyl tert-butyl (1 /?,3S)-cyclopentane1,3-diylbiscarbamate (prepared as described in WO2011/086053A1) (5.02 g, 15.0 mmol) in dichloromethane (75 mL) at room température. The reaction was stirred for 2 hours and was then concentrated to afford the title compound as a light brown oil (6.70 g, crude)
Step 2: Benzyl [(1R,3S)-3-(benzylamino)cyclopentyl]carbamate
Sodium triacetoxyhydroborate (4.38 g, 20.0 mmol) was added to a solution of benzyl [(1 /7,35)-3arninocyclopentyl]carbamate (5.23 g, 15.0 mmoî) and benzaldehyde (1.7 mL, 16.0 mmol) in dichloromethane (75 mL) at room température. The mixture was stirred for 21 hours and then aqueous 1 M sodi5 um hydroxide solution (75 mL) was added to make the solution basic. The aqueous layer was extracted with dichloromethane (2 x 25 mL). The combined organic layers were washed with brine (50 mL), dried over sodium sulfate and concentrated. The crude material was chromatographed on silica gel eluting with a mixture of dichloromethane and methanol (100:0 to 88:12) to afford the title compound as a yellow oil (3.47 g, 71%). 1H NMR (400 MHz, CDCb): δ 7.35-7.31 (m, 5 H), 7.30-7.26 (m, 5 H), 5.07 (s, 2 H), 4.1710 4.07 (m. 1 H) 3.76-3.68 (m. 2 H), 3.27-3.20 (m. 1 H), 2.02-1.51 (m, 6 H).
Step 3: (1S,3R)-N-Benzyl-N’-methylcyclopentane-1,3-diamine
Lithium aluminum hydride (1.02 g, 26.9 mmol) was added in portions to a solution of benzyl [(1R,3S)-3-(benzylamino)cyclopentyl]carbamate (3.47 g, 10.7 mmol) in tetrahydrofuran (70 mL) at room température. The reaction was heated to reflux for 3.5 hours. The mixture was then cooled in an ice bath and sequentially quenched with water (1.0 mL), aqueous 15% sodium hydroxide solution (1.0 mL) and water (3.0 mL). The suspension was diluted with ethyl acetate and filtered through Celite. The filtrate was concentrated and the residue was taken up in aqueous 0.5 M hydrochloric acid solution. The mixture was washed with diethyl ether (2 x 20 mL) and the aqueous solution was made basic (pH-11 ) with sodium hydroxide. The resulting mixture was extracted with dichloromethane (3 x 25 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude material was chromatographed on silica gel eluting with a mixture of dichloromethane and methanol (90:10) to afford the title compound as a yellow oil (204 mg. 9%). 1H NMR (400 MHz, CDCb): δ 7.33-7.20 (m, 5 H), 3.74 (s, 2 H), 3.19-3.13 (m. 1
H), 3.08-3.02 (m, 1 H) 2.39 (s, 3 H), 2.09-2.03 (m, 1 H),1.87-1.81 (m, 2 H), 1.67-1.54 (m, 2 H), 1.46-1.39 (m, 1 H). LC/MS (exact mass) calculated for CtaHîoNî; 204.163, found (M + H+); 205.1.
Example 12: 1-(3,3-Difluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-45 yl)amino]cyclobutyl)methanesuifonamide
Step 1 : Benzyl[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-dJpyrimidin-4yl}amino)cyclobutyl]carbamate
4-Chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-dIpyrimidine (15 g, 48.7 mmol) and benzyl [cis-3-(methylamino)cyclobutyl]carbamate (17.2 g, 63.5 mmol) were mixed with isopropyl alcohol (180 mL) and diisopropylethylamine (28 mL, 161 mmol). The resulting slurry was heated at 75°C for 6 hours.
The reaction was cooled to room température, filtered, washed with isopropyl alcohol (150 mL) and dried in an oven at 50°C to give the title compound (23.5 g, 95%) as a white solid. 1H NMR (400 MHz, DMSOde): δ 8.38 (s, 1 H), 8.03 (d, 2 H), 7.45 (d, 1 H). 7.38-7.28 (m. 4 H), 7.26 (s. 1 H), 7.25 (d, 1 H), 6.61 (d, 1
H), 5.08 (s, 2 H), 4.96 (d, 1 H), 4.77 (m, 1 H), 3.88 (m, 1 H), 3.23 (s, 3 H), 2.71 (m, 2 H), 2.36 (s, 3 H),
2.18 (m, 2 H).
Step 2: cis-N-Methyl-N-{7-[(4-methylphenyl)su!fonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}cyc!obutane-1,3diamine dihydrobromide
Benzyl [cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pynrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyf]carbamate (15.2 g, 30.1 mmol) was suspended in ethyl acetate (45 mL) and acetic acid (45 mL). To the slurry was slowly added a 4M solution of HBr in acetic acid (45 mL, 180 mmol), maintaining the température below 25°C. The resulting slurry was stirred at room température for 2 hours. The solids were collected by filtration, washed with ethyl acetate (450 mL), and dried at40°C to afford the title compound (16 g; 100%) as a white solid. ’H NMR (400 MHz, DMSO-de): δ 8.31 (s, 1 H). 8.20 (s, 2 H),
7.97 (d, 2 H), 7.72 (d, 1 H), 7.44 (d, 2 H), 7.08 (d, 1 H), 4.93 (m, 1 H), 3.54 (m, 1 H), 3.30 (s, 3 H), 2.50 (m, 4 H). 2,35 (s. 3 H). LC/MS (exact mass) calculated for C18H21N5O2S; 371.142, found (M + H*); 372.1. Step 3: ({[(3,3-Difluorocyclobutyi)methyl]thio}rnethyl)benzene
A mixture of (3,3-difluorocyclobutyl)methyl 4-methylbenzenesulfonate (see W02010/032200A1) (4 g, 14,5 mmol), benzyl imidothiocarbamate (3.53g, 17.4 mmol), sodium hydroxide solution (1.45 g, 36.2 mmol, dissolved in 16 mL water) and Ν,Ν-dimethylformamide (16 mL) was stirred at 60°C for 16 hours. Water (40 mL) and ethyl acetate (150 mL) were added. The organic layer was washed with water (40 mL), separated, dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 95:5) to afford the title com10 pound as colorless oil (3.2 g, 81%). NMR (400 MHz, CDCb): 5 7.34-7.24 (m, 5 H), 5.71 (s, 2 H), 2.712.61 (m, 2 H), 2.57-2.55 (m, 2 H), 2.30-2.14 (m, 3 H).
Step 4: (3,3-Difluorocyclobutyl)methanesulfonyl chtoride
This compound was prepared following the procedure of Example 8 Step 2, substituting S-[3(cyanomethyl)-3-methylcyclobutyl] ethanethioate for 2-(benzylthio)isonicotinonitri!e to afford the title com15 pound as a colorless oil (93%). 1H NMR (400 MHz, CDCb,}: δ 3.88-3.86 (m, 2 H), 3.03-2.94 (m, 3 H), 2.61-2.49 (m,2H).
Step 5: 1-(3,3-Difluorocyclobutyl)-N-[cis-3-(methyl{7-[(4-rnethylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin4-yl}amino)cyclobutyl]methanesulfonamide
A solution of (3,3-difluorocyclobutyf)methanesulfonyl chloride (2.5 g, 12,19 mmol) in 10 mL di20 chloromethane was added dropwise to a solution of cis-N-methyl-N-{7-[(4-methylphenyl)sulfony)]-7Hpyrrolo[2,3-d]pyrimidin-4-yl}cyclobutane-1,3-diamine dihydrobromide (3.25 g, 6.10 mmol) and triethylamine (3.08 g, 30.49 mmol) in dichloromethane (150 mL) at 0°C over 15 minutes. The reaction was stirred at room température for 4 hours. Water (50 mL) was added and the organic layer was separated. The
aqueous layer was extracted with dîchloromethane (2 x 150 mL), and the combined organic layers were separated dried over sodium sulfate. The crude compound was purified by chromatography on silica gel eluting with a gradient of dîchloromethane and methanol (100:0 to 90:10) to afford the title compound as a white solid (2.0 g, 61%). LC/MS (exact mass) calculated for C23H27F2N5O4S2; 539.147, found (M + H+);
540.1.
Step 6: 1-(3,3-Difîuorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimîdin-4yi)amino]cyclobutyl}methanesulfonamide
A solution of 1-(3,3-difluorocyclobutyl)-N-[cis-3-(methyl{7-[(4-methylphenyl)-sulfonyl]-7Hpyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]methanesulfonamide (2 g, 3.71 mmol) and lithium hydroxide 10 monohydrate (780 mg, 18.6 mmol) in éthanol (40 mL) and water (20 mL) was stirred at 60°C for 4 hours.
The éthanol was evaporated and the remaining aqueous layer was neutralized to pH 7 with hydrochloric acid and subsequently extracted with dîchloromethane (2 x 200 mL). The combined organic layers was dried over sodium sulfate, filtered, concentrated, and purified by préparative high performance liquid chromatography to afford the title compound (800 mg, 56%) as a white solid. 1H NMR (400 MHz, metha15 nol-d4): 68.15 (s, 1H), 8.13 (s, 1 H), 7.16-7.15 (m, 1 H), 6.73-6.62 (m, 1 H), 4.95-4.88 (m, 1 H), 3.73-3.71 (m, 1 H), 3.38 (s, 3 H), 3.28-3.26 (m, 2 H), 2.87-2.78 (m, 4 H), 2.63-2.61 (m, 1 H), 2.56-2.48 (m, 2 H), 2.35-2.28 (m, 2 H). LC/MS (exact mass) calculated for C16H21F2N5O2S; 385.138, found (M + H+); 386.1.
The following compounds, Examples 13-14, were prepared from cis-N-methyl-N-{7-[(420 methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}cyclobutane-1,3-diamine dihydrobromide (Example
12, Step 2) in a similar manner to that described in Example 12, Step 5, substituting the indicated sulfonyl chloride for (3,3-difluorocyc!obutyl)methanesulfonyl chloride and using the deprotection method îllustrated in Example 12, Step 6.
Example 13: 3,3-Difluoro-N-{cÎs-3-[methyl(7H-pyrmlo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}cyclobutanesulfonamide
This compound was prepared using 3,3-difluorocyclobutanesulfonyl chloride using the procedure in PCT Publication No. WO2011/068881.The crude compound was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (80:20 to 10:90) to afford the title compound as an off-white solid (22% over 2 steps). 1H NMR (400 MHz, methanol-d4): δ 8.13 (s, 1 H), 7,13 (d,
H), 6.70 (d, 1 H), 4.86-4.81 (m, 1H), 3.78-3.72 (m. 2 H), 3.35 (s, 3 H), 3.01-2.93 (m, 4 H), 2.78-2.76 (m,
H), 2.32-2.25 (m. 2 H). LC/MS (exact mass) calculated for CisHwFîNsOzS; 371.123, found (M + H*);
372.1.
Example 14: 1-Cyclopropyl-N-{cis-3-[meihyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}methanesulfonamide
This compound was prepared as a white solid using cyclopropylmethanesulfonyl chloride (73% over 2 steps). 1H NMR (400 MHz, DMSO-de): δ 11.64 (br. s„ 1 H), 8.11 (s, 1 H), 7.53 (d, 1 H), 7.12-7.19 (m. 1 H), 6.64 (m, 1 H), 4.84-4.97 (m, 1 H). 3.54-3.70 (m, 1 H), 3.26 (s, 3 H), 2.93 (d, 2 H), 2.55-2.66 (m, 2 H), 2.29-2.22 (m, 2 H), 0.96-1.09 (m, 1 H), 0.53-0.64 (m, 2 H). 0.29-0.39 (m, 2 H). LC/MS (exact mass) calculated for C15H19F2N5O2S; 335.142, found (M + H+); 336.0.
Example 15: 1-Cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2.3-d]pyrimidin-4-yl)amino]cyclobutyl}azetidine-3sulfonamide
Step 1 : tert-Buty! 3-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]amino}sulfonyl)azetidine-1-carboxylate
cis-N-Methyl-N-{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}cyclobutane-1,3diamin© dihydrobromide (7.0 g, 18.8 mmoi) was free-based by stirring in excess aqueous 1N sodium hydroxide solution for 3 minutes and then extracting into dichloromethane. The organic layer was dried over sodium sulfate and concentrated. The remaining free base was taken up in dichloromethane (200 mL), cooled to 0°C and treated with and triethylamine (13 mL, 94 mmol) and tert-butyl 3(chlorosulfonyl)azetidine-1-carboxylate, The reaction was allowed to stir at room température for 10 minutes. The crude mixture was washed with water and brine, then dried over sodium sulfate and concentrated to afford the crude product as a white solid. The solid was crystallized using a mixture of dichloromethane and diethyl ether to afford the title compound as a white solid (9.61 g, 90%). 1H NMR (400
MHz, methanol-di): S 8.19 (s, 1 H), 7.92-8.01 (m, 2 H), 7.54 (d, 1 H), 7.35 (d. 2 H). 6.86 (d, 1 H), 4.764.65 (m, 1 H), 4.18 (br. s., 2 H), 3.99-4.10 (m, 3 H), 3.66-3.78 (m, 1 H), 3.25 (s, 3 H), 2.64-2.78 (m, 2 H), 2.37 (s, 3 H), 2.10-2.25 (m, 2 H), 1.41 (s, 9 H). LC/MS (exact mass) calculated for CzbHmNgOgSî; 590.198, found (M + H+); 591.45.
Step 2: N-[cis-3-(Methyl{7-[(4-methylpheny!)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-415 yl}amino)cyclobutyl]azetidine-3-sulfonamide
Acetyl chloride (0.20 mL, 2.8 mmol) was added to a solution of tert-butyl 3-({[cis-3-(methyl{7-[(4methylphenyl)sulfony!]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]amino}sulfonyl)azetidine-1carboxylate (1.64 g, 2.78 mmol) in anhydrous dichloromethane (18 mL) and methanol (7 mL) at 0°C. The reaction mixture was stirred at room température for 16 hours. The white precipitate was filtered off and taken up in saturated aqueous sodium bicarbonate solution (20 mL). The resulting solution was extracted with dichloromethane (3 x 20 mL) and the combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (810 mg, 60%) as a white solid. LC/MS (exact mass) calculated forCïiHîeNeOiSz; 490.146, found (M + H*); 491.0.
Step 3: 1-Cyclopropyl-N-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]azetidine-3-sulfonamide
N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl)amino)cyclobutyl]azetidine-3-sulfonamide (810 mg, 1.65 mmol), methanol (10 mL), molecular sieves, and [(1-ethoxycyclopropyl)oxy](trimethyl)silane (0.53 mL, 2.64 mmol) were combined in a sealable reaction vessel. The vessel was purged with nitrogen and acetic acid (1.28 mL, 8.26 mmol) was added. The vessel was sealed and then heated at 80°C for 2 hours. After the mixture was cooled to room température, sodium cyanoborohydride (273 mg, 4.13 mmol) was added. The vessel was resealed and heated slowly to 40°C for 1.5 hours. The crude mixture was filtered over a bed of Celîte, rinsing with methanol.
The filtrate was concentrated and the residue was taken up an aqueous saturated sodium bicarbonate solution. The resulting solution was extracted with dichloromethane (5 x 20 mL), and the combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (576 mg, 74%) as a white solid. 1H NMR (400 MHz, methanol-d4): δ 8.23 (s, 1 H), 7.95-8.05 (m, 2 H), 7.58 (d, 1 H), 7.39 (d, 2 H), 6.90 (d, 1 H), 4.69-4.83 (m, 1 H), 3.94-4.09 (m, 1 H), 3.65-3.75 (m, 3 H), 3.54-3.64 (m, 2 H), 3.29 (s, 3 H), 2.67-2.79 (m, 2 H), 2.41 (s, 3 H). 2.15-2.29 (m, 2 H), 2.02-2.15 (m. 1 H), 0.43-0.51 (m, 2 H),
0.29-0.39 (m, 2 H). LC/MS (exact mass) calculated for C24H30N6O4S2; 530.177, found (M + H*); 531.0. Step 4: 1-Cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}azetidine-3sutfonamide
A solution of césium carbonate (976 mg, 3.0 mmol) in water (5 mL) was added to a solution of 120 cyclopropyl-N-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]azetidine-3-suîfonamide (530 mg, 1,0 mmol) in éthanol (10 mL). The reaction mixture was heated to reflux for 16 hours. After the solvent was removed, remaining material was taken up in water and extracted with a mixture of dichloromethane and methanol (96:4; 3x10 mL). The combined or-
ganic layers were dried over sodium sulfate and concentrated, The crude solid was crystallized from methanol to afford the title compound (225 mg, 59%) as a white solid, 1H NMR (400 MHz, methanol-d4): δ
8.10 (s, 1 H), 7.09 (d, 1 H), 6.66 (d, 1 H), 4.88-4.80 (m, 1 H), 4.03-3.96 (m, 1 H), 3.73-3.65 (m. 3 H), 3.613.57 (m, 2 H), 3.32 (s. 3 H), 2.77-2.68 (m, 2 H), 2.28-2.19 (m. 2 H), 2.08-2.03 (m, 1 H), 0.46-0.41 (m, 2
H), 0.34-0.31 (m, 2 H). LC/MS (exact mass) calculated for C17H24N6O2S; 376.168, found (M + H*); 377.0.
Example 16: N-(Cyclopropylmethyl)-N'-{cis-3-[methyÎ(7H-pyrrolo[2,3-d]pyn'midin-4yl)amino]cyclobutyl}sulfamide
Step 1 : N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin^l-yl}amino)cyclobutyl]-210 oxo-1,3-oxazolidine-3-sulfonamide
To a solution of chforosulfonyl isocyanate (1.76 mL, 20.6 mmol) in dichloromethane (150 mL) was added dropwise a solution of 2-bromoethanol (1.43 mL, 20.6 mmol) in dichloromethane (80 mL) at 0‘C. After 30 minutes at 0'C, a solution of cis-N-methyl-N-{7-[(4-methylphenyî)sulfonyl]-7H-pyrrolo[2,3d]pyrimidin-4-yl}cyclobutane-1,3-diamine dihydrobromide (11.0 g, 20.6 mmol) and triethylamine (10.42 g,
103.2 mmol) in dry dichloromethane (80 mL) was added dropwise, and the reaction mixture was allowed to warm to room température ovemight. The reaction solution was dissolved in dichloromethane (1 L), washed with aqueous 1M hydrochloric acid solution (2 x 800 mL) and brine (500 mL). The solution was dried over sodium sulfate and concentrated to afford the title compound as white solid (8.5 g, 79%). 1H NMR (400 MHz, CD3OD): δ 8.22 (s, 1 H), 8.00 (d, 2 H), 7.58 (d, 1 H), 7.38 (d, 2 H), 6.91 (d, 1 H), 4.88 (m,
1 H). 4.45-4.41 (m, 2 H), 4.06-4.02 (m, 2 H), 3.75 (m, 1 H), 3.29 (s, 3 H), 2.72-2.69 (m, 2 H), 2.40 (s, 3 H);
2.30-3.27 (m, 2 H). LC/MS (exact mass) calculated for C21H24N6O6S2; 520.120, found (M + H*); 521.4. Step 2: N-(Cyclopropy/methyl)-N'-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4y/}amino)cyclobutyl]sulfamide
A solution of N-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]-2-oxo-1,3-oxazoÎidine-3-sulfonamide (150 mg, 0.29 mmol), cyclopropanemethylamine (51 mg, 0.72 mmol) and triethylamine (116 mg, 1.15 mmol) in acetonitrile (3 mL) was stirred for 15 minutes at 100°C using microwave heating. The reaction mixture was concentrated to afford the crude title compound (146 mg, 100% crude yield) as a yellow oil. LC/MS (exact mass) calculated for C22H28N6O4S2; 504.161, found (M + H+); 505.2.
Step 3: N-(Cyclopropylmethy!)-N'fcis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4yl)amino]cyclobutyl}sulfamide
A solution of N-(cyclopropylmethyi)-N'-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,310 d]pyrimidin-4-yl)amrno)cyclobutyl]sulfamide (146 mg, 0.29 mmol), lithium hydroxide monohydrate (48 mg,
1.15 mmol) in éthanol (5 mL) and water (2.5 mL) was stirred at 100°C for 1 h. The reaction mixture was concentrated under vacuum and the crude product was purified by préparative high performance liquid chromatography to afford the title compound (14 mg, 14%) as a white solid. Ή NMR (400 MHz, methanol-d-»): δ 8.12 (s, 1 H), 7.13 (d, 1 H), 6.90 (d, 1 H), 4.90-4.86 (m, 1 H), 3.63-3.59 (m, 1 H), 3.37 (s, 3 H), 15 2.85-2.83 (m, 2 H), 2.78-271 (m, 2 H), 2.33-2.26 (m, 2 H), 1.05-1.03 (m. 1 H), 0.57-0.52 (m, 2 H); 0.300.25 (m, 2 H). LC/MS (exact mass) calculated for C15H22N6O2S; 350.152, found (M + H*); 351.2.
The following compounds, Examples 17-18, were prepared from N-[cÎs-3-(methyl{7-[(4methy!phenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo-1,3-oxazolidine-320 sulfonamide (Example 16, Step 1 ) in a similar manner to that described in Example 16, Step 2, substituting the indicated amine for cyclopropanemethylamine, and using the deprotection method illustrated in
Example 16, Step 3.
Example 17Aand 17B: (R)-and (S)-3-Cyano-N-{c!s-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4y/)amino]cyclobutyl}pyrrolidine- 1-sulfonamide
These compounds were prepared using racemic pyrrolidine-3-carbonitrile hydrochloride. The crude racemic mixture was purified by high performance liquid chromatography to afford a white solid (60 mg, 52% over 2 steps). The enantiomers were separated by supercritical fluid chromatography.
Enantîomer A (17A): 24 mg (21%): 1H NMR (400 MHz, CD3OD):Ô8.13 (s. 1 H), 7.13 (d, J = 3.2 Hz, 1 H),
6.69 (d, 1 H, J = 3.6 Hz, 1 H), 4.87-4.84 (m, 1 H), 3.73-3.67 (m, 1 H), 3.65-3.57 (m, 1 H), 3.53-3.50 (m, 2 H), 3.48-3.44 (m, 2 H), 3.405 (s, 3 H). 2.77-2.75 (m, 2 H), 2.42-2.20 (m, 4 H). LC/MS (exact mass) calculated for C16H21N7O2S; 375.148, found (M + H+); 376.1. Chiral HPLC rétention time = 5.97 minutes.
Enantîomer B (17B)r 25mg(21%)JH NMR (400 MHz, CDsOD): δ 8.13 (s, 1 H), 7.13 (d, J = 3.2 Hz, 1 H),
6.69 (d, 1 H, J = 3.6 Hz, 1 H), 4.87-4.84 (m, 1 H), 3.73-3.67 (m, 1 H), 3.65-3.57 (m, 1 H), 3.53-3.50 (m, 2 H). 3.48-3.44 (m, 2 H), 3.405 (s, 3 H), 2.77-2.75 (m, 2 H). 2.42-2.20 (m, 4 H). LC/MS (exact mass) calculated for C16H21N7O2S; 375.148, found (M + H+); 376.1. Chiral HPLC rétention time = 5.16 minutes.
I5 Example 18: 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}2,6· dihydropyrrolo[3.4-c]pyrazole-5(4H)-sulfonamide
This compound was prepared using 2-methyl-2,4,5,6-tetrahydropyrrolo[3,4-c]pyrazole hydrochloride. The crude compound was purified by high performance liquid chromatography to afford the title compound as an off-white solid (24% over 2 steps). 1H NMR (400 MHz, methanol-d4): δ 8.08 (s, 1 H),
7.38 (S, 1 H), 7.10 (d, 1 H). 6.66 (d, 1 H). 4.87-4.86 (m, 1 H), 4.42-4.41 (m, 4 H), 3.87 (s, 3 H), 3.71-3.67 (m, 1 H), 3.31 (s, 3 H), 2.68-2.61 (m, 2 H), 2.27-2.22 (m, 3H). LC/MS (exact mass) calculated for Ci7H22NeO2S; 402.159, found (M + H*); 403.2 and (M + Na); 425.1.
Example 19: N-{cis-3-[Methyl(7H-pyrro!o[2,3-d]pyrimidin-4-yl)amino]cyc!obutyl}-6-oxa-3azabicyclo[3.1.1]heptane-3-sulfonamide
Step 1 : N-[cis-3-(Methyl{7-[(4-methylpheny!)sulfonyl]-7H-pyrro!o[2,3-d]pyrimidinM-y!}amino)cyc!obutyl]-6oxa-3-azabicyc!o[3.1.1]heptane-3-sulfonamide
A mixture of N-[cis-3-(Methyl{7-[(4-methylphenyl)sutfonyl]-7H-pyrroioE2,3-d]pyrimidin-4yl}amino)cyclobutyl]-2-oxo-1,3-oxazolidine-3-sulfonamide (208 mg, 0.40 mmol), 6-oxa-3azabicycio[3.1.1lheptane (50 mg, 0.50 mmol), triethylamine (220 pL, 1.58 mmol) in acetonitrile (15 mL), was heated in a 20 mL microwave vial in a microwave reactorfor 1 hour at 120°C. Excess solvent was evaporated and the resulting oil was taken up in dichloromethane. The solution was washed with aque10 ous ammonium chloride and brine. The crude material was dried over sodium sulfate and concentrated to give an oil. This was chromatographed on silica gel eluting with a gradient methanol in dichloromethane (0:100 to 5:100) to afford the title compound as a foam (82 mg, 30%). 1H NMR (CDCh): 5 8.38 (s, 1 H), 8.04 (d. 2 H), 7.48 (d. 1 H), 7.28 (d, 2 H), 6.63 (d, 1 H), 4.78-4.69 (m, 1 H), 4.62 (d, 1 H), 4.47 (d, 1 H), 3.69-3.61 (m, 1 H). 3.58 (d, 3 H), 3.26-3.17 (m, 1 H), 3.24 (s, 3 H), 2.83-.275 (m, 2 H), 2.37 (s, 3 H), 2.18I5 2.11 (m, 2 H), 2.04 (d, 1 H), 1.18 (t, 1 H). LC/MS (exact mass) calculated for CasHaeNeOsSa; 532.156, found (M + H+); 533.
Step 2: N-{cis-3-[Methyl(7H-pyrro!o[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-6-oxa-3azabicydo[3.1.1]heptane-3-sulfonamide
N-[cis-3-(Methyl[7-[(4-methylphenyl)sulfonyl]-7H-pyrrola[2,3-d]pyrimidin-4-yl}amino)cyctobutyi]-620 oxa-3-azabicycîo[3.1.1 ]heptane-3-sulfonamide (229 mg, 0.43 mmol) was added to a solution of in 1M tetrabutylammonium fluoride in tetrahydrofuran (6.5 mL, 6.4 mmol). The reaction was stirred at room température for 10 hours. The mixture was concentrated and the remaining material was chromatographed on silica gel eluting with a mixture of methanol in ethyl acetate (1:9). A yellow oil was isolated that was
triturated with a mixture of ethyl acetate and heptane to give a yellow solid. The solid was partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over sodium sulfate and concentrated to afford an off-white solid. This was triturated with diethyl ether and then isopropyl alcohol to afford the title compound as a white solid (14 mg, 9%). Ή NMR (CDsOD) δ 8.08 (s, 1 H), 7.09 (d, 1 H), 6.66 (s, 1 H), 4.90-4.81 (m, 1H), 4.62 (d, 2 H), 3.86-8.84 (m , 1 H), 3.66 (t, 1 H), 3.56-3.49 (m, 3 H), 3.33 (s, 3 H), 3.19-3.13 (m, 1 H), 2.75-
2.70 (m, 2 H), 2.32-2.24 (m, 2 H), 2.05-2.03 (d, 1 H). LC/MS (exact mass) calculated for CwHzîNeOsS; 378.147, found (M + H*); 379.5.
The following compounds, Examples 20-24, were prepared from N-[cis-3-(methyl{7-[(4methylphenyl)suifonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo-1,3-oxazolidine-3sulfonamide (Example 16, Step 1) in a similar manner to that described in Exampte 16, Step 2, substituting the indicated amine for cyclopropanemethylamine, and using the deprotection method illustrated in Example 19, Step 2.
Exampfe 20: 3-Cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-cyclobutyfazetidine-1sulfonamide
This compound was prepared using azetidine-3-carbonitrile. The crude compound was purified by high performance liquid chromatography to afford the title compound as a white solid (23% over 2 steps).
Ή NMR (400 MHz, CDaOD): δ 8.10 (s, 1 H), 7.11 (d, 1 H), 6.68 (d, 1 H), 4.80 (m, 1 H), 4.02 (m. 2 H). 3.90 (m, 2 H), 3.58 (m, 2 H), 3.32 (s, 3 H), 2.72 (m, 2 H), 2.25 (m, 2 H). LC/MS (exact mass) calculated for C15H19N7O2S; 361.132, found (M + H+); 362.1.
Example 21 : N-{cis-3-[Methy!(7H-pyirolo[2,3-d]pyrimidin-4-y!)amino]cyclobutyl}-4-( 1H-pyrazo!-3yfpiperidine- 1-sulfonamide
This compound was prepared using 4-(1H-pyrazol-3-yl)piperidine. The crude compound was purified by chromatography on silica gel eluding with a mixture of dichloromethane and methanol (9:1 ). The isolated material was triturated with diethyl ether and then ethyl acetate to afford the title compound as a white solid (10% over 2 steps). 1H NMR (300 MHz, CD3OD): δ 8.09 (s, 1 H), 7.48 (s, 1 H), 7.12-7.05 (m, 1 H), 6.71-6.60 (m, 1 H), 6.22-6.08 (m, 1 H), 4.92-4.73 (m, 1H), 3.80-3.55 (m, 3H), 3.41 (s, 3H), 2.90-2.65 (m, 5 H), 2.38-2.19 (m, 2 H), 2.09-1.90 (m, 2 H) and 1.83-1.65 (m, 2 H). LC/MS (exact mass) calculated for CisHîeNaOîS; 430.190, found (M + H+); 431.1.
Example 22: N-(2-Cyanoethyl)-N-methy!-N'-{cis-3-[methyl(7H-pyrrolo[2,3-d]pynmidin-4yl)amino]cyclobutyl}su/famide
This compound was prepared using 3-methylaminopropionitrile. The crude compound was purified by high performance liquid chromatography to afford the title compound (7% over 2 steps). LC/MS (exact mass) calculated for C15H21N7O2S; 363.148, found (M + H+): 364.0.
Example 23 and 27: (1S,5S)-1-Cyano-N-{cis-3-[methyi(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}3-azabicyc!o[3.1.0]hexane-3-sulfonamide and ( 1R,5R)- 1-Cyano-N-fcis-3-[methyl(7H-pyirolo[2,3d]pyrimidin-4-yl)amino]cyclobutyl}-3-azabicyclo[3.1.0]hexane-3-sulfonamide
These compounds were prepared using racemic 3-azabicyclo[3.1.0]hexane-1-carbonitrile. The crude racemic compound was purified as a white solid (92 mg, 21 % over 2 steps) by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (30:1 to 5:1). The title enantiomers were separated by supercritical fluid chromatography.
Enantiomer 23: 41 mg (9%); SFC rétention time = 4.28 minutes; 1H NMR (400 MHz, methanol-ch): 5 8.13 (s, 1 H), 7.13 (d, 1 H), 6.70 (d, 1 H), 4.61 (s, 1 H), 3.57 - 3.72 (m, 2 H), 3.43 - 3.51 (m, 3 H), 3.36 (s, 3 H),
2.70 - 2.77 (m, 2 H), 2.24 - 2.38 (m, 3 H), 1.41 - 1.48 (m, 1 H), 1.32 (t, 1 H). LC/MS (exact mass) catculated forCnHiiNzOaS; 387.148, found (M + H+); 388.1.
Enantiomer 27: 40 mg (9%); SFC rétention time = 4.84 minutes 1H NMR (400 MHz, methanol-d^): δ 8.13 (s, 1 H), 7.13 (d, 1 H), 6.70 (d. 1 H), 4.61 (s, 1 H), 3.57 - 3.72 (m, 2 H), 3.43 - 3.51 (m, 3 H), 3.36 (s, 3 H),
2.70 - 2.77 (m, 2 H), 2.24 - 2.38 (m, 3 H), 1.41 -1.48 (m, 1 H), 1.32 (t, 1 H). LC/MS (exact mass) calculated for C17H21N7O2S; 387.148, found (M + H*); 388.1.
Racemic 3-azabicyclo[3.1.0]hexane-1-carbonitrile was prepared as follows.
Step 1: Racemic tert-butyl rac-1-formyI-3-azabicyclo[3.1.0]hexane-3-carboxylate
1,1,1-Triacetoxy-1,1-dihydro-1,2-benziodoxol-3(1H)-one (6.5 g, 15.2 mmol) was added to a solution of racemic tert-butyl-1-(hydroxymethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylate (Syn/eff2009, 921) (2.5 g, 11.7 mmol) in anhydrous dichloromethane (60 mL). The reaction mixture stirred for 2 hours at room température. The mixture was diluted with dichloromethane (60 mL), washed with a saturated aqueous solution of sodium sulfite, saturated sodium bicarbonate (30 mL) and brine (50 mL). The organic layer was dried over sodium sulfate and concentrated to give afford the title compound as a colorless oil (1.7 g, 68%). 1H NMR (400 MHz, CDCI3):ô9.01 (d. 1 H), 3.83 (d, 1 H), 3.68 (t. 1H), 3.59 (dd, 1 H), 3.503.36 (m, 1 H), 2.25-2.09 (m, 1 H), 1.63 (t, 1 H) , 1.43 (s, 9 H), 1.19-1.06 (m, 1 H).
Step 2: Racemic tert-butyl-1-[(hydroxyimino)methyl]-3-azabicyclo[3.1.0]hexane-3-carboxy!ate
Potassium carbonate (3.89 g, 28.2 mmol) and hydroxylamine hydrochloride (671 mg, 9.7 mmol) were added to a solution of tert-butyl racemic 1-formyl-3-azabicyclo[3.1,0]hexane-3-carboxylate (1.7 g, 8.05 mmol) in anhydrous dichloromethane (40 mL) at room température and then stirred for 16 hours. The mixture was diluted with ethyl acetate (80 mL), and washed with water (30 mL) and brine (30 mL).
SI
The organic layer was dried over sodium sulfate and concentrated, The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (0:100 to 83:17) to afford the title compound as a yellow oil (1.6 g, 88%). 1H NMR (400 MHz, CDCh): δ 7.18 (s, 1 H), 3.74-3.55 (m, 3 H), 3.44-3.40 (m, 1 H), 1.74-1.72 (m, 1 H), 1.44 (s, 9 H), 1.10 (t, 1 H), 0.86-0.83 (m, 1 H).
Step 3: Racemic tert-butyi-1-cyano-3-azabicyc!o[3.1.0]hexane-3-carboxy!ate
To a solution of racemic tert-buty!-1-[(hydroxyimino)methyl]-3-azabicyclo[3.1.0]hexane-3carboxylate (925 mg, 4.09 mmol) in anhydrous tetrahydrofuran (100 mL) was added methyl N(triethylammonium sulfonyl)carbamate (2.92 g, 12.3 mmol). The reaction mixture was heated to reflux for 3 hours. After évaporation of the solvent, the residue was chromatographed on silica gel eluting with a mixture of petroleum ether and ethyl acetate (5:1) to afford the title compound as a colorless oil (570 mg, 67%). iH NMR (400 MHz, CDCIs): δ 3.84 (dd, 1 H), 3.64 (dd, 1 H), 3.50 (d, 1 H), 3.46 (dd, 1 H), 2.21-2.12 (m, 1 H), 1.44 (s, 9 H), 0.96 (t, 1 H).
Step 4: Racemic 3-azabicycio[3.1.0]hexane-1-carbonitrile
A solution of racemic tert-butyl-1-cyano-3-azabicycio[3.1.0]hexane-3-carboxylate în trifluoroacetic acid (1 mL) and dichloromethane (10 mL) was stirred at room température for 1 hour. The solvent was removed to afford the title compound (205 mg, 100%) as a brown oil.
Exampte 24: Racemic 3-cyano-N-{trans-3-[methy/(7H-pyrroio[2,3-d]pyrimidin-4yl)amino]cyciobutyl}pyrrolidine-1-sulfonamide
This compound was synthesized following the procedure of Example 10, substituting 3cyanopyrrofidine-1-sulfonyl chloride for cyclopropylmethanesulfonyl chloride. The crude product was purified using préparative high performance liquid chromatography to afford the title compound as an offwhite solid (5%). 1H NMR (400 MHz, CDsOD): δ 8.13 (s, 1 H), 7.14 (d, 1 H), 6.67 (d, 1 H), 5.45-5.41 (m, 1
H), 4,00-3.64 (m, 1 H), 3.62-3.52 (m, 1 H), 3.51-3.47 (m, 2 H), 3.45-3.39 (m, 2 H), 3.369 (s, 3 H), 2.78-
2.70 (m, 2 H), 2.53-2.47 (m, 2 H); 2.39-2.36 (m, 1 H); 2,27-2.24 (m, 1 H). LC/MS (exact mass) calculated for C16H21N7O2S; 375.148, found (M + H+); 375.9.
Racemic 3-cyanopyrro!îdine- 1-sulfonyl chloride
A solution of racemic pyrrolidine-3-carbonitrile (53 mg, 0,4 mmol) and triethylamine (101 g, 1 mmol) in dry dichloromethane (1.0 mL) was added dropwise to a stirred solution of sulfuryl chloride (64.8 mg, 0.48 mmol) in dichloromethane (3.0 mL) was added at -78“C. The reaction was stirred at -78°C for 30 minutes, and then allowed to warm to room température over 1 hour. The reaction solution was washed with aqueous 1M hydrochloric acid (5 mL) and brine (5 mL), dried over sodium sulfate and con10 centrated to afford the title compound as colorless oil (68 mg, crude).
Example 25: N-(cis-3-{[(4,4-Difluoropiperidin- l-yOsulfonyljmethytycyclobutyli-N-methyl^H-pyrrolofé, 3d]pyrimidin-4-amine
Step 1 : cis/trans-Ethy! 3-[(tert-butoxycarbonyl)amino]cyclobutanecarboxylate
To a solution of a mixture of cis- and trans-ethyl 3-aminocyclobutanecarboxylate hydrochloride (cis/trans =10:1) (W02009/60278) (10 g, 55.7 mmol) and triethylamine (19.4 mL, 139.1 mmol.) in dichloromethane (370 mL) at 0°C was added dropwise di-terf-butyl dicarbonate (15.8 g, 72.3 mmol). After addition was complété, the mixture was stirred at room température overnight. The solvent was evaporated under reduced pressure and the resulting residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10:1 to 3:1) to afford the title mixture as a white solid (19 g, 92%). 1H NMR (400 MHz. CDCb): δ 4.77 (s, 1 H), 4.13 (q, 3 H), 2.68-2.82 (m, 1 H), 2.60 (d. 2 H), 1.99 2.17 (m, 2 H), 1.43 (s, 9 H), 1.25 (t, 3 H).
Step 2: cis/trans-[3-(Methylamino)cyclobuty!]methanol
Lithium aiuminum hydride (9.14 g, 240.4 mmol) was suspended in dry tetrahydrofuran (350 mL). The mixture was cooled to 0°C and a solution of cis/trans ethyl 3-[(tertbutoxycarbonyl)amino]cyclobutanecarboxylate (cis/trans = 10:1) (11.7 g, 48.1 mmol) in dry tetrahydrofuran (170 mL) was added drop wise. After addition was complété, the resulting mixture was heated to reflux ovemight. After it was cooled to room température, the reaction was diluted with tetrahydrofuran (1.5 L) and then cooled to 0~5°C. Small portions of NaïSCk.lOHzO were added until gas évolution had ceased. The mixture was filtered to remove the solids, which were washed with more tetrahydrofuran (500mL). The filtrate was concentrated to dryness affording the title mixture (cis/trans = 10:1) as an oil (10 g, >100%), 1H NMR (400 MHz, CDCb): δ 3.58 (d, J = 3.8 Hz, 2 H), 3.06-3.17 (m, 1 H),
2.34-2.43 (m, 3 H), 2.32 (s, 3 H), 1.48-1.57 (m, 2 H).
Step 3: cis/trans-[3-(Methy!{7-[(4-methylphenyl)sulfonyl]-7H-pyrro!o[2,3-d]pyrimîdin-4yl}amino)cyclobutyl}me‘hanol
Potassium iodide (173 mg) and triethylamine (13 mL, 93.8 mmol) were added to a solution of cis/trans-[3-(methylamino)cyclobutyl]methanol (6.0 g, 52.1 mmol) in acetone (250 mL). 4-Chloro-7-[(415 methylphenyl)su1fonyl]-7H-pyrrolo[2,3-d]pyrimidine (14.4 g, 46.9 mmol) was then added and the resulting mixture was heated to reflux ovemight. After évaporation of the solvent under reduced pressure, the residue was diluted with dichloromethane (500 mL). The solution was washed sequentially with water (300 mL), 2% aqueous citric acid (300 mL) and brine (300 mL), and then dried over sodium sulfate. After filtration, the solution was filtered and concentrated to afford the title mixture as a light solid (15.3 g, 85%). A portion (5.0 g) of the cis/trans-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)-cyclobutyl]methanol mixture was separated by supercritical fluid chromatography using a Chiralpak-AD column:
cis isomer, 4.6 g: 1H NMR (400 MHz, methanol-di): δ 8,20 (s, 1 H), 7.98 (d, 2 H), 7.53 (d, 1 H), 7,34 (d, 2 H), 6.83 (d, 2 H), 4.99-4.95 (m, 1 H), 3.56 (d, J = 5.6 Hz, 1 H), 3.24 (s, 3 H), 2.36 (s. 3 H), 2.34-2.28 (m, 2
H), 2.24-2.19 (m, 1 H), 2.11-2.03 (m, 2 H). LC/MS (exact mass) calculated for C19H22N4O3S: 386.14, found (M + H+): 387.3 trans isomer, 0.4 g: 1H NMR (400 MHz, methanol-d4): δ 8.20 (s, 1 H), 7.98 (d, 2 H), 7.55 (d, 1 H), 7.35 (d, 2H), 6.84 (d. 2H), 5.26-5.22 (m, 1H), 3.69 (d, 1H), 3.30 (s, 3H), 2.46-2.41 (m. 3H), 2.39 (s, 3H), 2.19-2.14 (m, 2H). LC/MS (exact mass) calculated for CtsH22N4O3S; 386.14, found (M + H*): 387.3
Step 4: ds-[3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin~4yl}amino)cydobutyl]methyl 4-methy/benzenesulfonate
To a solution of cis-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobuty1]methanol (20 g, 51.8 mmol) and N,N-dimethyiamjnopyridine (12.6 g, 103.6 mmol) in dichloromethane (500 mL) at 0°C was added p-toluenesulfonyl chloride (14.8 g, 77.7 mmol). The reaction mixture was stirred at room température for 16 hours and then washed with water (500 mL). The combined aqueous washes were extracted with dichloromethane (2 x 800 mL). The combined organic layers were dried, filtered and concentrated under vacuum. The residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 95:5) to afford the title compound (23 g, 82%) as a white solid. LC/MS (exact mass) calculated for C2SH28N4O5S2: 540.150, found (M + H+): 541.3. Step 5: S-{[cis-3-(Methyl{7-[(4-methylpheny/)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]methyl}ethanethioate
To a solution of potassium thioacetate (678 mg, 5.93 mmol) in N,N-dimethylformamide (5 mL) was added a solution of [cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]methyl 4-methylbenzenesulfonate (2.0 g, 3.70 mmol) in N,N-dimethylformamide (6 mL) dropwise over 5 minutes at room température. The mixture was then heated to at 50-55°C ovemight.
The mixture was cooled to room température and quenched by pouring into aqueous saturated sodium bicarbonate solution (60 mL). The mixture was extracted with ethyl acetate (3 x 30 mL) and the combined organic layers were washed with water (3 x 30 mL), brine (30 mL). After drying over NazSCU the solution was concentrated. The residue was chromatographed on silica gel eluting with a gradient of dichloro5 methane and methanol (100:0 to 80:20) to afford the title compound (1.2 g, 73%) as a yellow solid. 1H NMR (400 MHz, CDCh): 8 8.39 (s, 1 H), 8.04 (d, 2 H), 7.45 (d, 1 H), 7.27 (d, 2 H), 6.63 (d, 1 H), 4.98-4.88 (m, 1 H) 3.22 (s, 3 H) 3.02-3.00 (m, 2 H) 2.45-2.44 (m. 2 H), 2.47 (m, 3 H) 2.22 (m, 3 H) 2.21-2.24 (m, 1 H) 1.92-1.87 (m. 2 H). LC/MS (exact mass) calculated for C21H24N4O3S2: 444.129. found (M + H*): 445.1. Step 6: [ds-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-410 yl}amino)cyclobutyl]methanesulfonic add
To a solution of S-f[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl)amino)cyclobutyl]methyl} ethanethioate (580 mg, 1.31 mmol), in formic acid (10 mL) at room température was added 30% aqueous hydrogen peroxide solution (0.7 mL, 6.92 mmol). The resulting mixture was stirred at room température ovemight. The reaction was poured into an aqueous 33% aqueous sodium bisulfate solution (1,12 mL) and then stined for 10 minutes. Aqueous 33% sodium hydroxide solution (1.8 mL) was then added to adjust the pH to 5. The resulting mixture was stirred at room température for 1 hour. The solid was collected solid by filtration, washed with water (10 mL) and vacuum dried at about 60°C to afford the title compound (634 mg, crude) as a white solid. LC/MS (exact mass) calculated for C19H22N4O5S2; 450.103, found (M + H+); 451.3.
Step 7: ds-[3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]methanesulfonyl chloride
Thionyl chloride (0,3 ml, 3.33 mmol) was added dropwise over 5 minutes to a solution of cis-[3(methyl(7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]methanesulfonic
acid (150 mg, 0.33 mmol) in dichloromethane (20 mL) at 0°C. Two drops of N,N-dimethylformamide were added to the solution, which was then heated at 75°C for 2 hours. The mixture was cooled and the solvent was evaporated. The residue was washed with anhydrous dichloromethane (3 x 10 mL) to afford the crude title compound (170 mg) as a yellow solid. LC/MS (exact mass) calculated for C19H21CIN4O4S2;
468.069, found (M + H+); 469.2.
Step 8: N-(cis-3f[(4,4-Difluoropiperidin- 1-yl)sulfonyi]methyl}cyciobutyl)-N-methyl-7-[(4methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine
To a mixture of 4,4-difiuoropiperidine (77 mg, 0.64 mmol) and triethylamine (97 mg, 0.96 mmol) in tetrahydrofuran (20 mL) at 0DC was added dropwise a solution of cis-[3-(methyl{7-[(410 methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}arTiino)cyclobutyl]methanesulfonyl chloride (150 mg, 0.320 mmol) in tetrahydrofuran (10 mLJ.The mixture was alfowed to warm to room température overnight. The solvent was evaporated and the residue was taken up in ethyl acetate (80 mL). The solution was washed with brine (30 mL), dried over sodium sulfate and concentrated to afford the crude title compound (134 mg) as a white solid. LC/MS (exact mass) calculated for C24H29F2N5O4S2; 553.651, found (M + H+); 554.3.
Step 9: N-(cis-3-{[(4,4-difluoropiperidin- 1-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,3dJpyrîmidin-4-amine
N-(cis-3-{[(4,4-Difluoropiperidin-1-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7-[(4methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine (134 mg, 0.24 mmol) and lithium hydroxide monohydrate (51 mg, 1.21 mmol) were combined in a mixture of éthanol (14 mL) and water (7 mL) and then heated at 50°C overnight. The reaction was concentrated under vacuum and diluted with ethyl acetate (100 mL). The resulting solution was washed with brine (30 mL), dried over sodium sulfate and concentrated. The residue was chromatographed using préparative thin layer chromatography eluting with a
mixture of ethyl acetate and methanol (20:1) to afford the title compound (31 mg, 32,3 %) as a white solid.
’H NMR (400 MHz, methanol-do): δ 8.13 (s, 1H). 7.13 (d, J = 3.6, 1H),6.70 (d, J = 3.6, 1 H), 5.12 (m, 1H),
3.49-3.47 (m, 4H), 3.46 (m, 3H), 3.33 (m, 2H), 2.62-2.54 (m, 3H), 2.25-2.20 (m, 1H), 2.11-2.05 (m. 2H).
LC/MS (exact mass) calculated for C17H23F2N5O2S; 399.154, found (M + H*); 400.3.
Examples 26 to 29. The following compounds were made starting from cis-[3-(methyl{7-[(4methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyc!obutyl]methanesulfonyl chloride (Example 25, Step 7), according to the procedures of Exampie 25, Step 8 (sulfonylation) and step 9 (deprotection), substituting the appropriate amine for 4,4-difluoropiperidine in Step 8.
Example 26: 1-[({cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc!obutyl}-methyl)sulfonyl]Y(trifÎuoromethyl)piperidin-4-ol
The title compound (31 mg) was prepared using 4-(trifluoromethyl)piperidin-4-ol in the sulfonylation step and was deprotected using the method from Example 25, Step 9. The compound was purified using préparative thrn layer chromatography eluting with eluting with a mixture of ethyl acetate and methanol (20:1). ’H NMR (400 MHz, methanol-di): δ 8.13 (s, 1H), 7.13 (d, J = 3.6, 1 H), 6.70 (d, J = 3.6, 1H), 5.10-5.08 (m, 1 H), 3.74-3.71 (m, 2 H). 3.36 (m, 3 H), 3.32-3.27 (m, 2 H), 3.19-3,13 (m, 2 H), 2.62-2.54 (m, 3 H), 2.25-2.21 (m, 2 H), 1.86-1.84 (m, 4 H). LC/MS (exact mass) calculated for CiaH24F3N5O3S; 447.155, found (M + H+); 448.3.
Example 28 and 29: (3R) and (3S)- 1-[({cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4y!)amino]cyclobutyl}methy!)su!fonyl]pyrrolidine-3-carbonitrile
The titfe compounds (330 mg total) were prepared using pyrrolidine-3-carbonitrile enriched as an 80:20 scalemic mixture of (3/i)-pyrrotidine-3-carbonitrile and (3^-pyrrolidine-3-carbonitrile enantiomers in the sulfonylation step and was de-protected using the method from Example 19, Step 2. The compound was purified by chromatography on silica gel eluting with gradient of petroleum ether and ethyl acetate (10:1 to 1:10). LC/MS m/z=375.2 (M+1). The enantiomers were separated by préparative supercritical fluid chromatography:
3/?-enantiomer(28): 178 mg. ’H NMR (400 MHz, CDCh):58.31 (s, 1H), 7.06 (d. 1 H), 6.58 (d, 1 H), 5.195.10 (m, 1 H), 3.77-3.75 (m, 1 H), 3.61-3.54 (m, 3 H), 3.33 (s. 3 H). 3.21-3.19 (m, 3 H), 2.69-2.66 (m, 3 H), 2.36-2.31 (m, 2 H), 2.14-2.11 (m, 2 H). LC/MS (exact mass) calculated for C17H22NSO2S; 374.15, found (M + H+); 375.2. Chiral HPLC rétention time = 2.65 minutes
3S-enantiomer(29): 31 mg. 1H NMR (400 MHz, CDCI3): 5 8.31 (s, 1H),7.06(d, 1 H), 6.58 (d, 1 H). 5.195.10 (m, 1 H), 3.77-3.75 (m, 1 H), 3.61-3.54 (m, 3 H), 3.33 (s, 3 H), 3.21-3.19 (m, 3 H). 2.69-2.66 (m, 3 H), 2.36-2.31 (m, 2 H), 2.14-2.11 (m, 2 H). LC/MS (exact mass) calculated for C17H22N6O2S; 374.15, found (M + H+); 375.2 Chiral HPLC rétention time = 2.53 minutes
Example 30: N-{cis-3-[(Buty/sulfony/)methyl]cydobutyl}-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine
Step 1 : N-{cis-3-[(Butylthio)methyl]cyclobutyl}-N-methyl-7H-pyrroÎo[2,3-d]pyrimidin-4-amine
A solution of cis-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]methyl 4-methyibenzenesuifonate from Example 25, Step 4 (23 g, 42.6 mmol) was stirred in N-methylpyrrolidine (100 mL). Then 1,8-diazabicycloundec-7-ene (12.8 g, 85.2 mmol) and 1butanethiol (7.8 g, 85.2 mmol) was added to the reaction mixture. The reaction was stirred at room température for 16 hours. Water (200 mL) and ethyl acetate (500 mL) were added. The aqueous layer was extracted with ethyl acetate (2 x 500 mL) and the combined organic layers were dried and concentrated.
The residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 90:10) to afford the title compound (11.8 g, 91%). LC/MS (exact mass) calculated for C1GH24N4S; 304.172, found (M + H*); 305.3.
Step 2: N-(cis-3-[(Buty!sulfonyl)methyl]cyclobuty/}-N-methyl-7H-pyrro!o[2,3-d]pyrimidin-4-arnine
N-{cis-3-[(Butylthio)methyl]cyclobutyl}-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-arnirie (12 g, 39.5 mmol) was dissolved in a mixture of tetrahydrofuran (200 mL), éthanol (200 mL) and water (200 mL). Potassium peroxomonosulfate (48.6 g, 79.0 mmol) was added and the reaction was stirred at room température for 1 hour. The mixture was then filtered; the solids were washed with a mixture of tetrahydrofuran (40 mL), éthanol (40 mL) and water (20 mL). The filtrate was treated with aqueous 10% sodium bisulfite solution (200 mL) and stirred at room température for 20 minutes. A saturated solution of aqueous sodium bicarbonate was added to adjust the pH to -7. The mixture was extracted with dichloromethane (3 x 800 mL) and the combined organic layers were dried and concentrated under vacuum. The crude residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 95:5) to obtain the title compound (11.4g, 86%). ’H NMR (400 MHz, methanol-di): 8 8.13 (s,
1 H), 7.13-7.12 (m. 1 H), 6.70-6.69 (m, 1 H), 5.13-5.10 (m, 1 H), 3.42 (s, 3 H). 3.33 (m, 2 H), 3.11-3.07 (m, 2 H), 2.65-2.63 (m, 3 H), 2.29-2.25 (m, 2 H),1.86-1.78 (m, 2 H), 1.55-1.50 (m. 2 H), 1.03-0.99 (m, 3 H). LC/MS (exact mass) calculated for C16H24N4O2S; 336.162, found (M + H+); 337,3
Example 31 : N-Methyl-N-(trans-3-((propylsulfony!)methyl)cyclobutyl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine
The title compounds were made as a mixture of cis and trans isomers (50 mg) startîng from cis and trans-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyi]methyl 4-methylbenzene sulfonate (cis/trans = 10:1) (Example 25, Step 4), according to procedures similar to those of Example 30, Steps 1 and 2, using propane-1-thiol instead of butane-1 -thiol in Step 2. The mix-
ture of cis and trans tsomers was purified by reverse phase high performance liquid chromatography éluting with gradient of water and acetonitrile (95:5 to 5:95). LC/MS (exact mass) calculated for C15H22N4O2S; 322.15, found (M + H*); 323.2
The cis and trans isomers were then separated by préparative supercritical fluid chromatography.
trans isomer (31), 12 mg: 1H NMR (400 MHz, methanol-d4): δ 8.12 (s, 1 H), 7.13-7.12 (m, 1 H), 6.69-6.66 (m, 1 H), 5.45-5.41 (m, 1 H), 3.46-3.44 (m, 2 H), 3.36 (s, 3 H), 3.11-3.09 (m, 2 H), 2.88-2.86 (m. 1 H), 2.75-2.67 (m, 2 H), 2.40-2.38 (m, 2 H), 1.91-1.86 (m, 2 H), 1.12-1.10 (m, 3 H). LC/MS (exact mass) calculated for C15H22N4O2S; 322.15, found (M + H+); 323.2 cis isomer, 36 mg: 1H NMR (400 MHz, methanol-d4): δ 8.12 (s, 1 H), 7.13-7.12 (m, 1 H), 6.70-6.69 (m, 1
H), 5.10-5.20 (m. 1 H), 3.36 (s, 3 H), 3.33-3.32 (m. 2 H), 3.08-3.04 (m, 2 H), 2.64-2.61 (m, 3 H), 2.24-2.22 (m, 2 H), 1.90-1.84 (m, 2 H), 1.13-1.09 (m, 3 H). LC/MS (exact mass) calculated for C15H22N4O2S; 322.15, found (M + H+); 323.2
Example 32: N-(cis-3-{[(2-Cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,315 dJpyrimidin-4-amine
Step 1 : N-(cis-3-{[(2-Cyclopropylethyl)sulfanyl]methyl}cyclobutyl)-N-methyl-7-[(4-methy!pheny!)sulfonyl]7H-pyrroio[2,3-d]pyrimidin-4-amine
Nitrogen was bubbled through a mixture of S-{[cis-3-(methyl{7-[(4-methylphenyl)sulfonyt]-7Hpyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]methyl} ethanethioate Example 25, Step 5 (190 mg, 0.43 mmol) and potassium carbonate (129 mg, 0.94 mmol) in methanol (10 mL) at 0'C for 2 minutes. 2Cyclopropylethyl 4-methylbenzenesulfonate (159 mg, 1.53 mmol) was then added and the solution was stirred for 6 hours at room température. Dichloromethane (30 mL) and water (20 mL) were added and the aqueous layer was extracted with dichloromethane (2 x 20 mL). The combined organic layers were
washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by préparative thin layer chromatography using ethyl acetate-petroleum ether (1:2) to give the title compound as a white solid (62 mg, 31%). LC/MS (exact mass) calculated for C24H30N4O2S2; 470.18, found (M + H*); 471.1
Step 2: N-(cis-3-{[(2-Cyclopropylethyi)sulfonyl]mefhyi}cyclobutyl)-N-methyl-7-[(4-methyiphenyl)sulfonyl]7H-pyrro!o[2,3-d]pyrimidin-4-amine
The mixture of N-(cis-3-{[(2-cyclopropylethyl)sulfanyl]-methyl}cyc!obutyl)-N-methyl-7-[(4methyl phenyl)-sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine (24 mg, 0.051 mmol) and potassium peroxomonosulfate (49 mg, 0.079 mmol) in tetrahydrofuran (1.2 mL), water (0.6 mL) and éthanol (1.2 mL) was stirred at room température for 20 minutes. Aqueous sodium bisulfite was added, followed by dichloromethane (20 mL). The aqueous layer was extracted with dichloromethane (2 x 20 mL) and the combined organic layers were extracted with brine, dried over sodium sulfate and concentrated. The crude material was used directly in next step. LC/MS (exact mass) calculated for C24H30N4O4S2; 502.17, found (M + H*); 503.3
Step 3: N-(cis-3-{[(2-Cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,3-dJpyrimidin-4amine
The mixture of N-(cis-3-{K2-cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7-[(4methylphenyl)sulfonyl]-7H-pyrrofo[2,3-dlpyrimidin-4-amine (49 mg, 0.097 mmol) and lithium hydroxide (30 mg, 1.3 mmol) in water (5 mL) and éthanol (10 mL) was stirred at 50°C for 2 hours. Then, dichloro20 methane (20 mL) was added and the aqueous layer was extracted with dichloromethane. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using a gradient of water and acetonitrile gradient (95:5 to 5:95) to give the title compound (14 mg, 40%) as white solid. 1H NMR (400
MHz, methanol-di): δδ.12 (s, 1 H), 7.13 (d, 2 H), 6.70 (d, 2 H), 5.12-5,09 (m, 1 H), 3.34 (s, 3 H), 3.343.33 (m, 2 H), 3.20-3.17 (m, 2 H), 2.64-2.61 (m. 3 H), 2.26-2.22 (m, 2 H), 1.75-1.69 (m, 2 H), 0.89-0.86 (m, 2 H), 0.56-0.52 (m, 2 H), 0.18-0.17 (m, 2 H). LC/MS (exact mass) calculated for C17H24N4O2S; 348.16, found (M + H+); 349.1
Example 33: N-[ds-3f{[(3,3-Difluorocyclobutyl)methyl]sulfonyl}-methyl)cyc!obutyl]-N-methy/-7Hpyrro!o[2,3-d]pyrimidin-4-amine
Step 1 : N-[cis-3-({[(3,3-Difluorocyc!obutyl)methyl]sulfanyl}methyl)cyclobutyl]-N-methyl-7H-pyn-olo[2,3d]pyrimidin-4-amine
Nitrogen was bubbled through a mixture of S-{[cis-3-(methyl(7-[(4-methylphenyl)-sulfonyl]-7Hpymolo[2,3-d]pyrimidin-4-yl)amino)cyclobutyl]methyl} ethanethioate, Example 25, Step 5 (250 mg, 0.56 mmol) and potassium carbonate (194 mg, 1.41 mmol) in methanol (100 mL) for two minutes at 0°C followed by addition of (3,3-difluorocyclobutyl)methyl 4-methylbenzenesulfonate (prepared as described in W02004/032834) (310 mg, 1.12 mmol). The mixture stirred for 6 hours at room température, filtered, and concentrated to give the title compound (270 mg, crude) as white solid. LC/MS (exact mass) calculated for C17H22F2N4S; 352.15, found (M + H+); 353.2
Step 2: N-[cis-3-({[(3,3-Difluorocyciobutyl)methyl]sulfony!}methyl)cyclobLityl]-N-rnethyl-7H-pyn-oÎo[2,3d]pyrîmidin-4-amine
The mixture of N-[cis-3-(([(3,3-dif!uorocyclobutyl)methyl]sulfanyl}-methyl)cyclobutyl]-N-methyl-7H20 pyrrolo[2,3-d]pyrimidin-4-amtne (45 mg, 0.13 mmol) and potassium peroxomonosulfate (157 mg, 0.26 mmol) in a mixture of tetrahydrofuran (20 mL), water (10 mL) and éthanol (20 mL) was stirred at room température for 20 minutes. Aqueous sodium bisulfite was then added, followed by dichloromethane (20 mL). The aqueous layer was extracted with dichloromethane (2 x 20 mL) and the combined organic lay-
ers were extracted with brine, dried over sodium sulfate, and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using water-acetonitrrle gradient (95:5 to 5:95) to give the title compound as white solid (34 mg, 39%). 1H NMR (400 MHz, methanol-d4): δ 8.29 (s,
H), 7.42 (d, 1 H), 7.03 (d. 1 H). 4.86 (m, 1 H), 3.51 (s, 3 H), 3.39-3.33 (m, 4 H), 2.84 (m, 1 H), 2.76-2.71 (m, 4 H), 2.53 (m, 2 H), 2.37-2.34 (m, 2 H). LC/MS (exact mass) calculated for C17H22F2N4O2S; 384.14, found (M + H+); 385.1.
Example 34A and 34B: (1R, 3R)and(1S, 3S)-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidîn~4yl)amino]cyclobutyl}methyl)sulfonyl]cyclopentanecarbonitrile
The title compound mixture of (1 R, 3R) and (1 S, 3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3d]pyrimidin-4-yl)amino]cyclobutyl}-methyl)sulfonyl]cyclopentane-carbonitrile was prepared from S-{[cis-3(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyc1obutyl]methyl} ethanethioate Example 25, Step 5, according to the procedure of Example 30, steps 1 and 2.
The title compound (180 mg) was separated by préparative supercritical fluid chromatography us15 ing a Chiralpak AS column:
(1R, 3Ri enantiomer 34A: 60 mg, 1H NMR (400 MHz, methanol-d4): δ 8.12 (s, 1 H), 7.13-7.12 (d, 1 H), 6.70-6.69 (d, 1 H), 5.17-5.11 (m, 1H), 3.86-3.78 (m, 1H), 3.41-3.36 (m. 5 H), 3.15-3.11 (m, 1 H), 2.63-2.53 (m, 4 H), 2.37-2.13 (m, 6 H), 2.03-1.91 (m, 1 H). LC/MS (exact mass) calculated for C17H22F2N4O2S; 373.16, found (M + H+); 374.1 (1S, 3S) enantiomer 34B: 27 mg, LC/MS (exact mass) calculated for C17H22F2N4O2S; 373.16, found (M +
H+); 374.1
The intermediate 3-cyanocyclopentyl 4-methylbenzenesuifonate used in step-1 was prepared as shown below:
3-Cyanocydopentyi 4-methylbenzenesulfonate
4-Methylbenzene-1-sulfonyl chloride (6.9 g, 36 mmol) and N,N-dimethylpyridin-4-amine (100 mg) were added to a solution of compound 3-hydroxycyclopentane-carbonitrile (J. Org. Chem. 2007, 72, 7423) (2 g, 18 mmol) and triethylamine ( 5.5 g, 54 mmol) in dichloromethane (100 mL). The reaction was stirred at room température for 15 hours and then quenched mixture was quenched by addition of saturated aqueous sodium bicarbonate solution (20 mL). The mixture was extracted with dichloromethane (4 x 50 mL). The combined organic layers were dried over magnésium sulfate and concentrated. The residue was chromatographed on silica gel by eluting with a mixture of petroleum ether and ethyl acetate (1:1) to give the title compound as yellow oil (0.5 g, 11% yield). LC/MS (exact mass) calculated for
I0 C13H15NO3S; 265.08, found (M + 23); 287.9.
Example 35: RacemicN-methyl-N-[ds-3-({[1-(propan-2-yl)pyrrolldin-3-yl]sulfonyl}methyl)cydobutyl]-7Hpyrro!o[2,3-d]pyrimidin-4-amine
Step 1 : tert-Buty! 3-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyiTolo[2,3-d]pyrimidin~415 yl}amino)cydobutyl]methyl}sulfanyl)pyrrolidine- 1-carboxylate
The solution of cis-[3-(methyl{7-[(4-rnethylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4yl}amino)cyclobutyl]methyl 4-methylbenzenesulfonate from Example 25, Step 4 (2 g, 3.7 mmol) was stirred în N-methylpyrrolîdine (40 mL). LS-Diazabicycloundec-Z-ene (1.13 g, 7.4 mmol) and 3-mercapto20 pyrrolidine-1-carboxylic acid tert-butyl ester (1.13 g. 5.6 mmol) were then added to the reaction mixture. The reaction was stirred at room température for 16 hours. Water (200 mL) and ethyl acetate (500 mL) were added. The aqueous layer was extracted with ethyl acetate (2 x 500 mL).and the combined organic
layers were dried and concentrated under vacuum to give the title compound as a white solid (2.6 g, 118%). LC/MS (exact mass) calculated for C2BH37N5O4S2; 571.23, found (M + H+): 572.1.
Step 2: N-Methyl-7-[(4-methylphenyl)sulfony!]-N-{cis-3-[(pyrro)idin-3-yisulfanyl)methyl]cyclobutyl}-7Hpyrro!o[2,3-d]pyrimidin-4-amine
To a solution of tert-butyl 3-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3d]pyrimidin-4-yl}amino)cyclobutyl]methyl}sulfanyl)pyrrolidine-1-carboxylate (2.6 g, 4.5 mmol) in methanol (15 mL) was added 3M hydrochloricacid solution in methanol (40 mL). The resulting solution was stirred at room température for 1 hour. The solution was concentrated to give the crude product, which was purified by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 85:15) to give the title compound as colorless oil (1.7 g, 52%). 1H NMR (400 MHz, methanol^): δ 8.21 (s, 1 H), 7.99 (d, 2 H), 7.56 (d, 1 H), 7.37 (d, 2 H), 6,88 (d, 1 H), 4.95-4.87 (m, 1H), 3.29 (s, 1 H), 3.27 (s, 3 H), 3.21-3.17 (m, 1 H), 3.04-3.96 (m, 1 H), 2.92-29 (m, 1 H), 2.72-2.01 (m, 3 H), 2.50-2.43 (m, 2 H), 2.39 (s, 3 H). 2.29-2.15 (m. 2 H), 2.03-2.01 (m, 2 H), 1.98-1.65 (m, 1 H). LC/MS (exact mass) calculated for C23H29N5O2S2; 471.18, found (M + 23): 494.
Step 3: N-Methyl-7-[(4-methylphenyl)sulfonyl]~N-[cis-3-({[ 1-(propan-2-yl)pyrTolidin-3yl]sulfanyl}methyl)cyclobutyl]-7H~pyrrolo[2,3-d]pyrimidin-4-amine
To a solution of N-methyl-7-[(4-methylphenyl)sulfonyl]-N-{cis-3-[(pyrrolidin-3y[sulfanyl)methyl]cyclobutyl}-7H-pyrrolo[2,3-d]pyrimidin-4-amine (472 mg, 1 mmol) in dichloromethane (50 mL) was added acetone (174 mg, 3 mmol), 4Â molecular sieves (40 mg) and sodium cyanoborohydride (189 mg, 3 mmol). The resulting solution was stirred at room température for 1 hour, then diluted with dichloromethane (70 mL) and water (70 mL). The aqueous layer was extracted with dichloromethane (2 x 50 mL) and the combined organic layers were washed with brine (100 mL). The organic layer was dried
over anhydrous sodium sulfate and concentrated to give title compound (500 mg, 97% yield) as cotorless oil. LC/MS (exact mass) calculated for C26H35N5O2S2; 513.22, found (M + H+); 514.1.
Step 4: N-Methyl-7-[(4-methylphenyl)sulfony/]-N-[cis-3-({[ 1-(propan-2-yl)pyrro!idin-3yl]sulfonyl}methyl)cyclobutyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine
The mixture of N-methyl-7-[(4-methylphenyl)su!fonyl]-N-[cis-3-({[1-(propan-2-yl)pyrrolidin-3yl]sulfanyl}methyl)cyclobutyl]-7H-pyrTOlo[2,3-d]pyrimidin-4-amine_(500 mg, 1.0 mmol) and potassium peroxomonosulfate (1.23 g, 2.0 mmol) in tetrahydrofuran (20 mL), water (10 mL), and éthanol (20 mL) was stirred at room température for 30 minutes. The reaction solution was diluted with ethyl acetate (100 mL) and water (50 mL). The aqueous layer was extracted with ethyl acetate (3 x 50 mL) and the combined organic layers were washed with brine (100 mL), dried over sodium sulfate and concentrated to give title compound as colorless oil (420 mg, 90%). LC/MS (exact mass) calculated for C26H35N5O4S2; 545.21, found (M + H*): 546.3.
Step 5: N-Methyl-N-[cis-3-({[1-(propan-2-yl)pyn-olidin-3-yl]sulfonyl}methyl)-cyclobutyl]-7H-pyirolo[2,3d]pyrimidin-4-amine
The mixture of N-methyl-7-[(4-methylphenyl)sulfonyl]-N-[cis-3-({[1-(propan-2-yl)pyrrolidin-3yl]sulfonyl}methyl)cyclobutyl]-7H-pymjlo[2,3-d]pyrimidin-4-amine_(330 mg, 0.6 mmol) and lithium hydroxide (126 mg, 3 mmol) in a mixture of water (5 mL) and éthanol (10 mL) was stirred at 50°C for 2 hours. The mixture was then concentrated and the residue was taken up rn ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using a gradient of water and acetonitrile (95:5 to 5:95) to give the title compound (89 mg, 38%) as white solid. 1H NMR (400 MHz, CDCh): δ 8.32 (s, 1 H), 7.07 (d, 1 H), 6.57 (d, 1 H), 5.13 (m, 1H), 3.57 (m, 1H), 3.33 (s, 3 H) 3.05-3.22 (m, 3H), 2.92 (m, 1H), 2.78
- 2.87 (m, 1H), 2.58-2.77 (m, 4H), 2.50 (m, 1H). 2.19-2.34 (m, 2H), 2.06-2.19 (m, 2H), 1.12 (d, 6H).
LC/MS (exact mass) calculated for CiaHzgNsOîS; 391.20, found (M + H+); 392.3
Example 36: N-(cis-3-{[(3-Chloro-4-f!uorophenyl)sulfony/]methyl}cyclobutyl)-N-methy!-7H-pyrrolo[2,35 d]pyrimidin-4-amine
Step 1 : N-(cis-3-{[(3-Chloro-4-fluorophenyl)sulfanyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,3dJpyrimîdin-4-amine
To a solution of 3-chloro-4-fluorothiophenol (93 mg, 0.55 mmol) in tetrahydrofuran (1.5 mL) was added 50% aqueous sodium hydroxide (44 mg, 0.55 mmol) and éthanol (1.5 mL). The mixture was stirred at room température for 1 hour. A solution of cis-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyaolo[2,3d]pyrimidin-4-yl}amino)cyclobutyl]methyl 4-methylbenzenesulfonate from Example 25, Step 4 (200 mg, 0.37 mmol) in tetrahydrofuran (1.5 mL) was added to the reaction mixture. The combined mixture was heated at 40°C ovemight. The reaction was concentrated and purified by silica column eluting with a gradient of heptanes and ethyl acetate (90:10 to 0:100) to afford the title compound (69 mg, 49.6%). ’H NMR (400 MHz, CDCh): δ 8.18 (s, 1 H), 7.39 (dd, 1 H), 7.28 (s, 1 H), 7.03-7.08 (m, 1 H), 7.00 (d, 1 H), 6.52 (d,
H), 4.97-5.07 (m, 1 H), 3.35 (m, 2 H), 3.23 (s, 3 H), 2.89 (s, 1 H), 2.43-2.52 (m, 2 H), 2.19-2.30 (m, 2 H). Step 2: N-(cis-3-{[(3-Chloro-4-fluorophenyl)sulfonyl]methyl}cyclobutyÎ)-N-methy/-7H-pyrrolo[2,3d]pyrimidin-4-amine
To a solution of N-(cis-3-{[(3-chloro-4-fluorophenyl)sulfanyl]methyl}cyc1obutyl)-N-methyl-7H20 pyrrolo[2,3-d]pyrimidin-4-amine (75 mg, 0.2 mmol) in dichloromethane (10 mL) was added 3chlorobenzoperoxoic acid (107 mg). The reaction was stirred at room température ovemight and then concentrated. The crude residue was chromatographed on silica gel eluting with a gradient of dichloromethane and 2M ammonia in methanol (80:20) to obtain the title compound (48 mg, 59.2%). ’H NMR
(400 MHz, methanol-d4): 8 8.31 (s, 1H), 8.02 (m, 1H), 7.81-7.83 (m, 1H) 7.37-7.27 (m, 1H), 7.09 (d, 1H),
6.65 (s, 1 H). 5.18-5.10 (m, 1 H), 4.15-4.09 (m, 1 H), 3.32 (m, 5 H), 2.59-2.54 (m, 2 H), 2.44-2.42 (m, 2 H).
LC/MS (exact mass) calculated for C18H18CIFN4O2S; 408.08, found (M + H*); 409
Example 37: 2-[({cis-3-[Methy!(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl) sulfonyljpyridine4-carbonitrile
Step 1:2-({[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrOlo[2,3-d]pyrimidin-4yl}amino)cyclobuty!]methyl}su!fanyl)pyridine-4-carbonitrile
1,8-Dtazabicycloundec-7-ene (24.6 g, 161 mmol) and 2-mercapto-isonicotinonitrile (16.1 g, 118 mmol) were added to a solution of [cis-3-(methyl{7-[(4-methylphenyi)sulfonyl]-7H-pyrrolo[2,3-d]pyrim!din4-yl}amino)cyclobutyl]-methyl methanesulfonate (50 g, 110 mmol) in N-methylpyrrolidine (250 mL). The reaction was heated at 50°C ovemight. Additional 2-mercaptoisonicotinonitrile (8.1 g, 59 mmol) was added to drive the reaction to completion. The mixture was cooled to about 0°C and then the reaction was quenched by dropwise addition of water. The solids were collected by filtration, washed with water, and dried under vacuum at 50‘C to give the title compound as a bright yellow solid (45.8 g, 82.8%). LC/MS (exact mass) calculated for C25H24N6O2S2; 504.14, found (M + H+); 505.1
Step 2: 2-[({cis-3-[Methyi(7H-pyrro/o[2,3-d]pyrimidin-4-y!)amino]cyclobutyl}methyl)sulfanyl]pyndine-4carbonitri/e
To a solution of 2-({[cis-3-(methyl{7-[(4-methylpheny!)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-420 yl}amino)cycfobutyl]methyl}sulfanyl)pyridine-4-carbonitrile (45.3 g, 89.8 mmol) in tetrahydrofuran (180 mL) was added a solution of 1M tetrabutylammonium fluoride in tetrahydrofuran (269 mL). The reaction mixture was heated to reflux for 6 hours and then cooled to room température. Water was added dropwise over 45 minutes. The solids were collected by filtration and washed with a mixture of 20% tetrahydrofuran (33 mL) and water (97 mL). The wet cake was dried under vacuum at 50’C to give the title compound as a tan solid (25 g, 79%). LC/MS (exact mass) calculated for CisHiaNeSz 350.13, found (M + H+); 351.1 Step 3: 2-[({cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]pyridine~4carbonitriïe
Potassium peroxomonosulfate (236.8 g, 385.2 mmol) was added slowly to a mixture of 2-[({cis-3[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-methyl)sulfanyl]pyridine-4-carbonitrile (22.5 g, 64.2 mmol) in methanol (337 mL) and water (56 mL) at 0°C. The reaction was stirred at 3DC for 20 hours. The reaction was quenched using 10% aqueous sodium bisulfate solution (40 mL). and the resulting slurry was stirred at room température for 2 hours. Aqueous 10% potassium carbonate solution was added unti! the pH was 4 to 5. The material was filtered and rinsed with water. The wet filter cake was dried under vacuum at 40°C to give an off-white solid. This material was taken up in tetrahydrofuran (50 mL) and heated to reflux for 3 hours. The mixture was cooled to room température and filtered to collect the solid, which was dried under vacuum at 40'C to the title compound as a light tan powder (17.3 g, 70.46%). 1H NMR (400 MHz, CDCh): S 11.97 (s, 1 H), 8.95 (d, 1 H), 8.33-8.28 (m, 2 H), 7.81 (d, 1 H) 7.1 (d. 1 H), 6.54 (d, 1 H). 5.13-5.08 (m, 1 H), 3.63 (m, 2 H), 3.30 (s, 3 H), 2.54-2.48 (m. 3 H), 2.09-2.07 (m, 2 H). LC/MS (exact mass) calculated for CiaHieNeOaSï; 382.12, found (M + H+); 383.1.
Example 38. 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-1,3-thiazole5-su/fonamîde
Step 1: 2-Methyl-N-[cis-3-(methyl{7~[(4-rnethylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrÎmidin-4yl}amino)cyclobutylJ-1,3-thiazole-5-sulfonamide
Triethylamine (62.0g, 0.613 mol) is added to a solution of cis-N-methyl-N-7H-pyrrolo[2,3dlpyrimidin-4-ylcyclobutane-1,3-diamÎne hydrochloride (22.2.g, 0.102 mol) in dichloromethane (250 mL).
100
2-Methylthiazole-5-sulfonyl chloride (28,0g, 0,142 mol) in dichloromethane (250 mL) is added over 30 minutes at room température to the reaction mixture. After 1.5 hours the solvent is removed under reduced pressure and the résultant solid dissolved in 4:1 ethyacetate:dichloromethane (400 mL). The solution is fiîtered through a 40g silica plug, rinsing with ethylacetate (800 mL) and dichloromethane (100 mL).
The solvent from the filtrate is removed under reduced pressure to give solid (59g). The solid is purified using silica gel column chromatography eluting from 1:1 dichloromethane:ethylacetate to neat ethylacetate to give the title compound (44.4g, 81 %); m/z (Cl) 533 [M+H]+.
Step 2: 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidÎn-4-y/)amino]cyclobutyl}-1,3-thiazole-5sulfonamide
Lithium hydroxide (12.1g, 0.505 mol) in water (290 mL) is added to 2-methyl-N-[cis-3-(methyl{7[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyc!obutyl]-1,3-thiazole-5-sulfonamide (43.8g, 82.2 mmol) in isopropyl alcohol (435 mL) and the mixture heated to 60oC overnight. After cooling to room température the reaction mixture is fiîtered rinsing with water (145 mL). The filtrate is pH is adjusted to 6-7 using 6M aqueous hydrochloric acid. The reaction slurry is concentrated under reduced pressure. Water (370 mL) is added and the mixture cooled to 0°C. The solved is colîected by filtration, washed with cold water (150 ml) then vacuum dried at 60°C overnight to give the title compound (25.0g, 80%); ’H NMR (DMSO-d6); δ 11.66-11.71 (1 H), 8.44-8.47 (1 H), 8.11-8.08 (2 H). 7.16-7.17 (1 H),6.636.65 (1 H), 4.86-4.94 (1 H), 3.58-3.68 (1 H), 3.22 (3 H), 2,74 (3 H), 2.40-2.46 (2 H), 2.10-2.18 (2H). m/z (Cl) 379 [M+H]*.
Biological Evaluation
JAK Caliper Enzyme Assay at 1 mM ATP
ΙΟΙ
Test article was solubilized in dimethyl sulfoxide (DMSO) to a stock concentration of 30 mM. An 11-point hatf log dilution sériés was created in DMSO with a top concentration of 600 μΜ. The test compound plate also contained positive control wells containing a known inhibitor to define 100% inhibition and négative control wells containing DMSO to define no inhibition. The compound plates were diluted 1 to 60 resulting in a top final assay compound concentration of 10 μΜ and a 2% DMSO concentration.
Test article and assay controls were added to a 384-well plate. Reaction mixtures contained 20 mM HEPES, pH 7.4,10 mM magnésium chloride, 0.01% bovine sérum albumin (BSA), 0.0005% Tween 20, 1 mM ATP and 1 μΜ peptide substrate. The JAK1 and TYK2 assays contained 1 μΜ of the IRStide peptide (5FAM-KKSRGDYMTMQID) and the JAK2 and JAK3 assays contained 1 μΜ of the JAKtide pep10 tide (FITC-KGGEEEEYFELVKK). The assays were initiated by the addition of 20 nM JAK1,1 nM JAK2, 1 nM JAK3 or 1 nM TYK2 enzyme and were incubated at room température for three hours for JAK1,60 minutes for JAK2, 75 minutes for JAK3 or 135 minutes for TYK2. Enzyme concentrations and incubation times were optimized for each new enzyme preps and were modified sfightly over time to ensure 20%30% phosphorylation. The assays were stopped with a final concentration of 10 mM EDTA, 0.1 % Coat15 ing Reagent and 100 mM HEPES, pH=7.4. The assay plates were placed on a Caliper Life Science Lab
Chip 3000 (LC3000) instrument, and each well was sampled using appropriate séparation conditions to measure the unphosphorylated and phosphorylated peptide.
Table 1. Data for JAK Caliper™ Enzyme assay at 1 mM ATP.
Structure Example JAK1 IC50 (nM) JAK2 IC50 (nM) JAK3 IC5Q (nM) Tyk2 ICso (nM)
102
οΛος 'χ'-' 0 0 F N H 1 22 381 >9220 1020
O x.·...../......™ xQ 2 29 803 >10000 1250
O rxii CQ 3 14 542 >10000 479
.....O......Mo ' Λ ° Y M-γ l 4B 6 607 >10000 965
XJ > A ° VX VQ s 4A 18 1400 >10000 2710
H ,? _ N /V ôo 5 6 313 8090 878
103
.. .n'AVOo ÔQ 6 28 933 >10000 2380
Ο An ° vV I V w “ 7A 31 2020 >10000 5240
>.....Ο......Y Ύυ Ν 7B 16 750 >10000 2440
Η ο °Αη IL JO H ™ H 8 3 700 >10000 260
i I H O ... ..□ ° w· w 9 231 5630 >10000 6670
A> 10 1030 7180 >10000 >10000
104
.... Ο/Υ 11 29 574 5950 2040
Η Ρ Ία h 03 ’ ' 12 6 413 >9670 770
Η J-^r ^ο·Ύ Ύ 13 5 177 8840 323
ΖΫ 14 42 1200 >10000 1870
γ-Υ<° ?Υ_ Ϋ, ν Μ Η 15 6 597 >10000 4910
^,Ο <Γο ti» ™ Η 16 51 1100 >10000 1780
105
H H Y°ZQ W N H 17A 5 308 >10000 337
Aq CQ x- 17B 13 434 >9770 1120
//V ° o Ύ 18 46 1080 >10000 7380
y2...... VO 19 34 1150 >10000 2030
-Y ° n Y ÇQ N H 20 4 171 5500 332
h O r-YS? Λ Y 21 1 52 3120 365
106
A 22 22 412 >10,000 1190
AV w 23 8 551 >10000 565
-A w A 27 17 987 >10000 1970
N A 24 241 3370 >10000 7870
F p A γ-α^< ^....O <To A 25 9 373 >10000 713
_cA □ <?% ^hT' Ά 26 6 88 1880 358
107
-/G Μ.......... ά? 28 5 179 5270 444
29 17 372 >9930 1100
nz>c^ ^N' dx N H 30 9 220 >6710 553
.....o-7 ào 31 67 946 >10000 3610
Cq 32 14 426 >10000 1460
□O’C'O/ ^N F ÔX Η H 33 5 161 6570 582
108
34 B 9 309 >10000 840
34A 37 801 >10000 3280
ύο N H 35 106 3760 >10000 >10000
^xr: □ M 36 4 85 3190 242
/ / O O ôx ™ H 37 6 569 >8880 418
μ —f 0 0 N Ô3 38 4.6 512 >10000 546
109
HWB INF alpha induced STAT3 phosphorylation Assay
Test articles were prepared as 30 mM stocks in 100% DMSO, and then diluted to 5 mM, A 10point 2,5 dilution sériés was created in DMSO with a top concentration of 5 mM, Further dilution was done by adding 4 μ|_ of the above test article solutions into 96 μΙ_ of PBS with a top concentration of 200 μΜ.
To a 96-well polypropylene plate (VWR 82007-292) 90 μΙ of HWB was added per well, followed by addition of 5 μΙ test article solutions prepared above to give a top concentration of 10 μΜ, The plate was mixed and incubated for 45 minutes at 37°C. To each well was added 5 μΙ of human IFN alpha (Universal Type I IFN, R&D Systems #11200-2; final concentration of 5000U/ml) or D-PBS (unstimulated control), mixed and incubated 15 minutes at 37°C. The reaction was quenched by adding Lyse/Fix Buffer [BD Phosflow 5x Lyse/Fix Buffer (BD #558049)] to ali wells at 1000 μΙ/well and incubated for 20 minutes at 37°C; after washing with FACS buffer [D-PBS (Invitrogen cat# 14190) containing 0.1 % BSA and 0.1% sodium azide], 400 pl ice cold 90% methanol/water was added to each well and incubated on ice for 30 minutes. One more wash was done with cold FACS buffer and ail samples were finally resuspended in 250 μΙ/well of the desired Alexa Fluor 647 conjugated anti-phospho-STAT3 (pY705) antibody (BD #557815) at 1:125 dilution in FACS buffer. After overnight incubation at 4 degree ail the samples were transferred into a 96-well polypropylene U-bottom plate (Falcon #353077) and checked by flow cytometry machine. ICso values obtained for examples 1 to 9, 11-23, 25-38 were in the range of 22 to 2610 nM.
Canine în w&üT-cell prolifération assay
I ΙΟ
T-cell activation play s a key rôle in a variety of inflammatory and autoimmune disorders as well as asthma, allergies and pruritus. Since T-cell activation can, in part, can be triggered by cytokines that signal through the JAK-STAT pathway, a JAK inhibitor could be effective against such diseases involving aberrant T-cell activation.
Methods: Canine whole blood was collected in sodium heparin tubes from 29 beagle dogs and 23 mixed breed dogs. Whole blood (20 pL) was plated in 96-well plates (Costar 3598) with 180 pL of medium (RPMI 1640, Gibco #21870-076, with 1% heat inactivated fêtai bovine sérum, Gîbco #10082-39, 292 pg/ml L-glutamine, Gibco #250030-081, 100 u/ml penicillin and 100pg streptomycin per ml, Gibco #15140-122) contaîning vehicle control or test compound (0,001 to 10 μΜ), concanavalrn A (ConA; 1 pg/ml, Sigma C5275), and canine interleukrn-2 (IL-2; 50 ng/ml, R&D Systems 1815-CL/CF). Wells containing whole blood, medium with vehicle control and no ConA or IL-2 were used as background controls. Plates were incubated at 37° C for 48 hours. Tritiated thymidine, 0.4 pCi/well (Perkin Elmer, Net027A005MC), was added for 20 additional hours. Plates were frozen and then thawed, washed and filtered using a Brandel MLR-96 cell harvester and pre-wet filter mats (Wallac 1205-401, Perkin Elmer). Filters were dried at 60° C for one hour (Précision 16EG convection oven) and placed into filter sample bags (Wallac 1205-411, Perkin Elmer) with 10 mL of scintillant (Wallac 1205-440, Perkin Elmer). Sealed filters were counted on a LKB Wallac 1205 Betaplate liquid scintillation counter. Data were collected via Gterm Betaplate program v1.1 and transformed into percent inhibition, calculated using the following formula:
100[(Mean Drug Treatment cpm) — (Mean BCK cpm)] — — 1 χ [(Mean Non Drug Treatment cpm) — (Mean BCK cpm)]
100 = % Inhibition
Data were graphically displayed as percent inhibition using GraphPad Prison 4.0, and ICso curves were fitted using a point to point analysis.
ni
Example 38 had an ICao of 48.5 nM in this assay. This data suggests that the compounds of the présent invention are effective in inhibiting T-ceJI prolifération, a key feature in diseases resulting from
JAK dysrégulation.
112

Claims (2)

  1. What is claimed is:
    1. A compound of formula l having the structure:
    or a pharmaceutically acceptable sait thereof, wherein
    R1 is hydrogen or C1-C4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, hydroxy, methoxy, amino, CFs, and Ca-Ce
    10 cycloalkyl;
    R2 and R3 are each independently hydrogen, deuterium, C-i-Ce linear or branched chain alkyl, C3Ce cycloalkyl, Ci-Cs linear or branched chain perfiuoroalkyl, Ci-Ce linear or branched chain alkoxy, Ci-Ce linear or branched chain perfluoroalkoxy, halogen, cyano, hydroxyl, amino, carboxy, aminocarbonyl, aryl, 15 heteroaryl, (aryi)Ci-Ce linear or branched chain alkyl, (heteroaryl)Ci-Ce linear or branched chain alkyl, (heterocyclic)Ci-C<> linear or branched chain alkyl, (Ci-Ce linear or branched chain alkyljaryl, (Ci-Cs linear or branched chain alkyl)heteroaryl, (Ci-Ce linear or branched chain alkyl)heterocyclic, (C1-C6 linear or
    113 branched chain alkoxyl)carbonyl, (Ci-Ce linear or branched chain alkyl)amino-carbonylamino, or (Ci-Ce linear or branched chain alkyljaminocarbonyl;
    R4 is selected from hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, Ci-Cb linear or branched chain perfluoroalkyl, aryl, and alkylaryl;
    X is selected from -NH- and —CRaRb-, where (a) Ra and Rb are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, Ca-Ce cycloalkyl, aryl, (aryl)Ci-C6 linear or branched chain alkyl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, (heteroaryl)Ci-Cs linear or branched chain alkyl, (heterocyclic)Ci-C6 linear or branched chain alkyl, or (b) Ra and Rb together form a chain comprising — (CRcRd)/-, where Rc and Rd are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, CF3, hydroxyl, CONH2, or SO2CH3;
    Y rs -A-R5, where A rs a bond, -(CH2)k- or -(CD2)k- and R5 is Ci-Ce linear or branched chain alkyl, Ca-Ce cycloalkyl, aryl, or -NRaRb, or is an unsaturated, saturated or partially saturated monocyclic or brcyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Cb linear or branched chain alkyl, CN, hydroxyl, CF3, -ORe, -NReRf, -S(O)PRe and C3-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and SO2CH3, where (a) Ra· and Rb- are independently hydrogen, deuterium, Ci-Ce linear ll4 or branched chain alkyl, C3-Cs cycloalkyl, aryl, (Ci-Cs linear or branched chain alkyljaryl, heteroaryl, or (Ci-Ce linear or branched chain alkyljheteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc·, or (b) Ra- and Rb· together form a chain comprising —(CRc-Rtr)/-, where Rc- and R<r are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, aryl, (Ci-Cë linear or
    5 branched chain alkyljaryl, heteroaryl, (Ci-Ce linear or branched chain alkyljheteroaryl, halo, CN, hydroxyl, CF3, CONH2, --ORb, NReRt, or -S(O)PRe; where Re and Rf are independently hydrogen, deuterium, C1Cs linear or branched chain alkyl, or C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2;
    /is 2, 3, 4 or 5; /ris 1, 2; 3, or4; p is 0, 1 or 2; and, n is 1 or 2.
    15 2, The compound of claim 1 which is a compound of formula IA having the structure:
    or a pharmaceutically acceptable sait thereof, wherein Y is -A-R5, where A is a bond, --(CFLJk- or -(CD2)k- and R5 is Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, or -NRaRb·, or is an un-
    H5 saturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Cb linear or branched chain alkyl, CN, hydroxyl, CF3, --ORe, -NRBRf, -S(O)PRe and Cs-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and SO2CH3, where (a) Ra· and Rb- are independently hydrogen, deuterium, Ci-Cb linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (Ci-Cb linear or branched chain alky!)aryI, heteroaryl, or (Ci-Cb linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc-, or (b) Ra- and Rb· together form a chain comprising —(CRcRd·)/-, where Rc· and R<j· are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, aryl, (Ci-Cb linear or branched chain alkyljaryl, heteroaryl, (Ci-Cb linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2, —ORa, -NReRf, or -S(O)PRa; where Ra and Rr are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or Cs-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONHîj/ts 2, 3, 4 or 5; Aris 1,2; 3, or 4; and, p is 0, 1 or 2.
    3. A compound of claim 2 wherein A is a bond and R5 is a C1-C6 linear or branched chain alkyl, C3-C6 cycloalkyl or aryl.
    116
    4. A compound of claim 2 wherein A is a bond or --(CHî)^-, and Rs is Ca-Ce cycloalkyl wherein said C3-C6 cycloalkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, Ci-Cs linear or branched chain alkyl, and CN where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo,
    5 CN, hydroxyl, CONH2, and SO2CH3; where Ais 1, 2, or 3.
    5. A compound of claim 2 wherein A is a bond or -(CH2)k--, and R5 is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containîng a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen,
    10 and sulfur, wherein said alkyl, Ca-Cs cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyl, CN, hydroxyl, CF3, --NRa Rb·, -ORe, -S(O)PRe and Ca-Cs cycloalkyl: where Ais 1, 2, or 3,
    15 6. The compound of daim 1 which is a compound of formula IB having the structure:
    IB
    117 or a pharmaceutically acceptable sait thereof, wherein (a) Ra- and Rb· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, CaCe cycloalkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, or (Ci-Ce linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rcj (b) Ra· and Rb- together form a chain comprising —(CRcRd )/-, where Rc· and Rd· are independently hydrogen, deuterium, Ci-Cs linear or branched chain alkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2, --ORe, NRaRf, or --S(O)PRe; where Re and Rt are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consîsting of halo, CN, hydroxyl, CF3, and CONH2; or, (c) Ra· and Rb· together form an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyl, CF3, --NRa Rt>·, --OR0, -S(O)PRe and C3-C6 cycloalkyl; /is 2, 3, 4 or 5; and, p is 0, 1 or 2.
    7. The compound of cfaim 1 which îs a compound of formula IC having the structure:
    R.· or a pharmaceutically acceptable sait thereof, wherein (a) Ra- and Rb· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, C3Ce cycloalkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, or (Ci-Cb linear or branched
    5 chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc;
    (b) Ra· and Rb· together form a chain comprising — (CRc Rdj/-, where Rc· and Rd· are independently hydrogen, deuterium, Ci-Cs linear or branched chain alkyl, aryl, (Ci-Ce linear or branched chain alkyl)aryl, heteroaryl, (Ci-Cb linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CFa, CONH2, -ORe, -
    10 NReRr, or -S(O)PRe; where RB and Rt are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, or C3-C« cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; or, (c) Ra· and Rb· together form an unsaturated, saturated or partially saturated monocyclic or bicy-
    15 clic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic
    1I9 ring structure is further optionally substituted with one or more substituents selected from the group con sisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyl, CFa, --NRa'Rb·, --0Re, S(0)PRe and Ca-Ce cycloalkyl; jis 2, 3, 4 or 5; and, p is 0, 1 or 2,
    5 8. The compound of claim 1 which is a compound of formula ID having the structure:
    or a pharmaceutically acceptable sait thereof, wherein
    Y is -AR5, where A is a bond or --(CHîJn-, and R5 is Ci-Ce linear or branched chain alkyl, Ca-Ce
    10 cycloalkyl, aryl, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, Ca-Ce cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, Ci-Ce linear or branched chain alkyl, CN, hydroxyl, CF3, -NRaRt»·,
    15 -ORe, —S(O)pRe and C3-C6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and SO2CH3, where (a) Ra· and Rb· are independently hydrogen, deuterium, Ci-Ce linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (arylJCi-Ce linear or branched chain alkyl, heteroaryl, (Ci-Ce linear or branched chain al-
    120 kyl)heteroaryl, (heteroarylJCi-Ce lînear or branched chain alkyl, (heterocyclic)Ci-Cs lînear or branched chain alkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more Rc, or (b) Ra· and Rb· together form a chain comprising —(CRcRdj/-, where Rc· and Rd are independently hydrogen, deuterium, Ci-Ce lînear or branched chain alkyl, aryl, (Ci-Ce lînear or branched chain alkyljaryl, heteroaryl, (Ci-Ce linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONHz, -ORe, NReRf, or -S(O)PRe; where Re and Rt where are independently hydrogen, deuterium, Ci-Cs linear or branched chain alkyl, or C3-Ce cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; y is 2, 3, 4 or 5; kis 1, 2, or 3; and, p is 0, 1 or 2,
    9, The compound of claim 1 selected from the group consisting of: 4-cyano-N-{cis-3-[methyl(7H-pyrroio[2,3-d]pyrimidin-4-yl)aminoJcyclobutyl)pyridine-2-sulfonamide; 2,2,2-trifl uoro- N-{cis-3-[methyl(7 H-pyrrolo[2,3 - d ] py ri m idîn-4-yl)amino]cyclobuty I}ethanesulfonamide;
  2. 2-methyl·N-{cis-3-[methyl(7l·l-pyrΓOlo[2t3-d]pyrimidin-4-yl)amino]cyclobutyl}-pΓopaπe-1sulfonamide;
    N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)aminoîcyclobutyl}propane-1-sulfonamide,·
    1-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methanesulfonamide;
    N-{ciS'3-[(butylsu1fonyl)methyl]cyclobutyl}-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine; 1-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin~4-yl)amino]cyclobutyl}-azetidine-3sulfonamide;
OA1201500328 2013-02-22 2014-02-11 Pyrrolo [2, 3 - D]pyrimidine derivatives as inhibitors of Janus-Related Kinases (JAK). OA17460A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/767,947 2013-02-22

Publications (1)

Publication Number Publication Date
OA17460A true OA17460A (en) 2016-12-22

Family

ID=

Similar Documents

Publication Publication Date Title
US9549929B2 (en) Pyrrolo[2,3-D]pyrimidine derivatives
EP3180344B1 (en) Pyrrolo[2,3-d]pyrimidine derivatives useful for inhibiting janus kinase
ES2859510T3 (en) Pyridazinone fused tricyclic compounds useful for treating orthomyxovirus infections
ES2655971T9 (en) Aminopyrimidinyl compounds as JAK inhibitors
WO2019046318A1 (en) Spirocycle compounds and methods of making and using same
EP3946606A1 (en) Bicyclic jak inhibitors and uses thereof
WO2022217118A1 (en) Pyrimidine based ras modulators and uses thereof
OA17460A (en) Pyrrolo [2, 3 - D]pyrimidine derivatives as inhibitors of Janus-Related Kinases (JAK).