WO2023091707A1 - Bicyclic inhibitors of jak and methods of use - Google Patents

Bicyclic inhibitors of jak and methods of use Download PDF

Info

Publication number
WO2023091707A1
WO2023091707A1 PCT/US2022/050466 US2022050466W WO2023091707A1 WO 2023091707 A1 WO2023091707 A1 WO 2023091707A1 US 2022050466 W US2022050466 W US 2022050466W WO 2023091707 A1 WO2023091707 A1 WO 2023091707A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
salt
membered heterocycle
carbocycle
Prior art date
Application number
PCT/US2022/050466
Other languages
French (fr)
Inventor
Robert Murray Mckinnell
Tom M. LAM
Philip GERKEN
Cameron Smith
Christina OWENS
Luke BORALSKY
Original Assignee
Theravance Biopharma R&D Ip, Llc
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 Theravance Biopharma R&D Ip, Llc filed Critical Theravance Biopharma R&D Ip, Llc
Publication of WO2023091707A1 publication Critical patent/WO2023091707A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • Asthma is a chronic disease of the airways for which there is no prevention or cure.
  • the disease is characterized by inflammation, fibrosis, hyper-responsiveness, and remodeling of the airways, all of which contribute to airflow limitation.
  • An estimated 300 million people worldwide suffer from asthma and it is estimated that the number of people with asthma will grow by more than 100 million by 2025.
  • asthma afflicts about 6 to 8% of the population, making it one of the most common chronic diseases in the country.
  • most patients can achieve control of asthma symptoms with the use of inhaled corticosteroids that may be combined with a leukotriene modifier and/or a long-acting beta agonist, there remains a subset of patients with severe asthma whose disease is not controlled by conventional therapies.
  • Severe persistent asthma is defined as disease that remains uncontrolled on high doses of inhaled corticosteroids. While patients with severe asthma are estimated to account for approximately 5% of all asthma patients, they have a high risk of morbidity and mortality and are responsible for a disproportionate share of health care resource utilization among patients with asthma. There remains a need for novel therapies to treat these patients.
  • Cytokines are intercellular signaling molecules which include chemokines, interferons, interleukins, lymphokines, and tumor necrosis factor. Cytokines are critical for normal cell growth and immunoregulation but also drive immune-mediated diseases and contribute to the growth of malignant cells. Elevated levels of many cytokines have been implicated in the pathology of asthma inflammation. For example, antibody-based therapies targeted at interleukins (IL)-5 and 13 have been shown to provide clinical benefit in subsets of severe asthma patients.
  • IL interleukins
  • Cytokines implicated in asthma inflammation which signal through the JAK-STAT pathway include IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-11 , IL-13, IL-23, IL-31, IL-27, thymic stromal lymphopoietin (TSLP), interferon-y (IFNy) and granulocyte-macrophage colony-stimulating factor (GM-CSF).
  • JAK-STAT Signal Transducer and Activator of Transcription
  • Cytokines implicated in asthma inflammation which signal through the JAK-STAT pathway include IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-11 , IL-13, IL-23, IL-31, IL-27, thymic stromal lymphopoietin (TSLP), interferon-y (IFNy) and granulocyte-macrophage colony-sti
  • the JAK family comprises four members, JAKI, JAK2, JAK3, and tyrosine kinase 2 (TYK2). Binding of cytokine to a JAK-dependent cytokine receptor induces receptor dimerization which results in phosphorylation of tyrosine residues on the JAK kinase, effecting JAK activation. Phosphoiylated JAKs, in turn, bind and phosphorylate various STAT proteins which dimerize, internalize in the cell nucleus and directly modulate gene transcription, leading, among other effects, to the downstream effects associated with inflammatory disease.
  • the JAKs usually associate with cytokine receptors in pairs as homodimers or heterodimers.
  • Specific cytokines are associated with specific JAK pairings.
  • Each of the four members of the JAK family is implicated in the signaling of at least one of the cytokines associated with asthma inflammation. Consequently, a chemical inhibitor with pan-activity against all members of the JAK family could modulate a broad range of pro-inflammatory pathways that contribute to severe asthma.
  • Chronic obstructive pulmonary disease COPD
  • cystic fibrosis CF
  • pneumonitis interstitial lung diseases (including idiopathic pulmonary fibrosis), acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, and sarcoidosis are also respiratory tract diseases in which the pathophysiology is believed to be related to JAK-signaling cytokines.
  • Local administration of a JAK inhibitor to the lungs by inhalation offers the potential to be therapeutically efficacious by delivering a potent anti-cytokine agent directly to the site of action, limiting systemic exposure and therefore limiting the potential for adverse systemic immunosuppression.
  • the need remains for a potent JAK inhibitor suitable for local administration to the lungs for treatment of respiratory disease.
  • JAK-signaling cytokines also play a major role in the activation of T cells, a sub-type of immune cells that is central to many immune processes.
  • Pathological T cell activation is critical in the etiology of multiple respiratory diseases.
  • Autoreactive T cells play a role in bronchiolitis obliterans organizing pneumonia (also termed COS). Similar to COS, the etiology of lung transplant rejections is linked to an aberrant T cell activation of the recipient s T cells by the transplanted donor lung.
  • Lung transplant rejections may occur early as Primary Graft Dysfunction (PGD), organizing pneumonia (OP), acute rejection (AR) or lymphocytic bronchiolitis (LB) or they may occur years after lung transplantation as Chronic Lung Allograft Dysfunction (CLAD).
  • CLAD was previously known as bronchiolitis obliterans (BO) but now is considered a syndrome that can have different pathological manifestations including BO, restrictive CLAD (rCLAD or RAS) and neutrophilic allograft dysfunction.
  • Chronic lung allograft dysfunction (CLAD) is a major challenge in longterm management of lung transplant recipients as it causes a transplanted lung to progressively lose functionality (Gauthier et al., Curr. Transplant. Rep. 2016, 3(3), 185—191).
  • CLAD is poorly responsive to treatment and therefore, there remains a need for effective compounds capable of preventing or treating this condition.
  • JAK-dependent cytokines such as IFNy and IL-5 are up-regulated in CLAD and lung transplant rejection (Berastegui et al., Clin. Transplant. 2017, 31, el2898).
  • high lung levels of CXCR3 chemokines such as CXCL9 and CXCL10, which are downstream of JAK-dependent IFN signaling, are linked to worse outcomes in lung transplant patients (Shino et al, PLOS One, 2017, 12 (7), eOl 80281).
  • JAK inhibition has been shown to be effective in kidney transplant rejection (Vicenti et al., American Journal of Transplantation 2012, 12, 2446-56). Therefore, JAK inhibitors have the potential to be effective in treating or preventing lung transplant rejection and CLAD. Similar T cell activation events as described as the basis for lung transplant rejection also are considered the main driver of lung graft-versus- host disease (GVHD) which can occur post hematopoietic stem cell transplants. Similar to CLAD, lung GVHD is a chronic progressive condition with extremely poor outcomes and no treatments are currently approved.
  • JAK inhibitors have been associated with adverse immunosuppression resulting from significant systemic exposure from oral administration.
  • the present disclosure provides these and other related advantages.
  • One objective of the present disclosure is to deliver a potent JAK inhibitor locally with minimal systemic exposure in order to address any unintended and unwanted systemic side effects of JAK inhibition during treatment.
  • the present disclosure provides inhaled, long-acting and lung-selective JAK inhibitors for the treatment of a respiratory disease, such as asthma, chronic obstructive pulmonary disease, or lung transplant rejection.
  • a respiratory disease such as asthma, chronic obstructive pulmonary disease, or lung transplant rejection.
  • Compounds of the present disclosure may be used as a monotherapy or co-dosed with other therapies, whether delivered by inhalation, orally, intravenously, subcutaneously, or topically.
  • R 1 is independently selected at each occurrence from halogen, -OH, -O(C 1-4 alkyl), -O(C 1-4 haloalkyl), and C 1-4 alkyl;
  • R 2 is selected from hydrogen, halogen, and C 1-4 alkyl
  • R 3 is independently selected at each occurrence from halogen and C 1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond; when is a double bond, then A is CR 4 R 3 ; when is a triple bond, then A is CR 6 ;
  • R 4 and R 5 are each independently selected from hydrogen and R A ; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from hydrogen and R A ;
  • R 10 is independently selected at each occurrence from hydrogen; and C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 1- to 6-membered heteroalkyl, Co- 3 alkyl-(C 3-12 carbocycle), and Co- 3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN,
  • R 11 and R 12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R 10 ;
  • R 7 is hydrogen or C 1-6 alkyl.
  • R 1 is independently selected at each occurrence from halogen, -OH, -O(C 1-4 alkyl), -O( C 1-4 haloalkyl), and C 1-4 alkyl;
  • R 2 is selected from hydrogen, halogen, and C 1-4 alkyl
  • R 3 is independently selected at each occurrence from halogen and C 1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond; when is a double bond, then A is CR 4 R 5 ; when is a triple bond, then A is CR 6 ;
  • R 4 and R 5 are each independently selected from hydrogen and R A ; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from hydrogen and R A ;
  • R 11 and R 12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R 10 .
  • n 0.
  • a compound of Formula (I) or (I’) may be a compound of Formula (I-A): (I-A), or a pharmaceutically acceptable salt thereof.
  • R 1 may independently be selected at each occurrence from halogen, -OH, and -CH 2 CH 3 .
  • a compound of Formula (I) or (I’) may be a compound of Formula (I-D): (I-D), or a pharmaceutically acceptable salt thereof.
  • R 4 is selected from R A and R 5 is selected from hydrogen and CH 3 ;
  • R 4 is selected from hydrogen and CH 3 and R 5 is selected from R A ;
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from R A ;
  • halogen -NH2, -NHCH 3 ,
  • R A is independently selected at each occurrence from:
  • a compound or salt of Formula (I), (I’), (I -A) or (I-D) may be a double bond, wherein A is CR 4 R 5 .
  • a compound of Formula (I) may be a compound of Formula (I-E): (I-E), or a pharmaceutically acceptable salt thereof.
  • R 4 is selected from R A and R 5 is selected from hydrogen and CH 3 ;
  • R 4 is selected from hydrogen and CH 3 and R 5 is selected from R A ;
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R A is independently selected at each occurrence from:
  • each 4- to 10-membered heterocycle in R A is independently optionally substituted with one or more substituents selected from -OCH 3 , and C 1-4 alkyl;
  • a compound or salt of Formula (I), (I’), (I-A) or (I-D) may be a triple bond, wherein A is CR".
  • a compound of Formula (I) may be a compound of Formula (I-F): (I-F), or a pharmaceutically acceptable salt thereof
  • R 6 is selected from: C 1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R 10 ) 2 , C 3 -10 carbocycle, and 4- to 10-membered heterocycle; and C 3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C 3 -10 carbocycle and 4- to 10-membered heterocycle in R 6 is independently optionally substituted with one or more substituents selected from halogen, -OR 10 , -N(R 10 ) 2 , and C 1-4 alkyl; and
  • R 10 is independently selected at each occurrence from hydrogen and C 1-4 alkyl.
  • R 2 may be selected from hydrogen and halogen, such as R 2 is hydrogen. In some embodiments, R 2 is F.
  • R 2 is selected from hydrogen and F
  • R 4 is selected from R A and R 5 is selected from hydrogen and CH 3 : R 4 is selected from hydrogen andCH 3 and R 5 is selected from R A ; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from R A ;
  • R A is independently selected at each occurrence from:
  • R 2 is selected from hydrogen and F;
  • R 2 is selected from hydrogen and F
  • R 2 is selected from hydrogen and F
  • R 2 is selected from hydrogen and F
  • the present disclosure provides a substantially pure stereoisomer of a compound or salt disclosed herein, such as a compound or salt of Formula (I), (I’), (I-A), (I-D), (I-E) or (I-F), or a compound disclosed in Table 1.
  • the stereoisomer may be provided in at least 90% enantiomeric excess.
  • the present disclosure provides a compound selected from Table 1, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises one or more additional therapeutic agents.
  • the pharmaceutical composition may be formulated for inhalation.
  • the present disclosure provides a method of inhibiting JAK, comprising contacting JAK with an effective amount of a compound or salt disclosed herein.
  • the present disclosure provides a method of treating a JAK-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein.
  • the disease or condition may be a respiratory disease.
  • the present disclosure provides a compound or salt disclosed herein for use in treating a respiratory disease.
  • the present disclosure provides the use of a compound or salt disclosed herein for the manufacture of a medicament for treating a respiratory disease.
  • the respiratory disease may be selected from asthma, chronic obstructive pulmonary disease, cystic fibrosis, pneumonitis, idiopathic pulmonary fibrosis, acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, sarcoidosis, an eosinophilic disease, a helminthic infection, pulmonary arterial hypertension, lymphangioleiomyomatosis, bronchiectasis, an infiltrative pulmonary disease, drag-induced pneumonitis, fungal induced pneumonitis, allergic bronchopulmonary aspergillosis, hypersensitivity pneumonitis, eosinophilic granulomatosis with polyangiitis, idiopathic acute eosinophilic pneumonia, idiopathic chronic eosinophilic pneumonia, hypereosinophilic syndrome, Loffler syndrome, bronchiolitis obliterans organizing pneumonia, lung graf
  • the respiratory disease is selected from asthma and chronic obstructive pulmonary disease.
  • the asthma may be selected from T2-dominant (eosinophilic) asthma and non-T2-dominant (non- eosinophilic) asthma.
  • the present disclosure provides a method of treating lung transplant rejection in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein.
  • the present disclosure provides a compound or salt disclosed herein for use in treating lung transplant rejection.
  • the present disclosure provides the use of a compound or salt disclosed herein for the manufacture of a medicament for treating lung transplant rejection.
  • the lung transplant rejection may be selected from primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction.
  • the lung transplant rejection is acute lung transplant rejection.
  • the lung transplant rejection is chronic lung allograft dysfunction.
  • the lung transplant rejection is selected from bronchiolitis obliterans, restrictive chronic lung allograft dysfunction, and neutrophilic allograft dysfunction.
  • Any of the subject methods may further comprise administering a second therapeutic agent.
  • a compound or salt disclosed herein may be administered by inhalation.
  • C x-y or “C x -C y ” when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl, is meant to include groups that contain from x to y carbons in the chain.
  • C x.y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, that contain from x to y carbons in the chain.
  • Alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including linear and branched alkyl groups.
  • An alkyl group may contain from one to twelve carbon atoms (e.g., C1-12 alkyl), such as one to eight carbon atoms (C 1-8 alkyl) or one to six carbon atoms (C 1-6 alkyl).
  • Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, and decyl.
  • An alkyl group is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Haloalkyl refers to an alkyl group that is substituted by one or more halogens.
  • exemplary haloalkyl groups include trifluoromethyl, difluoromethyl, trichloro methyl, 2,2,2-trifluoroethyl, 1,2 -difluoroethyl, 3- bromo-2-fluoropropyl, and 1 ,2-dibromoethyl.
  • alkenyl refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkenyl groups, containing at least one double bond.
  • An alkenyl group may contain from two to twelve carbon atoms (e.g., C 2-12 alkenyl), such as two to eight carbon atoms (C 2-8 alkenyl) or two to six carbon atoms (C 2-6 alkenyl).
  • Exemplary alkenyl groups include ethenyl (i.e., vinyl), prop-l-enyl, but-l-enyl, pent-l-enyl, penta- 1, 4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Alkynyl refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkynyl groups, containing at least one triple bond.
  • An alkynyl group may contain from two to twelve carbon atoms (e.g., C 2-12 alkynyl), such as two to eight carbon atoms (C 2-8 alkynyl) or two to six carbon atoms (C 2-6 alkynyl).
  • Exemplary alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Alkylene or “alkylene chain” refers to substituted or unsubstituted divalent saturated hydrocarbon groups, including linear alkylene and branched alkylene groups, that contain from one to twelve carbon atoms (e.g., C 1-12 alkylene), such as one to eight carbon atoms (C1-8 alkylene) or one to six carbon atoms (C 1-6 alkylene).
  • Exemplary alkylene groups include methylene, ethylene, propylene, and n-butylene.
  • alkenylene and alkynylene refer to alkylene groups, as defined above, which comprise one or more carbon-carbon double or triple bonds, respectively.
  • alkylene, alkenylene or alkynylene chain can be through one carbon or any two carbons of the chain.
  • an alkylene, alkenylene, or alkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
  • Heteroalkyl refers to substituted or unsubstituted alkyl, alkenyl and alkynyl groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quatemized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkyl group has a chain length of 3 to 8 atoms.
  • Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl, heteroalkenyl or heteroalkynyl chain.
  • a heteroalkyl, heteroalkenyl, or heteroalkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Hetero alkylene refers to substituted or unsubstituted alkylene, alkenylene and alkynylene groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quatemized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8 -membered heteroalkylene group has a chain length of 3 to 8 atoms.
  • the points of attachment of the heteroalkylene, hetero alkenylene or heteroalkynylene chain to the rest of the molecule can be through either one heteroatom or one carbon, or any two heteroatoms, any two carbons, or any one heteroatom and any one carbon in the heteroalkylene, hetero alkenylene or heteroalkynylene chain.
  • a heteroalkylene, heteroalkenylene, or heteroalkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
  • Carbocycle refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is a carbon atom.
  • Carbocycle may include C 3 -10 monocyclic rings, C 2-6 bicyclic rings, C 2-6 spirocyclic rings, and C 6-12 bridged rings.
  • Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings.
  • the carbocycle is a C 2-6 aryl group, such as C 6-10 aryl.
  • the carbocycle is a C 2-6 cycloalkyl group.
  • the carbocycle is a C 2-6 cycloalkenyl group.
  • cycloalkenyl refers to a non-aromatic ring containing at least one double bond, wherein each atom of the ring is a carbon atom.
  • an aromatic ring e.g., phenyl
  • a saturated or unsaturated ring e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, are included in the definition of carbocycle.
  • Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl. Unless stated otherwise specifically in the specification, a carbocycle is optionally substituted by one or more substituents such as those substituents described herein.
  • Heterocycle refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms, for example 1, 2 or 3 heteroatoms selected from O, S and N. Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, 6- to 12-membered spirocyclic rings, and 6- to 12- membered bridged rings. Each ring of a bicyclic heterocycle may be selected from saturated, unsaturated, and aromatic rings. The heterocycle may be attached to the rest of the molecule through any atom of the heterocycle, valence permitting, such as a carbon or nitrogen atom of the heterocycle.
  • the heterocycle is a 5 - to 10-membered heteroaryl group, such as 5- or 6-membered heteroaryl.
  • the heterocycle is a 3- to 12-membered heterocycloalkyl group.
  • the heterocycle is a 3- to 12-membered heterocycloalkyl group (i.e. a saturated or partially unsaturated cyclic ring having one or more (e.g., 1 to 5) heteroatoms).
  • a heterocycle e.g., pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene.
  • heterocycles include pyrrolidinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, piperidinyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, thiophenyl, oxazolyl, thiazolyl, morpholinyl, indazolyl, indolyl, and quinolinyl.
  • a heterocycle is optionally substituted by one or more substituents such as those substituents described herein.
  • Heteroaryl refers to a 5- to 12-membered aromatic ring that comprises at least one heteroatom, such as 1, 2 or 3 heteroatoms, selected from O, S and N.
  • the heteroaryl ring may be selected from monocyclic or bicyclic — including fused, spirocyclic and bridged ring systems — wherein at least one of the rings in the ring system is aromatic.
  • the heteroatom(s) in the heteroaryl may optionally be oxidized.
  • One or more nitrogen atoms, if present, are optionally quatemized.
  • the heteroaryl may be attached to the rest of the molecule through any atom of the heteroaryl, valence permitting, such as a carbon or nitrogen atom of the heteroaryl.
  • heteroaryl groups include, but are not limited to, azepinyl, benzimidazolyl, benzisothiazolyl, benzisoxazolyl, benzofuranyl, benzothiazolyl, benzothiophenyl, benzoxazolyl, furanyl, imidazolyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, purinyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyridazolyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroquinolinyl, thiadiazolyl, thiazolyl, and thienyl groups. Unless stated otherwise,
  • a waved line drawn across a bond is used herein to denote where a bond disconnection or attachment occurs.
  • R a is attached to the para position of a fluorophenyl ring through a single bond. If R a is 2-pyridine as in , then R a may be depicted as “
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons or heteroatoms of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • heteroatoms such as nitrogen may have any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • R 11 and R 12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R 10 .
  • bivalent substituent groups are specified herein by their conventional chemical formulae, written from left to right, they are intended to encompass the isomer that would result from writing the structure from right to left, e.g., -CH 2 O- is also intended to encompass to -OCH 2 -.
  • “Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not.
  • an “optionally substituted” group may be either unsubstituted or substituted.
  • Compounds of the present disclosure also include crystalline and amorphous forms of those compounds, pharmaceutically acceptable salts, and active metabolites having the same type of activity, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, amorphous forms of the compounds, and mixtures thereof.
  • the compounds described herein may exhibit their natural isotopic abundance, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.
  • hydrogen has three naturally occurring isotopes, denoted (protium), 2 H (deuterium), and 3 H (tritium). Protium is the most abundant isotope of hydrogen in nature.
  • Enriching for deuterium may afford certain therapeutic advantages, such as increased in vivo half-life and/or exposure, or may provide a compound useful for investigating in vivo routes of drug elimination and metabolism.
  • isotopes that may be incorporated into compounds of the present disclosure include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 0, 35 S, 36 C1, and 18 F.
  • compounds of Formula (I) enriched in tritium or carbon-14 which can be used, for example, in tissue distribution studies; compounds of the disclosure enriched in deuterium especially at a site of metabolism, resulting, for example, in compounds having greater metabolic stability; and compounds of Formula (I) enriched in a positron emitting isotope, such as 11 C, 18 F, 15 O and 13 N, which can be used, for example, in Positron Emission Topography (PET) studies.
  • Isotopically-enriched compounds may be prepared by conventional techniques well known to those skilled in the art.
  • Certain compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms, the asymmetric centers of which can be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the carbon atoms in order to optimize the therapeutic activity of the compounds of the disclosure, e.g., to treat asthma, it may be desirable that the carbon atoms have a particular configuration (e.g., (R,R), (S,S), (S,R), or (R,S)) or are enriched in a stereoisomeric form having such configuration.
  • the compounds of the disclosure may be provided as racemic mixtures.
  • the disclosure relates to racemic mixtures, pure stereoisomers (e.g., enantiomers and diastereoisomers), stereoisomer-enriched mixtures, and the like, unless otherwise indicated.
  • pure stereoisomers e.g., enantiomers and diastereoisomers
  • stereoisomer-enriched mixtures and the like, unless otherwise indicated.
  • a chemical structure is depicted herein without any stereochemistry, it is understood that all possible stereoisomers are encompassed by such structure.
  • a particular stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the disclosure unless otherwise indicated, provided that the utility of the composition as a whole is not eliminated by the presence of such other isomers.
  • stereoisomers may be obtained by numerous methods that are known in the art, including preparation using chiral synthons or chiral reagents, resolution using chiral chromatography using a suitable chiral stationary phase or support, or by chemically converting them into diastereoisomers, separating the diastereoisomers by conventional means such as chromatography or recrystallization, then regenerating the original stereoisomer.
  • tautomer refers to each of two or more isomers of a compound that exist in equilibrium and which ready interconvert.
  • 1,2, 3 -triazole exists in two tautomeric forms:
  • pharmaceutically acceptable refers to a material that is not biologically or otherwise unacceptable when used in the subject compositions and methods.
  • pharmaceutically acceptable carrier refers to a material — such as an adjuvant, excipient, glidant, sweetening agent, diluent, preservative, dye, colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent or emulsifier — that can be incorporated into a composition and administered to a patient without causing unacceptable biological effects or interacting in an unacceptable manner with other components of the composition.
  • Such pharmaceutically acceptable materials typically have met the required standards of toxicological and manufacturing testing, and include those materials identified as suitable inactive ingredients by the U.S. Food and Drug Administration.
  • salts and “pharmaceutically acceptable salt” refer to a salt prepared from a base or an acid.
  • Pharmaceutically acceptable salts are suitable for administration to a patient, such as a mammal (for example, salts having acceptable mammalian safety for a given dosage regime). Salts can be formed from inorganic bases, organic bases, inorganic acids and organic acids.
  • a compound contains both a basic moiety, such as an amine, pyridine or imidazole, and an acidic moiety, such as a carboxylic acid or tetrazole, zwitterions may be formed and are included within the term “salt” as used herein.
  • Salts derived from inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, and zinc salts, and the like.
  • Salts derived from organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, A. A'-dibcnzylcthylcncdiaminc.
  • Salts derived from inorganic acids include salts of boric, carbonic, hydrohalic (hydrobromic, hydrochloric, hydrofluoric or hydroiodic), nitric, phosphoric, sulfamic and sulfuric acids.
  • Salts derived from organic acids include salts of aliphatic hydroxyl acids (for example, citric, gluconic, glycolic, lactic, lactobionic, malic, and tartaric acids), aliphatic monocarboxylic acids (for example, acetic, butyric, formic, propionic and trifluoro acetic acids), amino acids (for example, aspartic and glutamic acids), aromatic carboxylic acids (for example, benzoic, p-chlorobenzoic, diphenylacetic, gentisic, hippuric, and triphenylacetic acids), aromatic hydroxyl acids (for example, o- hydroxybenzoic, p-hydroxybenzoic, l-hydroxynaphthalene-2-carbox
  • a therapeutically effective amount refers to that amount of a compound described herein that is sufficient to affect treatment when administered to a subject in need thereof.
  • a therapeutically effective amount for treating asthma is an amount of compound needed to, for example, reduce, suppress, eliminate, or prevent one or more symptoms of asthma in a subject, or to treat the underlying cause of the asthma.
  • the therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the specific dose will vary depending on the particular compound chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the term “effective amount” refers to an amount sufficient to obtain a desired result, which may not necessarily be a therapeutic result.
  • an “effective amount” may be the amount needed to inhibit an enzyme.
  • treating refers to an approach for obtaining beneficial or desired results with respect to a disease, disorder, or medical condition (such as asthma) in a subject, including but not limited to the following: (a) preventing the disease or medical condition from occurring, e.g., preventing the reoccurrence of the disease or medical condition or prophylactic treatment of a subject that is pre-disposed to the disease or medical condition; (b) ameliorating the disease or medical condition, e.g., eliminating or causing regression of the disease or medical condition in a subject; (c) suppressing the disease or medical condition, e.g., slowing or arresting the development of the disease or medical condition in a subject; or (d) alleviating symptoms of the disease or medical condition in a subject.
  • “treating asthma” would include preventing pulmonary inflammation from occurring, ameliorating pulmonary inflammation, suppressing pulmonary inflammation, and alleviating the symptoms of asthma (for example, improved lung function tests). Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function (e.g., activity, expression, binding, protein-protein interaction) of a target protein (e.g., JAK). Accordingly, the terms “antagonist” and “inhibitor” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
  • selective inhibition refers to the ability of a biologically active agent to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
  • subject and “patient” refer to an animal, such as a mammal, for example a human.
  • the methods described herein can be useful in both human therapeutics and veterinary applications.
  • the subject is a mammal, and in some embodiments, the subject is human.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of Formula (I)).
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug is inactive when administered to a subject but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.
  • prodrug is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of an active compound are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound, and the like.
  • in vivo refers to an event that takes place in a subject's body.
  • in vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject.
  • in vitro assays encompass cell-based assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • the disclosure is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the disclosure includes compounds produced by a process comprising administering a compound of this disclosure to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the disclosure in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to a human
  • the present disclosure provides compounds that are capable of selectively binding to and/or modulating JAK.
  • the compounds modulate JAK by binding to or interacting with one or more amino acids and/or one or more metal ions. The binding of these compounds may disrupt JAK downstream signaling.
  • the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is independently selected at each occurrence from halogen, -OH, -O(C 1-4 alkyl), -O(C 1-4 haloalkyl), and C 1-4 alkyl;
  • R 2 is selected from hydrogen, halogen, and C 1-4 alkyl
  • R 3 is independently selected at each occurrence from halogen and C 1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond; when is a double bond, then A is CR 4 R 3 ; when a is a triple bond, then A is CR":
  • R 4 and R 5 are each independently selected from hydrogen and R A ; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from hydrogen and R A ;
  • R 11 and R 12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R 10 ;
  • R 7 is hydrogen or C 1-6 alkyl.
  • R 7 is hydrogen. In some embodiments, R 7 is C 1-6 alkyl. In some embodiments, R 7 is C 1-3 alkyl. In some embodiments, R 7 is -CH 3 .
  • n is 0 or 1, such as n is 0.
  • the compound of Formula (I) or (I’) is a compound of Formula (I-A): such as a compound of Formula (I-B) or (I-C): or a pharmaceutically acceptable salt thereof.
  • R 1 is independently selected at each occurrence from halogen, -OH, -O(C 1-2 alkyl), and C 1-2 alkyl. In some embodiments, R 1 is independently selected at each occurrence from halogen, -OH, and C 1-2 alkyl. In some embodiments, R 1 is independently selected at each occurrence from halogen, -OH, and -CH 2 CH 3 . In some embodiments, R 1 is independently selected at each occurrence from -OH, and -CH 2 CH 3 .
  • the compound of Formula (I) or (I’) is a compound of Formula (I-D): such as a compound of Formula (I-E) or (I-F): or a pharmaceutically acceptable salt thereof.
  • R 4 is selected from R A and R 5 is selected from hydrogen and CH 3 ;
  • R 4 is selected from hydrogen and CH 3 and R 5 is selected from R A ;
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from R A ;
  • R A is independently selected at each occurrence from:
  • a compound of Formula (I), (F), (I -A) or (I-D) is a double bond and A is CR 4 R 5 . In some embodiments, is a triple bond and A is CR ft .
  • R 4 is selected from R A and R 5 is hydrogen; or
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 4 is selected from R A and R 5 is selected from hydrogen and CH 3 ;
  • R 4 is selected from hydrogen and CH 3 and R 5 is selected from R A ;
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R A is independently selected at each occurrence from:
  • each 4- to 10-membered heterocycle in R A is independently optionally substituted with one or more substituents selected from -OCH 3 , and C 1-4 alkyl;
  • R 4 is selected from R A and R 5 is hydrogen; or
  • R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R A is independently selected at each occurrence from:
  • each 4- to 10-membered heterocycle in R A is independently optionally substituted with one or more substituents selected from -OCH 3 , and C 1-4 alkyl;
  • R 4 is selected from:
  • R 5 is hydrogen
  • R 10 is independently selected at each occurrence from hydrogen; and C 1-4 alkyl and 3- to 6-membered heterocycle.
  • R 6 is selected from R A ;
  • R 6 is selected from: C 1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R 10 ) 2 , C 3 -10 carbocycle, and 4- to 10-membered heterocycle; and C 3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C 3 -10 carbocycle and 4- to 10-membered heterocycle in R 6 is independently optionally substituted with one or more substituents selected from halogen, -OR 10 , -N(R 10 ) 2 , and C 1-4 alkyl; and
  • R 10 is independently selected at each occurrence from hydrogen and C 1-4 alkyl.
  • R 6 is selected from:
  • -CH 2 (5- to 7-membered heterocycle), optionally substituted with -CH 3 ;
  • R 2 is selected from hydrogen and halogen. In some embodiments, R 2 is selected from hydrogen and F. In some embodiments, R 2 is hydrogen. In some embodiments, R 2 is F.
  • R 2 is selected from hydrogen and F
  • R 4 is selected from R A and R 5 is selected from hydrogen and -CH 3 : R 4 is selected from hydrogen and -CH 3 and R 5 is selected from R A ; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from R A ;
  • R A is independently selected at each occurrence from:
  • R 2 is selected from hydrogen and -F
  • the present disclosure provides a compound of Formula (I-D): or a pharmaceutically acceptable salt thereof, wherein:
  • R 2 is selected from hydrogen and F; represents a double bond or triple bond; when is a double bond, then A is CR 4 R 5 ; when is a triple bond, then A is CR 6 ;
  • R 4 is selected from R A and R 5 is hydrogen; or R 4 and R 5 are taken together with the carbon atom to which they are attached to form a C 3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more R A ;
  • R 6 is selected from R A ;
  • R A is independently selected at each occurrence from:
  • the present disclosure provides a compound of Formula (I-D): or a pharmaceutically acceptable salt thereof, wherein:
  • R 2 is selected from hydrogen and F
  • references herein to “a compound of Formula (I)” implicitly also include the compound of Formula (I’), (I-A), (I-B), (I-C), (I-D), (I-E), and (I-F).
  • the skilled person will understand that the context of a passage may exclude one or more of Formulas (I’), (I-A), (I-B), (I-C), (I-D), (I-E), and (I-F), for example, if the passage provides an embodiment of a particular variable (e.g., R 1 ) different than what is depicted in the formula.
  • a compound of Formula (I) is provided as a substantially pure stereoisomer.
  • the stereoisomer is provided in at least 80% enantiomeric excess, such as at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or at least 99.9% enantiomeric excess.
  • reaction times and conditions are intended to be approximate, e.g. taking place at about atmospheric pressure within a temperature range of about -10 °C to about 110 °C over a period of about 1 to about 24 hours; reactions left to run overnight average a period of about 16 hours.
  • a compound of Formula Ih may be prepared according to Scheme 1.
  • acyl chloride la can be coupled to a suitably protected imidazole to provide lb, which can be reacted with hydrazine monohydrate to form the pyrazole ring of 1c, optionally with one or more protecting group manipulations following the ring formation.
  • Installation of a substituted phenyl ring may proceed via a Suzuki reaction using boronic ester Id to afford intermediate le. Formation of bromoimidazole If can be followed by another Suzuki reaction with boronic ester 1g, and optionally one or more coupling reactions and/or protecting group manipulations, to provide an alkene of Formula Ih.
  • a compound of Formula 2e may be prepared according to Scheme 2.
  • imidazole 2a can be iodinated to provide diiodo imidazole 2b.
  • Selective removal of one iodo group to give 2c can be followed by a Sonogashira coupling with alkyne 2d, and optionally one or more coupling reactions and/or protecting group manipulations, to provide an alkyne of Formula 2e.
  • a compound of the present disclosure for example, a compound of a formula given in Table 1, is synthesized according to one of the general routes outlined in Schemes 1 and 2, Examples 1-9, or by methods generally known in the art.
  • exemplary compounds may include, but are not limited to, a compound or salt thereof selected from Table 1.
  • exemplary compounds may include, but are not limited to, a compound or salt thereof selected from:
  • the present disclosure provides a method of inhibiting JAK signaling, comprising contacting a cell with an effective amount of a compound disclosed herein, such as a compound of Formula (I).
  • the present disclosure provides a method of inhibiting a JAK protein, comprising contacting the JAK protein with an effective amount of a compound disclosed herein. Inhibition of JAK signaling can be assessed by a variety of methods known in the art.
  • Non-limiting examples include a showing of (a) a decrease in kinase activity of JAK; (b) a decrease in binding affinity between JAK and one or more of its respective ligands, such as a cytokine; (c) a decrease in the levels of phosphorylated intracellular signaling molecules downstream in the JAK signaling pathway, such as a decrease in pSTAT3 or pSTAT6 levels; (d) a decrease in binding of JAK to downstream signaling molecules; and/or (e) an increase in ATP levels or a decrease in ADP levels. Kits and commercially available assays can be utilized for determining one or more of the above.
  • the present disclosure provides a method of treating a JAK-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein.
  • the disease or condition is a respiratory disease, such as asthma, chronic obstructive pulmonary disease or lung transplant rejection.
  • the disease or condition is asthma, such as T2-dominant (eosinophilic) asthma or non-T2-dominant (non-eosinophilic) asthma.
  • the disease or condition is lung transplant rejection, such as primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction.
  • the JAK inhibitors of the present disclosure have been designed for the treatment of inflammatory and fibrotic disease of the respiratory tract.
  • the compounds have been designed to enable delivery of a potent anti-cytokine agent directly to the site of action of respiratory disease in the lung while limiting systemic exposure.
  • Example 10 As shown in Example 10 and Table 2, the compounds of the present disclosure have been shown to be potent inhibitors of the JAK family of enzymes: JAK1, JAK2, JAK3, and TYK2.
  • Certain compounds disclosed herein are expected to inhibit an effect of the pro-inflammatory cytokine IL-13 in mouse lung tissue. Specifically, compounds of the present disclosure are expected to inhibit IL-13-induced phosphorylation of STAT6 in lung tissue, which would provide evidence of local lung JAK target engagement in vivo. This effect is expected when the pro-inflammatory cytokine IL-13 is administered 8 hours after administration of the test compound, which would provide further evidence of significant retention in the lung.
  • Cytokines implicated in asthma inflammation which signal through the JAK-STAT pathway include IL-2, IL- 3, IL-4, IL-5, IL-6, IL-9, IL-11, IL-13, IL-23, IL-31 , IL-27, thymic stromal lymphopoietin (TSLP), interferon- y (IFNy) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Accordingly, the compounds of the present disclosure are expected to be useful for the treatment of inflam matory respiratory disorders, such as asthma.
  • TSLP thymic stromal lymphopoietin
  • IFNy interferon- y
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • the present compounds are also expected to be useful for the treatment of chronic obstructive pulmonary disease, cystic fibrosis, pneumonitis, interstitial lung diseases (including idiopathic pulmonary’ fibrosis), acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, and sarcoidosis.
  • asthma endotypes may be broadly regarded as type 2 (T2) high or T2-low (Kuravilla et al., Clin Rev Allergy Immunol 2019, 56(2), 219-233). Based on their mechanism of action, the compounds of the disclosure have the potential to treat both endotypes, T2-high and T2-low.
  • Eosinophilic airway inflammation is a characteristic feature of diseases collectively termed eosinophilic lung diseases (Cottin et al., Clin. Chest. Med. 2016, 37(3), 535-56). Eosinophilic diseases have been associated with IL-4, IL-13 and IL-5 signaling.
  • Eosinophilic lung diseases include infections (especially helminthic infections), drug-induced pneumonitis (induced for example by therapeutic drags such as antibiotics, phenytoin, or 1 -tryptophan), fungal-induced pneumonitis (e.g., allergic bronchopulmonary aspergillosis), hypersensitivity pneumonitis and eosinophilic granulomatosis with polyangiitis (formerly known as Churg-Strauss syndrome).
  • Eosinophilic lung diseases of unknown etiology include idiopathic acute eosinophilic pneumonia, idiopathic chronic eosinophilic pneumonia, hypereosinophilic syndrome, and Loffler syndrome.
  • C ytokines such as IFNy, IL-12 and IL-6 have been implicated in a range of non-allergic lung diseases such as sarcoidosis and lymphangioleiomyomatosis (El-Hashemite et al.. Am. J. Respir. Cell. Mol. Biol. 2005, 33, 227-230, and El-Hashemite et al., Cancer Res. 2004, 64, 3436-3443).
  • Bronchiectasis and infiltrative pulmonary diseases are diseases associated with chronic neutrophilic inflammation.
  • Pathological T cell activation is critical in the etiology of multiple respiratory diseases.
  • autoreactive T cells play a role in bronchiolitis obliterans organizing pneumonia (also termed COS). Similar to COS, the etiology of lung transplant rejections is linked to an aberrant T cell activation of the recipient’s T cells by the transplanted donor lung.
  • Lung transplant rejections may occur early as Primary Graft Dysfunction (PGD), organizing pneumonia (OP), acute rejection (AR) or lymphocytic bronchiolitis (LB) or they may occur years after lung transplantation as Chronic Lung Allograft Dysfunction (CLAD). Therefore, JAK inhibitors have the potential to be effective in treating or preventing lung transplant rejection and CLAD.
  • PGD Primary Graft Dysfunction
  • OP organizing pneumonia
  • AR acute rejection
  • LB lymphocytic bronchiolitis
  • CLAD Chronic Lung Allograft Dysfunction
  • GVHD lung graft-versus-host disease
  • systemic JAK inhibition is associated with serious adverse events and a small therapeutic index, the need remains for an inhaled lung-directed, non-systemic JAK inhibitor to prevent and/or treat lung transplant rejection or lung GVHD.
  • the compounds of the present disclosure have the characteristics required to meet this need, [0124] Therefore, provided herein is a method of treating or preventing lung transplant rejection in a human in need thereof, comprising administering to the human a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • the lung transplant rejection is selected from the group consisting of primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is acute lung transplant rejection. In some embodiments, the lung transplant rejection is chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is selected from the group consisting of bronchiolitis obliterans, restrictive chronic lung allograft dysfunction, and neutrophilic allograft dysfunction.
  • immune-checkpoint inhibitor induced pneumonitis another T ceil mediated lung disease emerged with the increased use of immune-checkpoint inhibitors. In cancer patients treated with these T cell stimulating agents, fatal pneumonitis can develop.
  • the compounds of the disclosure possess biological activity’ allowing inhibition of IFNy secretion.
  • the present disclosure provides a method of treating a respiratory disease in a mammal (e.g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • a mammal e.g., a human
  • the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • the respiratory disease is asthma, chronic obstructive pulmonary disease (COPD), pneumonitis, cystic fibrosis (CF), interstitial lung diseases (including idiopathic pulmonary fibrosis), acute lung injury’, acute respiratory’ distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, or sarcoidosis.
  • the respiratory’ disease is asthma or chronic obstructive pulmonary disease.
  • the asthma is T2-high asthma.
  • the asthma is T2-low asthma.
  • the respiratory disease is viral induced acute lung injury (VI-ALI).
  • the respiratory disease is chronic lung allograft rejection (CLAD).
  • the respiratory disease is pulmonary hypertension associated interstitial lung disease (PH-ILD).
  • the respiratory disease is scleroderma associated interstitial lung disease (Sc-ILD).
  • the respiratory disease is a lung infection, an eosinophilic disease, a helminthic infection, pulmonary arterial hypertension, lymphangioleiomyomatosis, bronchiectasis, an infiltrative pulmonary disease, drug-induced pneumonitis, fungal induced pneumonitis, allergic bronchopulmonary' aspergillosis, hypersensitivity pneumonitis, eosinophilic granulomatosis with polyangiitis, idiopathic acute eosinophilic pneumonia, idiopath ic chronic eosinophilic pneumonia, hy pereosinophilic syndrome.
  • the present disclosure further provides a method of treating asthma in a mammal (e.g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically- acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • a mammal e.g., a human
  • the compounds of the present disclosure will typically be administered in a single daily dose or in multiple doses per day, although other forms of administration may be used.
  • the amount of active agent administered per dose or the total amount administered per day will typically be determined by a physician, in light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the compounds of the present disclosure When used to treat a respiratory disease (including but not limited to a disease described herein), the compounds of the present disclosure will typically be administered in a single daily dose or in multiple doses per day, although other forms of administration may be used.
  • the amount of active agent administered per dose or the total amount administered per day will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient’s symptoms, and the like.
  • Human coronavirus is a common respiratoiy pathogen and typically induces mild upper respiratory disease.
  • SARS-CoV-1 severe acute respiratory syndrome-associated coronavirus
  • MERS-CoV Middle East respiratory syndrome-associated coronavirus
  • COVID-19 patients Similar to SARS-CoV-1 and MERS-CoV, a subset of COVID-19 patients (about 16%) can develop a severe respiratory illness manifested by acute lung injury (AL1) leading to ICU admission (about 5%), respiratory failure (about 6.1%) and death (Wang et al., JAMA 2020, 323, 11, 1061-1069; Guan et al., N Engl J Med. 2020, 382, 1708-1720; Huang et al., The Lancet 2020, 395 (10223), 497-506; Chen et al., The Lancet 2020, 395(10223), 507-13).
  • a subgroup of patients with CO VID-19 appears to have a hyperinflammatory “cytokine storm” resulting in acute lung injury and acute respiratory distress syndrome (ARDS).
  • ARDS acute respiratory distress syndrome
  • This cytokine storm may also spill over into the systemic circulation and produce sepsis and ultimately, multi-organ dysfunction syndrome.
  • the dysregulated cytokine signaling that appears in COVID- 19 is characterized by increased expression of interferons (IFNs), interleukins (Ils), and chemokines, resulting in ALT and associated mortality.
  • IFNs interferons
  • Ils interleukins
  • chemokines resulting in ALT and associated mortality.
  • This hyperinflammatory response can potentially be modulated and treated by a lung-selective pan-Janus Kinase (JAK) inhibitor.
  • Monoclonal antibodies directed against IL-6 (tocilizumab) appear to be effective in treating patients with AL1 from COVID-19 (Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al.
  • JAK inhibitors have also been shown to be beneficial in mouse models of lipopolysaccharide- or ganciclovir-induced ALL
  • EUA emergency use authorization
  • compounds of formula (1) which are lung-selective inhaled pan-JAK inhibitors, could be uniquely suited to dampen the cytokine storm associated with coronaviruses such as COVID-19.
  • coronaviruses such as COVID-19.
  • additional infections that lead to worsened mortality may also be avoided. This is particularly true in those patients requiring ventilatory 7 support.
  • an inhaled medication may be a way to avoid systemic immunosuppression that would pre-dispose patients to these risks .
  • the present disclosure provides a method of treating a mammal (or patient) infected with a coronavirus, such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV, or the symptoms thereof, the method comprising adm inistering to the mammal (or patient) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically - acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • a coronavirus such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV
  • the present disclosure also provides a method of treating ALT and/or ARDS in a mammal (or a patient) caused by a coronavirus infection (such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV), the method comprising adm inistering to the mammal (or patient) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically - acceptable earner and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • a coronavirus infection such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV
  • JAK inhibitors have been linked to the treatment of nasal inflammatory diseases (Therapeutic Effects of Intranasal Tofacitintb on Chronic Rhinosinusitis with Nasal Polyps in Mice, Joo et al., The Laryngoscope 2020, https://doi.org/10.1002/laiy.29129). Further, Dupilumab, which acts by blocking the IL-4 and IL-13 signaling pathways, has been approved for the treatment of chrome rhinosinusitis with nasal poly ps.
  • nasal inflammatory diseases in a mammal (e g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the present disclosure, or a pharmaceutically' acceptable salt thereof.
  • the nasal inflammatory disease is selected from the group consisting of chronic rhinosinusitis with or w ithout nasal poly ps, nasal polyposis, sinusitis with nasal polyps, and rhinitis (non-allergic, allergic, perennial, and vasomotor rhinitis).
  • the compounds of the present disclosure may' also be useful for a variety of other diseases.
  • the compounds of the present disclosure may' be useful for a variety' of gastrointestinal inflammatory indications that include, but are not limited to, inflammatory bowel disease, ulcerative colitis (proctosigmoiditis, pancolitis, ulcerative proctitis and left-sided colitis), Crohn’s disease, collagenous colitis, lymphocytic colitis, Behcet’s disease, celiac disease, immune checkpoint inhibitor induced colitis, ileitis, eosinophilic esophagitis, graft versus host disease-related colitis, and infectious colitis.
  • ulcerative colitis proctosigmoiditis, pancolitis, ulcerative proctitis and left-sided colitis
  • Crohn’s disease collagenous colitis
  • lymphocytic colitis Behcet’s disease
  • celiac disease immune checkpoint inhibitor induced colitis
  • ileitis eosinophilic es
  • the compounds of the present disclosure may be useful for the induction and maintenance of remission of ulcerative colitis, and for the treatment of Crohn’s disease, immune checkpoint inhibitor induced colitis, and the gastrointestinal adverse effects in graft versus host disease.
  • the present disclosure provides a method of treating a gastrointestinal inflammatory disease in a mam mal (e.g., a human), the method comprising administering to the mammal a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
  • Atopic dermatitis and other inflammatory skin diseases have been associated with elevation of proinflammatory cytokines that rely on the JAK-STAT pathway. Therefore, the compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may be beneficial in a number of dermal inflammatory or pruritic conditions that include, but are not limited to atopic dermatitis, alopecia areata, vitiligo, psoriasis, dermatomyositis, cutaneous T ceil lymphoma and subtypes (Sezary syndrome, mycosis fungoides, pagetoid reticulosis, granulomatous slack skin, lymphomatoid papulosis, pityriasis lichenoides chronica, pityriasis lichenoides et varioliformis acuta, CD30+ cutaneous T-cell lymphoma, secondary cutaneous CD30+ large cell lymphoma, non-mycosis fungoides
  • atopic dermatitis, alopecia areata, vitiligo, prurigo nodularis, lichen planus, primary 7 localized cutaneous amyloidosis, bullous pemphigoid, and dermal manifestations of graft versus host disease are characterized by elevation of certain cytokines that signal via JAK activation.
  • compounds of the present disclosure, or a pharmaceutically acceptable salt thereof may be able to alleviate associated dermal inflammation or pruritus driven by these cytokines.
  • compounds of the present disclosure, or a pharmaceutically' acceptable salt thereof may be expected to be useful for the treatment of atopic dermatitis and other inflammatory skin diseases.
  • the present disclosure provides a method of treating an inflammatory skin disease in a mammal (e.g., a human), the method comprising applying a pharmaceutical composition comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof and a pharmaceutical carrier to the skin of the mammal.
  • a mammal e.g., a human
  • the inflammatory skin disease is atopic dermatitis.
  • ocular diseases have been shown to be associated with elevations of proinflainmatory cytokines that rely on the JAK-STAT pathway.
  • the compounds of the present disclosure, or a pharmaceutically acceptable salt thereof may be useful for the treatment of ocular diseases that include, but are not limited to, uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, age-related macular degeneration, and atopic keratoconjunctivitis.
  • uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, and age-related macular degeneration are characterized by elevation of certain pro- inflammatory cytokines that signal via the JAK-STAT pathway.
  • the present disclosure provides a method of treating an ocular disease in a mammal (e.g., a human), the method comprising administering a pharmaceutical composition comprising a compound of the present disclosure or a pharmaceutically-acceptable salt thereof and a pharmaceutical carrier to the eye of the mammal (or human).
  • a mammal e.g., a human
  • the ocular disease is uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, age-related macular degeneration, or atopic keratoconjunctivitis.
  • the method comprises administering the compound of the present disclosure, or a pharmaceutically acceptable salt thereof by intravitreal injection.
  • Compounds of the present disclosure, or a pharmaceutically acceptable salt thereof may also be used in combination with one or more compound useful to ocular diseases.
  • the compounds of the present disclosure, or a pharmaceutically acceptable salt thereof may also be useful to treat other diseases such as other inflammatory diseases, autoimmune diseases or cancers.
  • the compounds of the present disclosure, or a pharmaceutically acceptable salt thereof may be useful to treat one or more of cytokine release syndrome (CRS), arthritis, rheumatoid arthritis, juvenile rheumatoid arthritis, transplant rejection, xerophthalmia, psoriatic arthritis, diabetes, insulin dependent diabetes, motor neuron disease, myelodysplastic syndrome, pain, sarcopenia, cachexia, septic shock, systemic lupus erythematosus, leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, ankylosing spondylitis, myelofibrosis, B-cell lymphoma, hepatocellular carcinoma, Hodgkin’s disease, breast cancer
  • the present disclosure provides a pharmaceutical composition.
  • the pharmaceutical composition may comprise a compound disclosed herein, such as a compound of Formula (I), and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated for oral administration.
  • the pharmaceutical composition is formulated for inhalation.
  • the pharmaceutical composition comprises a compound disclosed herein and an additional therapeutic agent. Non-limiting examples of such therapeutic agents are described herein below.
  • Pharmaceutical compositions typically include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Formula (I), or a compound provided in Table 1 — described herein as the active agent.
  • the active agent may be provided in any form suitable for the particular mode of administration, such as a free base, a free acid, or a pharmaceutically acceptable salt.
  • the methods and pharmaceutical compositions of the present disclosure include the use of N-oxides, crystalline forms (e.g., polymorphs), as well as metabolites of these compounds having similar activity. All tautomers of the compounds described herein are included within the scope of the present disclosure. Additionally, the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol and the like.
  • Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, vaginal, aerosol, pulmonary, nasal, transmucosal, topical, transdermal, otic, ocular, and parenteral modes of administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation.
  • a long acting formulation is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection.
  • a compound described herein is provided in the form of a rapid release formulation, an extended release formulation, or an intermediate release formulation.
  • a compound described herein is provided in the form of a nebulized formulation.
  • a compound described herein is administered locally to the lungs by inhalation.
  • Compounds of the present disclosure are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, 0.5 to 100 mg, 1 to 50 mg, or from 5 to 40 mg per day may be administered to a subject in need thereof. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • a compound of the present disclosure may be administered in a single dose.
  • a compound of the disclosure is administered in multiple doses, such as about once, twice, three times, four times, five times, six times, or more than six times per day.
  • dosing is about once a month, once every two weeks, once a week, or once every other day.
  • a compound of the disclosure and an additional therapeutic agent are administered together about once per day to about 6 times per day.
  • the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or more than about one year.
  • a dosing schedule is maintained as long as necessary.
  • a compound of the present disclosure may be administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • compositions of the present disclosure typically contain a therapeutically effective amount of a compound of the present disclosure.
  • a pharmaceutical composition may contain more than a therapeutically effective amount, e.g., bulk compositions, or less than a therapeutically effective amount, e.g., individual unit doses designed for coadministration to achieve a therapeutically effective amount.
  • compositions of the present disclosure contain from about 0.01 to about 95% by weight of the active agent; including, for example, from about 0.05 to about 30% by weight; and from about 0. 1 % to about 10% by weight of the active agent.
  • a pharmaceutical composition of the present disclosure may contain from 0.1 mg to 100 mg of the active agent, such as 1 mg to 20 mg or 1 mg to 10 mg of the active agent.
  • any conventional carrier or excipient may be used in the pharmaceutical compositions of the present disclosure.
  • the choice of a particular carrier or excipient, or combinations of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state. Additionally, the carriers or excipients used in the pharmaceutical compositions of this disclosure may be commercially-available. Conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7 th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
  • Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as com starch and potato starch; cellulose, such as microcrystalline cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic
  • compositions are typically prepared by thoroughly and intimately mixing or blending the active agent with a pharmaceutically-acceptable carrier and one or more optional ingredients. The resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills and the like using conventional procedures and equipment.
  • the pharmaceutical composition is suitable for inhaled administration.
  • Pharmaceutical compositions for inhaled administration are typically in the form of an aerosol or a powder.
  • Such compositions are generally administered using inhaler delivery devices, such as a dry powder inhaler (DPI), a metered-dose inhaler (MDI), a nebulizer inhaler, or a similar delivery device.
  • DPI dry powder inhaler
  • MDI metered-dose inhaler
  • nebulizer inhaler or a similar delivery device.
  • the pharmaceutical composition is administered by inhalation using a dry powder inhaler.
  • dry powder inhalers typically administer the pharmaceutical composition as a free- flowing powder that is dispersed in a patient’s air-stream during inspiration.
  • the therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PLA), polylactide-co-glycolide (PLGA) or combinations thereof.
  • the therapeutic agent is micronized and combined with a suitable carrier to form a composition suitable for inhalation.
  • a representative pharmaceutical composition for use in a dry powder inhaler comprises lactose and a micronized form of a compound disclosed herein.
  • a dry powder composition can be made, for example, by combining dry milled lactose with the therapeutic agent and then dry blending the components. The composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
  • Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (IV AX); ClickHaler (Innovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Ellipta (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaler (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like.
  • a pharmaceutical composition of the present disclosure may be administered by inhalation using a metered-dose inhaler.
  • metered-dose inhalers typically discharge a measured amount of a therapeutic agent using a compressed propellant gas.
  • pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant.
  • Any suitable liquefied propellant may be employed, including hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-w-propane, (HFA 227); and chlorofluorocarbons, such as CCI3F.
  • the propellant is a hydrofluoroalkane.
  • the hydrofluoroalkane formulation contains a co-solvent, such as ethanol or pentane, and/or a surfactant, such as sorbitan trioleate, oleic acid, lecithin, and glycerin.
  • a representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01% to about 5% by weight of a compound of the present disclosure; from about 0% to about 20% by weight ethanol; and from about 0% to about 5% by weight surfactant; with the remainder being an HFA propellant.
  • Such compositions are typically prepared by adding chilled or pressurized hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic agent is micronized and then combined with the propellant. The composition is then loaded into an aerosol canister, which typically forms a portion of a metered-dose inhaler device.
  • Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
  • a pharmaceutical composition of the present disclosure may be administered by inhalation using a nebulizer inhaler.
  • nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition to spray as a mist that is carried into the patient’s respiratory tract.
  • the therapeutic agent when formulated for use in a nebulizer inhaler, can be dissolved in a suitable carrier to form a solution.
  • the therapeutic agent can be micronized or nanomilled and combined with a suitable carrier to form a suspension.
  • a representative pharmaceutical composition for use in a nebulizer inhaler comprises a solution or suspension comprising from about 0.05 pg/mL to about 20 mg/mL of a compound of the present disclosure and excipients compatible with nebulized formulations.
  • the solution has a pH of about 3 to about 8.
  • Nebulizer devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative nebulizer devices or products include the Respimat® Softmist TM Inhalaler (Boehringer Ingelheim); the AERx® Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus® Reusable Nebulizer or PARI eFlow®rapid Nebulizer System (Pari GmbH); and the like.
  • a pharmaceutical composition of the present disclosure may be prepared in a dosage form intended for oral administration.
  • Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present disclosure as an active ingredient.
  • the pharmaceutical compositions of the disclosure will typically comprise the active agent and one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate.
  • such solid dosage forms may also comprise: fillers or extenders, binders, humectants, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the present pharmaceutical compositions.
  • Alternative formulations may include controlled release formulations, liquid dosage forms for oral administration, transdermal patches, and parenteral formulations. Conventional excipients and methods of preparation of such alternative formulations are described, for example, in the reference by Remington, supra. [0166] The following non-limiting examples illustrate representative pharmaceutical compositions of the present disclosure.
  • a micronized compound of the present disclosure (1 g) is blended with milled lactose (25 g). This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide between about 0. 1 mg to about 4 mg of the compound per dose. The contents of the blisters are administered using a dry powder inhaler.
  • a micronized compound of the present disclosure (1 g) is blended with milled lactose (20 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1:20.
  • the blended composition is packed into a dry powder inhalation device capable of delivering between about 0.1 mg to about 4 mg of the compound per dose.
  • a micronized compound of the present disclosure (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL). The resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 pm. The micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1,1,2-tetrafluoroethane in an amount sufficient to provide about 0.1 mg to about 4 mg of the compound per dose when administered by the metered dose inhaler.
  • a representative nebulizer composition is as follows. A compound of the present disclosure (2 g of free-base equivalents) is dissolved in a solution containing 80 mL reverse-osmosis water, 0.1-1% by weight of anhydrous citric acid, and 0.5 -1.5 equivalents of hydrochloric acid, followed by addition of sodium hydroxide to adjust the pH to 3.5 to 5.5. Thereafter, between 4-6% by weight of D-mannitol is added and solution q.s. to 100 mL. The solution is then filtered through a 0.2 pm filter and stored at room temperature prior to use. The solution is administered using a nebulizer device that provides about 0.1 mg to about 4 mg of the compound per dose. [0175] Kits
  • the present disclosure provides a kit comprising one or more unit doses of a compound or pharmaceutical composition described herein, optionally wherein the kit further comprises instructions for using the compound or pharmaceutical composition.
  • the kit comprises a carrier, package, or container that is compartmentalized to receive one or more containers, such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials, such as glass or plastic.
  • the articles of manufacture provided herein may contain packaging materials.
  • Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • the container(s) may include one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein.
  • the container(s) may optionally have a sterile access port (for example, the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • kits may optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit includes one or more containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein.
  • materials include, but are not limited to, buffers, diluents, filters, needles, syringes, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • the pharmaceutical composition is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein.
  • the pack may contain metal or plastic foil, such as a blister pack.
  • the compounds and pharmaceutical compositions of the disclosure may be used in combination with one or more therapeutic agents which act by the same mechanism or by a different mechanism to treat a disease.
  • the one or more agents may be administered sequentially or simultaneously, in separate compositions or in the same composition.
  • Useful classes of agents for combination therapy include, but are not limited to, a beta 2 adrenoceptor agonist, a muscarinic receptor antagonist, a glucocorticoid agonist, a G- protein coupled receptor-44 antagonist, a leukotriene D4 antagonist, a muscarinic M3 receptor antagonist, a histamine Hl receptor antagonist, an immunoglobulin E antagonist, a PDE 4 inhibitor, an IL-4 antagonist, a muscarinic Ml receptor antagonist, a histamine receptor antagonist, an IL-13 antagonist, an IL-5 antagonist, a 5 -Lipoxygenase inhibitor, a beta adrenoceptor agonist, a CCR3 chemokine antagonist, a CFTR stimulator, an immunoglobulin modulator, an interleukin 33 ligand inhibitor, a PDE 3 inhibitor, a phosphoinositide-3 kinase delta inhibitor, a thromboxane A2 antagonist, an
  • a JAK inhibitor and a second therapeutic agent can be administered sequentially, wherein the two agents are introduced into a subject at two different time points.
  • the two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, 1 or more months, or according to any intermittent regimen schedule disclosed herein.
  • the JAK inhibitor and the second therapeutic agent are administered simultaneously.
  • the two agents may form part of the same composition, or the two agents may be provided in one or more unit doses. When in separate doses, the two agents may be administered via different routes.
  • the JAK inhibitor or the second therapeutic agent are administered parenterally, orally, inhalatively, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally.
  • a therapeutically effective amount of a combination of a JAK inhibitor and a second therapeutic agent refers to a combination of a JAK inhibitor and a second therapeutic agent, wherein the combination is sufficient to affect the intended application, including but not limited to, disease treatment, as defined herein.
  • Also contemplated in the subject methods is the use of a sub-therapeutic amount of a JAK inhibitor and a second therapeutic agent in combination for treating an intended disease condition.
  • the individual components of the combination though present in sub-therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
  • the amount of the JAK inhibitor and the second therapeutic agent administered may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • Measuring an immune response and/or the inhibition of biological effects of JAK can comprise performing an assay on a biological sample, such as a sample from a subject. Any of a variety of samples may be selected, depending on the assay. Examples of samples include, but are not limited to blood samples (e.g. blood plasma or serum), exhaled breath condensate samples, bronchoalveolar lavage fluid, sputum samples, urine samples, and tissue samples.
  • blood samples e.g. blood plasma or serum
  • exhaled breath condensate samples e.g. blood plasma or serum
  • bronchoalveolar lavage fluid e.g. bronchoalveolar lavage fluid
  • sputum samples e.g. urine samples, and tissue samples.
  • a subject being treated with a JAK inhibitor and a second therapeutic agent may be monitored to determine the effectiveness of treatment, and the treatment regimen may be adjusted based on the subject’s physiological response to treatment. For example, if inhibition of a biological effect of JAK inhibition is above or below a threshold, the dosing amount or frequency may be decreased or increased, respectively. Alternatively, the treatment regimen may be adjusted with respect to an immune response.
  • the methods can further comprise continuing the therapy if the therapy is determined to be efficacious.
  • the methods can comprise maintaining, tapering, reducing, or stopping the administered amount of a compound or compounds in the therapy if the therapy is determined to be efficacious.
  • the methods can comprise increasing the administered amount of a compound or compounds in the therapy if it is determined not to be efficacious.
  • the methods can comprise stopping therapy if it is determined not to be efficacious.
  • treatment with a JAK inhibitor and a second therapeutic agent is discontinued if inhibition of the biological effect is above or below a threshold, such as in a lack of response or an adverse reaction.
  • the biological effect may be a change in any of a variety of physiological indicators.
  • agents that may be used in combination with the compounds disclosed herein include, but are not limited to, abatacept, abediterol, aclidinium, aclidinium bromide, ACT-774312, Actair, Actimmune, acumapimod, adalimumab, ADC-3680, Adipocell, AG-1321001, AG-NPP709, alicaforsen, Allergovac depot, alpha-cyclodextrin-stabilized sulforaphane, AM-211, ambroxol + erdosteine, Ampion, anakinra, ANB-020, andecaliximab, APL-1, apremilast, arformoterol, ARRY-502, ASM-024, ASM-8, aviptadil, AXP-1275, AXP- E, AZD-1419, AZD-7594, AZD-8871, AZD-9898, azithromycin
  • a compound disclosed herein is administered in combination with a second therapeutic agent, optionally wherein the second therapeutic agent is administered by inhalation.
  • the present disclosure provides a method of treating a JAK mediated disease or condition, such as a respiratory disease, in a subject, comprising administering to the subject a JAK inhibitor, such as a compound disclosed in Table 1, and one or more second therapeutic agents.
  • the present disclosure provides a method of treating asthma, such as T2-dominant (eosinophilic) asthma or non- T2-dominant (non-eosinophilic) asthma, in a subject, comprising administering to the subject a JAK inhibitor, such as a compound disclosed in Table 1, and a second therapeutic agent.
  • a JAK inhibitor such as a compound disclosed in Table 1
  • a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
  • the therapeutic agent may be selected from the classes of agents specified above and from the lists of specific agents described above.
  • the pharmaceutical composition is suitable for delivery to the lungs.
  • the pharmaceutical composition is suitable for inhaled or nebulized administration.
  • the pharmaceutical composition is a dry powder or a liquid composition.
  • the disclosure provides a method of treating a disease or disorder in a mammal comprising administering to the mammal a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
  • the agents When used in combination therapy, the agents may be formulated in a single pharmaceutical composition, or the agents may be provided in separate compositions that are administered simultaneously or at separate times, by the same or by different routes of administration. Such compositions can be packaged separately or may be packaged together as a kit. The two or more therapeutic agents in the kit may be administered by the same route of administration or by different routes of administration.
  • the disclosure also includes compounds and pharmaceutical compositions as described herein for use in treating a disease or disorder as described herein.
  • the disclosure includes the manufacture of a medicament comprising a compound or pharmaceutical composition for the treatment of a disease or disorder as described herein.
  • BoczO di-tert-butyl dicarbonate
  • BSA bovine serum albumin
  • Fraction V d day(s)
  • DCM dichloromethane or methylene chloride
  • DIPEA or DIEA A'. A'-diisopropylcthylaminc
  • EDTA ethylenediaminetetraacetic acid
  • EGTA ethylene glycol-bis(P-aminoethyl ether)-
  • HEPES 4-(2-hyrdroxyethyl)-l -piperazine ethanesulfonic acid
  • NBS N-bromosuccinimide
  • Pd(dppf)C1 2 [l,l'-Z>A(diphenylphosphino)ferrocene]- dichloropalladium(II)
  • Pd(PPhs) 4 tetrakis(triphenylphosphine)palladium(0)
  • PE petroleum ether
  • SiO 2 silicon dioxide or silica
  • reaction mixtures were worked up as described specifically in each preparation; commonly, reaction mixtures were purified by extraction and other purification methods such as temperature- and solvent-dependent crystallization, and precipitation.
  • reaction mixtures were routinely purified by preparative HPLC, typically using Microsorb C18 and Microsorb BDS column packings and conventional eluents.
  • Progress of reactions was typically monitored by liquid chromatography mass spectrometry (LCMS). Characterization of isomers was typically done by Nuclear Overhauser effect spectroscopy (NOE). Characterization of reaction products was routinely carried out by mass spectrometry and/or 1 H-NM R spectroscopy.
  • Example 1 Synthesis of (E)-3-ethyl-4-(3-(4-(3-(isopropylamino)prop-1-en-i-y1l)-1H- imidazol-2-yl)- 1H-indazol-6-yl)phenol (also known as 3-ethyl-4-[3-[4-[(E)-3-(isopropylamino)prop-l-enyl]-1H-imidazol-2- yl]- 1H-indazol-6-yl]phenol) (23).
  • Step A Preparation of (2-((3-ethylphenoxy)metboxy)etbyl)trimethylsilane (1-2).
  • Step B Preparation of (2-((4-bromo-3 -ethylphenoxy )methoxy)ethyl)trim ethylsilane (1-3).
  • 1-2 200 g, 792 mmol
  • ACN 1.40 L
  • NBS 141 g, 792 mmol
  • the reaction mixture was stirred at room temperature for 2 h. TLC showed complete consumption of starting material.
  • the reaction mixture was poured into ice cold water (IL) and extracted with EtOAc (2 x IL).
  • Step C Preparation of (2-((3-ethyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)methoxy)ethyl)trimethylsilane (1-4).
  • 1-3 100 g, 302 mmol
  • 1 ,4- dioxane 1 .00 L
  • bis(pinacolato)diboron 76.6 g, 302 mmol
  • potassium acetate 59.2 g, 604 mmol
  • reaction mixture was stirred and heated at 110 °C for 16 hours under nitrogen. TLC indicated the complete consumption of the starting material.
  • the reaction mixture was diluted with EtOAc (1 L) and washed with water (1 L). The combined organic layers were separated, dried over Na 2 SO 4 , and concentrated. The crude mixture was purified by silica gel column chromatography (0-10% EtOAc in heptane) to afford the desired product as a yellow liquid (75.0 g, 66% yield).
  • Step D Preparation of 4-bromo-2-fluorobenzoyl chloride (1-6).
  • 4-bromo ⁇ 2- fluorobenzoic acid 50.0 g, 228 mmol
  • DCM 300 mL
  • DMF 4.0 mL
  • oxalyl chloride 96.57 mL, 913 mmol
  • the resulting reaction mixture was stirred at room temperature for 1 h.
  • TLC monitoring checked by quenching in MeOH
  • the reaction was concentrated under reduced pressure (under nitrogen) to afford an off-white solid (54.2 g) which w’as used in the next step without further purification.
  • Step E Preparation of (l-benzyI-1H-imidazoI-2-yl)(4-bromo-2-fluorophenyI)methanone (1-7).
  • 1 -benzyl- IH-imidazole 30.0 g, 190 mmol
  • acetonitrile 165 mL
  • triethylamine 133.4 mL, 949 mmol
  • Compound 1-6 54.2 g, 228 mmol
  • reaction wms quenched with cold water (500 mL) and extracted with ethyl acetate (2 x 600 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to get crude product which was purified by silica gel column chromatography (10% EtOAc in Heptane) to obtain the desired product as an off-white solid (79.0 g, 58% yield).
  • Step F Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-1H-indazole (1-8).
  • 1-7 53.0 g, 147.5 mmol
  • DMSO 105 mL
  • dropwdse hydrazine hydrate 72.5 mL, 1475.5 mmol
  • the reaction mixture was allowed to stir at 90 °C for 3 h. After 3 h, TLC showed complete consumption of starting material.
  • the reaction mixture was diluted with ice cold water (800 mL) and precipitation was observed.
  • Step G Preparation of 3-(l-ben2y4-1H-imidazol-2-yl)-6-bromo-l-(tetrahydro-2H-pyran-2-yl)-1H- indazole (1-9).
  • 1-8 (47.0 g, 133.1 mmol) in ethyl acetate (350 mL) at 0 °C was added TFA (30.5 mL, 399.1 mmol).
  • Dihydropyran (60.8 mL, 665.3 mmol) was added dropwise. The reaction mixture was then heated to 80 °C and stirred for 2 days. After 2 days, TLC showed complete consumption of starting material.
  • the reaction mixture was diluted with water (400 mL) and extracted with ethyl acetate (2 x 900 mL). The combined organic layers were further washed with saturated aq. NaHCO 3 solution (800 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude product was purified by silica gel column chromatograph ⁇ ' (5% EtOAc in heptane) to afford the desired product as an off white solid (40 g, 69% yield).
  • Step H Preparation of 3-(l-benzyI-1H-imidazoI-2-yl)-6-(2-ethyl-4-((2- (trimethy1silyl)ethoxy)methoxy)pheny1)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (1-10).
  • 1-9 (60.0 g, 137 mmol) and 1-4 (62.3 g, 165 mmol) in dioxane (360 mL) and water (90.0 mL) was added K 3 PO 4 (87.4 g, 412 mmol).
  • the reaction mixture was purged with argon for 15 minutes then Pd/PPlrfo (15.9 g, 13.7 mmol) added to it. The reaction was then heated to 110 °C and stirred for 3 h. TLC showed consumption of the starting material.
  • the reaction mixture was diluted with water (600 mL) and extracted w’ith ethyl acetate (2 x 500 mL). The combined organics were washed with brine (600 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography (10% EtOAC in heptane). The desired product was isolated as a clear liquid (65 g, 78% yield).
  • Step I Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-3-(1H-imidazol-2-yl)- l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (1-11).
  • the reaction mixture was subjected to hydrogenation using a FL balloon and was allowed to stir at room temperature for 16 h. TLC showed complete consumption of starting material.
  • reaction mixture was filtered through a pad of Celite, washed with EtOAc (500 mL), and the filtrate was concentrated under reduced pressure to afford the crude desired product (53.0 g, 96% yield) as a colorless liquid that was used directly in the next step without further purification.
  • Step J Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-l-(tetrahydro-2H- pyran-2-yl)-3-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-2-yl)-1H-indazole (1-12).
  • reaction mixture was then allowed to stir at 0 °C for 20 min, after which SEMC1 (17.6 mL, 99.5 mmol) was added dropwise at 0 °C.
  • the reaction mixture was allowed to warm to room temperature and stirred for 1 h. TLC showed complete consumption of the starting material.
  • the reaction mixture was quenched with ice-water (1 L) and extracted ethyl acetate (3 x 500 mL). The combined organic layers were washed with water (800 mL) and brine (800 mL), dried over Na 2 SO 4 , and concentrated under reduced pressure to afford the crude compound which was purified by silica gel column chromatography (15% EtOAc in heptane).
  • Step K Preparation of 3-(4-bromo-l-((2-(trimethylsilyl)etihoxy)methyl)-1H-imidazol-2-yl)-6-(2- ethy l-4-((2-(trimethylsilyl)ethoxy )methoxy)phenyl) ⁇ 1 -(tetrahydro-2H-pyran-2-y 1)- 1 H-indazole (1-13).
  • NBS (2.74 g, 15.4 mmol) was taken up in DCM (100 mL) and added dropwise to a stirred solution of 1-12 (10.0 g, 15.4 mmol) in DCM (400 mL) at 0 °C.
  • reaction mixture was stirred at 0 °C for 5 minutes. TLC showed complete consumption of the starting material.
  • the reaction mixture was quenched with ice water (300 mL) and extracted with DCM (2 x 250 mL). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and evaporated to afford crude product which was purified by silica gel column chromatography (8- 10% EtOAc in heptane). The desired product was isolated as a colorless amorphous solid (9.10 g, 81% yield).
  • Step L Preparation of 4-(3-(4-bromo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazol-6- yl)-3 -ethylphenol (1-14).
  • TBAF IM in THF
  • Step M Preparation of tert-butyl (E)-(3-(2-(6-(2-ethyl-4-hydroxyphenyl)-l-(tetrahydro-2H -pyran-2- yl)-1H- indazol-3-yl)-1H- imidazol-4-yl)allyl)carbamate (1-15).
  • Step N Preparation of (E)-4-(3-(4-(3-aminoprop-l-en-l-yl)- 1H-imidazol-2-yI)-1H -indazol-6-yl)-3- ethylphenol (1-16).
  • Step O Preparation of (E)-3-ethyl-4-(3-(4-(3-(isopropylamino)prop-l-en-l-yl)-1H -imidazol-2-yl)-1H- indazol-6-yl)phenol (23).
  • acetone 100 pi, 1.36 mmol
  • sodium cyanoborohydride 9 mg, 0.139 mmol
  • Step A Preparation of 4-bromo-2,3 -difluorobenzoyl chloride (2-2).
  • 4-bromo- 2,3 -difluorobenzoic acid (28.0 g, 118 mmol) (2-1) in DCM (300 mL) and DMF (915 ⁇ L, 0.1 eq., 11.8 mmol)
  • oxalyl chloride (40.5 mL, 473mmol) dropwise ai room temperature.
  • the resulting reaction mixture was stirred at room temperature for 2h. After completion of reaction (TLC monitoring, checked by quenching in MeOH), the reaction was concentrated under reduced pressure (under nitrogen) to afford an off-white solid (31.0 g) which was used in the next step without further purification.
  • Step B Preparation of (1-benzyl-1H-imidazol-2-yl)(4-bromo-2,3-difluorophenyl)methanone (2-3).
  • compound 2-2 (16.0 g, 101 mmol) in acetonitrile (100 ml) was added triethylamine (51 .2 g, 506 mmol).
  • 1 -benzyl- 1H-imidazole (31.0 g, 121 mmol) was dissolved in acetonitrile (100 mL) separately and added to the reaction mixture at room temperature. The reaction was allowed to stir at room temperature for 2 h. TLC showed consumption of starting material.
  • reaction was quenched with cold water (500 mL) and extracted with ethyl acetate (2 x 600 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to get crude product which was purified by silica gel column chromatography (10-15% EtOAc in Heptane) to obtain the desired product as a light yellow solid (28.0 g, 73% yield).
  • Step C Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-7-fluoro-1H-indazole (2-4).
  • DMSO 120 mL
  • hydrazine hydrate 28.0 mL, 575 mmol
  • the reaction mixture was allowed to stir at 90 °C for 3 h. After 3 h, TLC showed complete consumption of SM.
  • the reaction mixture was diluted with ice cold water (2 x 500 mL) and precipitation was observed.
  • Step D Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-7-fluoro-l-(tetraliydro-2H-pyran-2- yl)-1H-indazole (2-5).
  • TFA ethyl acetate
  • Dihydropyran 23.6 mL, 269 mmol
  • the reaction mixture was diluted with water (400 mL) and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were further washed -with saturated aq. NaHCOs solution (800 mL), dried over anhydrous NazSCfi, filtered and concentrated.
  • the crude product vras purified by silica gel column chromatography (8-10% EtOAc in heptane) to afford the desired product as an off white solid (19.5 g, 78% yield).
  • Step E Preparation of 3-(1-benzyl-1H-imidazol-2-yl)-6-(2-ethyl-4-((2-
  • reaction was then heated to 100 °C and stirred for 16 h. TLC showed consumption of the starting material.
  • the reaction mixture was filtered through a pad of Celite and the residue washed with ethyl acetate (2 x 200 mL). The combined organics were washed with cold wmter (300 mL) and brine (300 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography (12% EtOAC in heptane). The desired product was isolated as a clear amorphous solid (20.0 g, 73% yield).
  • Step F Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-3-(1 H- imidazol-2-yl)-1 -(tetrahydro-2H-pyran-2-yl)-1 H-indazole (2-7).
  • 2-6 (20.0 g, 31.9 mmol) in isopropanol (200 mL) and THF (50.0 mL) was added 20% Pd(OH) 2 /C (20.0 g, 163 mmol).
  • the reaction mixture was subjected to hydrogenation using a H 2 balloon and was allowed to stir at room temperature for 5 h.
  • Step G Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l- (tetrahydro-2H-pyran-2-yl)-3-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-2-yl)-1H-indazole (2-8).
  • 2-7 (18.0 g, 33.5 mmol) in DMF (180 mL) was added sodium hydride (2.81 g, 70.3 mmol) at 0 °C.
  • reaction mixture was then allowed to stir at 0 °C for 30 min, after which SEMC1 (8.39 g, 50.3 mmol) was added dropwise at 0 °C.
  • SEMC1 8.39 g, 50.3 mmol
  • the reaction mixture was allowed to warm to room temperature and stirred for 2 h. TLC showed complete consumption of the s tarting material.
  • the reaction mixture was quenched with ice-water (300 mL) and extracted in ethyl acetate (2 x 300 mL). The combined organic layers were washed with water (400 mL) and brine (400 mL), dried over Na2 S SO 4 , and concentrated under reduced pressure to afford the crude compound which w ; as purified by silica gel column chromatography (20-25% EtOAc in heptane).
  • Step H Preparation of 3-(4-bromo-l-((2-(trimethylsilyl)ethoxyjmethyl)-1H-imidazol-2-yl)-6-(2- ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (2-9).
  • NBS (1 .60 g, 9.0 mmol) was taken up in DCM (60 mL) and added dropwise to a stirred solution of 2-8 (6.0 g, 9.0 mmol) in DCM (240 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 5 minutes. TLC showed complete consumption of the starting material. The reaction mixture was quenched with water (100 mL) and extracted with DCM (2 x 300 mL). The combined organic layers were dried over anhydrous filtered and evaporated to afford crude product which wrns purified by silica gel column chromatography (8-10% EtOAc in heptane).
  • Step 1 Preparation of 4-(3-(4-bromo-1H-imidazol-2-yl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H- indazol-6-yT)-3 -ethylphenol (2-10).
  • TBAF IM in THF
  • the reaction mixture was diluted with water (500 mL) and extracted with ethyl acetate (2 x 500 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (20-25% EtOAc in heptane) to afford the desired product as an off-white solid (5.25 g, 53% yield).
  • Step J Preparation of tert-butyl (E)-(3-(2-(6-(2-ethyl-4-hydroxyphenyl)-7-fluoro-l -(tetrahydro-2H- pyran-2-yl)-1H-indazol-3-yl)-1H-imidazol-4-yl)allyl)carbamate (2-11).
  • Step K Preparation of (E)-4-(3-(4-(3-aminoprop-l-en-l-yl)-1 H-imidazol-2-yl)-7-fluoro-1 H-indazol- 6-yl)-3 -ethylphenol (2-12).
  • To 2-11 (81 mg, 0.144 mmol) was added trifluoroacetic acid (1 mL) and the reaction mixture was stirred at 50 °C for 30 minutes. The reaction mixture was then concentrated under reduced pressure and purified by preparative HPLC chromatography (5-70% ACN/water gradient with 0.05% trifluoroacetic acid) to obtain the TFA salt of the title compound as a white solid (49 mg, 69. 1% yield), (m/z): [M+H] + calcd for C 21 H 20 FN 5 O378.17, found 378.02.
  • Step L Preparation of (E)-3-ethyl-4-(7-fluoro-3-(4-(3-((tetrahydro-2H-pyran-4-yl)amino)prop-l-en- 1-yl)-1H-imidazol-2-yl)- 1H-indazol-6-yl)phenol (64).
  • Example 3 Synthesis of (S,E)-3-ethy1-4-(3-(4-(2-(morpholin-3-yl)vinyl)-1H-imidazol-2-yl)-1H- indazol-6-yl)phenol (6).
  • Step A Preparation of ethyl 1-(N,N-dimethylsulfanioyl)-1H-imidazole-4-carboxylate (3-2).
  • ethyl 1H-imidazole-4-carboxylate 30 g, 214.2 mmol
  • DMF 300 mL
  • NaH 12.8 g, 321.4 mmol
  • dimethylsulfamoyl chloride 27.7 mL, 257 mmol
  • Step B Preparation of ethyl l-(dimethylsulfamoyl)-2-iodo-1H-imidazole ⁇ 4-carboxylate (3-3).
  • LDA 2M in THF, 17 mL, 34 mmol
  • Iodine 7.9 g, 31 .1 mmol
  • anhydrous THF 40 mL
  • Step C Preparation of l-(benzyloxy)-3 -ethylbenzene (3-5).
  • 3-ethylphenol (25.0 g, 204.0 mmol) (3-4) in ACM (250 mL, 10 vol) was added potassium carbonate (42.0 g, 306 mmol) at room temperature.
  • the resulting reaction mass was stirred at room temperature for 15 minutes, followed by the addition of benzyl bromide (24.0 mL, 204 mmol) in dropwise manner.
  • the resulting reaction mixture was stirred for 6 hours at room temperature. After completion of the reaction (TLC monitoring), the resulting reaction mass was poured into water (1.0 L) followed by extraction of compound with EtOAc (2 x 2L).
  • Step D Preparation of 4-(benzyloxy)-l -bromo-2 -ethylbenzene (3-6).
  • 1 -(benzyloxy )-3 -ethylbenzene 35.0 g, 164 mmol
  • ACN 525 mL, 15 vol
  • N- bromosuccinimide 32.0 g 181 mmol
  • the resulting reaction mixture was stirred for 1 hour at room temperature. After completion of reaction (TLC monitoring), the resulting reaction mass was poured into ice cold water (1 .50 L) followed by the extraction of compound with EtOAc (2 x IL).
  • Step E Preparation of 2-(4-(benzyloxy)-2-ethylphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (3- 7).
  • a stirred solution of 4-(benzyioxy)-l-bromo-2-ethylbenzene (42.0 g, 144 mmol) (3-6), bis(pinacolato) diboron (44.0 g, 173 mmol), and potassium acetate (28 g, 288 mmol) in dioxane (440 mL) was degassed by purging N2 (g) for 15 min followed by addition of PdC12(dppf)-DCM complex (11.0 g, 15 mmol).
  • the resulting reaction mixture was heated to 80 °C for 16h. After completion of the reaction (TLC monitoring), the reaction mass was filtered through celite bed and mother liquor was evaporated under reduced pressure to obtain the crude product. Crude residue was purified by column chromatography over silica gel (100-200M) by using eluents 1% EtOAc in hexane to get the desired product as light yellow oily compound (32.0 g, 66%).
  • Step F Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-l-(oxan-2-yl)-1H-indazole (3-9).
  • 6-bromo ⁇ l -(oxan-2-yl)-1H-indazole (20 g, 71.1 mmol) and 3-7 (26.5 g, 78.2 mmol) in dioxane (150 mL) and water (20 mL) w’as added sodium carbonate (15.1 g, 142 mmol).
  • the reaction vessel was purged with argon for 20 minutes, then bis(diphenylphosphino)ferrocene paliadium(ll)dichloride dichloromethane adduct (5.81 g, 7.11 mmol) was added.
  • the reaction mixture was stirred at 100 °C for 16 h, until judged complete by TLC and LCMS.
  • the reaction mixture was then cooled to room temperature, filtered through a Celite pad, and the pad was further washed with ethyl acetate (2x200 mL).
  • the combined filtrate was then washed with cold water (2x100 mL) and brine (100 mL), after which the organic layer was dried over sodium sulfate and concentrated under reduced pressure.
  • Step G Preparation of 6-[4-(benzyloxy)-2-etbylphenylj-1H-indazole (3-10).
  • 3-9 43 g, 104 mmol
  • methanol 215 mL
  • aqueous hydrochloric acid 6N, 60 mL
  • the resulting solution was first warmed to room temperature, then heated at 90 °C for 16 h until judged complete by TLC.
  • the reaction mixture was partially concentrated under reduced pressure, then neutralized by slow addition of saturated aqueous sodium bicarbonate solution (200 mL) at 0 °C. This solution was extracted with ethyl acetate (2x300 mL).
  • Step H Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-3-iodo-1H-indazole (3-11).
  • 3-10 36 g, 110 mmol
  • DMF 120 mL
  • N-iodosuccinimide 49.3 g, 219 mmol
  • the reaction mixture was then diluted with water (200 mL) and extracted with ethyl acetate (3x100 mL).
  • Step 1 Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-3-iodo-l-(oxan-2-yl)-1H-indazole (3-12).
  • 3-11 46 g, 101 mmol
  • DCM 250 mL
  • p-toluenesulfonic acid 1.74 g, 10.1 mmol
  • 3,4-dihydro-2H-pyran (13.9 mL, 152 mmol
  • Step J Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-l-(oxan-2-yl)-3-(trimethylstannyl)-1H- indazole (3-13). A solution of 3-12 (20 g, 37.
  • Step K Preparation of ethyl 2- ⁇ 6-[4-(benzyloxy)-2-ethylphenyl]-1-(oxan-2-yT)-1H-indazol-3-yl ⁇ -1 - (dimethylsulfamoyl)-1H-imidazole-4-carboxylate (3-14).
  • 3-3 3.0 g, 8.04 mmol
  • toluene 40 ml,
  • 3-13 4.63 g, 80.4 mmol
  • Step L Preparation of ethyl 1-(dimethylsulfamoyl)-2-[6 ⁇ (2-ethyl-4-hydroxyphenyl)-l-(oxan-2-yl)- 1H-indazol-3-yl]-1H -imidazole-4-carboxylate (3-15).
  • 3-14 5.50 g, 8.36 mmol
  • THF trifluorofuran
  • isopropyl alcohol (30 mL) was added 10% palladium on carbon (10.0 g, 94 mmol).
  • the reaction mixture was then subjected to hydrogenation using a hydrogen balloon and allowed to stir at room temperature for 2 days.
  • Step M Preparation of ethyl l-(dimethylsulfamoyl)-2-[6-(2-ethyl-4- ⁇ [2-
  • Step N Preparation of 2-[6-(2-ethyl-4- ⁇ [2-(trimethylsityl)ethoxy]methoxy ⁇ phenyI)-l -(oxan-2-yl)- l H-indazoi-3-yl]-4-(hydroxymethyi)-N,N-dimethyl-1H-imidazole-l-sulfonamide (3-17).
  • 3-16 4.0 g, 5.73 mmol
  • THF 15 mL
  • lithium borohydride 2M in THF, 14.3 mL, 28.7 mmol
  • Step 0 Preparation of 4-(chloromethyl)-2-(6-(2-etbyl-4-((2.-(triniethylsilyl)ethoxy)metboxy)pbenyl)- l-(tetrahydro-2fi r -pj ⁇ ran-2-yl)-1H -indazol-3-yl)-N,N-dimethyl-1H- imidazole-l -sulfonamide (3-18). To a 100 ml flask was added 3-17 (1.6 g, 2.44 mmol) in DCM (20 mL).
  • Step P Preparation of diethyl ((l-(N,N-dimethylsulfamoyl)-2-(6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy iphenyl)- 1 -(tetrahydro-2H -pyran ⁇ 2-yl)- l/J-indazol-3 -yl)- 1H-imidazol-4- yl)metbyl)phosphonate (3-19). To a vial of 3-18 (1.108 g, 1 .64 mmol) was added neat triethyl phosphite (1 mL, 5.83 mmol).
  • reaction mixture was heated to 110 °C for 16 b. Following this, the reaction mixture was cooled to room temperature, concentrated under reduced pressure, and purified by silica gel chromatography (30-100% EtOAc/hexanes gradient) to afford the desired product (1.03 g, 81% yield), (m/z): [M+H] + calcd for C 36 H 54 N 5 O 8 SSi776.32, found 775.85.
  • Step Q Preparation of tert-butyl (3S)-3-((E)-2-(l -(N,N-dimethylsulfamoyl)-2-(6-(2-ethyl-4-((2- (trimethylsily l)ethoxy)methoxy )phenyl)- 1 -(tetrahydro-2H-pyran-2-yl)- 1H-indazol-3 -yl)- 1H-imidazol-4- yl)vinyl)morpholme-4-carboxylate (3-20).
  • a solution of 3-19 150 mg.
  • Step R Preparation of tert-butyl (3S)-3-((E)-2-(2-(6-(2-ethyl-4-hydroxyphenyl)-l-(tetrahydro-2H- pyran-2-yl)-1H-indazol-3-yl)-1H-imidazol-4-yl)vinyi)morpholine-4-carboxylate (3-21).
  • 3-20 55 mg, 0.066 mmol
  • methanol (1 mL
  • water 1 mL
  • reaction mixture was heated at 45 °C for 4 h, until complete removal of both SEM and sulfonyl urea protecting groups w*as observed.
  • the reaction mixture was then cooled to room temperature, diluted with 1 :1 ACN/water (10 mL) and the solution was frozen and lyophilized.
  • the dried residue was dissolved in THF (2 mL).
  • a solution of sodium borohydride (2.0 mg) in 1 : 1 THF/ethanol (2 mL) was slowly added, and the reaction mixture was stirred for 30 minutes. The volatiles were removed under reduced pressure and the crude product was used directly in the next step without further purification, (m/z): [M+H] * calcd for C 34 H 41 N 5 O 5
  • Step S Preparation of (AE)-3-ethyl-4-(3-(4-(2-(morphohn-3-yl)vinyl)- 1H-imidazoi-2-yl)-1H- indazol- 6-yl)phenol (6).
  • the reaction mixture was heated to 45 °C and stirred for 3 h.
  • the reaction mixture was then concentrated under reduced pressure, and the residue was purified by preparative HPLC chromatography (5-40% ACN/water gradient with 0.05% trifluoroacetic acid) to afford the TEA. salt of the title compound as a white solid (2 mg, 5.9% yield), (m/z): [M+H] + calcd for C 24 H 25 N 5 O 2 416.20, found 416.10.
  • Example 4 Synthesis of (S,E)-3-ethyl-4-(7-fluoro-3-(4-(2-(l-methylpyrrolidin-2-yl)vinyl)-1H- imidazol-2-yl)-1H-indazol-6-yl)phenol (40).
  • Step A Preparation of tert-butyl (S,E )-2-(2-(4,4.5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)vinyl)pyrrolidine-l -carboxylate (4-2).
  • 2,2,6,6-tetramethylpiperidine 508 ⁇ L, 3.01 mmol
  • THF 3 mL
  • n-Butyllithium 2.5 M in hexanes, 1.205 mL, 3.01 mmol
  • Step B Preparation of tert-butyl (2S)-2-((E)-2-(2-(6-(2-ethyl-4-hydroxyphenyl)-7-fluoro-1- (tetrahydro-2H -pyran-2-yl)-1H -indazol-3-yl)-1H -imidazol-4-yl)vinyl)pyrrolidine-l-carboxylate (4-3).
  • Step C Preparation of (S,E )-3-ethyl-4-(7-fluoro-3-(4-(2-(pyrrolidin-2-yl)vinyl)-1H -imidazol-2-yl)- 1H -indazol-6-yl)phenol (4-4).
  • 4-3 124 mg, 0.206 mmol
  • DCM dimethyl sulfoxide
  • trifluoroacetic acid 2 mL
  • Step D Preparation of (S,E)-3-ethyL4-(7-fluoro-3-(4-(2-(l-methylpyrrolidin-2-yl)vinyl )-1H - imidazol-2-yl)-1H -indazol-6-yl)phenol (40).
  • 4-4 10 mg, 0.024 mmol
  • formaldehyde 37 wt% in water, 100 ⁇ L, 0.077 mmol
  • sodium cyanoborohydride 7.5 mg, 0.120 mmol.
  • Example 5 Synthesis of 3-ethyl-4-(7-fluoro-3-(4-(piperidin-4-ylidenemethyl)-lH-imidazol-2-yl)- lH-indazol-6-yl)phenol (47).
  • Step A Preparation of tert-butyl 4-((4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)methylene)piperidine-1 -carboxylate (5-2).
  • 2,2,6,6-tetramethylpiperidine 508 ⁇ L, 3.01 mmol
  • THF 3 mL
  • n-Butyllithium 2.5 M in hexanes, 1.205 mL, 3.01 mmol
  • Step B Preparation of tert-butyl 4-((2-(6-(2-ethyl-4-hydroxy phenyl)- 7-fluoro-1 -(tetrahydro -2H - pyran-2-yl)-1H -indazol-3-yl)-1H -imidazol-4-yl)methylene)piperidine-l -carboxylate (5-3).
  • To a vial was added 2-10 (230 mg, 0.474 mmol) and 5-2 (153 mg, 0.474 mmol) in dioxane (3 mL), followed by a solution of sodium carbonate (151 mg, 1.422 mmol) in water (1 mL).
  • Step C Preparation of 3-ethyl-4-(741uoro-3-(4-(piperidin-4-ylidenemethyl)4#-imidazol-2-yi)-1H - indazol-6-yl)phenol (47).
  • Example 6 Synthesis of 3-ethyl-4-(3-(4-(3-(4-methylpiperazin-l-yl)prop-l -yn-l-yl)-1H -imidazol-2- y1)-1H-indazol-6-yl)phenol (66).
  • Step A Preparation of 3-(4,5-diiodo-1H -imidazol-2-yl)-6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy)phenyr)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (6-1).
  • Step B Preparation of 4-(3-(4,5-diiodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazol- 6-yl)-3 -ethylphenol (6-2).
  • Compound 6-1 (1.95 g, 2.53 mmol) was dissolved in a mixture of THF (30 mL), methanol (10 mL), and water (10 mL), then trifluoro acetic acid (3.90 mL, 50,6 mmol) was added and the reaction mixture was stirred at 50 °C for 6 h.
  • Step C Preparation of 3-ethyl-4-(3-(4-iodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran ⁇ 2-yl)-1H- indazol-6-yl)phenol (6-3).
  • Sodium sulfite (3.63 g, 28.8 mmol) was added to a solution of 6-2 (1 .23 g, 1.92 mmol) in ethanol (26 mL) and water (13 mL), then the reaction vessel was sealed and heated with stirring at 110 °C for 2 days. The reaction was then cooled, water (20 mL) was added, and the mixture was extracted with ethyl acetate (2x60 mL).
  • Step D Preparation of 3-ethyl-4-(3 -(4-(3 -(4-methylpiperazin- 1 -y l)prop-1 -yn- 1 -y 1)- 1H-imidazol-2- yl)-1H-indazol ⁇ 6-yl)phenol (66).
  • 1-methyl-4 ⁇ (prop-2-yn ⁇ l-yl)piperazine (18.8 mg, 0.136 mmol), 6-3 (35 mg, 0.068 mmol), and triethylamine (28 uL, 0.204 mmol) were dissolved in DMF (1.2 mL), then the reaction was degassed with nitrogen for 10 minutes.
  • Cuprous iodide (2.6 mg, 0,014 mmol) and bis(triphenylphosphine)- palladium(II)chloride (4.8 mg, 0.0068 mmol) were added, the reaction was again degassed with nitrogen for 5 minutes, then the reaction was sealed and stirred at 80 °C for 1 h. The reaction mixture was concentrated and the residue was dissolved in DCM (1 mL), then trifluoroacetic acid (0.5 mL) was added and the reaction mixture was stirred at 45 °C for 40 minutes.
  • Example 7 Synthesis of 4-(3-(4-(azetidm-3-ylethynyl)-1H-imidazol-2-yl)-1H-indazol-6-yl)-3- ethylphenol (120).
  • Example 8 Synthesis of (R)-3-ethyl-4-(7-fluoro-3-(4-(morpholin-3-ylethynyl)-1H-imidazol-2-yl)- 1H-indazol-6-yl)phenol (22).
  • Step A Preparation of tert-butyl (R)-3-ethynylmorpholine-4-carboxylate (8-2).
  • (S)-N-Boc-3- morpholmecarbaldehyde (30 mg, 0.139 mmol) was dissolved in methanol (2.0 mL), then potassium carbonate (38,5 mg, 0.279 mmol) was added, followed by dimethyl (2-oxopropanimidoy1)phosphonate (32.3 mg, 0,167 mmol), and the reaction mixture was stirred at room temperature. After judged complete by TLC (24 h), the reaction mixture was concentrated, then the residue was partitioned between ethyl acetate (3 mL) and water (3 mL).
  • Step B Preparation of 3-(4,5-diiodo-1H-imidazol-2-yl)-6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yI)-1H-indazole (8-3).
  • a solution of 2-7 (1 .0 g, 1.86 mmol) was dissolved in DCM (23 ml.) and the solution was cooled to 0 °C.
  • N- iodosuccinamide (0.92 g, 4.10 mmol) in DCM (4 mL) and THF (3 mL) was then added dropwise, and the reaction mixture was stirred at 0 °C for 3 h, then at ambient temperature for 16 h.
  • the reaction was quenched by addition of saturated aqueous sodium sulfite (40 mL).
  • the mixture was then extracted with DCM (40 mL), and the organic layer was washed with brine (40 mL), dried over sodium sulfate, and concentrated under reduced pressure.
  • the crude product was used without further purification.
  • Step C Preparation of 4-(3-(4,5-diiodo-1H-imidazol-2-yl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H- indazol-6-yl)-3 -ethylphenol (8-4).
  • Compound 8-3 (1.56 g, 1.98 mmol) was dissolved in a mixture of THF (22.5 mL), methanol (7.5 mL), and water (7.5 mL), then trifluoroacetic acid (3.05 mL, 39.6 mmol) was added and the reaction mixture was stirred at 50 °C for 6 h.
  • Step D Preparation of 3-ethyl-4-(7-fluoro-3-(4-iodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2- yl)-1H-indazol-6-yl)phenol (8-5).
  • Sodium sulfite (3.58 g, 28.4 mmol) was added to a solution of 8-4 (0.93 g, 1.42 mmol) in ethanol (19 mL) and water (9.5 mL), then the reaction vessel was sealed and heated with stirring at 110 °C for 2 days.
  • Step E (R)-3-ethyl-4-(7-fluoro-3-(4-(morpholin-3-ylethynyl)-1H -imidazol-2-yl)-1H-indazol-6- yl)phenol (22).
  • Compound 8-5 25 mg, 0.047 mmol
  • tert-butyl (R)-3-ethynylmorpholine-4-carboxyiate (8-2) 24,80 mg, 0.117 mmol
  • triethylamine 0.020 ml, 0. 141 mmol
  • Example 9 Synthesis of 3-etliyl-4-(7-fluoro-3-(4-(((2S,4S)-4-methoxypyiToIidin-2-yl)ethynyI)-1H- imidazol-2-; rl)-1H-indazoi-6-yl)phenol (49).
  • Step A Preparation of tert-butyl (2S,4S)-2-ethynyl-4-hydroxypyrrolidine-l -carboxylate (9-2).
  • DIBAL-H IM in toluene, 20.4 mL, 20.4 mmol
  • Potassium carbonate (2.25 g, 16.3 mmol) was added, followed by the addition of dimethyl (l-diazo-2-oxopropyl)phosphonate (1.88 g, 9,78 mmol), and the resulting mixture w'as allowed to warm to room temperature and stir for 16 h.
  • the organic layer was separated, washed with brine (30 mL), dried over sodium sulfate, and concentrated under reduced pressure.
  • Step B Preparation of tert-butvl (2S,4S)-2-ethynyl-4-methoxypyrrolidine-l -carboxylate (9-3).
  • 9-2 (0.75 g, 3.55 mmol) in DMF (5 mL)
  • sodium hydride (60% dispersion in mineral oil, 156 mg, 3.91 mmol) at -20 °C and the resulting solution was stirred for 30 minutes.
  • Methyl iodide (265 ⁇ L, 4.26 mmol) was then added and the reaction mixture wrns allowed to warm to room temperature and stir for 5 h.
  • Step C Preparation of 3-ethyl-4-(7-fluoro-3-(4-(((2S,4S)-4-methoxypyrrolidin-2-yl)ethynyl)-1H- itnidazol-2-yl)-TH-indazol-6-yl)phenol (49).
  • Compound 8-5 50 mg, 0.094 mmol
  • 9-3 52.9 mg, 0.235 mmol
  • triethylamine 0.039 ml, 0.282 mmol
  • Example 10 Biochemical JAK Kinase Assays to Measure pKi.
  • a panel of four LanthaScreen JAK biochemical assays (JAK1, 2, 3 and Tyk2) were carried in a common kinase reaction buffer (50 mM HEPES, pH 7.5, 0.01% Brij-35, 10 mM MgCL, and 1 mM EGTA).
  • Recombinant GST-tagged JAK enzymes and a GFP-tagged ST ATI peptide substrate were obtained from Life Technologies.
  • Table 2 shows biological activities of selected compounds in a biochemical JAK assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
  • Example 11 Cellular JAKI Potency Assay to Measure pICso-
  • the JAKI cellular potency assay was carried out by measuring inhibition of interleukin- 13 (IL-13, R&D Systems) induced STAT6 phosphorylation in BEAS-2B human lung epithelial ceils (ATCC), BEAS-2B cells were grown at 37°C in a 5% CO2 humidified incubator in 50% DMEM/50% F-12 medium (Life Technologies) supplemented with 10% FBS (Hyclone), 100 U/mL penicillin, 100 ⁇ g/mL streptomycin (Life Technologies), and 2 mM GlutaMAX (Life Technologies).
  • IL-13 interleukin- 13
  • R&D Systems induced STAT6 phosphorylation in BEAS-2B human lung epithelial ceils
  • ceils were seeded at a 7,500 cells/well density in white poly-D-lysine-coated 384-weil plates (Coming) with 25 ⁇ L medium and were allowed to adhere overnight in the incubator.
  • the medium was removed and replaced with 12 ⁇ L of assay buffer (Hank's Balanced Salt Solution/HBSS, 25mM HEPES, and 1 mg/ml bovine serum album in/BSA) containing dose-responses of test compounds.
  • assay buffer Hank's Balanced Salt Solution/HBSS, 25mM HEPES, and 1 mg/ml bovine serum album in/BSA
  • Cells were incubated with test compounds at 37 °C for 1 h and followed by the addition of 12 ⁇ l of pre-wanned IL-13 (80 ng/mL in assay buffer) for stimulation. After incubating at 37 °C for 30 min, the assay buffer (containing compound and IL- 13) was removed, and 10 ⁇ L of cell lysis buffer added (25 mM HEPES, 0.1 % SDS, 1 % NP-40, 5 mM MgCh, 1.3 mM EDTA, 1 mM EGTA, supplemented with Complete Ultra mini protease inhibitors and PhosSTOP from Roche Diagnostics). The plates were shaken at ambient temperature for 30min before the addition of detection reagents.
  • cell lysis buffer 25 mM HEPES, 0.1 % SDS, 1 % NP-40, 5 mM MgCh, 1.3 mM EDTA, 1 mM EGTA, supplemented with Complete Ultra mini protease inhibitors and PhosST
  • pSTAT6 levels of pSTAT6 were measured using the AlphaLISA SureFire Ultra pSTAT6 (Tyr641) assay kit from PerkinElmer. For dose-response analy sis, percent inhibition data were plotted vs. compound concentrations, and ICso values were determined from a 4-parameter robust fit model with the Graphpad Prism software.
  • Data are expressed as pICLu (negative decadic logarithm IC50) values. Certain compounds disclosed herein exhibited pICso values of greater than 6 or greater than 7 when tested in BEAS-2B cells. Test compounds having a lower IC50 value or higher pICso value in this assay show greater inhibition of IL-13 induced STAT6 phosphorylation.
  • Table 3 shows biological activities of selected compounds in a cellular JAK potency assay.
  • Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
  • Example 12 Cytotoxicity Measured by Premature Chromosome Condensation [15] (pCC’15).
  • ATP cellular adenosine triphosphate
  • Beas2B cells were grown in 50% DMEM (Life Technologies) and 50% F-12 (Life Technologies) media, supplemented with 10% Fetal Bovine Serum (ATCC), 25 mM HEPES (Life Technologies), and lx Pen-Strep (Life Technologies). Cells were cultured in a humidified incubator set at 37 °C, 5% CO2, and trypsinized using 0.25% Trypsin with 0.5% polyvinylpyrrolidone (PVP). [0285] For the assay, Beas2B cells were seeded at 500 cells/well (25
  • PVP polyvinylpyrrolidone
  • the effect of the compound on cell viability was determined through analysis of dose-dependent quantified changes in ATP from baseline (non-compound treated cells) and wells treated with 60 pM AT9283, a well-characterized cytotoxic compound. Data are expressed as pCCi5 (negative decadic logarithm CC15) values. Certain compounds disclosed herein exhibited pCCi5 values of less than 6, or less than 5.7, or less than 5.5 when tested in Beas2B cells.
  • Table 4 shows cytotoxicities of selected compounds in a premature chromosome condensation assay.
  • Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
  • Example 13 Murine model of IL-13 induced pSTAT6 induction in lung tissue.
  • IL-13 binds to cell surface receptors activating members of the Janus family of kinases (JAK) which then phosphorylate STAT6 and subsequently activate further transcription pathways.
  • JK Janus family of kinases
  • a dose of IL-13 was delivered locally into the lungs of mice to induce the phosphorylation of STAT6 (pSTAT6) which is then measured as the endpoint.
  • Selected compounds of the present disclosure were tested in the assay. Activity in the model is evidenced by a decrease in the level of pSTAT6 present in the lungs of treated animals at 9 hours compared to the vehicle treated, IL-13 challenged control animals. The difference between the control animals which were vehicle treated, IL-13 challenged and the control animals which were vehicle treated, vehicle challenged dictated the 0% and 100% inhibitory effect, respectively, in any given experiment. Exemplary’ compounds were tested in the assay and exhibited inhibition of STAT6 phosphoiylation at 9 hours after IL- 13 challenge as documented below.
  • Table 5 shows inhibition of STAT6 phosphorylation of selected compounds in the murine model.
  • Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
  • Example 14 Pharmacokinetics in Plasma and Lung in Mouse After Oral Aspiration of Test Compounds.
  • Plasma and lung concentrations of test compounds were quantified and pharmacokinetic parameters were calculated in the following manner.
  • Male CD1 mice from Charles River Laboratories were used in the pharmacokinetic studies.
  • Test compounds were individually formulated in 20% propylene glycol in pH 4 citrate buffer at a concentration of 0.2 mg/mL.
  • Test compounds were administered in two, 25 gL increments introduced into the trachea of each mouse by oral aspiration using a calibrated pipette once the animal was anesthetized using isoflurane. Blood samples were collected as terminal collections via cardiac puncture at 0.167, 1, 4, 8, and 24 hr post-dosing.
  • Plasma and lung concentrations of test compounds were determined by LC-MS/MS analysis against analytical standards constructed into a standard curee in the test matrix.
  • the pharmacokinetic parameters of test compounds were determined by non -com partmental analysis. For concentrations below the limit of quantification, zero was used for mean calculations. Mean values were not reported if more than 50% of the samples were below the limit of quantification at a timepoint, or if more than 50% of a calcuiated pharmacokinetic parameter was not reportable.
  • AUQo-inf AUCH + Ciw / k
  • AUCc&p the area under the concentration-time curve from the time of dosing to the last measurable concentration calculated by the linear trapezoidal rule
  • Ciast the last measurable concentration
  • k the first order rate constant associated with the terminal elimination phase, estimated by linear regression of time versus log concentration.
  • the lung-to-plasma AUC ratio was determined as the ratio of the lung AUC(o-inf) in ⁇ g*hr/g to the plasma AUC(o-inf) in ⁇ g*hr/mL. Certain compounds disclosed herein exhibited lung-to-plasma AUC ratios of greater than 100, such as greater than 200, in male CD1 mice.
  • Table 6 shows plasma AUC(o-24) values, lung tissue AUC(o-24) values and the lung-to-plasma AUC ratios of selected compounds as assessed in accordance with this Example.
  • H- H- denotes a value ⁇ 0.5
  • +++ denotes a value from 0.6 to 1 .0
  • ++ denotes a value from 1 .1 to 1 .5.
  • ++++ denotes a value from 101 to 200
  • +++ denotes a value from 51 to 100
  • ++ denotes a value from 10 to 50.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Transplantation (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides inhibitors of the Janus family of tyrosine kinases (JAK). Also disclosed are methods to modulate the activity of JAK and methods of treatment of disorders mediated by JAK.

Description

BICYCLIC INHIBITORS OF JAK AND METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of United States Provisional Application
Serial Number 63/281,218, filed November 19, 2021, the contents of which are hereby incorporated by reference in their entirety.
BACKGROUND
[0002] Asthma is a chronic disease of the airways for which there is no prevention or cure. The disease is characterized by inflammation, fibrosis, hyper-responsiveness, and remodeling of the airways, all of which contribute to airflow limitation. An estimated 300 million people worldwide suffer from asthma and it is estimated that the number of people with asthma will grow by more than 100 million by 2025. In the United States, asthma afflicts about 6 to 8% of the population, making it one of the most common chronic diseases in the country. Although most patients can achieve control of asthma symptoms with the use of inhaled corticosteroids that may be combined with a leukotriene modifier and/or a long-acting beta agonist, there remains a subset of patients with severe asthma whose disease is not controlled by conventional therapies. Severe persistent asthma is defined as disease that remains uncontrolled on high doses of inhaled corticosteroids. While patients with severe asthma are estimated to account for approximately 5% of all asthma patients, they have a high risk of morbidity and mortality and are responsible for a disproportionate share of health care resource utilization among patients with asthma. There remains a need for novel therapies to treat these patients.
[0003] Cytokines are intercellular signaling molecules which include chemokines, interferons, interleukins, lymphokines, and tumor necrosis factor. Cytokines are critical for normal cell growth and immunoregulation but also drive immune-mediated diseases and contribute to the growth of malignant cells. Elevated levels of many cytokines have been implicated in the pathology of asthma inflammation. For example, antibody-based therapies targeted at interleukins (IL)-5 and 13 have been shown to provide clinical benefit in subsets of severe asthma patients. Among the cytokines implicated in asthma inflammation, many act through signaling pathways dependent upon the Janus family of tyrosine kinases (JAKs), which signal through the Signal Transducer and Activator of Transcription (STAT) family of transcription factors. Cytokines implicated in asthma inflammation which signal through the JAK-STAT pathway include IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-11 , IL-13, IL-23, IL-31, IL-27, thymic stromal lymphopoietin (TSLP), interferon-y (IFNy) and granulocyte-macrophage colony-stimulating factor (GM-CSF).
[0004] The JAK family comprises four members, JAKI, JAK2, JAK3, and tyrosine kinase 2 (TYK2). Binding of cytokine to a JAK-dependent cytokine receptor induces receptor dimerization which results in phosphorylation of tyrosine residues on the JAK kinase, effecting JAK activation. Phosphoiylated JAKs, in turn, bind and phosphorylate various STAT proteins which dimerize, internalize in the cell nucleus and directly modulate gene transcription, leading, among other effects, to the downstream effects associated with inflammatory disease. The JAKs usually associate with cytokine receptors in pairs as homodimers or heterodimers. Specific cytokines are associated with specific JAK pairings. Each of the four members of the JAK family is implicated in the signaling of at least one of the cytokines associated with asthma inflammation. Consequently, a chemical inhibitor with pan-activity against all members of the JAK family could modulate a broad range of pro-inflammatory pathways that contribute to severe asthma.
[0005] However, the broad anti-inflammatory effect of such inhibitors could suppress normal immune cell function, potentially leading to increased risk of infection. Evidence of increased infection risk has been observed with the JAK inhibitor tofacitinib, which is dosed orally for the treatment of rheumatoid arthritis. In asthma, inflammation is localized to the respiratory' tract. Inflammation of the airways is characteristic of other respiratory diseases in addition to asthma. Chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), pneumonitis, interstitial lung diseases (including idiopathic pulmonary fibrosis), acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, and sarcoidosis are also respiratory tract diseases in which the pathophysiology is believed to be related to JAK-signaling cytokines. Local administration of a JAK inhibitor to the lungs by inhalation offers the potential to be therapeutically efficacious by delivering a potent anti-cytokine agent directly to the site of action, limiting systemic exposure and therefore limiting the potential for adverse systemic immunosuppression. The need remains for a potent JAK inhibitor suitable for local administration to the lungs for treatment of respiratory disease.
[0006] JAK-signaling cytokines also play a major role in the activation of T cells, a sub-type of immune cells that is central to many immune processes. Pathological T cell activation is critical in the etiology of multiple respiratory diseases. Autoreactive T cells play a role in bronchiolitis obliterans organizing pneumonia (also termed COS). Similar to COS, the etiology of lung transplant rejections is linked to an aberrant T cell activation of the recipient s T cells by the transplanted donor lung. Lung transplant rejections may occur early as Primary Graft Dysfunction (PGD), organizing pneumonia (OP), acute rejection (AR) or lymphocytic bronchiolitis (LB) or they may occur years after lung transplantation as Chronic Lung Allograft Dysfunction (CLAD). CLAD was previously known as bronchiolitis obliterans (BO) but now is considered a syndrome that can have different pathological manifestations including BO, restrictive CLAD (rCLAD or RAS) and neutrophilic allograft dysfunction. Chronic lung allograft dysfunction (CLAD) is a major challenge in longterm management of lung transplant recipients as it causes a transplanted lung to progressively lose functionality (Gauthier et al., Curr. Transplant. Rep. 2016, 3(3), 185—191). CLAD is poorly responsive to treatment and therefore, there remains a need for effective compounds capable of preventing or treating this condition. Several JAK-dependent cytokines such as IFNy and IL-5 are up-regulated in CLAD and lung transplant rejection (Berastegui et al., Clin. Transplant. 2017, 31, el2898). Moreover, high lung levels of CXCR3 chemokines such as CXCL9 and CXCL10, which are downstream of JAK-dependent IFN signaling, are linked to worse outcomes in lung transplant patients (Shino et al, PLOS One, 2017, 12 (7), eOl 80281). Systemic JAK inhibition has been shown to be effective in kidney transplant rejection (Vicenti et al., American Journal of Transplantation 2012, 12, 2446-56). Therefore, JAK inhibitors have the potential to be effective in treating or preventing lung transplant rejection and CLAD, Similar T cell activation events as described as the basis for lung transplant rejection also are considered the main driver of lung graft-versus- host disease (GVHD) which can occur post hematopoietic stem cell transplants. Similar to CLAD, lung GVHD is a chronic progressive condition with extremely poor outcomes and no treatments are currently approved. A retrospective, multicenter survey study of 95 patients with steroid-refractory acute or chronic GVHD who received the systemic JAK inhibitor ruxolitinib as salvage therapy demonstrated complete or partial response to ruxolitinib in the majority of patients including those with lung GVHD (Zeiser et al., Leukemia 2015, 29, 10, 2062-68). As systemic JAK inhibition is associated with serious adverse events and a small therapeutic index, the need remains for an inhaled lung-directed, non-systemic JAK inhibitor to prevent and/or treat lung transplant rejection or lung GVHD,
SUMMARY
[0007] One of the main problems with JAK inhibitors developed to date is that these molecules have been associated with adverse immunosuppression resulting from significant systemic exposure from oral administration. In view of the foregoing, a need exists for small molecules that target JAK and for use of such compounds in the treatment of various JAK-mediated diseases, such as asthma, chronic obstructive pulmonary disease, and lung transplant rejection, while limiting adverse side effects. The present disclosure provides these and other related advantages. One objective of the present disclosure is to deliver a potent JAK inhibitor locally with minimal systemic exposure in order to address any unintended and unwanted systemic side effects of JAK inhibition during treatment. Therefore, in some aspects, the present disclosure provides inhaled, long-acting and lung-selective JAK inhibitors for the treatment of a respiratory disease, such as asthma, chronic obstructive pulmonary disease, or lung transplant rejection. Compounds of the present disclosure may be used as a monotherapy or co-dosed with other therapies, whether delivered by inhalation, orally, intravenously, subcutaneously, or topically.
[0008] In certain aspects the present disclosure provides a compound of Formula (I):
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected at each occurrence from halogen, -OH, -O(C1-4 alkyl), -O(C1-4 haloalkyl), and C1-4 alkyl;
R2 is selected from hydrogen, halogen, and C1-4 alkyl;
R3 is independently selected at each occurrence from halogen and C1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond; when is a double bond, then A is CR4R3; when is a triple bond, then A is CR6;
R4 and R5 are each independently selected from hydrogen and RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from hydrogen and RA;
RA is independently selected at each occurrence from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =0, =S, =N(R10);
Ci-10 alkyl, C2.io alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =0, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and
C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10,
-NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =0, =S, =N(R10), R10, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, Co-3 alkyl-(C3-12 carbocycle), and Co-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN,
-NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =0, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -OCH2CH2-OCH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10; and
R7 is hydrogen or C1-6 alkyl.
[0009] In certain aspects the present disclosure provides a compound of Formula (I’):
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected at each occurrence from halogen, -OH, -O(C1-4 alkyl), -O( C1-4 haloalkyl), and C1-4 alkyl;
R2 is selected from hydrogen, halogen, and C1-4 alkyl;
R3 is independently selected at each occurrence from halogen and C1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond; when is a double bond, then A is CR4R5; when
Figure imgf000006_0002
is a triple bond, then A is CR6;
R4 and R5 are each independently selected from hydrogen and RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from hydrogen and RA;
RA is independently selected at each occurrence from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10,
-S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10,
-NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10,
-OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10);
C1-10 alkyl, C2-10 alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10,
-NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -0C(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), R10, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)0H, -C(O)NH2, =O, -OH, -CH20H, -CH2CH2OH, -0CH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10.
[0010] For a compound or salt of Formula (I) or (I’), m may be an integer from 1 to 3, such as m is 2. In some embodiments, n is 0.
[0011] A compound of Formula (I) or (I’) may be a compound of Formula (I-A):
Figure imgf000007_0001
(I-A), or a pharmaceutically acceptable salt thereof. [0012] For a compound or salt of Formula (I), (I’), or (I-A), R1 may independently be selected at each occurrence from halogen, -OH, and -CH2CH3.
[0013] A compound of Formula (I) or (I’) may be a compound of Formula (I-D):
Figure imgf000008_0001
(I-D), or a pharmaceutically acceptable salt thereof.
[0014] In some embodiments, for a compound or salt of Formula (I), (F), (I-A), or (I-D):
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA; and
R6 is selected from RA;
RA is independently selected at each occurrence from: halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12;
C1-10 alkyl, optionally substituted at each occurrence with one or more substituents selected from halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR 11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, R10, C1-6 alkyl, and C1-6 haloalkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)2, C3-6 carbocycle, and 3- to 6-membered heterocycle. [0015] In some embodiments, for a compound or salt of Formula (I), (I’), (I-A) or (I-D):
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-6 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C0-3 alkyl-(C3-i2 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
[0016] For a compound or salt of Formula (I), (I’), (I -A) or (I-D),
Figure imgf000009_0001
may be a double bond, wherein A is CR4R5. For example, a compound of Formula (I) may be a compound of Formula (I-E):
Figure imgf000009_0002
(I-E), or a pharmaceutically acceptable salt thereof.
[0017] In some embodiments, for a compound or salt of Formula (I-E):
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more RA; and
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NHS(=O)2R10, -NHC(O)R10, and 4- to 10-membered heterocycle; and
4- to 10-membered heterocycle, wherein each 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from -OCH3, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-6 carbocycle), and C0-3 alkyl-(3- to 6-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -N(CH3)2, =O, -OH, and -CH3.
[0018] For a compound or salt of Formula (I), (I’), (I-A), (I-D) or (I-E), may be selected from
Figure imgf000010_0001
Figure imgf000011_0001
[0019] For a compound or salt of Formula (I), (I’), (I-A) or (I-D), may be a triple bond, wherein A is CR". For example, a compound of Formula (I) may be a compound of Formula (I-F):
Figure imgf000011_0002
(I-F), or a pharmaceutically acceptable salt thereof
[0020] In some embodiments, for a compound or salt of Formula (I-F):
R6 is selected from: C1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, C3 -10 carbocycle, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in R6 is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen and C1-4 alkyl.
[0021] For a compound or salt of Formula (I), (I’), (I-A), (I-D) or (I-F),
Figure imgf000011_0004
may be selected from
Figure imgf000011_0003
Figure imgf000012_0001
[0022] For a compound or salt of Formula (I), (F), (I-A), (I-D), (I-E) or (I-F), R2 may be selected from hydrogen and halogen, such as R2 is hydrogen. In some embodiments, R2 is F.
[0023] In some embodiments, for a compound or salt of Formula (I-D):
R2 is selected from hydrogen and F;
R4 is selected from RA and R5 is selected from hydrogen and CH3: R4 is selected from hydrogen andCH3 and R5 is selected from RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-io carbocycle), and C0-3 alkyl-(4- to 10-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
[0024] In some embodiments, for a compound or salt of Formula (I-D): R2 is selected from hydrogen and F; and
Figure imgf000013_0001
[0025] In some embodiments, for a compound or salt of Formula (I-D):
R2 is selected from hydrogen and F; and
Figure imgf000013_0002
[0026] In some embodiments, for a compound or salt of Formula (I-D):
R2 is selected from hydrogen and F; and
Figure imgf000013_0003
Figure imgf000014_0001
[0027] In some embodiments, for a compound or salt of Formula (I-D):
R2 is selected from hydrogen and F; and
Figure imgf000014_0002
[0028] In certain aspects, the present disclosure provides a substantially pure stereoisomer of a compound or salt disclosed herein, such as a compound or salt of Formula (I), (I’), (I-A), (I-D), (I-E) or (I-F), or a compound disclosed in Table 1. The stereoisomer may be provided in at least 90% enantiomeric excess. In certain aspects, the present disclosure provides a compound selected from Table 1, or a pharmaceutically acceptable salt thereof.
[0029] In certain aspects, the present disclosure provides a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition further comprises one or more additional therapeutic agents. The pharmaceutical composition may be formulated for inhalation.
[0030] In certain aspects, the present disclosure provides a method of inhibiting JAK, comprising contacting JAK with an effective amount of a compound or salt disclosed herein. In certain aspects, the present disclosure provides a method of treating a JAK-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein. The disease or condition may be a respiratory disease. In certain aspects, the present disclosure provides a compound or salt disclosed herein for use in treating a respiratory disease. In certain aspects, the present disclosure provides the use of a compound or salt disclosed herein for the manufacture of a medicament for treating a respiratory disease. The respiratory disease may be selected from asthma, chronic obstructive pulmonary disease, cystic fibrosis, pneumonitis, idiopathic pulmonary fibrosis, acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, sarcoidosis, an eosinophilic disease, a helminthic infection, pulmonary arterial hypertension, lymphangioleiomyomatosis, bronchiectasis, an infiltrative pulmonary disease, drag-induced pneumonitis, fungal induced pneumonitis, allergic bronchopulmonary aspergillosis, hypersensitivity pneumonitis, eosinophilic granulomatosis with polyangiitis, idiopathic acute eosinophilic pneumonia, idiopathic chronic eosinophilic pneumonia, hypereosinophilic syndrome, Loffler syndrome, bronchiolitis obliterans organizing pneumonia, lung graft-versus-host disease, coronavirus infections (e.g., COVID-19, SARS, MERS), chronic rhinosinusitis with or without nasal polyps, nasal polyposis, sinusitis with nasal polyps, rhinitis, and immune-checkpoint-inhibitor induced pneumonitis. In some embodiments, the respiratory disease is selected from asthma and chronic obstructive pulmonary disease. The asthma may be selected from T2-dominant (eosinophilic) asthma and non-T2-dominant (non- eosinophilic) asthma.
[0031] In certain aspects, the present disclosure provides a method of treating lung transplant rejection in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein. In certain aspects, the present disclosure provides a compound or salt disclosed herein for use in treating lung transplant rejection. In certain aspects, the present disclosure provides the use of a compound or salt disclosed herein for the manufacture of a medicament for treating lung transplant rejection. The lung transplant rejection may be selected from primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is acute lung transplant rejection. In some embodiments, the lung transplant rejection is chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is selected from bronchiolitis obliterans, restrictive chronic lung allograft dysfunction, and neutrophilic allograft dysfunction. [0032] Any of the subject methods may further comprise administering a second therapeutic agent. In practicing any of the subject methods, a compound or salt disclosed herein may be administered by inhalation.
INCORPORATION BY REFERENCE
[0033] All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
DETAILED DESCRIPTION
[0034] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs.
[0035] Chemical structures are named herein according to IUPAC conventions as implemented in ChemDraw® software (Perkin Elmer, Inc., Cambridge, MA).
[0036] As used in the specification and claims, the singular forms “a”, “an” and “the” include plural references unless the context clearly dictates otherwise.
[0037] The term “Cx-y” or “Cx-Cy” when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl, is meant to include groups that contain from x to y carbons in the chain. For example, the term “Cx.y alkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, that contain from x to y carbons in the chain. [0038] “Alkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including linear and branched alkyl groups. An alkyl group may contain from one to twelve carbon atoms (e.g., C1-12 alkyl), such as one to eight carbon atoms (C1-8 alkyl) or one to six carbon atoms (C1-6 alkyl). Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, and decyl. An alkyl group is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0039] “Haloalkyl” refers to an alkyl group that is substituted by one or more halogens. Exemplary haloalkyl groups include trifluoromethyl, difluoromethyl, trichloro methyl, 2,2,2-trifluoroethyl, 1,2 -difluoroethyl, 3- bromo-2-fluoropropyl, and 1 ,2-dibromoethyl.
[0040] “Alkenyl” refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkenyl groups, containing at least one double bond. An alkenyl group may contain from two to twelve carbon atoms (e.g., C2-12 alkenyl), such as two to eight carbon atoms (C2-8 alkenyl) or two to six carbon atoms (C2-6 alkenyl). Exemplary alkenyl groups include ethenyl (i.e., vinyl), prop-l-enyl, but-l-enyl, pent-l-enyl, penta- 1, 4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0041] “Alkynyl” refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkynyl groups, containing at least one triple bond. An alkynyl group may contain from two to twelve carbon atoms (e.g., C2-12 alkynyl), such as two to eight carbon atoms (C2-8 alkynyl) or two to six carbon atoms (C2-6 alkynyl). Exemplary alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0042] “Alkylene” or “alkylene chain” refers to substituted or unsubstituted divalent saturated hydrocarbon groups, including linear alkylene and branched alkylene groups, that contain from one to twelve carbon atoms (e.g., C1-12 alkylene), such as one to eight carbon atoms (C1-8 alkylene) or one to six carbon atoms (C1-6 alkylene). Exemplary alkylene groups include methylene, ethylene, propylene, and n-butylene. Similarly, “alkenylene” and “alkynylene” refer to alkylene groups, as defined above, which comprise one or more carbon-carbon double or triple bonds, respectively. The points of attachment of the alkylene, alkenylene or alkynylene chain to the rest of the molecule can be through one carbon or any two carbons of the chain. Unless stated otherwise specifically in the specification, an alkylene, alkenylene, or alkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
[0043] “Heteroalkyl”, “heteroalkenyl” and “hetero alkynyl” refer to substituted or unsubstituted alkyl, alkenyl and alkynyl groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quatemized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkyl group has a chain length of 3 to 8 atoms. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl, heteroalkenyl or heteroalkynyl chain. Unless stated otherwise specifically in the specification, a heteroalkyl, heteroalkenyl, or heteroalkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0044] “Hetero alkylene”, “hetero alkenylene” and “heteroalkynylene” refer to substituted or unsubstituted alkylene, alkenylene and alkynylene groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quatemized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8 -membered heteroalkylene group has a chain length of 3 to 8 atoms. The points of attachment of the heteroalkylene, hetero alkenylene or heteroalkynylene chain to the rest of the molecule can be through either one heteroatom or one carbon, or any two heteroatoms, any two carbons, or any one heteroatom and any one carbon in the heteroalkylene, hetero alkenylene or heteroalkynylene chain. Unless stated otherwise specifically in the specification, a heteroalkylene, heteroalkenylene, or heteroalkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
[0045] “Carbocycle” refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is a carbon atom. Carbocycle may include C3 -10 monocyclic rings, C2-6 bicyclic rings, C2-6 spirocyclic rings, and C6-12 bridged rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings. In some embodiments, the carbocycle is a C2-6 aryl group, such as C6-10 aryl. In some embodiments, the carbocycle is a C2-6 cycloalkyl group. In some embodiments, the carbocycle is a C2-6 cycloalkenyl group. As used herein, “cycloalkenyl” refers to a non-aromatic ring containing at least one double bond, wherein each atom of the ring is a carbon atom. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, are included in the definition of carbocycle. Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl. Unless stated otherwise specifically in the specification, a carbocycle is optionally substituted by one or more substituents such as those substituents described herein.
[0046] “Heterocycle” refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms, for example 1, 2 or 3 heteroatoms selected from O, S and N. Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, 6- to 12-membered spirocyclic rings, and 6- to 12- membered bridged rings. Each ring of a bicyclic heterocycle may be selected from saturated, unsaturated, and aromatic rings. The heterocycle may be attached to the rest of the molecule through any atom of the heterocycle, valence permitting, such as a carbon or nitrogen atom of the heterocycle. In some embodiments, the heterocycle is a 5 - to 10-membered heteroaryl group, such as 5- or 6-membered heteroaryl. In some embodiments, the heterocycle is a 3- to 12-membered heterocycloalkyl group. In some embodiments, the heterocycle is a 3- to 12-membered heterocycloalkyl group (i.e. a saturated or partially unsaturated cyclic ring having one or more (e.g., 1 to 5) heteroatoms). In an exemplary embodiment, a heterocycle, e.g., pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Exemplary heterocycles include pyrrolidinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, piperidinyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, thiophenyl, oxazolyl, thiazolyl, morpholinyl, indazolyl, indolyl, and quinolinyl. Unless stated otherwise specifically in the specification, a heterocycle is optionally substituted by one or more substituents such as those substituents described herein.
[0047] “Heteroaryl” refers to a 5- to 12-membered aromatic ring that comprises at least one heteroatom, such as 1, 2 or 3 heteroatoms, selected from O, S and N. As used herein, the heteroaryl ring may be selected from monocyclic or bicyclic — including fused, spirocyclic and bridged ring systems — wherein at least one of the rings in the ring system is aromatic. The heteroatom(s) in the heteroaryl may optionally be oxidized. One or more nitrogen atoms, if present, are optionally quatemized. The heteroaryl may be attached to the rest of the molecule through any atom of the heteroaryl, valence permitting, such as a carbon or nitrogen atom of the heteroaryl. Examples of heteroaryl groups include, but are not limited to, azepinyl, benzimidazolyl, benzisothiazolyl, benzisoxazolyl, benzofuranyl, benzothiazolyl, benzothiophenyl, benzoxazolyl, furanyl, imidazolyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, purinyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyridazolyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroquinolinyl, thiadiazolyl, thiazolyl, and thienyl groups. Unless stated otherwise specifically in the specification, a heteroaryl is optionally substituted by one or more substituents such as those substituents described herein.
[0048] Unless stated otherwise, hydrogen atoms are implied in structures depicted herein as necessary to satisfy the valence requirement.
[0049] A waved line drawn across a bond is used herein to denote where a bond disconnection or
Figure imgf000018_0001
attachment occurs. For example, in the structure
Figure imgf000018_0002
Rais attached to the para position of a fluorophenyl ring through a single bond. If Ra is 2-pyridine as in
Figure imgf000018_0003
, then Ra may be depicted as “
Figure imgf000018_0004
[0050] The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons or heteroatoms of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, heteroatoms such as nitrogen may have any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
[0051] A compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1, 2 or 3 substituents selected from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10);
C1-10 alkyl, C2-10 alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), R10, C1-6 alkyl, Cw haloalkyl, C2.6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, Co-3 alkyl-(C3-12 carbocycle), and Co-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH20H, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10.
[0052] In some embodiments, a compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1, 2 or 3 substituents selected from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10);
Ci-io alkyl, C2-10 alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, =O, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, =O, =S, =N(R10), C1-6 alkyl, C1-6 haloalkyl, C2.6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH20H, -CH2CH2OH, -0CH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10.
[0053] In some embodiments, a compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1, 2 or 3 substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -NHCH2CH3, =O, -OH, -OCH3, and -OCH2CH3; and C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is optionally substituted by halogen, - CN, -NO2, -NH2, -NHCH3, -NHCH2CH3, =O, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, - C(CH3)3, C3-12 carbocycle, or 3- to 6-membered heterocycle.
[0054] It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted”, references to chemical moieties herein are understood to include substituted variants. For example, reference to a “heteroaryl” group or moiety implicitly includes both substituted and unsubstituted variants.
[0055] Where bivalent substituent groups are specified herein by their conventional chemical formulae, written from left to right, they are intended to encompass the isomer that would result from writing the structure from right to left, e.g., -CH2O- is also intended to encompass to -OCH2-.
[0056] “Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, an “optionally substituted” group may be either unsubstituted or substituted. [0057] Compounds of the present disclosure also include crystalline and amorphous forms of those compounds, pharmaceutically acceptable salts, and active metabolites having the same type of activity, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, amorphous forms of the compounds, and mixtures thereof.
[0058] The compounds described herein may exhibit their natural isotopic abundance, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure. For example, hydrogen has three naturally occurring isotopes, denoted
Figure imgf000021_0001
(protium), 2H (deuterium), and 3H (tritium). Protium is the most abundant isotope of hydrogen in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increased in vivo half-life and/or exposure, or may provide a compound useful for investigating in vivo routes of drug elimination and metabolism. Examples of isotopes that may be incorporated into compounds of the present disclosure include, but are not limited to, 2H, 3H, 13C, 14C, 15N, 18O, 170, 35S, 36C1, and 18F. Of particular interest are compounds of Formula (I) enriched in tritium or carbon-14, which can be used, for example, in tissue distribution studies; compounds of the disclosure enriched in deuterium especially at a site of metabolism, resulting, for example, in compounds having greater metabolic stability; and compounds of Formula (I) enriched in a positron emitting isotope, such as 11C, 18F, 15O and 13N, which can be used, for example, in Positron Emission Topography (PET) studies. Isotopically-enriched compounds may be prepared by conventional techniques well known to those skilled in the art.
[0059] As used herein, the phrase “of the formula” or “having the formula” or “having the structure” is not intended to be limiting and is used in the same way that the term “comprising” is commonly used. For example, if one structure is depicted, it is understood that all stereoisomer and tautomer forms are encompassed, unless stated otherwise.
[0060] Certain compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms, the asymmetric centers of which can be defined, in terms of absolute stereochemistry, as (R)- or (S)-. In some embodiments, in order to optimize the therapeutic activity of the compounds of the disclosure, e.g., to treat asthma, it may be desirable that the carbon atoms have a particular configuration (e.g., (R,R), (S,S), (S,R), or (R,S)) or are enriched in a stereoisomeric form having such configuration. The compounds of the disclosure may be provided as racemic mixtures. Accordingly, the disclosure relates to racemic mixtures, pure stereoisomers (e.g., enantiomers and diastereoisomers), stereoisomer-enriched mixtures, and the like, unless otherwise indicated. When a chemical structure is depicted herein without any stereochemistry, it is understood that all possible stereoisomers are encompassed by such structure. Similarly, when a particular stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the disclosure unless otherwise indicated, provided that the utility of the composition as a whole is not eliminated by the presence of such other isomers. Individual stereoisomers may be obtained by numerous methods that are known in the art, including preparation using chiral synthons or chiral reagents, resolution using chiral chromatography using a suitable chiral stationary phase or support, or by chemically converting them into diastereoisomers, separating the diastereoisomers by conventional means such as chromatography or recrystallization, then regenerating the original stereoisomer.
[0061] Additionally, where applicable, all cis-trans or E/Z isomers (geometric isomers), tautomeric forms and topoisomeric forms of the compounds of the invention are included within the scope of the invention unless otherwise specified.
[0062] The term “tautomer”, as used herein, refers to each of two or more isomers of a compound that exist in equilibrium and which ready interconvert. For example, one skilled in the art would readily understand that 1,2, 3 -triazole exists in two tautomeric forms:
Figure imgf000022_0001
Unless otherwise specified, chemical entities described herein are intended to include all possible tautomers, even when a structure depicts only one of them. For example, even though a single tautomer of a compound of Formula (I-D) may be depicted herein, the disclosure is intended to include all possible tautomers, including:
Figure imgf000022_0002
[0063] The term “pharmaceutically acceptable” refers to a material that is not biologically or otherwise unacceptable when used in the subject compositions and methods. For example, the term “pharmaceutically acceptable carrier” refers to a material — such as an adjuvant, excipient, glidant, sweetening agent, diluent, preservative, dye, colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent or emulsifier — that can be incorporated into a composition and administered to a patient without causing unacceptable biological effects or interacting in an unacceptable manner with other components of the composition. Such pharmaceutically acceptable materials typically have met the required standards of toxicological and manufacturing testing, and include those materials identified as suitable inactive ingredients by the U.S. Food and Drug Administration.
[0064] The terms “salt” and “pharmaceutically acceptable salt” refer to a salt prepared from a base or an acid. Pharmaceutically acceptable salts are suitable for administration to a patient, such as a mammal (for example, salts having acceptable mammalian safety for a given dosage regime). Salts can be formed from inorganic bases, organic bases, inorganic acids and organic acids. In addition, when a compound contains both a basic moiety, such as an amine, pyridine or imidazole, and an acidic moiety, such as a carboxylic acid or tetrazole, zwitterions may be formed and are included within the term “salt” as used herein. Salts derived from inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, and zinc salts, and the like. Salts derived from organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, A. A'-dibcnzylcthylcncdiaminc. diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, A-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like. Salts derived from inorganic acids include salts of boric, carbonic, hydrohalic (hydrobromic, hydrochloric, hydrofluoric or hydroiodic), nitric, phosphoric, sulfamic and sulfuric acids. Salts derived from organic acids include salts of aliphatic hydroxyl acids (for example, citric, gluconic, glycolic, lactic, lactobionic, malic, and tartaric acids), aliphatic monocarboxylic acids (for example, acetic, butyric, formic, propionic and trifluoro acetic acids), amino acids (for example, aspartic and glutamic acids), aromatic carboxylic acids (for example, benzoic, p-chlorobenzoic, diphenylacetic, gentisic, hippuric, and triphenylacetic acids), aromatic hydroxyl acids (for example, o- hydroxybenzoic, p-hydroxybenzoic, l-hydroxynaphthalene-2-carboxylic and 3-hydroxynaphthalene-2- carboxylic acids), ascorbic, dicarboxylic acids (for example, fumaric, maleic, oxalic and succinic acids), glucoronic, mandelic, mucic, nicotinic, orotic, pamoic, pantothenic, sulfonic acids (for example, benzenesulfonic, camphorsulfonic, edisylic, ethanesulfonic, isethionic, methanesulfonic, naphthalenesulfonic, naphthalene- 1,5 -disulfonic, naphthalene-2,6-disulfonic and p-toluenesulfonic acids), xinafoic acid, and the like.
[0065] The term “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to affect treatment when administered to a subject in need thereof. For example, a therapeutically effective amount for treating asthma is an amount of compound needed to, for example, reduce, suppress, eliminate, or prevent one or more symptoms of asthma in a subject, or to treat the underlying cause of the asthma. The therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The specific dose will vary depending on the particular compound chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried. The term “effective amount” refers to an amount sufficient to obtain a desired result, which may not necessarily be a therapeutic result. For example, an “effective amount” may be the amount needed to inhibit an enzyme.
[0066] As used herein, “treating” or “treatment” refers to an approach for obtaining beneficial or desired results with respect to a disease, disorder, or medical condition (such as asthma) in a subject, including but not limited to the following: (a) preventing the disease or medical condition from occurring, e.g., preventing the reoccurrence of the disease or medical condition or prophylactic treatment of a subject that is pre-disposed to the disease or medical condition; (b) ameliorating the disease or medical condition, e.g., eliminating or causing regression of the disease or medical condition in a subject; (c) suppressing the disease or medical condition, e.g., slowing or arresting the development of the disease or medical condition in a subject; or (d) alleviating symptoms of the disease or medical condition in a subject. For example, “treating asthma” would include preventing pulmonary inflammation from occurring, ameliorating pulmonary inflammation, suppressing pulmonary inflammation, and alleviating the symptoms of asthma (for example, improved lung function tests). Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder
[0067] A “therapeutic effect”, as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[0068] The terms “antagonist” and “inhibitor” are used interchangeably, and they refer to a compound having the ability to inhibit a biological function (e.g., activity, expression, binding, protein-protein interaction) of a target protein (e.g., JAK). Accordingly, the terms “antagonist” and “inhibitor” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
[0069] The term “selective inhibition” or “selectively inhibit” refers to the ability of a biologically active agent to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
[0070] The terms “subject” and “patient” refer to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human. “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
[0071] “Prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of Formula (I)). Thus, the term “prodrug” refers to a precursor of a biologically active compound that is pharmaceutically acceptable. In some aspects, a prodrug is inactive when administered to a subject but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam); Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” (1987) A.C.S. Symposium Series, Vol. 14; and Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press) each of which is incorporated in full by reference herein. The term “prodrug” is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of an active compound, as described herein, are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and benzamide derivatives of an amine functional group in the active compound, and the like.
[0072] The term “in vivo” refers to an event that takes place in a subject's body.
[0073] The term “in vitro” refers to an event that takes places outside of a subject's body. For example, an in vitro assay encompasses any assay run outside of a subject. In vitro assays encompass cell-based assays in which cells alive or dead are employed. In vitro assays also encompass a cell-free assay in which no intact cells are employed.
[0074] The disclosure is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the disclosure includes compounds produced by a process comprising administering a compound of this disclosure to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the disclosure in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
[0075] The present disclosure provides compounds that are capable of selectively binding to and/or modulating JAK. In some embodiments, the compounds modulate JAK by binding to or interacting with one or more amino acids and/or one or more metal ions. The binding of these compounds may disrupt JAK downstream signaling.
[0076] In certain aspects, the present disclosure provides a compound of Formula (I):
Figure imgf000026_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected at each occurrence from halogen, -OH, -O(C1-4 alkyl), -O(C1-4 haloalkyl), and C1-4 alkyl;
R2 is selected from hydrogen, halogen, and C1-4 alkyl;
R3 is independently selected at each occurrence from halogen and C1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; represents a double bond or triple bond;
Figure imgf000026_0002
when is a double bond, then A is CR4R3; when a is a triple bond, then A is CR":
R4 and R5 are each independently selected from hydrogen and RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from hydrogen and RA;
RA is independently selected at each occurrence from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10);
Ci-10 alkyl, C2-10 alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10,
-NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), R10, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, 1- to 6-membered heteroalkyl, Co-3 alkyl-(C3-12 carbocycle), and Co-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH20H, -CH2CH2OH, -0CH3, -OCH2CH3, -OCH2CH2-OCH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10; and
R7 is hydrogen or C1-6 alkyl.
[0077] In some embodiments, R7 is hydrogen. In some embodiments, R7 is C1-6 alkyl. In some embodiments, R7 is C1-3 alkyl. In some embodiments, R7 is -CH3.
[0078] In some embodiments, for a compound of Formula (I) or (I’), m is an integer from 1 to 3, such as m is 2. In some embodiments, n is 0 or 1, such as n is 0.
[0079] In some embodiments, the compound of Formula (I) or (I’) is a compound of Formula (I-A):
Figure imgf000027_0001
such as a compound of Formula (I-B) or (I-C):
Figure imgf000028_0003
or a pharmaceutically acceptable salt thereof.
[0080] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B) or (I-C), R1 is independently selected at each occurrence from halogen, -OH, -O(C1-2 alkyl), and C1-2 alkyl. In some embodiments, R1 is independently selected at each occurrence from halogen, -OH, and C1-2 alkyl. In some embodiments, R1 is independently selected at each occurrence from halogen, -OH, and -CH2CH3. In some embodiments, R1 is independently selected at each occurrence from -OH, and -CH2CH3.
[0081] In some embodiments, the compound of Formula (I) or (I’) is a compound of Formula (I-D):
Figure imgf000028_0001
such as a compound of Formula (I-E) or (I-F):
Figure imgf000028_0002
or a pharmaceutically acceptable salt thereof.
[0082] In some embodiments, for a compound of Formula (I), (I’), (I-A) or (I-D):
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from RA;
RA is independently selected at each occurrence from: halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2,
-S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12,
-NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12;
C1-10 alkyl, optionally substituted at each occurrence with one or more substituents selected from halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, R10, C1-6 alkyl, and C1-6 haloalkyl; and R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-6 carbocycle, and 3- to 6-membered heterocycle.
[0083] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B), (I-C), (I-D), (I-E) or (I-F):
RA is independently selected at each occurrence from:
-N(R10)2;
Ci-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, - OR10, -N(R10)2, and C1-6 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
|0084] In some embodiments, for a compound of Formula (I), (I’), (I -A) or (I-D),
Figure imgf000029_0002
is selected from :
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
[0088] In some embodiments, for a compound of Formula (I), (F), (I -A) or (I-D), is a double bond and
Figure imgf000034_0002
A is CR4R5. In some embodiments,
Figure imgf000034_0003
is a triple bond and A is CRft.
[0089] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B), (I-D) or (I-E):
R4 is selected from RA and R5 is hydrogen; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
RA is independently selected at each occurrence from: halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12;
Ci-10 alkyl, optionally substituted at each occurrence with one or more substituents selected from halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, R10, C1-6 alkyl, and C1-6 haloalkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-6 carbocycle, and 3- to 6-membered heterocycle.
[0090] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B), (I-D) or (I-E):
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NHS(=O)2R10, -NHC(O)R10, and 4- to 10-membered heterocycle; and
4- to 10-membered heterocycle, wherein each 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from -OCH3, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-6 carbocycle), and C0-3 alkyl-(3- to 6-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -N(CH3)2, =O, -OH, and -CH3.
[0091] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B), (I-D) or (I-E):
R4 is selected from RA and R5 is hydrogen; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NHS(=O)2R10, -NHC(O)R10, and 4- to 10-membered heterocycle; and
4- to 10-membered heterocycle, wherein each 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from -OCH3, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-6 carbocycle), and C0-3 alkyl-(3- to 6-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -N(CH3)2, =O, -OH, and -CH3.
[0092] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-B), (I-D) or (I-E):
R4 is selected from:
-CH2N(R10)2; and
5- to 7-membered heterocycle, optionally substituted with -CH3;
R5 is hydrogen; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl and 3- to 6-membered heterocycle.
Figure imgf000036_0001
Figure imgf000037_0001
[0094] In some embodiments, for a compound of Formula (I), (F), (I-A), (I-C), (I-D) or (I-F):
R6 is selected from RA;
RA is independently selected at each occurrence from: C1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, R10, C1-6 alkyl, and C1-6 haloalkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-6 carbocycle, and 3- to 6-membered heterocycle.
[0095] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-C), (I-D) or (I-F):
R6 is selected from: C1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, C3 -10 carbocycle, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in R6 is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen and C1-4 alkyl.
[0096] In some embodiments, for a compound of Formula (I), (I’), (I-A), (I-C), (I-D) or (I-F), R6 is selected from:
-CH2(5- to 7-membered heterocycle), optionally substituted with -CH3; and
5- to 7-membered heterocycle.
[0097] In some embodiments for a compound of Formula (I-C) or (I-F), is selected from
Figure imgf000038_0001
Figure imgf000038_0002
Figure imgf000039_0001
[0098] In some embodiments, for a compound of Formula (I), (F), (I-A), (I-B), (I-C), (I-D), (I-E) or (I-F), R2 is selected from hydrogen and halogen. In some embodiments, R2 is selected from hydrogen and F. In some embodiments, R2 is hydrogen. In some embodiments, R2 is F.
[0099] In some embodiments, for a compound of Formula (I), (I-A) or (I-D):
R2 is selected from hydrogen and F;
R4 is selected from RA and R5 is selected from hydrogen and -CH3: R4 is selected from hydrogen and -CH3 and R5 is selected from RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-10 carbocycle), and C0-3 alkyl-(4- to 10-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
[0100] In some embodiments, for a compound of Formula (I), (I’), (I-A) or (I-D):
R2 is selected from hydrogen and -F; and
Figure imgf000039_0002
Figure imgf000040_0001
[0101] In certain aspects, the present disclosure provides a compound of Formula (I-D):
Figure imgf000040_0002
or a pharmaceutically acceptable salt thereof, wherein:
R2 is selected from hydrogen and F; represents a double bond or triple bond; when is a double bond, then A is CR4R5; when is a triple bond, then A is CR6;
R4 is selected from RA and R5 is hydrogen; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-6 alkyl; and R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
[0102] In certain aspects, the present disclosure provides a compound of Formula (I-D):
Figure imgf000041_0001
or a pharmaceutically acceptable salt thereof, wherein:
R2 is selected from hydrogen and F; and
Figure imgf000041_0002
[0103] References herein to “a compound of Formula (I)” implicitly also include the compound of Formula (I’), (I-A), (I-B), (I-C), (I-D), (I-E), and (I-F). The skilled person will understand that the context of a passage may exclude one or more of Formulas (I’), (I-A), (I-B), (I-C), (I-D), (I-E), and (I-F), for example, if the passage provides an embodiment of a particular variable (e.g., R1) different than what is depicted in the formula.
[0104] In some embodiments, a compound of Formula (I) is provided as a substantially pure stereoisomer. In some embodiments, the stereoisomer is provided in at least 80% enantiomeric excess, such as at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or at least 99.9% enantiomeric excess.
[0105] The chemical entities described herein can be synthesized according to one or more illustrative schemes herein and/or techniques known in the art. Materials used herein are either commercially available or prepared by synthetic methods generally known in the art. These schemes are not limited to the compounds listed in the examples or by any particular substituents, which are employed for illustrative purposes.
Although various steps are described and depicted in Schemes 1 and 2 and Examples 1-9, the steps in some cases may be performed in a different order than the order shown in Schemes 1 and 2 and Examples 1-9. Various modifications to these synthetic reaction schemes may be made and will be suggested to one skilled in the art having referred to the present disclosure. Numberings or R groups in each scheme typically have the same meanings as those defined elsewhere herein unless otherwise indicated. The abbreviation PG refers to a suitable protecting group, and it may be the same or a different protecting group when used more than once in a given scheme.
[0106] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure, generally within a temperature range from -10 °C to 200 °C. Further, except as otherwise specified, reaction times and conditions are intended to be approximate, e.g. taking place at about atmospheric pressure within a temperature range of about -10 °C to about 110 °C over a period of about 1 to about 24 hours; reactions left to run overnight average a period of about 16 hours.
[0107] In general, compounds of the disclosure may be prepared by the following reaction schemes:
Scheme 1
Figure imgf000042_0001
[0108] In some embodiments, a compound of Formula Ih may be prepared according to Scheme 1. For example, acyl chloride la can be coupled to a suitably protected imidazole to provide lb, which can be reacted with hydrazine monohydrate to form the pyrazole ring of 1c, optionally with one or more protecting group manipulations following the ring formation. Installation of a substituted phenyl ring may proceed via a Suzuki reaction using boronic ester Id to afford intermediate le. Formation of bromoimidazole If can be followed by another Suzuki reaction with boronic ester 1g, and optionally one or more coupling reactions and/or protecting group manipulations, to provide an alkene of Formula Ih.
Figure imgf000043_0001
[0109] In some embodiments, a compound of Formula 2e may be prepared according to Scheme 2. For example, imidazole 2a can be iodinated to provide diiodo imidazole 2b. Selective removal of one iodo group to give 2c can be followed by a Sonogashira coupling with alkyne 2d, and optionally one or more coupling reactions and/or protecting group manipulations, to provide an alkyne of Formula 2e.
[0110] In some embodiments, a compound of the present disclosure, for example, a compound of a formula given in Table 1, is synthesized according to one of the general routes outlined in Schemes 1 and 2, Examples 1-9, or by methods generally known in the art. In some embodiments, exemplary compounds may include, but are not limited to, a compound or salt thereof selected from Table 1.
Table 1
Figure imgf000043_0002
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0002
[0111] In some embodiments, exemplary compounds may include, but are not limited to, a compound or salt thereof selected from:
Figure imgf000068_0001
[0112] Methods
[0113] In some aspects, the present disclosure provides a method of inhibiting JAK signaling, comprising contacting a cell with an effective amount of a compound disclosed herein, such as a compound of Formula (I). In some embodiments, the present disclosure provides a method of inhibiting a JAK protein, comprising contacting the JAK protein with an effective amount of a compound disclosed herein. Inhibition of JAK signaling can be assessed by a variety of methods known in the art. Non-limiting examples include a showing of (a) a decrease in kinase activity of JAK; (b) a decrease in binding affinity between JAK and one or more of its respective ligands, such as a cytokine; (c) a decrease in the levels of phosphorylated intracellular signaling molecules downstream in the JAK signaling pathway, such as a decrease in pSTAT3 or pSTAT6 levels; (d) a decrease in binding of JAK to downstream signaling molecules; and/or (e) an increase in ATP levels or a decrease in ADP levels. Kits and commercially available assays can be utilized for determining one or more of the above.
[0114] In some aspects, the present disclosure provides a method of treating a JAK-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein. In some embodiments, the disease or condition is a respiratory disease, such as asthma, chronic obstructive pulmonary disease or lung transplant rejection. In some embodiments, the disease or condition is asthma, such as T2-dominant (eosinophilic) asthma or non-T2-dominant (non-eosinophilic) asthma. In some embodiments, the disease or condition is lung transplant rejection, such as primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction.
[0115] The JAK inhibitors of the present disclosure have been designed for the treatment of inflammatory and fibrotic disease of the respiratory tract. In particular, the compounds have been designed to enable delivery of a potent anti-cytokine agent directly to the site of action of respiratory disease in the lung while limiting systemic exposure.
[0116] As shown in Example 10 and Table 2, the compounds of the present disclosure have been shown to be potent inhibitors of the JAK family of enzymes: JAK1, JAK2, JAK3, and TYK2.
[0117] It has been recognized that the broad anti-inflammatory effect of JAK inhibitors could suppress normal immune cell function, potentially leading to increased risk of infection. The present compounds have therefore been optimized to limit absorption from the lung into the plasma, thus minimizing the risk of immunosuppression. As described in the experimental section below, the absorption and distribution of select compounds have been profiled in preclinical assays. Several compounds were tested in mice, in Example 14, and showed post-dosing high concentration in lung tissue and low absorption into plasma.
[0118] Certain compounds disclosed herein are expected to inhibit an effect of the pro-inflammatory cytokine IL-13 in mouse lung tissue. Specifically, compounds of the present disclosure are expected to inhibit IL-13-induced phosphorylation of STAT6 in lung tissue, which would provide evidence of local lung JAK target engagement in vivo. This effect is expected when the pro-inflammatory cytokine IL-13 is administered 8 hours after administration of the test compound, which would provide further evidence of significant retention in the lung.
[0119] The anti-inflammatory activity of JAK inhibitors has been robustly demonstrated in preclinical models of asthma (Malaviya et al,, Ini. Immunophamacol. 2010, 10, 829-836; Matsunaga et al., Biochem. and Biophys. Res. Commun. 2011, 404, 261-267; Kudlacz et al., Eur. J Pharmacol 2008, 582, 154-161). Cytokines implicated in asthma inflammation which signal through the JAK-STAT pathway include IL-2, IL- 3, IL-4, IL-5, IL-6, IL-9, IL-11, IL-13, IL-23, IL-31 , IL-27, thymic stromal lymphopoietin (TSLP), interferon- y (IFNy) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Accordingly, the compounds of the present disclosure are expected to be useful for the treatment of inflam matory respiratory disorders, such as asthma. The present compounds, therefore, are also expected to be useful for the treatment of chronic obstructive pulmonary disease, cystic fibrosis, pneumonitis, interstitial lung diseases (including idiopathic pulmonary’ fibrosis), acute lung injury, acute respiratory distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, and sarcoidosis. Further, asthma endotypes may be broadly regarded as type 2 (T2) high or T2-low (Kuravilla et al., Clin Rev Allergy Immunol 2019, 56(2), 219-233). Based on their mechanism of action, the compounds of the disclosure have the potential to treat both endotypes, T2-high and T2-low. [0120] The compounds of the present disclosure possess biological activity involved in the inhibition of cytokines associated with inflammation. Therefore, the compounds of the present disclosure are expected to be useful for the treatment of certain specific respiratory diseases, as detailed below.
[0121] Eosinophilic airway inflammation is a characteristic feature of diseases collectively termed eosinophilic lung diseases (Cottin et al., Clin. Chest. Med. 2016, 37(3), 535-56). Eosinophilic diseases have been associated with IL-4, IL-13 and IL-5 signaling. Eosinophilic lung diseases include infections (especially helminthic infections), drug-induced pneumonitis (induced for example by therapeutic drags such as antibiotics, phenytoin, or 1 -tryptophan), fungal-induced pneumonitis (e.g., allergic bronchopulmonary aspergillosis), hypersensitivity pneumonitis and eosinophilic granulomatosis with polyangiitis (formerly known as Churg-Strauss syndrome). Eosinophilic lung diseases of unknown etiology include idiopathic acute eosinophilic pneumonia, idiopathic chronic eosinophilic pneumonia, hypereosinophilic syndrome, and Loffler syndrome.
[0122] C ytokines such as IFNy, IL-12 and IL-6 have been implicated in a range of non-allergic lung diseases such as sarcoidosis and lymphangioleiomyomatosis (El-Hashemite et al.. Am. J. Respir. Cell. Mol. Biol. 2005, 33, 227-230, and El-Hashemite et al., Cancer Res. 2004, 64, 3436-3443). Bronchiectasis and infiltrative pulmonary diseases are diseases associated with chronic neutrophilic inflammation.
[0123] Pathological T cell activation is critical in the etiology of multiple respiratory diseases. Autoreactive T cells play a role in bronchiolitis obliterans organizing pneumonia (also termed COS). Similar to COS, the etiology of lung transplant rejections is linked to an aberrant T cell activation of the recipient’s T cells by the transplanted donor lung. Lung transplant rejections may occur early as Primary Graft Dysfunction (PGD), organizing pneumonia (OP), acute rejection (AR) or lymphocytic bronchiolitis (LB) or they may occur years after lung transplantation as Chronic Lung Allograft Dysfunction (CLAD). Therefore, JAK inhibitors have the potential to be effective in treating or preventing lung transplant rejection and CLAD. Similar T cell activation events as described as the basis for lung transplant rejection also are considered the main driver of lung graft-versus-host disease (GVHD) which can occur post hematopoietic stem cell transplants. As systemic JAK inhibition is associated with serious adverse events and a small therapeutic index, the need remains for an inhaled lung-directed, non-systemic JAK inhibitor to prevent and/or treat lung transplant rejection or lung GVHD. The compounds of the present disclosure have the characteristics required to meet this need, [0124] Therefore, provided herein is a method of treating or preventing lung transplant rejection in a human in need thereof, comprising administering to the human a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In some embodiments, the lung transplant rejection is selected from the group consisting of primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is acute lung transplant rejection. In some embodiments, the lung transplant rejection is chronic lung allograft dysfunction. In some embodiments, the lung transplant rejection is selected from the group consisting of bronchiolitis obliterans, restrictive chronic lung allograft dysfunction, and neutrophilic allograft dysfunction.
[0125] More recently, immune-checkpoint inhibitor induced pneumonitis, another T ceil mediated lung disease emerged with the increased use of immune-checkpoint inhibitors. In cancer patients treated with these T cell stimulating agents, fatal pneumonitis can develop. The compounds of the disclosure possess biological activity’ allowing inhibition of IFNy secretion.
[0126] Accordingly, in one embodiment, the present disclosure provides a method of treating a respiratory disease in a mammal (e.g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
[0127] In some embodiments, the respiratory disease is asthma, chronic obstructive pulmonary disease (COPD), pneumonitis, cystic fibrosis (CF), interstitial lung diseases (including idiopathic pulmonary fibrosis), acute lung injury’, acute respiratory’ distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, or sarcoidosis. In some embodiments, the respiratory’ disease is asthma or chronic obstructive pulmonary disease. In some embodiments, the asthma is T2-high asthma. In some embodiments, the asthma is T2-low asthma. In some embodiments, the respiratory disease is viral induced acute lung injury (VI-ALI). In some embodiments, the respiratory disease is chronic lung allograft rejection (CLAD). In some embodiments, the respiratory disease is pulmonary hypertension associated interstitial lung disease (PH-ILD). In some embodiments, the respiratory disease is scleroderma associated interstitial lung disease (Sc-ILD).
[0128] In some embodiments, the respiratory disease is a lung infection, an eosinophilic disease, a helminthic infection, pulmonary arterial hypertension, lymphangioleiomyomatosis, bronchiectasis, an infiltrative pulmonary disease, drug-induced pneumonitis, fungal induced pneumonitis, allergic bronchopulmonary' aspergillosis, hypersensitivity pneumonitis, eosinophilic granulomatosis with polyangiitis, idiopathic acute eosinophilic pneumonia, idiopath ic chronic eosinophilic pneumonia, hy pereosinophilic syndrome. Loftier syndrome, bronchiolitis obliterans organizing pneumonia, acute and chronic lung transplant rejections (including PGD, OP, LB, AR and CLAD, BO, restrictive CLAD and neutrophilic allograft dysfunction), lung graft-versus-host disease, or immune-checkpoint-inhibitor induced pneumonitis.
[0129] The present disclosure further provides a method of treating asthma in a mammal (e.g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically- acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. [0130] When used to treat asthma, the compounds of the present disclosure will typically be administered in a single daily dose or in multiple doses per day, although other forms of administration may be used. The amount of active agent administered per dose or the total amount administered per day will typically be determined by a physician, in light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[0131] When used to treat a respiratory disease (including but not limited to a disease described herein), the compounds of the present disclosure will typically be administered in a single daily dose or in multiple doses per day, although other forms of administration may be used. The amount of active agent administered per dose or the total amount administered per day will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient’s symptoms, and the like.
[0132] Human coronavirus is a common respiratoiy pathogen and typically induces mild upper respiratory disease. The two highly pathogenic viruses, severe acute respiratory syndrome-associated coronavirus (SARS-CoV-1) and Middle East respiratory syndrome-associated coronavirus (MERS-CoV), caused severe respiratory syndromes resulting in more than 10% and 35% mortality, respectively (Assiri et al., N Engl J Med. 2013, 369, 407-1). Similar to SARS-CoV-1 and MERS-CoV, a subset of COVID-19 patients (about 16%) can develop a severe respiratory illness manifested by acute lung injury (AL1) leading to ICU admission (about 5%), respiratory failure (about 6.1%) and death (Wang et al., JAMA 2020, 323, 11, 1061-1069; Guan et al., N Engl J Med. 2020, 382, 1708-1720; Huang et al., The Lancet 2020, 395 (10223), 497-506; Chen et al., The Lancet 2020, 395(10223), 507-13). A subgroup of patients with CO VID-19 appears to have a hyperinflammatory “cytokine storm” resulting in acute lung injury and acute respiratory distress syndrome (ARDS). This cytokine storm may also spill over into the systemic circulation and produce sepsis and ultimately, multi-organ dysfunction syndrome. The dysregulated cytokine signaling that appears in COVID- 19 is characterized by increased expression of interferons (IFNs), interleukins (Ils), and chemokines, resulting in ALT and associated mortality. This hyperinflammatory response can potentially be modulated and treated by a lung-selective pan-Janus Kinase (JAK) inhibitor. Monoclonal antibodies directed against IL-6 (tocilizumab) appear to be effective in treating patients with AL1 from COVID-19 (Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective Treatment of Severe COVID-19 Patients with Tocilizumab, 2020, PNAS, https://doi.org/10.1073/pnas.2005615117). Infection with mouse adapted strains of the 2003 SARS-CoV-1 and 2012 MERS-CoV, as well as a transgenic mouse expressing the human SARS-CoV-1 receptor hACE2 infected with human SARS-CoV-1, demonstrate elevations of JAK-dependent cytokines, such as IFNy, IL-6, and IL- 12, and downstream chemokines, such as chemokine (C-C motif) ligand 10 (CCL10), CCL2, and CCL7. JAK inhibitors have also been shown to be beneficial in mouse models of lipopolysaccharide- or ganciclovir-induced ALL Finally, based on the results of clinical trials, baricitinib, a JAK inhibitor, has received an emergency use authorization (EUA) in combination with remdesivir, for the treatment of COVID- 19 in patients requiring supplemental oxygen, invasive mechanical ventilation, or extracorporeal membrane oxygenation.
[0133] Therefore, compounds of formula (1), which are lung-selective inhaled pan-JAK inhibitors, could be uniquely suited to dampen the cytokine storm associated with coronaviruses such as COVID-19. By delivering to the lung and avoiding systemic immunosuppression, additional infections that lead to worsened mortality may also be avoided. This is particularly true in those patients requiring ventilatory7 support. As major causes of death in subjects with COVID-19 appear to be comorbidities and superinfection, an inhaled medication may be a way to avoid systemic immunosuppression that would pre-dispose patients to these risks .
[0134] Therefore, the present disclosure provides a method of treating a mammal (or patient) infected with a coronavirus, such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV, or the symptoms thereof, the method comprising adm inistering to the mammal (or patient) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically - acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. The present disclosure also provides a method of treating ALT and/or ARDS in a mammal (or a patient) caused by a coronavirus infection (such as SARS-CoV-1, SARS-CoV-2, and MERS-CoV), the method comprising adm inistering to the mammal (or patient) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically - acceptable earner and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof. [0135] The mechanism of action of JAK inhibitors has been linked to the treatment of nasal inflammatory diseases (Therapeutic Effects of Intranasal Tofacitintb on Chronic Rhinosinusitis with Nasal Polyps in Mice, Joo et al., The Laryngoscope 2020, https://doi.org/10.1002/laiy.29129). Further, Dupilumab, which acts by blocking the IL-4 and IL-13 signaling pathways, has been approved for the treatment of chrome rhinosinusitis with nasal poly ps.
[0136] Therefore, also provided herein is a method of treating nasal inflammatory diseases in a mammal (e g., a human), the method comprising administering to the mammal (or human) a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the present disclosure, or a pharmaceutically' acceptable salt thereof. In some embodiments, the nasal inflammatory disease is selected from the group consisting of chronic rhinosinusitis with or w ithout nasal poly ps, nasal polyposis, sinusitis with nasal polyps, and rhinitis (non-allergic, allergic, perennial, and vasomotor rhinitis).
[0137] As JAK inhibitors, the compounds of the present disclosure may' also be useful for a variety of other diseases. The compounds of the present disclosure may' be useful for a variety' of gastrointestinal inflammatory indications that include, but are not limited to, inflammatory bowel disease, ulcerative colitis (proctosigmoiditis, pancolitis, ulcerative proctitis and left-sided colitis), Crohn’s disease, collagenous colitis, lymphocytic colitis, Behcet’s disease, celiac disease, immune checkpoint inhibitor induced colitis, ileitis, eosinophilic esophagitis, graft versus host disease-related colitis, and infectious colitis. Ulcerative colitis, Crohn’s disease collagenous colitis, lymphocytic colitis, eosinoph ilic esophagitis, graft versus host disease- related colitis, infectious colitis, Behcet’s disease, celiac disease, immune checkpoint inhibitor induced colitis (e.g., CTLA-4 inhibitor-induced colitis, PD-1- or PD-L1 -inhibitor-induced colitis), and ileitis are characterized by elevation of certain pro-inflamm atony cytokine levels. As many pro -inflammatory cytokines signal via JAK activation, compounds described in this application may be able to alleviate the inflammation and provide symptom relief. In particular, the compounds of the present disclosure may be useful for the induction and maintenance of remission of ulcerative colitis, and for the treatment of Crohn’s disease, immune checkpoint inhibitor induced colitis, and the gastrointestinal adverse effects in graft versus host disease. In one embodiment, therefore, the present disclosure provides a method of treating a gastrointestinal inflammatory disease in a mam mal (e.g., a human), the method comprising administering to the mammal a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the present disclosure, or a pharmaceutically acceptable salt thereof.
[0138] Atopic dermatitis and other inflammatory skin diseases have been associated with elevation of proinflammatory cytokines that rely on the JAK-STAT pathway. Therefore, the compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may be beneficial in a number of dermal inflammatory or pruritic conditions that include, but are not limited to atopic dermatitis, alopecia areata, vitiligo, psoriasis, dermatomyositis, cutaneous T ceil lymphoma and subtypes (Sezary syndrome, mycosis fungoides, pagetoid reticulosis, granulomatous slack skin, lymphomatoid papulosis, pityriasis lichenoides chronica, pityriasis lichenoides et varioliformis acuta, CD30+ cutaneous T-cell lymphoma, secondary cutaneous CD30+ large cell lymphoma, non-mycosis fungoides CD30- cutaneous large T-cell lymphoma, pleomorphic T-cell lymphoma, Lennert lymphoma, subcutaneous T-cell lymphoma, angiocentric lymphoma, blastic NK-cell lymphoma), prurigo nodularis, lichen planus, primary localized cutaneous amyloidosis, bullous pemphigoid, skin manifestations of graft versus host disease, pemphigoid, discoid lupus, granuloma annulare, lichen simplex chronicus, vulvar/scrotal/perianal pruritus, lichen sclerosus, post herpetic neuralgia itch, lichen planopilaris, and follicnlitisdecalvans. In particular, atopic dermatitis, alopecia areata, vitiligo, prurigo nodularis, lichen planus, primary7 localized cutaneous amyloidosis, bullous pemphigoid, and dermal manifestations of graft versus host disease are characterized by elevation of certain cytokines that signal via JAK activation. Accordingly, compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may be able to alleviate associated dermal inflammation or pruritus driven by these cytokines. In particular, compounds of the present disclosure, or a pharmaceutically' acceptable salt thereof, may be expected to be useful for the treatment of atopic dermatitis and other inflammatory skin diseases. In one embodiment, therefore, the present disclosure provides a method of treating an inflammatory skin disease in a mammal (e.g., a human), the method comprising applying a pharmaceutical composition comprising a compound of the present disclosure, or a pharmaceutically acceptable salt thereof and a pharmaceutical carrier to the skin of the mammal. In some embodiments, the inflammatory skin disease is atopic dermatitis.
[0139] Many ocular diseases have been shown to be associated with elevations of proinflainmatory cytokines that rely on the JAK-STAT pathway. The compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, therefore, may be useful for the treatment of ocular diseases that include, but are not limited to, uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, age-related macular degeneration, and atopic keratoconjunctivitis. In particular, uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, and age-related macular degeneration are characterized by elevation of certain pro- inflammatory cytokines that signal via the JAK-STAT pathway. Accordingly, compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may be able to alleviate the associated ocular inflammation and reverse disease progression or provide symptom relief. In one embodiment, therefore, the present disclosure provides a method of treating an ocular disease in a mammal (e.g., a human), the method comprising administering a pharmaceutical composition comprising a compound of the present disclosure or a pharmaceutically-acceptable salt thereof and a pharmaceutical carrier to the eye of the mammal (or human). In some embodiments, the ocular disease is uveitis, diabetic retinopathy, diabetic macular edema, dry eye disease, age-related macular degeneration, or atopic keratoconjunctivitis. In some embodiments, the method comprises administering the compound of the present disclosure, or a pharmaceutically acceptable salt thereof by intravitreal injection. Compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may also be used in combination with one or more compound useful to ocular diseases.
[0140] The compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may also be useful to treat other diseases such as other inflammatory diseases, autoimmune diseases or cancers. The compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, may be useful to treat one or more of cytokine release syndrome (CRS), arthritis, rheumatoid arthritis, juvenile rheumatoid arthritis, transplant rejection, xerophthalmia, psoriatic arthritis, diabetes, insulin dependent diabetes, motor neuron disease, myelodysplastic syndrome, pain, sarcopenia, cachexia, septic shock, systemic lupus erythematosus, leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, ankylosing spondylitis, myelofibrosis, B-cell lymphoma, hepatocellular carcinoma, Hodgkin’s disease, breast cancer, multiple myeloma, melanoma, non-Hodgkin lymphoma, non-small-cell lung cancer, ovarian clear cell carcinoma, ovary tumor, pancreas tumor, polycythemia vera, Sjogren’s syndrome, soft tissue sarcoma, sarcoma, splenomegaly, T-cell lymphoma, and thalassemia major.
[0141] Pharmaceutical Compositions
[0142] In some aspects, the present disclosure provides a pharmaceutical composition. The pharmaceutical composition may comprise a compound disclosed herein, such as a compound of Formula (I), and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated for oral administration. In some embodiments, the pharmaceutical composition is formulated for inhalation. In some embodiments, the pharmaceutical composition comprises a compound disclosed herein and an additional therapeutic agent. Non-limiting examples of such therapeutic agents are described herein below. [0143] Pharmaceutical compositions typically include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Formula (I), or a compound provided in Table 1 — described herein as the active agent. The active agent may be provided in any form suitable for the particular mode of administration, such as a free base, a free acid, or a pharmaceutically acceptable salt. Additionally, the methods and pharmaceutical compositions of the present disclosure include the use of N-oxides, crystalline forms (e.g., polymorphs), as well as metabolites of these compounds having similar activity. All tautomers of the compounds described herein are included within the scope of the present disclosure. Additionally, the compounds described herein encompass unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol and the like.
[0144] Suitable routes of administration include, but are not limited to, oral, intravenous, rectal, vaginal, aerosol, pulmonary, nasal, transmucosal, topical, transdermal, otic, ocular, and parenteral modes of administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
[0145] In certain embodiments, a compound described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained release formulation. In some embodiments, a long acting formulation is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. In some embodiments, a compound described herein is provided in the form of a rapid release formulation, an extended release formulation, or an intermediate release formulation. In some embodiments, a compound described herein is provided in the form of a nebulized formulation. In some embodiments, a compound described herein is administered locally to the lungs by inhalation.
[0146] Compounds of the present disclosure are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, 0.5 to 100 mg, 1 to 50 mg, or from 5 to 40 mg per day may be administered to a subject in need thereof. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
[0147] A compound of the present disclosure may be administered in a single dose. In some embodiments, a compound of the disclosure is administered in multiple doses, such as about once, twice, three times, four times, five times, six times, or more than six times per day. In some embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In some embodiments, a compound of the disclosure and an additional therapeutic agent are administered together about once per day to about 6 times per day. In some embodiments, the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or more than about one year. In some embodiments, a dosing schedule is maintained as long as necessary. A compound of the present disclosure may be administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[0148] Pharmaceutical compositions of the present disclosure typically contain a therapeutically effective amount of a compound of the present disclosure. Those skilled in the art will recognize, however, that a pharmaceutical composition may contain more than a therapeutically effective amount, e.g., bulk compositions, or less than a therapeutically effective amount, e.g., individual unit doses designed for coadministration to achieve a therapeutically effective amount.
[0149] Typically, pharmaceutical compositions of the present disclosure contain from about 0.01 to about 95% by weight of the active agent; including, for example, from about 0.05 to about 30% by weight; and from about 0. 1 % to about 10% by weight of the active agent. A pharmaceutical composition of the present disclosure may contain from 0.1 mg to 100 mg of the active agent, such as 1 mg to 20 mg or 1 mg to 10 mg of the active agent.
[0150] Any conventional carrier or excipient may be used in the pharmaceutical compositions of the present disclosure. The choice of a particular carrier or excipient, or combinations of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state. Additionally, the carriers or excipients used in the pharmaceutical compositions of this disclosure may be commercially-available. Conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
[0151] Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as com starch and potato starch; cellulose, such as microcrystalline cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer’s solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical compositions.
[0152] Pharmaceutical compositions are typically prepared by thoroughly and intimately mixing or blending the active agent with a pharmaceutically-acceptable carrier and one or more optional ingredients. The resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills and the like using conventional procedures and equipment.
[0153] In one aspect, the pharmaceutical composition is suitable for inhaled administration. Pharmaceutical compositions for inhaled administration are typically in the form of an aerosol or a powder. Such compositions are generally administered using inhaler delivery devices, such as a dry powder inhaler (DPI), a metered-dose inhaler (MDI), a nebulizer inhaler, or a similar delivery device.
[0154] In a particular embodiment, the pharmaceutical composition is administered by inhalation using a dry powder inhaler. Such dry powder inhalers typically administer the pharmaceutical composition as a free- flowing powder that is dispersed in a patient’s air-stream during inspiration. In order to achieve a free-flowing powder composition, the therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PLA), polylactide-co-glycolide (PLGA) or combinations thereof. Typically, the therapeutic agent is micronized and combined with a suitable carrier to form a composition suitable for inhalation.
[0155] A representative pharmaceutical composition for use in a dry powder inhaler comprises lactose and a micronized form of a compound disclosed herein. Such a dry powder composition can be made, for example, by combining dry milled lactose with the therapeutic agent and then dry blending the components. The composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
[0156] Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (IV AX); ClickHaler (Innovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Ellipta (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaler (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like. [0157] A pharmaceutical composition of the present disclosure may be administered by inhalation using a metered-dose inhaler. Such metered-dose inhalers typically discharge a measured amount of a therapeutic agent using a compressed propellant gas. Accordingly, pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant. Any suitable liquefied propellant may be employed, including hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HFA 134a) and 1,1,1,2,3,3,3-heptafluoro-w-propane, (HFA 227); and chlorofluorocarbons, such as CCI3F. In a particular embodiment, the propellant is a hydrofluoroalkane. In some embodiments, the hydrofluoroalkane formulation contains a co-solvent, such as ethanol or pentane, and/or a surfactant, such as sorbitan trioleate, oleic acid, lecithin, and glycerin.
[0158] A representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01% to about 5% by weight of a compound of the present disclosure; from about 0% to about 20% by weight ethanol; and from about 0% to about 5% by weight surfactant; with the remainder being an HFA propellant. Such compositions are typically prepared by adding chilled or pressurized hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic agent is micronized and then combined with the propellant. The composition is then loaded into an aerosol canister, which typically forms a portion of a metered-dose inhaler device.
[0159] Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
[0160] A pharmaceutical composition of the present disclosure may be administered by inhalation using a nebulizer inhaler. Such nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition to spray as a mist that is carried into the patient’s respiratory tract. Accordingly, when formulated for use in a nebulizer inhaler, the therapeutic agent can be dissolved in a suitable carrier to form a solution. Alternatively, the therapeutic agent can be micronized or nanomilled and combined with a suitable carrier to form a suspension.
[0161] A representative pharmaceutical composition for use in a nebulizer inhaler comprises a solution or suspension comprising from about 0.05 pg/mL to about 20 mg/mL of a compound of the present disclosure and excipients compatible with nebulized formulations. In one embodiment, the solution has a pH of about 3 to about 8.
[0162] Nebulizer devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative nebulizer devices or products include the Respimat® Softmist ™ Inhalaler (Boehringer Ingelheim); the AERx® Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus® Reusable Nebulizer or PARI eFlow®rapid Nebulizer System (Pari GmbH); and the like.
[0163] A pharmaceutical composition of the present disclosure may be prepared in a dosage form intended for oral administration. Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present disclosure as an active ingredient.
[0164] When intended for oral administration in a solid dosage form, the pharmaceutical compositions of the disclosure will typically comprise the active agent and one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate. Optionally or alternatively, such solid dosage forms may also comprise: fillers or extenders, binders, humectants, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the present pharmaceutical compositions.
[0165] Alternative formulations may include controlled release formulations, liquid dosage forms for oral administration, transdermal patches, and parenteral formulations. Conventional excipients and methods of preparation of such alternative formulations are described, for example, in the reference by Remington, supra. [0166] The following non-limiting examples illustrate representative pharmaceutical compositions of the present disclosure.
[0167] Dry Powder Composition
[0168] A micronized compound of the present disclosure (1 g) is blended with milled lactose (25 g). This blended mixture is then loaded into individual blisters of a peelable blister pack in an amount sufficient to provide between about 0. 1 mg to about 4 mg of the compound per dose. The contents of the blisters are administered using a dry powder inhaler.
[0169] Dry Powder Composition
[0170] A micronized compound of the present disclosure (1 g) is blended with milled lactose (20 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1:20. The blended composition is packed into a dry powder inhalation device capable of delivering between about 0.1 mg to about 4 mg of the compound per dose.
[0171] Metered-Dose Inhaler Composition
[0172] A micronized compound of the present disclosure (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL). The resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 pm. The micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1,1,2-tetrafluoroethane in an amount sufficient to provide about 0.1 mg to about 4 mg of the compound per dose when administered by the metered dose inhaler.
[0173] Nebulizer Composition
[0174] A representative nebulizer composition is as follows. A compound of the present disclosure (2 g of free-base equivalents) is dissolved in a solution containing 80 mL reverse-osmosis water, 0.1-1% by weight of anhydrous citric acid, and 0.5 -1.5 equivalents of hydrochloric acid, followed by addition of sodium hydroxide to adjust the pH to 3.5 to 5.5. Thereafter, between 4-6% by weight of D-mannitol is added and solution q.s. to 100 mL. The solution is then filtered through a 0.2 pm filter and stored at room temperature prior to use. The solution is administered using a nebulizer device that provides about 0.1 mg to about 4 mg of the compound per dose. [0175] Kits
[0176] In certain aspects, the present disclosure provides a kit comprising one or more unit doses of a compound or pharmaceutical composition described herein, optionally wherein the kit further comprises instructions for using the compound or pharmaceutical composition. In some embodiments, the kit comprises a carrier, package, or container that is compartmentalized to receive one or more containers, such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials, such as glass or plastic.
[0177] The articles of manufacture provided herein may contain packaging materials. Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. For example, the container(s) may include one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein. The container(s) may optionally have a sterile access port (for example, the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits may optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
[0178] In some embodiments, a kit includes one or more containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Nonlimiting examples of such materials include, but are not limited to, buffers, diluents, filters, needles, syringes, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. In certain embodiments, the pharmaceutical composition is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack may contain metal or plastic foil, such as a blister pack.
[0179] Combination Therapy
[0180] The compounds and pharmaceutical compositions of the disclosure may be used in combination with one or more therapeutic agents which act by the same mechanism or by a different mechanism to treat a disease. The one or more agents may be administered sequentially or simultaneously, in separate compositions or in the same composition. Useful classes of agents for combination therapy include, but are not limited to, a beta 2 adrenoceptor agonist, a muscarinic receptor antagonist, a glucocorticoid agonist, a G- protein coupled receptor-44 antagonist, a leukotriene D4 antagonist, a muscarinic M3 receptor antagonist, a histamine Hl receptor antagonist, an immunoglobulin E antagonist, a PDE 4 inhibitor, an IL-4 antagonist, a muscarinic Ml receptor antagonist, a histamine receptor antagonist, an IL-13 antagonist, an IL-5 antagonist, a 5 -Lipoxygenase inhibitor, a beta adrenoceptor agonist, a CCR3 chemokine antagonist, a CFTR stimulator, an immunoglobulin modulator, an interleukin 33 ligand inhibitor, a PDE 3 inhibitor, a phosphoinositide-3 kinase delta inhibitor, a thromboxane A2 antagonist, an elastase inhibitor, a Kit ty rosine kinase inhibitor, a leukotriene E4 antagonist, a leukotriene antagonist, a PGD2 antagonist, a TNF alpha ligand inhibitor, a TNF binding agent, a complement cascade inhibitor, an eotaxin ligand inhibitor, a glutathione reductase inhibitor, an histamine H4 receptor antagonist, an IL-6 antagonist, an IL2 gene stimulator, an immunoglobulin gamma Fc receptor IIB modulator, an interferon gamma ligand, an interleukin 13 ligand inhibitor, an interleukin 17 ligand inhibitor, a L-Selectin antagonist, a leukocyte elastase inhibitor, a leukotriene C4 antagonist, a Leukotriene C4 synthase inhibitor, a membrane copper amine oxidase inhibitor, a metalloprotease-12 inhibitor, a metalloprotease-9 inhibitor, a mite allergen modulator, a muscarinic receptor modulator, a nicotinic acety lcholine receptor agonist, a nuclear factor kappa B inhibitor, a p-Selectin antagonist, a PDE 5 inhibitor, a PDGF receptor antagonist, a phosphoinositide-3 kinase gamma inhibitor, a TLR-7 agonist, a TNF antagonist, an Abl tyrosine kinase inhibitor, an acetylcholine receptor antagonist, an acidic mammalian chitinase inhibitor, an ACTH receptor agonist, an actin polymerization modulator, an adenosine Al receptor antagonist, an adenylate cyclase stimulator, an adrenoceptor antagonist, an adrenocorticotrophic hormone ligand, an alcohol dehydrogenase 5 inhibitor, an alpha 1 antitrypsin stimulator, an alpha 1 proteinase inhibitor, an androgen receptor modulator, an angiotensin converting enzyme 2 stimulator, an ANP agonist, a Bcr protein inhibitor, a beta 1 adrenoceptor antagonist, a beta 2 adrenoceptor antagonist, a beta 2 adrenoceptor modulator, a beta amyloid modulator, a BMP 10 gene inhibitor, a BMP 15 gene inhibitor, a calcium channel inhibitor, a cathepsin G inhibitor, a CCL26 gene inhibitor, a CCR3 chemokine modulator, a CCR4 chemokine antagonist, a cell adhesion molecule inhibitor, a chaperonin stimulator, a chitinase inhibitor, a collagen I antagonist, a complement C3 inhibitor, a CSF-1 antagonist, a CXCR2 chemokine antagonist, a cytokine receptor common beta chain modulator, a cytotoxic T-lymphocyte protein-4 stimulator, a deoxyribonuclease I stimulator, a deoxyribonuclease stimulator, a dipeptidyl peptidase I inhibitor, a DNA gyrase inhibitor, a DP prostanoid receptor modulator, an E-Selectin antagonist, an EGFR family tyrosine kinase receptor inhibitor, an elastin modulator, an Endothelin ET-A antagonist, an Endothelin ET-B antagonist, an epoxide hydrolase inhibitor, a FGF3 receptor antagonist, a Fyn tyrosine kinase inhibitor, a GAT A 3 transcription factor inhibitor, a Glucosylceramidase modulator, a Glutamate receptor modulator, a GM-CSF ligand inhibitor, a Guanylate cyclase stimulator, a H+ K+- ATPase inhibitor, an hemoglobin modulator, an Heparin agonist, an Histone deacetylase inhibitor, an Histone deacetylase-2 stimulator, an HMG CoA reductase inhibitor, an I-kappa B kinase beta inhibitor, an ICAM1 gene inhibitor, an IL-17 antagonist, an IL-17 receptor modulator, an IL-23 antagonist, an IL-4 receptor modulator, an Immunoglobulin G modulator, an Immunoglobulin G1 agonist, an Immunoglobulin G1 modulator, an Immunoglobulin epsilon Fc receptor LA antagonist, an Immunoglobulin gamma Fc receptor IIB antagonist, an Immunoglobulin kappa modulator, an Insulin sensitizer, an Interferon beta ligand, an Interleukin 1 like receptor antagonist, an Interleukin 18 ligand inhibitor, an Interleukin receptor 17A antagonist, an Interleukin- 1 beta ligand inhibitor, an Interleukin- 5 ligand inhibitor, an Interleukin-6 ligand inhibitor, a KCNA voltage-gated potassium channel-3 inhibitor, a Kit ligand inhibitor, a Laminin -5 agonist, a Leukotriene CysLTl receptor antagonist, a Leukotriene CysLT2 receptor antagonist, a LOXL2 gene inhibitor, a Lyn tyrosine kinase inhibitor, a MARCKS protein inhibitor, a MDR associated protein 4 inhibitor, a Metalloprotease-2 modulator, a Metalloprotease-9 modulator, a Mineralocorticoid receptor antagonist, a Muscarinic M2 receptor antagonist, a Muscarinic M4 receptor antagonist, a Muscarinic M5 receptor antagonist, a Natriuretic peptide receptor A agonist, a Natural killer cell receptor modulator, a Nicotinic Ach receptor alpha 7 subunit stimulator, a NK cell receptor modulator, a Nuclear factor kappa B modulator, an opioid growth factor receptor agonist, a P-Glycoprotein inhibitor, a P2X3 purinoceptor antagonist, a p38 MAP kinase inhibitor, a Peptidase 1 modulator, a phospholipase A2 inhibitor, a phospholipase C inhibitor, a plasminogen activator inhibitor 1 inhibitor, a platelet activating factor receptor antagonist, a PPAR gamma agonist, a prostacyclin agonist, a protein tyrosine kinase inhibitor, a SH2 domain inositol phosphatase 1 stimulator, a signal transduction inhibitor, a sodium channel inhibitor, a STAT-3 modulator, a Stem cell antigen-1 inhibitor, a superoxide dismutase modulator, a T cell surface glycoprotein CD28 inhibitor, a T-cell surface glycoprotein CD8 inhibitor, a TGF beta agonist, a TGF beta antagonist, a thromboxane synthetase inhibitor, a thymic stromal lymphoprotein ligand inhibitor, a thymosin agonist, a thymosin beta 4 ligand, a TLR-8 agonist, a TLR-9 agonist, a TLR9 gene stimulator, a Topoisomerase IV inhibitor, a Troponin I fast skeletal muscle stimulator, a Troponin T fast skeletal muscle stimulator, a Type 1 IL-1 receptor antagonist, a Type II TNF receptor modulator, an ion channel modulator, a uteroglobin stimulator, and a VIP agonist.
[0181] In practicing any of the subject methods, a JAK inhibitor and a second therapeutic agent can be administered sequentially, wherein the two agents are introduced into a subject at two different time points. The two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, 1 or more months, or according to any intermittent regimen schedule disclosed herein.
[0182] In some embodiments, the JAK inhibitor and the second therapeutic agent are administered simultaneously. The two agents may form part of the same composition, or the two agents may be provided in one or more unit doses. When in separate doses, the two agents may be administered via different routes.
[0183] In some embodiments, the JAK inhibitor or the second therapeutic agent are administered parenterally, orally, inhalatively, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally. As used herein, a therapeutically effective amount of a combination of a JAK inhibitor and a second therapeutic agent refers to a combination of a JAK inhibitor and a second therapeutic agent, wherein the combination is sufficient to affect the intended application, including but not limited to, disease treatment, as defined herein. Also contemplated in the subject methods is the use of a sub-therapeutic amount of a JAK inhibitor and a second therapeutic agent in combination for treating an intended disease condition. The individual components of the combination, though present in sub-therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
[0184] The amount of the JAK inhibitor and the second therapeutic agent administered may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
[0185] Measuring an immune response and/or the inhibition of biological effects of JAK can comprise performing an assay on a biological sample, such as a sample from a subject. Any of a variety of samples may be selected, depending on the assay. Examples of samples include, but are not limited to blood samples (e.g. blood plasma or serum), exhaled breath condensate samples, bronchoalveolar lavage fluid, sputum samples, urine samples, and tissue samples.
[0186] A subject being treated with a JAK inhibitor and a second therapeutic agent may be monitored to determine the effectiveness of treatment, and the treatment regimen may be adjusted based on the subject’s physiological response to treatment. For example, if inhibition of a biological effect of JAK inhibition is above or below a threshold, the dosing amount or frequency may be decreased or increased, respectively. Alternatively, the treatment regimen may be adjusted with respect to an immune response. The methods can further comprise continuing the therapy if the therapy is determined to be efficacious. The methods can comprise maintaining, tapering, reducing, or stopping the administered amount of a compound or compounds in the therapy if the therapy is determined to be efficacious. The methods can comprise increasing the administered amount of a compound or compounds in the therapy if it is determined not to be efficacious. Alternatively, the methods can comprise stopping therapy if it is determined not to be efficacious. In some embodiments, treatment with a JAK inhibitor and a second therapeutic agent is discontinued if inhibition of the biological effect is above or below a threshold, such as in a lack of response or an adverse reaction. The biological effect may be a change in any of a variety of physiological indicators.
[0187] Specific agents that may be used in combination with the compounds disclosed herein include, but are not limited to, abatacept, abediterol, aclidinium, aclidinium bromide, ACT-774312, Actair, Actimmune, acumapimod, adalimumab, ADC-3680, Adipocell, AG-1321001, AG-NPP709, alicaforsen, Allergovac depot, alpha-cyclodextrin-stabilized sulforaphane, AM-211, ambroxol + erdosteine, Ampion, anakinra, ANB-020, andecaliximab, APL-1, apremilast, arformoterol, ARRY-502, ASM-024, ASM-8, aviptadil, AXP-1275, AXP- E, AZD-1419, AZD-7594, AZD-8871, AZD-9898, azithromycin, bambuterol, batefenterol, beclomethasone, beclomethasone dipropionate, bedoradrine, bencycloquidium bromide, benralizumab, bertilimumab, BI- 1060469, BI-1467335, BI-443651, bilastine, BIO-11006, bosentan, brodalumab, BTT-1023, budesonide, budesonide + salmeterol, canakinumab, carbon dioxide + perfluorooctyl bromide, CCI-15106, CDX-0158, CG-201, CHF-5992, CHF-6001, ciclesonide, ciprofloxacin, citrulline, CJM-112, CK-2127107, CLBS-03, CMP-001, CNTO-6785, colforsin daropate, CRTH2 antagonist, CYP-001, dalazatide, danirixin, Dectrekumab, dectrekumab + VAK-694, deflazacort, dexamethasone cipecilate, dexamethasone sodium phosphate, dexpramipexole, dociparstat, domase alfa, doxofylline, DS-102, DSP-3025, dupilumab, dust mite vaccine, duvelisib, ebastine, EC-18, eicosapentaenoic acid monoglycerides, Emedastine, emeramide, enobosarm, epinastine, Epsi-gam, erdosteine, esomeprazole, etanercept, fevipiprant, fexofenadine, Flunisolide, fluticasone, fluticasone + formoterol, fluticasone furoate, fluticasone propionate, fluticasone propionate + salmeterol xinafoate, formoterol, formoterol fumarate, FP-025, Gamunex, GC-1112, gefapixant, gefitinib, Gemilukast, gerilimzumab, glycopyrronium, glycopyrronium bromide, golimumab, GSK-2245035, GSK-2256294, GSK-2292767, GSK-2586881, GSK-3772847, hdm-ASIT+, HI- 1640V, histamine human immunoglobulin, HMP-301, human allogenic adipose-derived mesenchymal progenitor cell therapy, hyaluronic acid, iloprost, imatinib, IMD-1041, imidafenacin, indacaterol, indacaterol maleate, infliximab, inhaled interferon beta, INS- 1007, interferon gamma, ipratropium + fenoterol, ipratropium + salbutamol, ipratropium bromide, ivacaftor, lebrikizumab, lenzilumab, levalbuterol, levocetirizine, LH-011, ligelizumab, LT-4001, Lucinactant, lysine acetylsalicylate, mannitol, masilukast, masitinib, MEDI-3506, MEDI-9314, mepolizumab, metenkefalin acetate, methylprednisolone suleptanate, Mitizax, mizolastine, MK-1029, mogamulizumab, mometasone, mometasone furoate, monosodium alpha luminol, montelukast, MPC-300-IV, mugwort pollen allergen vaccine, MV-130, N-6022, nadolol, nedocromil, nemiralisib, nitric oxide, NKTT- 120, NOV-14, OATD-01, OligoG-COPD-5/20, olodaterol, olopatadine, omalizumab, OPK-0018, oxis Turbuhaler, Ozagrel, parogrelil, PBF-680, pemirolast, pemirolast sodium, PIN-201104, pitavastatin, PL-3994, plecanatide, POL-6014, pranlukast, prednisolone, PrEP-001, procaterol, PRS-060, PT-007, PUR-1800, QBKPN, QBW-251, ramatroban, RBx-10017609, recombinant midismase, REGN-3500, ReJoin, remestemcel-L, repirinast, reslizumab, revefenacin, RG-6149, RG-7990, RGN-137, risankizumab, rivipansel, RNS-60, roflumilast, rosiglitazone, rosiptor acetate, RP-3128, RPC-4046, RPL-554, R-TPR-022, rupatadine, RUTI, RV-1729, salbutamol, salbutamol HFA, salbutamol sulfate, salmeterol, salmeterol xinafoate, SB-010, SCM-CGH, secukinumab, SENS-111, seratrodast, setipiprant, SHP-652, sirukumab, sodium chromoglycate, sodium pyruvate, solithromycin, Stempeucel, suplatast, TA-270, tadekinig alfa, TBS-5, TD-5471, TEV- 46017, TEV-48107, tezepelumab, theophylline, timapiprant, tiotropium bromide, tiotropium bromide monohydrate, tipelukast, TR-4, tralokinumab, tranilast, trantinterol, tregalizumab, triamcinolone acetonide, tridecactide acetate, TRN-157, tulobuterol, UCB-4144, udenafil, umeclidinium, umeclidinium bromide, VALERGEN-DS, vamorolone, vilanterol, vilanterol trifenatate, VR-096, VR-179, VTX-1463, VX-371, VX- 561, WIN-901X, YHD-001, YPL-001, Zafirlukast, zileuton, ZPL-389, and ZPL-521.
[0188] In some embodiments, a compound disclosed herein is administered in combination with a second therapeutic agent, optionally wherein the second therapeutic agent is administered by inhalation. In some embodiments, the present disclosure provides a method of treating a JAK mediated disease or condition, such as a respiratory disease, in a subject, comprising administering to the subject a JAK inhibitor, such as a compound disclosed in Table 1, and one or more second therapeutic agents. In some embodiments, the present disclosure provides a method of treating asthma, such as T2-dominant (eosinophilic) asthma or non- T2-dominant (non-eosinophilic) asthma, in a subject, comprising administering to the subject a JAK inhibitor, such as a compound disclosed in Table 1, and a second therapeutic agent.
[0189] Also provided herein is a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents. The therapeutic agent may be selected from the classes of agents specified above and from the lists of specific agents described above. In some embodiments, the pharmaceutical composition is suitable for delivery to the lungs. In some embodiments, the pharmaceutical composition is suitable for inhaled or nebulized administration. In some embodiments, the pharmaceutical composition is a dry powder or a liquid composition.
[0190] Further, in a method aspect, the disclosure provides a method of treating a disease or disorder in a mammal comprising administering to the mammal a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
[0191] When used in combination therapy, the agents may be formulated in a single pharmaceutical composition, or the agents may be provided in separate compositions that are administered simultaneously or at separate times, by the same or by different routes of administration. Such compositions can be packaged separately or may be packaged together as a kit. The two or more therapeutic agents in the kit may be administered by the same route of administration or by different routes of administration.
[0192] The disclosure also includes compounds and pharmaceutical compositions as described herein for use in treating a disease or disorder as described herein. In addition, the disclosure includes the manufacture of a medicament comprising a compound or pharmaceutical composition for the treatment of a disease or disorder as described herein.
EXAMPLES
[0193] The following examples are given for the purpose of illustrating various embodiments of the disclosure and are not meant to limit the present disclosure in any fashion. The present examples, along with the methods and compositions described herein, are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the disclosure. Changes therein and other uses which are encompassed within the spirit of the disclosure as defined by the scope of the claims will occur to those skilled in the art.
[0194] The following abbreviations have the following meanings unless otherwise indicated and any other abbreviations used herein and not defined have their standard, generally accepted meaning:
ACN = acetonitrile
BoczO = di-tert-butyl dicarbonate
BSA = bovine serum albumin, Fraction V d = day(s) DCM = dichloromethane or methylene chloride
DIPEA or DIEA = A'. A'-diisopropylcthylaminc
DMF = A'. A'-dimcthylformamidc
DM SO = dimethyl sulfoxide
DPPF = l,l’-bis(diphenylphosphino)ferrocene
EDTA = ethylenediaminetetraacetic acid
EGTA = ethylene glycol-bis(P-aminoethyl ether)-
N,N,N’,N’ -tetraacetic acid
EtOAc or EA = ethyl acetate g = gram(s) h = hour(s)
HEPES = 4-(2-hyrdroxyethyl)-l -piperazine ethanesulfonic acid LDA = lithium diisopropylamide min = minute(s)
NBS = N-bromosuccinimide
Pd(dppf)C12 = [l,l'-Z>A(diphenylphosphino)ferrocene]- dichloropalladium(II) Pd(PPhs)4 = tetrakis(triphenylphosphine)palladium(0)
Pd/C = palladium on activated carbon, 10% loading
PE = petroleum ether
RT, rt, or r.t. = room temperature
SEMC1 = 2-(trimethylsilyl)ethoxymethyl chloride
SiO2 = silicon dioxide or silica
TFA = trifluoro acetic acid
THF = tetrahydrofuran
THP = tetrahydropyran
XPhos Pd G4 = Buchwald 4th generation palladacycle
[0195] Unless noted otherwise, all materials, such as reagents, starting materials and solvents, were purchased from commercial suppliers, such as Sigma- Aldrich, Fluka Riedel-de Haen, and the like, and were used without further purification.
[0196] Reactions were run under nitrogen atmosphere, unless noted otherwise. The progress of reactions was monitored by thin layer chromatography (TLC), analytical high-performance liquid chromatography (anal. HPLC), and mass spectrometry, the details of which are given in specific examples.
[0197] Reactions were worked up as described specifically in each preparation; commonly, reaction mixtures were purified by extraction and other purification methods such as temperature- and solvent-dependent crystallization, and precipitation. In addition, reaction mixtures were routinely purified by preparative HPLC, typically using Microsorb C18 and Microsorb BDS column packings and conventional eluents. Progress of reactions was typically monitored by liquid chromatography mass spectrometry (LCMS). Characterization of isomers was typically done by Nuclear Overhauser effect spectroscopy (NOE). Characterization of reaction products was routinely carried out by mass spectrometry and/or 1 H-NM R spectroscopy. For NMR measurement, samples were dissolved in deuterated solvent (CD3OD, CDC13, or DMSO-d6), and 1 H-NMR spectra were acquired with a Varian Gemini 2000 instrument (400 MHz) under standard observation conditions. Mass spectrometric identification of compounds was typically conducted using an electrospray ionization method (ESMS) with an Applied Biosystems (Foster City, CA) model API 150 EX instrument or an Agilent (Palo Alto, CA) model 1200 LC/MSD instrument.
[0198] Example 1 : Synthesis of (E)-3-ethyl-4-(3-(4-(3-(isopropylamino)prop-1-en-i-y1l)-1H- imidazol-2-yl)- 1H-indazol-6-yl)phenol (also known as 3-ethyl-4-[3-[4-[(E)-3-(isopropylamino)prop-l-enyl]-1H-imidazol-2- yl]- 1H-indazol-6-yl]phenol) (23).
Figure imgf000088_0001
Figure imgf000089_0001
[0199] Step A: Preparation of (2-((3-ethylphenoxy)metboxy)etbyl)trimethylsilane (1-2). To a stirred solution of 3-ethylphenol (200 g, 1.64 mol) (1-1) in DMF (1.50 L) cooled to 0 ºC was added NaH (78.6 g, 1.96 mol) portion wise. The reaction mixture was then stirred at 0 °C for Ih. SEMC1 (300 g, 1.80 mol) was then added drop-wise at 0 °C, and the reaction mixture was allowed to stir at room temperature for 2 h. TLC showed complete consumption of starting material. The reaction mixture was quenched with ice-water (2.0 L) and extracted with ethyl acetate (2 X 1.0 L). The combined organic layers were washed w ith brine solution (1.0 L), dried over and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography (5-10% EtOAc in heptane) to afford the desired product as a clear liquid (305 g, 74% yield). ‘HNMR (400 MHz, DMSO-d6) δ 7.17 (t, J = 7.8 Hz, IH), 6.88 - 6.77 (m, 3H), 5.20 (s, 2H), 3.69 (t, J = 8.0 Hz, 2H), 2.56 (q, .7 = 7.6 Hz, 211), 1.16 (t, J = 7.6 Hz, 3H), 0.88 (t, J = 8.1 Hz, 2.H), 0.00 (s, 9H).
[0200] Step B: Preparation of (2-((4-bromo-3 -ethylphenoxy )methoxy)ethyl)trim ethylsilane (1-3). To a stirred solution of 1-2 (200 g, 792 mmol) in ACN (1.40 L) cooled to 0 °C was added NBS (141 g, 792 mmol) portion wise over a period of 30 minutes. The resulting reaction mixture was stirred at room temperature for 2 h. TLC showed complete consumption of starting material. The reaction mixture was poured into ice cold water (IL) and extracted with EtOAc (2 x IL). The combined organic layers were washed with water (IL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude mixture was purified by silica gel column chromatography (100% heptane) to afford the desired product as a clear oil (230 g, 88% yield). !H NMR (400 MHz, DMSO-d6) δ 7.44 (dd, J = 8.8, 1 .9 Hz, IH), 6.99 (d, J = 2.9 Hz, 1H), 6.81 (dt, J = 8.8, 2.6 Hz, IH), 5.21 (d, J = 2.0 Hz, 2.H), 3.73 - 3.64 (m, 2H), 2.63 (qd, J = 7.5, 2.0 Hz, 2H), 1.14 (td, 7.6, 1.9 Hz, 3H ), 0.95 - 0.79 (m, 2H), 0.00 (s, 9H).
[0201] Step C: Preparation of (2-((3-ethyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)methoxy)ethyl)trimethylsilane (1-4). To a stirred solution of 1-3 (100 g, 302 mmol) in 1 ,4- dioxane (1 .00 L) were added bis(pinacolato)diboron (76.6 g, 302 mmol) and potassium acetate (59.2 g, 604 mmol). The reaction mixture was degassed with nitrogen for 15 minutes after which PdC12(dppf)-DCM (24.6 g, 30.2 mmol) was added. The reaction mixture was stirred and heated at 110 °C for 16 hours under nitrogen. TLC indicated the complete consumption of the starting material. The reaction mixture was diluted with EtOAc (1 L) and washed with water (1 L). The combined organic layers were separated, dried over Na2SO4, and concentrated. The crude mixture was purified by silica gel column chromatography (0-10% EtOAc in heptane) to afford the desired product as a yellow liquid (75.0 g, 66% yield). 1H NMR (400 MHz, DMSO-d6) 8 7.57 (d, J = 8.1 Hz, IH), 6.87 - 6.77 (m, 2H), 5.21 (d, J = 13.2 Hz, 2H), 3.69 (t, J = 8.0 Hz, 211 ). 2.80 (q, J === 7.5 Hz, 2H ), 1.27 (s, 12H), 1.11 (t . J = 7.5 Hz, 3H), 0.91 - 0.84 (m, 2H), 0.00 (s, 9H).
[0202] Step D: Preparation of 4-bromo-2-fluorobenzoyl chloride (1-6). To a stirred solution of 4-bromo~2- fluorobenzoic acid (50.0 g, 228 mmol) (1-5), in DCM (300 mL) and DMF (4.0 mL) was added oxalyl chloride (96.57 mL, 913 mmol) dropwise at 0 °C. The resulting reaction mixture was stirred at room temperature for 1 h. After completion of reaction (TLC monitoring, checked by quenching in MeOH), the reaction was concentrated under reduced pressure (under nitrogen) to afford an off-white solid (54.2 g) which w’as used in the next step without further purification.
[0203] Step E: Preparation of (l-benzyI-1H-imidazoI-2-yl)(4-bromo-2-fluorophenyI)methanone (1-7). To a stirred solution of 1 -benzyl- IH-imidazole (30.0 g, 190 mmol) in acetonitrile (165 mL) was added triethylamine (133.4 mL, 949 mmol) at room temperature. Compound 1-6 (54.2 g, 228 mmol) was taken up separately in acetonitrile (165 ml) and added to the reaction mixture. The reaction was allowed to stir at room temperature for 2 h. TLC showed consumption of starting material. The reaction wms quenched with cold water (500 mL) and extracted with ethyl acetate (2 x 600 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to get crude product which was purified by silica gel column chromatography (10% EtOAc in Heptane) to obtain the desired product as an off-white solid (79.0 g, 58% yield). 1H NMR (400 MHz, DMSO-d6) δ 7.80 (s, 1H), 7.68 (dd, J = 9.7, 1.8 Hz, 1H), 7.61 (t, J = 7.8 Hz, 1H), 7.54 (dd, J= 8.3, 1.8 Hz, 1H), 7.36 (dd, J = 8.1, 6.5 Hz, 2H), 7.32 - 7.26 (m, 1H), 7.25 (s, 1H), 7.22 (dd, J = 6.9, 1.8 Hz, 2H), 5.70 (s, 2.H). (m/z): [M+H]+ calcd for C17H13BrFN2O 359.02. found 358.97.
[0204] Step F : Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-1H-indazole (1-8). To a stirred solution of 1-7 (53.0 g, 147.5 mmol) in DMSO (105 mL) wrns added dropwdse hydrazine hydrate (72.5 mL, 1475.5 mmol) at room temperature. The reaction mixture was allowed to stir at 90 °C for 3 h. After 3 h, TLC showed complete consumption of starting material. The reaction mixture was diluted with ice cold water (800 mL) and precipitation was observed. The reaction was filtered and washed with ice cold water (500 mL) to afford the desired product as an off-white solid (47.0 g, 90% yield). *H NMR (400 MHz, DMSO-tfe) 8 8.36 (d, J= 8.6 Hz, 1H), 7.79 (d, J= 1.6 Hz, 1H), 7.40 (s, HI), 7.35 (dd, J = 8.6, 1.7 Hz, 1H), 7.29 (dd, J = 8.1, 6.5 Hz, 2H), 7.26 - 7.22 (m, 1H), 7.21 - 7.17 (m, 2H), 7.16 (s, 1 H), 5.84 (s, 2H). (m/z): [M+H]+ calcd for C 17H14BrN4353.04 found 353.03.
[0205] Step G: Preparation of 3-(l-ben2y4-1H-imidazol-2-yl)-6-bromo-l-(tetrahydro-2H-pyran-2-yl)-1H- indazole (1-9). To a stirred solution of 1-8 (47.0 g, 133.1 mmol) in ethyl acetate (350 mL) at 0 °C was added TFA (30.5 mL, 399.1 mmol). Dihydropyran (60.8 mL, 665.3 mmol) was added dropwise. The reaction mixture was then heated to 80 °C and stirred for 2 days. After 2 days, TLC showed complete consumption of starting material. The reaction mixture was diluted with water (400 mL) and extracted with ethyl acetate (2 x 900 mL). The combined organic layers were further washed with saturated aq. NaHCO3 solution (800 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude product was purified by silica gel column chromatograph}' (5% EtOAc in heptane) to afford the desired product as an off white solid (40 g, 69% yield). 1 H NMR (400 MHz, DMSO-d6) δ 8.37 (d, J 8.6 Hz, 1H), 8.07 (d, J = 1.6 Hz, 1H), 7.48 (d, J = 1.2 Hz, 1 H), 7.42 (dd, J = 8.6, 1.6 Hz, 1H), 7.33 - 7.25 (m, 2H), 7.25 - 7.19 (m, 3H), 7.19 (d, J = 1.2 Hz, 1H), 5.94 (dd, J = 8.9, 2.5 Hz, 1H), 5.86 (d, J = 15.3 Hz, 1H), 5.77 (d, J= 15.3 Hz, 1H), 3.78 (tp, J = 1 1.6, 3.8 Hz, 2H), 2.38 - 2.25 (m, 1H), 2.04 - 1.89 (m, 2H), 1.70 (dtt, 11.5, 8.5, 4.0 Hz, 1H), 1.62 - 1.45 (m, 2H). (m/z): [M+H]+ calcd for C22H22BrN4O 437.10 found 437.11.
[0206] Step H: Preparation of 3-(l-benzyI-1H-imidazoI-2-yl)-6-(2-ethyl-4-((2- (trimethy1silyl)ethoxy)methoxy)pheny1)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (1-10). To a stirred solution of 1-9 (60.0 g, 137 mmol) and 1-4 (62.3 g, 165 mmol) in dioxane (360 mL) and water (90.0 mL) was added K3PO4 (87.4 g, 412 mmol). The reaction mixture was purged with argon for 15 minutes then Pd/PPlrfo (15.9 g, 13.7 mmol) added to it. The reaction was then heated to 110 °C and stirred for 3 h. TLC showed consumption of the starting material. The reaction mixture was diluted with water (600 mL) and extracted w’ith ethyl acetate (2 x 500 mL). The combined organics were washed with brine (600 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography (10% EtOAC in heptane). The desired product was isolated as a clear liquid (65 g, 78% yield). 1H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 8.4 Hz, 1H), 7.61 (s, 1H), 7.47 (s, 1H), 7.34 - 7.27 (m, 2H), 7.27 - 7.20 (m, 3H), 7.20 - 7.14 (m, 3H), 7.01 (d, J = 2.6 Hz, 1H), 6.94 (dd, J = 8.4, 2.6 Hz, 1H), 5.98 - 5.87 (m, 2H), 5.85 - 5.74 (m, 1H), 5.27 (s, 2H), 3.82 (d, J = 1 1.4 Hz, 1H), 3.72 (q, J = 10.1 , 9.0 Hz, 3H), 2.55
Figure imgf000091_0001
. .7 7.5 Hz, 2.H), 2.35 (s, 1H), 1.98 (s, 2.H), 1.79 - 1.64 (m, 1H), 1.55 (s, 2.H), 1.04 (t, J--- 7.5 Hz, 3H), 0.92 (t, J--- 8.1 Hz, 2H), 0.00 (s, 9H). (m/z): [M+H]+ calcd for C36H45N4O3Si 609.33 found 609.38.
[0207] Step I: Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-3-(1H-imidazol-2-yl)- l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (1-11). To a stirred solution of 1-10 (65.0 g, 107 mmol) in isopropanol (450 mL) and THF (150.0 mL) was added 20% Pd(OH)2/C (60.0 g, 84.5 mmol). The reaction mixture was subjected to hydrogenation using a FL balloon and was allowed to stir at room temperature for 16 h. TLC showed complete consumption of starting material. The reaction mixture was filtered through a pad of Celite, washed with EtOAc (500 mL), and the filtrate was concentrated under reduced pressure to afford the crude desired product (53.0 g, 96% yield) as a colorless liquid that was used directly in the next step without further purification. *H NMR (400 MHz, DMSO-d6) δ 12.68 (s, 1H), 8.38 (d, J--- 8.3 Hz, 1H), 7.65 (s, 1H), 7.23 (s, HI), 7.19 (dd, J = 8.3, 3.7 Hz, 2H), 7.14 (s, 1H), 7.01 (s, HI), 6.95 (d, J = 9.4 Hz, HI), 5.95 (d, J = 9.8 Hz, 1H), 5.27 (s, 2H), 3.93 (d, J= 12.0 Hz, 1H), 3.74 (t, J = 8.1 Hz, 3H), 2.56 (d, J= 7.6 Hz, 2H), 2.03 (s, 2H), 1.76 (s, 1H), 1.58 (s, 2H), 1.05 (t, 7.5 Hz, 3H), 0.92 (t, J = 8.1 Hz, 2.H), 0.00 (s, 9H). (m/z): [M+H]+ calcd for C29H39N4O3Si 519.28 found 519.28.
[0208] Step J: Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-l-(tetrahydro-2H- pyran-2-yl)-3-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-2-yl)-1H-indazole (1-12). To a stirred solution of 1-11 (43.0 g, 82.9 mmol) in DMF (400 mL) -was added sodium hydride 60% w/w (4.97 g, 124 mmol) at 0 °C. The reaction mixture was then allowed to stir at 0 °C for 20 min, after which SEMC1 (17.6 mL, 99.5 mmol) was added dropwise at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 1 h. TLC showed complete consumption of the starting material. The reaction mixture was quenched with ice-water (1 L) and extracted ethyl acetate (3 x 500 mL). The combined organic layers were washed with water (800 mL) and brine (800 mL), dried over Na2SO4, and concentrated under reduced pressure to afford the crude compound which was purified by silica gel column chromatography (15% EtOAc in heptane). The desired product was isolated as an off white solid (42.0 g, 66% yield). 1H NMR (400 MHz, Chloroform-d) δ 8.50 (d, J= 8.4 Hz, 1H), 7.50 (s, 1H), 7.26 (s, 2H), 7.25 - 7.19 (m, 2H), 7.02 (s, 1H), 6.96 (d, J = 8.5 Hz, 1H), 6.09 (d, J = 10.5 Hz, 1H), 5.93 (d, J = 10.5 Hz, 1H), 5.75 (d, J = 9.0 Hz, 1H), 5.28 (s, 2H), 4.03 (d, J = 11.5 Hz, 1H), 3.81 (t , J = 8.4 Hz, 2H), 3.74 (t, J = 10.0 Hz, 2H), 3.63 (d, J = 8.5 Hz, 1H), 3.58 (t, J = 8.2 Hz, 2H), 2.59 (q, J = 7.5 Hz, 3H), 2.13 (d, J = 16.5 Hz, 2H), 1.83 - 1.70 (m, 2H), 1.10 (t, J = 7.5 Hz, 2H), 1.00 (t, ./ = 8.3 Hz, 2H), 0.88 (q, J = 7.3, 6.5 Hz, 2H), 0.00 (s, 9H), -0.09 (s, 9H). (m/z): [M+H]+ calcd for C35H53N4O4Si2 649.36 found 649.49.
[0209] Step K: Preparation of 3-(4-bromo-l-((2-(trimethylsilyl)etihoxy)methyl)-1H-imidazol-2-yl)-6-(2- ethy l-4-((2-(trimethylsilyl)ethoxy )methoxy)phenyl)~ 1 -(tetrahydro-2H-pyran-2-y 1)- 1 H-indazole (1-13). NBS (2.74 g, 15.4 mmol) was taken up in DCM (100 mL) and added dropwise to a stirred solution of 1-12 (10.0 g, 15.4 mmol) in DCM (400 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 5 minutes. TLC showed complete consumption of the starting material. The reaction mixture was quenched with ice water (300 mL) and extracted with DCM (2 x 250 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and evaporated to afford crude product which was purified by silica gel column chromatography (8- 10% EtOAc in heptane). The desired product was isolated as a colorless amorphous solid (9.10 g, 81% yield). 1H NMR (400 MHz, DMSO--d6) δ 8.31 (d, J = 8.3 Hz, 1H), 7.68 (s, 1H), 7.32 (s, 1H), 7.19 (dd, J = 13.4, 8.4 Hz, 2H), 7.01 id. J - ---- 2.5 Hz, 1 H ), 6.95 (dd, J - ---- 8.4, 2.5 Hz, 1H), 6.09 - 5.94 (m, 3H), 5.27 (s, 2H), 3.89 (d, J ----- 11.4 Hz, 1H), 3.78 (d, J = 5.7 Hz, 1 H), 3.74 (t, J--- 8.1 Hz, 2H), 3.52 (dt, J 16.0, 8.0 Hz, 2H), 2.55 (t, J - ---- 7.5 Hz, 2H), 2.44 (s, 1H), 2.05 (d, J = 11.1 Hz, 2H), 1.77 (s, 1H), 1.59 (s, 2H), 1.23 (s, 1 H), 1.03 (t, J = 7.5 Hz, 2H), 0.92 (t, J = 8.1 Hz, 2H), 0.84 (td, J = 10.5, 9.0, 5.4 Hz, 2H), 0.00 (m, 9H), -0.19 (s, 9H). (m/z): [M+H]+ calcd for C35H52N4O4Si2 72.7.2.7 found 72.7.58.
[0210] Step L: Preparation of 4-(3-(4-bromo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazol-6- yl)-3 -ethylphenol (1-14). To a stirred solution of 1-13 (32.0 g, 44.0 mmol) in THF (100 mL) was added TBAF (IM in THF) (448 mL, 448 mmol) at room temperature. The reaction mixture was then heated to 80 °C and stirred for 2 days. TLC indicated complete consumption of the starting material. The reaction mixture was diluted with ethyl acetate (500 mL) and washed with water (3 x 300 mL) and brine (300 mL). The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (20% EtOAc in heptane) to afford the desired product as an off- white solid (13.7 g, 66% yield). 1H NMR (400 MHz, DMSO-d6) δ 13.10 (s, 1H), 9.43 (s, 1H), 8.26 (d, J = 8.3 Hz, 1H), 7.64 (s, 1H), 7.39 (d, J = 1.9 Hz, 1H), 7.20 (dd. J = 8.4, 1.3 Hz, 1 H), 7.07 id. J = 8.2 Hz, 1 H), 6.76 (d, J = 2.5 Hz, 1H), 6.69 (dd, J = 8.2, 2.5 Hz, 1H), 5.96 (dd, J = 10.0, 2.4 Hz, 1H), 3.92 (d, J = 11.4 Hz, 1H), 3.77 (dt, .7 = 1 1.5, 6.9 Hz, 1 H), 2.07 (s, 2H), 2.03 (s, 1 H), 1.76 (s, 1H), 1.58 (p, J = 5.0 Hz, 2H), 1.04 (t, J = 7.5 Hz, 3H). (m/z): [M+H]+ calcd for C22H24BrN4O2 469.11 found 469.36.
[0211] Step M: Preparation of tert-butyl (E)-(3-(2-(6-(2-ethyl-4-hydroxyphenyl)-l-(tetrahydro-2H -pyran-2- yl)-1H- indazol-3-yl)-1H- imidazol-4-yl)allyl)carbamate (1-15). To a solution of 1-14 (180 mg, 0.385 mmol) in dioxane (3 mL) was added both tert-butyl (E)-(3-(4,4,5-trimethyl-l,3,2-dioxaborolan-2-yl)allyl)carbamate (218 mg, 0.770 mmol) and a solution of sodium carbonate (122 mg, 1 .155 mmol) in water (1 mL). Following this, the reaction mixture was sparged with nitrogen for 10 minutes. Next, Xphos Pd G3 (19.2 mg, 0.019 mmol) was added and the reaction mixture was sparged with nitrogen for an additional 10 minutes. The reaction mixture was put on a heating mantle set to 110 °C and stirred for 1 h. The reaction mixture was cooled to room temperature and the volatiles were removed under reduced pressure. The erode product was used directly in the next step without further purification, (m/z): [M+H]+ calcd for C31H37N5O4 544.28, found 544.00.
[0212] Step N: Preparation of (E)-4-(3-(4-(3-aminoprop-l-en-l-yl)- 1H-imidazol-2-yI)-1H -indazol-6-yl)-3- ethylphenol (1-16). To a solution of 1-15 (209 mg, 0.384 mmol) in DCM (2 mL) was added trifluoro acetic acid (2 mL). The reaction mixture was stirred at room temperature for 2 h, after which volatiles were removed under reduced pressure. The crude product was purified by preparative HPLC chromatography (10-50% ACN/water gradient with 0.05% trifluoroacetic acid) to afford the TFA salt of the product as a white solid (36 mg, 26.1% yield), (m/z): [M+H]+ calcd for C21H21N5O 360.17, found 360.20.
[0213] Step O: Preparation of (E)-3-ethyl-4-(3-(4-(3-(isopropylamino)prop-l-en-l-yl)-1H -imidazol-2-yl)-1H- indazol-6-yl)phenol (23). To a solution of 1-16 (10 mg, 0,028 mmol) in methanol (2 mL) was added acetone (100 pi, 1.36 mmol), followed by sodium cyanoborohydride (9 mg, 0.139 mmol). The reaction mixture was stirred at room temperature for 1.5 h. The reaction mixture was concentrated under reduced pressure, and the residue was purified by preparative HPLC chromatography' to afford the TFA salt of the title compound as a white solid (3.4 mg, 24% yield), (m/z): [M+H]+ calcd for C24H27N5O 402.22, found 402.20. [0214] Example 2: Synthesis of (E)-3-ethyl-4-(7-fluoro-3-(4-(3-((tetrahydro-2H-pyran-4-yl)amino)prop-1- en- 1 -y1)- 1 H-imidazol-2-y1)- 1H-indazol-6-yl)phenol (64).
Figure imgf000094_0001
[0215] Step A: Preparation of 4-bromo-2,3 -difluorobenzoyl chloride (2-2). To a stirred solution of 4-bromo- 2,3 -difluorobenzoic acid (28.0 g, 118 mmol) (2-1) in DCM (300 mL) and DMF (915 μL, 0.1 eq., 11.8 mmol)) was added oxalyl chloride (40.5 mL, 473mmol) dropwise ai room temperature. The resulting reaction mixture was stirred at room temperature for 2h. After completion of reaction (TLC monitoring, checked by quenching in MeOH), the reaction was concentrated under reduced pressure (under nitrogen) to afford an off-white solid (31.0 g) which was used in the next step without further purification.
[0216] Step B: Preparation of (1-benzyl-1H-imidazol-2-yl)(4-bromo-2,3-difluorophenyl)methanone (2-3). To a stirred solution of compound 2-2 (16.0 g, 101 mmol) in acetonitrile (100 ml) was added triethylamine (51 .2 g, 506 mmol). 1 -benzyl- 1H-imidazole (31.0 g, 121 mmol) was dissolved in acetonitrile (100 mL) separately and added to the reaction mixture at room temperature. The reaction was allowed to stir at room temperature for 2 h. TLC showed consumption of starting material. The reaction was quenched with cold water (500 mL) and extracted with ethyl acetate (2 x 600 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to get crude product which was purified by silica gel column chromatography (10-15% EtOAc in Heptane) to obtain the desired product as a light yellow solid (28.0 g, 73% yield). 1H NMR (400 MHz, DMSO-d6) δ 7.83 (s, IH), 7.66 (ddd, J = 8.1, 5.9, 1.8 Hz, IH), 7.46 tddd, J = 8.4, 6.4, 2.0 Hz, IH), 7.36 (t, ./ = 7.4 Hz, 2H), 7.30 (t, J= 7.0 Hz, IH), 7.28 (s, 2H), 7.23 (d, J = 6.9 Hz, 2H), 5.70 (s, 2.H). (m/z): [M+H]+ caicd for CnHizBrFjNzO 377.01 found 374.94.
[0217] Step C: Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-7-fluoro-1H-indazole (2-4). To a stirred solution of 2-3 (21.7 g, 57.5 mmol) in DMSO (120 mL) was added dropwise hydrazine hydrate (28.0 mL, 575 mmol) at room temperature. The reaction mixture was allowed to stir at 90 °C for 3 h. After 3 h, TLC showed complete consumption of SM. The reaction mixture was diluted with ice cold water (2 x 500 mL) and precipitation was observed. The reaction was filtered and washed with ice cold water (500 mL) to afford the desired product as an off-white solid (20.0 g, 87% yield). 5H NMR (400 MHz, DMSO-cfc) 5 8.19 (d, J = 8.7 Hz, 1 H), 7.43 (s, IH), 7.40 (dd, J = 8.6, 5.8 Hz, IH), 7.30 (!. J =- 7.3 Hz, 2H), 7.24 (d, J = 7.1 Hz, IH), 7.20 (d, J= 1.7 Hz, IH), 7.18 (s, 2H), 5.84 (s, 2H). (m/z): [M+H]+ caicd for CnHuBrFN* 373.03 found 372.94.
[0218] Step D: Preparation of 3-(l-benzyl-1H-imidazol-2-yl)-6-bromo-7-fluoro-l-(tetraliydro-2H-pyran-2- yl)-1H-indazole (2-5). To a stirred solution of 2-4 (20.0 g, 53.9 mmol) in ethyl acetate (350 mL) -was added TFA (12.4 mL, 162 mmol). Dihydropyran (23.6 mL, 269 mmol) was added dropwise at 0 °C. The reaction mixture vras then heated to 80 °C and stirred for 2 days. After 2 days, TLC showed complete consumption of starting material. The reaction mixture was diluted with water (400 mL) and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were further washed -with saturated aq. NaHCOs solution (800 mL), dried over anhydrous NazSCfi, filtered and concentrated. The crude product vras purified by silica gel column chromatography (8-10% EtOAc in heptane) to afford the desired product as an off white solid (19.5 g, 78% yield).1H NMR (400 MHz, DMSCMs) 5 8.23 (d, J = 8.6 Hz, IH), 7.52 (s, IH), 7.49 (dd, J = 8.6, 5.6 Hz, IH), 7.30 (dd, J--- 8.0, 6.5 Hz, 2H), 7.24 (d, J = 6.6 Hz, IH), 7.21 (d, J = 2.9 Hz, 2H), 7.19 (s, IH), 5.87 (dd, J
6.9, 2.4 Hz, HI), 5.86 id. J--- 15.3 Hz, IH), 5.77 (d, J = 15.3 Hz, IH), 3.87 - 3.80 (m, IH), 3.65 (td, J = 11.1, 3.3 Hz,1H), 2.36 - 2.27 (m, 1H), 2.01 (s, 1H), 1.75 - 1.67 (m, 1H ), 1.61 - 1.42 (m, 1H), (m/z): [ M +H ] +calcd for C22H21BrFN4O 455.09 found 455.06.
[0219] Step E: Preparation of 3-(1-benzyl-1H-imidazol-2-yl)-6-(2-ethyl-4-((2-
(trim ethylsilyl)ethoxy)m ethoxy )phenyl)-7-fluoro-1-(tetrahydro-2H-pyran -2 -yl)- 1H-indazole (2-6). To a stirred solution of 2-5 (19.5 g, 42.8 mmol) and 1-4 (17.8 g, 47.1 mmol) in dioxane (200 mL) and water (20.0 mL) was added K3PO4 (27.3 g, 128 mmol). The reaction mixture was purged with argon for 5 minutes, and PdCl2(dppf).-DCM (3,49 g, 4.28 mmol) was then added to it. The reaction was then heated to 100 °C and stirred for 16 h. TLC showed consumption of the starting material. The reaction mixture was filtered through a pad of Celite and the residue washed with ethyl acetate (2 x 200 mL). The combined organics were washed with cold wmter (300 mL) and brine (300 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography (12% EtOAC in heptane). The desired product was isolated as a clear amorphous solid (20.0 g, 73% yield).1 H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J = 8.4 Hz, 1H), 7.52 (s, 1H), 7.30 (d, 7.2 Hz, 2H), 7.25 (d, J = 6.3 Hz, 2H),
7.22 (d, J = 3.6 Hz, 2H), 7.16 (d, J = 8.4 Hz, 1H), 7.11 (t, J = 7.2 Hz, 1H), 7.03 (d, J = 2.6 Hz, 1H), 6.95 (dd, J-= 8.5, 2.6 Hz, 1H), 5.94 - 5.84 (m, 2H), 5.80 (d, J = 15.4 Hz, IH), 5.28 (s, 2H), 3.84 (s, 1H), 3.74 (t, J= 8.1 Hz, 2H), 3.58 (d, J = 11.9 Hz, IH), 2.04 (d, J = 14.0 Hz, 2H), 1.69 (s, 1H), 1.53 (s, 2H), 1.44 (s, 1H), 1.24 (s, 1H), 0.99 (t, J = 7.5 Hz, 3H), 0.92 (t, J = 8.1 Hz, 2H), 0.88 -- 0.79 (m, 2H), 0.00 (s, 9H). [ M+H ]+ calcd for C36H44FN4O3Si 627.32 found 627.54.
[0220] Step F: Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-3-(1 H- imidazol-2-yl)-1 -(tetrahydro-2H-pyran-2-yl)-1 H-indazole (2-7). To a stirred solution of 2-6 (20.0 g, 31.9 mmol) in isopropanol (200 mL) and THF (50.0 mL) was added 20% Pd(OH)2/C (20.0 g, 163 mmol). The reaction mixture was subjected to hydrogenation using a H2 balloon and was allowed to stir at room temperature for 5 h. TLC showed complete consumption of starting material. The reaction mixture was filtered through a pad of Celite and the filtrate was concentrated under reduced pressure to afford the crude desired product (18.0 g, 71% yield) as a transparent amorphous solid that was used directly in the next step without further purification. 1H NMR (400 MHz, DMSO-d6) δ 8.23 (d, J = 8.2 Hz, IH), 7.25 (s, 2H), 7. 18 (d, J = 8.3 Hz, 1H), 7.12 (dd, J = 8.3, 5.9 Hz, 1 H), 7.04 (d, J= 2.6 Hz, 1H), 6.96 (dd, J= 8.4, 2.6 Hz, IH), 5.90 (d, J= 9.4 Hz, 1H), 5.76 (s, 1H), 5.29 (s, 2H), 3.95 (d, J= 11.6 Hz, 1 H), 3.75 (t, J= 8.1 Hz, 2H), 3.65 (s, 1H), 2.44 (s, 2H), 2.09 (d, J= 13.8 Hz, 2H), 1.75 (s, IH), 1.57 (s, 2H), 1.44 (s, IH), 1.01 (t, J= 7.5 Hz, 3H), 0.91 (d, J 8.1 Hz, 2H), 0.00 (s, 9H). (m/z): [M+H]+ calcd for C29H38FN4O3Si 537.27 found 537.36.
[0221] Step G: Preparation of 6-(2-ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l- (tetrahydro-2H-pyran-2-yl)-3-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-2-yl)-1H-indazole (2-8). To a stirred solution of 2-7 (18.0 g, 33.5 mmol) in DMF (180 mL) was added sodium hydride (2.81 g, 70.3 mmol) at 0 °C. The reaction mixture was then allowed to stir at 0 °C for 30 min, after which SEMC1 (8.39 g, 50.3 mmol) was added dropwise at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 2 h. TLC showed complete consumption of the s tarting material. The reaction mixture was quenched with ice-water (300 mL) and extracted in ethyl acetate (2 x 300 mL). The combined organic layers were washed with water (400 mL) and brine (400 mL), dried over Na2SSO4, and concentrated under reduced pressure to afford the crude compound which w;as purified by silica gel column chromatography (20-25% EtOAc in heptane). The desired product was isolated as an off white solid (18.0 g, 71% yield). 1H NMR (400 MHz, DMSO-d6) δ 8.22 (d, .J = 8.3 Hz, 1H), 7.53 (s, 1H), 7.21 (s, 1H), 7.17 (d, J = 8.3 Hz, 1H), 7.12 (dd, J = 8.3, 5.9 Hz, 1H), 7.04 (d, J= 2.6 Hz, 1H), 6.96 (dd, J = 8.4, 2.6 Hz, IH), 5.99 - 5.88 (m, 3H), 5.2.9 (s, 2H), 3.92. (d, J = 11.4 Hz, 1H), 3.75 (t, J= 8.1 Hz, 2H), 3.67 (d, J- 12.9 Hz, IH), 3.51 (t, J= 8.0 Hz, 2H), 2.43 (s, 3H), 2.19 -- 2.11 (m, 1H), 2.06 (s, IH), 1.76 id. J= 13.7 Hz, 2H), 1.57 (s, 2H), 1.00 (t, J = 7.5 Hz, 3H), 0.92 (t, J= 8.1 Hz, 211), 0.80 (t, J= 8.0 Hz, 2H), 0.00, (s, 9H), -0.15 (s, 9H). (m/z): [M+H]+ calcd for C 35 H52FN 4O4Si2 667.35 found 667.47. [0222] Step H: Preparation of 3-(4-bromo-l-((2-(trimethylsilyl)ethoxyjmethyl)-1H-imidazol-2-yl)-6-(2- ethyl-4-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (2-9). NBS (1 .60 g, 9.0 mmol) was taken up in DCM (60 mL) and added dropwise to a stirred solution of 2-8 (6.0 g, 9.0 mmol) in DCM (240 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 5 minutes. TLC showed complete consumption of the starting material. The reaction mixture was quenched with water (100 mL) and extracted with DCM (2 x 300 mL). The combined organic layers were dried over anhydrous
Figure imgf000097_0001
filtered and evaporated to afford crude product which wrns purified by silica gel column chromatography (8-10% EtOAc in heptane). The desired product wrns isolated as a colorless amorphous solid (5.0 g, 60% yield). 1H NMR (400 MHz, DMSO-O 8 8.17 (d, J = 8.4 Hz, 1H), 7.35 (s, 1H), 7.14 (q, J = 7.6, 6.7 Hz, 2H), 7.03 (d, J- ---- 2.4 Hz, 1H), 6.96 (d, J - ---- 7.9 Hz, 1H), 5.98 (s, 1 H), 5.92 (d, J 9.6 Hz, 2H), 5.28 (s, 2H), 3.92 (d, J - ---- 11.1 Hz, HI), 3.74 (t, ,7= 8.0 Hz, 2H), 3.66 (s, 1H), 3.51 {t. .7 8.1 Hz, 2H), 2.44 (d, J = 8.1 Hz, 3H), 2.15 (d. J 12.9 Hz, 1H), 2.06 (s, 1H), 1.77 (s, 1H), 1.57 (s, 2H), 0.99 (t, .7 = 7.5 Hz, 3H), 0.92 (t, J = 8.0 Hz, 2H), 0.78 (t, J = 8.0 Hz, 3H), 0.00 (s, 9H), -0.17 (d, J = 1 .9 Hz, 9H). (m/z): [M+H]+ calcd for C 35 H51FN 4O4Si2 747.26 found 747.26.
[0223] Step 1: Preparation of 4-(3-(4-bromo-1H-imidazol-2-yl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H- indazol-6-yT)-3 -ethylphenol (2-10). To a stirred solution of 2-9 (15.0 g, 20.1 mmol) in THF (30 mL) was added TBAF (IM in THF) (52.0 mL, 18.5 mmol) at 0 °C. The reaction mixture was then heated to 80 °C and stirred for 2 days, TLC indicated complete consumption of the starting material. The reaction mixture was diluted with water (500 mL) and extracted with ethyl acetate (2 x 500 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (20-25% EtOAc in heptane) to afford the desired product as an off-white solid (5.25 g, 53% yield). 1H NMR (400 MHz, DMSO-d6) 9.53 (s, 1H), 8.12 (d, J = 8.2 Hz, 1H), 7.44 (s, 1H), 7.13 (dd, J = 8.3, 6.0 Hz, 1H), 7.04 (d, J =8.2Hz, 1 H ), 6.78 (d, J = 2.6 Hz, 1H), 6.70 (dd, J = 8.2, 2.6 Hz, 1H), 5.93 -- 5.86 (m, 1H), 3.95 (d, J= 1 1.3Hz,1H), 3.65 (dt, J= 11.8, 6.6 Hz, 1H), 2.39 (d, J - 7.7 Hz, 3H), 2.09 (q, J = 10.0, 7.3 Hz, 3H), 1.75 (s, 1H), 1.57 (d, J = 8.6 Hz, 2H), 0.99 (t, J = 7.5 Hz, 3H). (m/z): [M+H]+ calcd for t . :H; :Bri .\ :O- 487.10 found 487.35.
[0224] Step J: Preparation of tert-butyl (E)-(3-(2-(6-(2-ethyl-4-hydroxyphenyl)-7-fluoro-l -(tetrahydro-2H- pyran-2-yl)-1H-indazol-3-yl)-1H-imidazol-4-yl)allyl)carbamate (2-11). To a solution of 2-10 (70 mg, 0.144 mmol) in dioxane (0.57 mL) was added both tert-butyl (£)-(3-(4,4,5-trimethyl-l,3,2-dioxaborolan-2- yl)allyl)carbamate (82 mg, 0.288 mmol) and a solution of potassium phosphate, tribasic (92 mg, 0.433 mmol) in water (0.14 mL). The reaction mixture was then sparged with nitrogen for 10 minutes. XPhos Pd G4 (6 mg, 7.2 pmol) was added and the reaction mixture was heated to 110 °C overnight. Following this, the reaction mixture was cooled to room temperature and passed through a plug of Celite to afford the crude product, which was used without further purification, (m/z): [M+H] ' calcd for C 31 H36FN 5O4562.28, found 562.05.
[0225] Step K: Preparation of (E)-4-(3-(4-(3-aminoprop-l-en-l-yl)-1 H-imidazol-2-yl)-7-fluoro-1 H-indazol- 6-yl)-3 -ethylphenol (2-12). To 2-11 (81 mg, 0.144 mmol) was added trifluoroacetic acid (1 mL) and the reaction mixture was stirred at 50 °C for 30 minutes. The reaction mixture was then concentrated under reduced pressure and purified by preparative HPLC chromatography (5-70% ACN/water gradient with 0.05% trifluoroacetic acid) to obtain the TFA salt of the title compound as a white solid (49 mg, 69. 1% yield), (m/z): [M+H]+ calcd for C 21 H20FN 5O378.17, found 378.02.
[0226] Step L: Preparation of (E)-3-ethyl-4-(7-fluoro-3-(4-(3-((tetrahydro-2H-pyran-4-yl)amino)prop-l-en- 1-yl)-1H-imidazol-2-yl)- 1H-indazol-6-yl)phenol (64). To a solution of 2-12 (25 mg, 0.051 mmol) in methanol (1 mL) was added tetrahydro-4H-pyran-4-one (7.5 mg, 0.076 mmol), followed by sodium cyanoborohydride (12,8 mg, 0.203 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was then concentrated under reduced pressure and purified by preparative HPLC chromatography (5-70% ACN/water gradient with 0.05% trifluoroacetic acid) to obtain the TFA salt of the title compound as a white solid (28 mg, 96% yield), (m/z): [ M i l ; calcd for C 26 H28FN 5O4462.22, found 462. 10.
[0227] Example 3: Synthesis of (S,E)-3-ethy1-4-(3-(4-(2-(morpholin-3-yl)vinyl)-1H-imidazol-2-yl)-1H- indazol-6-yl)phenol (6).
Figure imgf000098_0001
Figure imgf000099_0001
[0228] Step A: Preparation of ethyl 1-(N,N-dimethylsulfanioyl)-1H-imidazole-4-carboxylate (3-2). To a stirred solution of ethyl 1H-imidazole-4-carboxylate (30 g, 214.2 mmol) (3-1) in DMF (300 mL) was added NaH (12.8 g, 321.4 mmol) portion wise at 0 °C. Then, dimethylsulfamoyl chloride (27.7 mL, 257 mmol) was added and the reaction mixture was stirred at room temperature for 2 h. After the reaction was judged complete by TLC, the reaction mixture was quenched by addition of cold water (600 mL) and extracted with ethyl acetate (2x600 mL). The combined organic layers were dried over sodium sulfate and concentrated, then the crude product was purified by silica gel chromatography (50% EtOAc/Hexanes) to afford the desired product as an off-white solid (34,5 g, 65.4% yield).
Figure imgf000099_0002
NMR (400 MHz, DMSO-ds) 8.29 (s, 1H), 8.24 (s, 1H), 4.27 (q, J = 6.8 Hz, 2H), 2.87 (s, 6H), 1.29 (t, J = 6.8 Hz, 3 H).
[0229] Step B: Preparation of ethyl l-(dimethylsulfamoyl)-2-iodo-1H-imidazole~4-carboxylate (3-3). To a stirred solution of 3-2 (7 g, 28.3 mmol) in anhydrous THF (60 mL) was added LDA (2M in THF, 17 mL, 34 mmol) at -78 °C and the reaction was stirred for 10 minutes. Iodine (7.9 g, 31 .1 mmol) in anhydrous THF (40 mL) was added, and the reaction mixture was stirred at -78 °C for 5 minutes. After the reaction was judged complete by TLC, the reaction mixture w as quenched by addition of saturated ammonium chloride solution (200 mL), extracted wdth ethyl acetate (2x300 mL), and washed with aqueous sodium thiosulfate solution (200 mL). The organic layer was then dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (30% EtOAc/Heptane) to afford the desired product as a light yellow solid (3.5 g, 33.1% yield). 1H NMR (400 MHz, DMSO-d6) 8.20 (s, 1H), 4.24 (q, J = 7 Hz, 2H), 2.99 (s, 6H), 1.27 (t, J = 7 Hz, 3H).
[0230] Step C: Preparation of l-(benzyloxy)-3 -ethylbenzene (3-5). To a stirred solution of 3-ethylphenol (25.0 g, 204.0 mmol) (3-4) in ACM (250 mL, 10 vol) was added potassium carbonate (42.0 g, 306 mmol) at room temperature. The resulting reaction mass was stirred at room temperature for 15 minutes, followed by the addition of benzyl bromide (24.0 mL, 204 mmol) in dropwise manner. The resulting reaction mixture was stirred for 6 hours at room temperature. After completion of the reaction (TLC monitoring), the resulting reaction mass was poured into water (1.0 L) followed by extraction of compound with EtOAc (2 x 2L). The combined organics were washed with cold water and brine solution, then dried over sodium sulfate, filtered and evaporated under reduced pressure. The crude product was purified by column chromatography’ over silica gel (100-200M) by using eluents 2% EtOAc in hexane to get the desired product as light yellow oily compound (35.0 g, 81%). 1H NMR (400 MHz, chloroform -d) δ 7.46-7.44 (m, 2H), 7.39 (t, J = 7.6 Hz, 2H), 7.34-7.31 (m, 1H), 7.21 (t, J = 7.6 Hz), 6.86-6.80 (m, 3H), 5.07 (s, 2H), 2.64 (q, J - 7.6 Hz, 2H), 1.24 (t, J = 7.6 Hz, 3H).
[0231] Step D: Preparation of 4-(benzyloxy)-l -bromo-2 -ethylbenzene (3-6). To an ice cold stirred solution of 1 -(benzyloxy )-3 -ethylbenzene (35.0 g, 164 mmol) (3-5) in ACN (525 mL, 15 vol) was added N- bromosuccinimide (32.0 g 181 mmol) in portions over a period of 15 minutes. The resulting reaction mixture was stirred for 1 hour at room temperature. After completion of reaction (TLC monitoring), the resulting reaction mass was poured into ice cold water (1 .50 L) followed by the extraction of compound with EtOAc (2 x IL). The combined organics were washed with water and dried over sodium sulfate, filtered and evaporated under reduced pressure to obtain the crude product. n-Hexane (250 mL) was added to the crude material, resulting in a shiny, followed by filtration through a sintered funnel. Mother liquor was evaporated under reduced pressure to obtain the desired product as light yellow oily compound (42.0 g, 87%). 1H NMR (400 MHz, chloroform-d) δ 7.52-7.29 (m, 7H), 6.88 (s, TH), 6.68 (d, J = 6.0 Hz, 1H), 5.04 (s, 2H), 2.69 (q, J = 7.6 Hz, 2H), 1.2.0 (t, J = 7.5 Hz, 3H).
[0232] Step E: Preparation of 2-(4-(benzyloxy)-2-ethylphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (3- 7). A stirred solution of 4-(benzyioxy)-l-bromo-2-ethylbenzene (42.0 g, 144 mmol) (3-6), bis(pinacolato) diboron (44.0 g, 173 mmol), and potassium acetate (28 g, 288 mmol) in dioxane (440 mL) was degassed by purging N2 (g) for 15 min followed by addition of PdC12(dppf)-DCM complex (11.0 g, 15 mmol). The resulting reaction mixture was heated to 80 °C for 16h. After completion of the reaction (TLC monitoring), the reaction mass was filtered through celite bed and mother liquor was evaporated under reduced pressure to obtain the crude product. Crude residue was purified by column chromatography over silica gel (100-200M) by using eluents 1% EtOAc in hexane to get the desired product as light yellow oily compound (32.0 g, 66%).
H XMR (400 MHz, chloroform-ri) 5 7.74 (d, J = 8.4 Hz, 1H), 7.45-7.36 (m, 5H), 6.84-6.78 (m, 2H), 5.08 (s, 2.H), 2.91 (q, J = 7.6 Hz), 1.33 (s, 12H), 1.19 (t, J = 7.6 Hz, 3H).
[0233] Step F: Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-l-(oxan-2-yl)-1H-indazole (3-9). To a stirred solution of 6-bromo~l -(oxan-2-yl)-1H-indazole (20 g, 71.1 mmol) and 3-7 (26.5 g, 78.2 mmol) in dioxane (150 mL) and water (20 mL) w’as added sodium carbonate (15.1 g, 142 mmol). The reaction vessel was purged with argon for 20 minutes, then bis(diphenylphosphino)ferrocene paliadium(ll)dichloride dichloromethane adduct (5.81 g, 7.11 mmol) was added. The reaction mixture was stirred at 100 °C for 16 h, until judged complete by TLC and LCMS. The reaction mixture was then cooled to room temperature, filtered through a Celite pad, and the pad was further washed with ethyl acetate (2x200 mL). The combined filtrate was then washed with cold water (2x100 mL) and brine (100 mL), after which the organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was then purified by silica gel chromatography (12% EtOAc/Heptane) to afford the desired product as a pale yellow semi-solid (23 g, 78.4% yield). 1HNMR (400 MHz, DMSO-d6) 8.12 (s, 1H), 7.77 (d, J = 8.3 Hz, 1H), 7.58 (s, 1H), 7.48 (d, J = 7.2 Hz, 2H), 7.41 (t, .7 = 8.2 Hz, 2H), 7.35 (t, .7 = 7.2 Hz, 1H), 7.15 (d, 8.4 Hz, 1H), 7.08 (dd, J = 8.2, 1.2
Hz, 1H), 7.0 (d. J = 2.6 Hz, 1H), 6.92 (dd, J--- 8.4, 2.6 Hz, 1H), 5.87 (dd, J 10.1, 2.3 Hz, 1H), 5.16 (s, 2H), 3.86 (m, 1H), 3.72 (m, 1H), 2.53 (q, .7 = 7.6 Hz, 2H), 2.40 (m, 1H), 1.95 (m, 2H), 1.70 (m, 1 H), 1.55 (m, 2H), 1.04 (t, J = 7.5 Hz, 3H).
[0234] Step G: Preparation of 6-[4-(benzyloxy)-2-etbylphenylj-1H-indazole (3-10). To a stirred solution of 3-9 (43 g, 104 mmol) in methanol (215 mL) was added aqueous hydrochloric acid (6N, 60 mL) at 0 °C dropwise. The resulting solution was first warmed to room temperature, then heated at 90 °C for 16 h until judged complete by TLC. The reaction mixture was partially concentrated under reduced pressure, then neutralized by slow addition of saturated aqueous sodium bicarbonate solution (200 mL) at 0 °C. This solution was extracted with ethyl acetate (2x300 mL). ’The combined organic layers were washed with brine (100 mL), dried over sodium sulfate, and concentrated under reduced pressure to afford the desired product as an off-white solid that was used without further purification (36 g, 75.7% yield). 1H NMR (400 MHz, DMSO-d6) 13.0 (s, 1H), 8.09 (s, 1H), 7.76 (d, .7 = 8.3 Hz, 1H), 7.48 (d, J = 7.2 Hz, 2H), 7.41 (t, J = 8.2 Hz, 2H), 7.35 (m, 2.H), 7.15 (d, J = 8.4 Hz, 1H), 7.00 (m, 2H), 6.91 (dd,J=8.4, 2.6 Hz, 1H), 5.15 (s, 2H), 2.53 (q, J = 7.6 Hz, 2H), 1.02 (t, J = 7.5 Hz, 3H).
[0235] Step H: Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-3-iodo-1H-indazole (3-11). To a stirred solution of 3-10 (36 g, 110 mmol) in DMF (120 mL) w'as added N-iodosuccinimide (49.3 g, 219 mmol) portionwise over 15 minutes at 0 °C, then the reaction mixture w’as stirred at room temperature for 2 h until judged complete by TLC. The reaction mixture was then diluted with water (200 mL) and extracted with ethyl acetate (3x100 mL). The combined organic layers were washed with sodium thiosulfate solution (200 mL), brine (200 mL), then dried over sodium sulfate and concentrated under reduced pressure to afford the desired product as a light-brown semi-solid that was used without further purification (46 g, 46% purity, 42.5% yield). bH NMR (400 MHz, DMSO-ty.) 13.5 (s, 1H), 7.50-7.33 (m, 7H), 7.12 (m, 2H), 7.00 (s, 1H), 6.91 (dd, J = 8.0, 2.0 Hz, 1H), 5.15 (s, 2H), 2.53 (q, J = 7,6 Hz, 2H), 1.02 (t, .7 = 7.5 Hz, 3H).
[0236] Step 1: Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-3-iodo-l-(oxan-2-yl)-1H-indazole (3-12). To a stirred solution of 3-11 (46 g, 101 mmol) in DCM (250 mL) was added p-toluenesulfonic acid (1.74 g, 10.1 mmol) and 3,4-dihydro-2H-pyran (13.9 mL, 152 mmol) dropwise at 0 °C, then the reaction mixture was stirred at room temperature for 16 h until judged complete by TLC. The reaction mixture was then diluted with DCM (400 mL), extracted with saturated aqueous sodium bicarbonate solution (100 mL), washed with brine (100 mL), then the organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (15% EtOAc/heptane) to afford the desired product as an off-white solid (40 g, 68.2% yield), (m/z): [M-rH]" calcd for C27H27IN2O2 539.11, found 539.20.
[0237] Step J: Preparation of 6-[4-(benzyloxy)-2-ethylphenyl]-l-(oxan-2-yl)-3-(trimethylstannyl)-1H- indazole (3-13). A solution of 3-12 (20 g, 37. 1 mmol) in toluene (200 mL) was purged with argon for 20 minutes, then 1,1,1 ,2,2,2-hexa-methyldistannane (9.31 mL, 44.6 mmol) was added, followed by paliadium(0)tetrakis(triphenylphosphine) and the reaction mixture was stirred at 110 °C for 2 h, until the reaction was judged complete by TLC, The reaction mixture was filtered through a pad of Celite, which was then washed with ethyl acetate (2x200 mL). The combined filtrate w’as concentrated under reduced pressure, then the crude product was purified by column chromatography with neutral alumina gel (5% EtOAc/heptane) to afford the desired product as an off-white solid (15.5 g, 61.7% yield), (m/z): fM+HJ* calcd for C 30 H36N2O2Sn 577.18, found 577.18.
[0238] Step K: Preparation of ethyl 2-{6-[4-(benzyloxy)-2-ethylphenyl]-1-(oxan-2-yT)-1H-indazol-3-yl}-1 - (dimethylsulfamoyl)-1H-imidazole-4-carboxylate (3-14). To a stirred solution of 3-3 (3.0 g, 8.04 mmol) in toluene (40 ml,) was added 3-13 (4.63 g, 80.4 mmol) and the mixture was degassed with argon for 15 minutes. To this mixture copper (I) iodide (306 mg, 1.61 mmol) and pailadium(0)tetrakis(triphenylphosphine) (929 mg, 0.80 mmol) were added and the solution -was again degassed -with argon for 5 minutes. The reaction mixture was then heated at 120 °C for 2 h. After the reaction was judged complete by TLC and LCMS, the reaction mixture was filtered through Celite and the filtrate was concentrated under vacuum . The crude product was then purified by silica gel chromatography (30% EtOAc/heptane) to afford the desired product as a light yellow solid (2.4 g, 45.4% yield), (m/z): [M+H]+ calcd for C 35 H39N 5O6S 658.26, found 658.37.
[0239] Step L: Preparation of ethyl 1-(dimethylsulfamoyl)-2-[6~(2-ethyl-4-hydroxyphenyl)-l-(oxan-2-yl)- 1H-indazol-3-yl]-1H -imidazole-4-carboxylate (3-15). To a stirred solution of 3-14 (5.50 g, 8.36 mmol) in THF (30 mL) and isopropyl alcohol (30 mL) was added 10% palladium on carbon (10.0 g, 94 mmol). The reaction mixture was then subjected to hydrogenation using a hydrogen balloon and allowed to stir at room temperature for 2 days. After the reaction was judged complete by TLC and LCMS the reaction mixture was filtered through Celite and washed with ethyl acetate. The filtrate was concentrated to afford the desired product as an off-white solid, which was used without further purification (4.52 g, 95.2% yield), (m/z): [M+H]+ calcd for C 28 H33N 5O6S 568.22, found 568.32.
[0240] Step M: Preparation of ethyl l-(dimethylsulfamoyl)-2-[6-(2-ethyl-4-{[2-
(trimethylsily l)ethoxy]methoxy }phenyl)- 1 -(oxan-2-yl)- 1 H-indazol -3 -y 1] - 1 H-im idazole-4-carboxylate (3-16). To a stirred solution of 3-15 (4.5 g, 7.93 mmol) in dimethylacetamide (40 mL) was added sodium hydride (60% in mineral oil, 476 mg, 11.9 mmol) at 0 °C, and the reaction mixture was stirred for 10 minutes. SEM- C1 (1.54 mL, 8.72 mmol) was then added and the reaction mixture was stirred at room temperature for 1 h. After the reaction was judged complete by TLC and LCMS the reaction mixture -was quenched by addition of ice cold water (500 mL) and extracted with ethyl acetate (600 mL), The organic layer was then dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (40% EtOAc/Heptane) to afford the desired product as an off-white solid (4.05 g, 73.2% yield), (m/z): [M+H]+ calcd for C 34 H47N 5O7SSi698.30, found 698.41.
[0241] Step N: Preparation of 2-[6-(2-ethyl-4-{ [2-(trimethylsityl)ethoxy]methoxy}phenyI)-l -(oxan-2-yl)- l H-indazoi-3-yl]-4-(hydroxymethyi)-N,N-dimethyl-1H-imidazole-l-sulfonamide (3-17). To a stirred solution of 3-16 (4.0 g, 5.73 mmol) in THF (15 mL) was added lithium borohydride (2M in THF, 14.3 mL, 28.7 mmol) at 0 °C, after which the reaction mixture was stirred at room temperature for 16 h. After consumption of starting material was observed by TLC, the reaction mixture was quenched by addition of methanol (30 mL), and the solution was heated at 60 CC for 2 h. The reaction mixture was then diluted with water (300 mL) and extracted with ethyl acetate (2x300 mL). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (5% MeOH/DCM) to afford the desired product as an off-white semi-solid (3.35 g, 89.1% yield), (m/z): [M+H]+ calcd for Cs^sNjOsSSi 656.29, found 656.58.
[0242] Step 0: Preparation of 4-(chloromethyl)-2-(6-(2-etbyl-4-((2.-(triniethylsilyl)ethoxy)metboxy)pbenyl)- l-(tetrahydro-2fir-pj<ran-2-yl)-1H -indazol-3-yl)-N,N-dimethyl-1H- imidazole-l -sulfonamide (3-18). To a 100 ml flask was added 3-17 (1.6 g, 2.44 mmol) in DCM (20 mL). To this, DIPEA (1 .92 mL, 10.98 mmol) was added and the reaction flask was cooled to 0 °C. Next, methanesulfonyl chloride (0.49 mL, 6.34 mmol) was added. The reaction mixture was stirred on ice for 20 minutes and then warmed to room temperature and stirred for 18 h. The reaction mixture was then diluted with water (30 mL) and extracted with DCM (2x20 mL). The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (10-35% EtOAc/hexanes gradient) to afford the desired product as a light yellow solid (1.1 1 g, 67.4% yield), (m/z): [M+H] 1 calcd for C 32 H44C1N 5O5SSi 6i7i4.25, found 673.95.
[0243] Step P: Preparation of diethyl ((l-(N,N-dimethylsulfamoyl)-2-(6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy iphenyl)- 1 -(tetrahydro-2H -pyran~2-yl)- l/J-indazol-3 -yl)- 1H-imidazol-4- yl)metbyl)phosphonate (3-19). To a vial of 3-18 (1.108 g, 1 .64 mmol) was added neat triethyl phosphite (1 mL, 5.83 mmol). The reaction mixture was heated to 110 °C for 16 b. Following this, the reaction mixture was cooled to room temperature, concentrated under reduced pressure, and purified by silica gel chromatography (30-100% EtOAc/hexanes gradient) to afford the desired product (1.03 g, 81% yield), (m/z): [M+H]+ calcd for C 36 H54N 5O8SSi776.32, found 775.85.
[0244] Step Q: Preparation of tert-butyl (3S)-3-((E)-2-(l -(N,N-dimethylsulfamoyl)-2-(6-(2-ethyl-4-((2- (trimethylsily l)ethoxy)methoxy )phenyl)- 1 -(tetrahydro-2H-pyran-2-yl)- 1H-indazol-3 -yl)- 1H-imidazol-4- yl)vinyl)morpholme-4-carboxylate (3-20). A solution of 3-19 (150 mg. 0.193 mmol) in THF (2.5 mL) was cooled to 0 °C, then 15-crown-5 (127.5 mg, 0.58 mmol) was added, followed by sodium hydride (60% dispersion in mineral oil, 11.6 mg, 0.29 mmol). The reaction mixture was warmed to room temperature and stirred for 30 minutes. The reaction mixture was then cooled to 0 °C and a solution of (R)-N-boc-3- morpholinecarbaldehyde (41.5 mg, 0.193 mmol) in THF (2 mL) was added slowly. The reaction mixture was gradually allowed to warm to room temperature and stirred for 4 h. The reaction m ixture was then quenched with methanol (0.5 mL) and water (0.5 mL). The volatiles were removed under reduced pressure and the crude product was purified by silica gel chromatography (5-70% EtOAc/bexanes gradient) to afford the desired product as a yellow oil (37 mg, 22.9% yield), (m/z): [M+H] + calcd for C 42 H60N 6O8SSi837.40, found
837.30.
[0245] Step R: Preparation of tert-butyl (3S)-3-((E)-2-(2-(6-(2-ethyl-4-hydroxyphenyl)-l-(tetrahydro-2H- pyran-2-yl)-1H-indazol-3-yl)-1H-imidazol-4-yl)vinyi)morpholine-4-carboxylate (3-21). To a solution of 3-20 (55 mg, 0.066 mmol) in THF (3 mL), methanol (1 mL), and water (1 mL) was added trifluoro ace tic acid (0.5 mL). The reaction mixture was heated at 45 °C for 4 h, until complete removal of both SEM and sulfonyl urea protecting groups w*as observed. The reaction mixture was then cooled to room temperature, diluted with 1 :1 ACN/water (10 mL) and the solution was frozen and lyophilized. The dried residue was dissolved in THF (2 mL). A solution of sodium borohydride (2.0 mg) in 1 : 1 THF/ethanol (2 mL) was slowly added, and the reaction mixture was stirred for 30 minutes. The volatiles were removed under reduced pressure and the crude product was used directly in the next step without further purification, (m/z): [M+H] * calcd for C 34 H41N5O5
600.31 , found 600.25.
[0246] Step S: Preparation of (AE)-3-ethyl-4-(3-(4-(2-(morphohn-3-yl)vinyl)- 1H-imidazoi-2-yl)-1H- indazol- 6-yl)phenol (6). To a solution of 3-21 (39 mg, 0.065) in DCM (2 mL) was added trifluoroacetic acid (2 mL). The reaction mixture was heated to 45 °C and stirred for 3 h. The reaction mixture was then concentrated under reduced pressure, and the residue was purified by preparative HPLC chromatography (5-40% ACN/water gradient with 0.05% trifluoroacetic acid) to afford the TEA. salt of the title compound as a white solid (2 mg, 5.9% yield), (m/z): [M+H]+ calcd for C24H25N5O2416.20, found 416.10.
[0247] Example 4: Synthesis of (S,E)-3-ethyl-4-(7-fluoro-3-(4-(2-(l-methylpyrrolidin-2-yl)vinyl)-1H- imidazol-2-yl)-1H-indazol-6-yl)phenol (40).
Figure imgf000104_0001
Figure imgf000105_0001
[0248] Step A: Preparation of tert-butyl (S,E )-2-(2-(4,4.5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)vinyl)pyrrolidine-l -carboxylate (4-2). To an oven dried round bottom flask cooled under nitrogen was added 2,2,6,6-tetramethylpiperidine (508 μL, 3.01 mmol) in THF (3 mL) and the solution was cooled to -78 °C. n-Butyllithium (2.5 M in hexanes, 1.205 mL, 3.01 mmol) was added dropwise over 10 minutes, and the reaction mixture was stirred at -78 °C for 30 minutes. A solution of bis[(pinacolato)boryl]methane (670 mg, 2.51 mmol) in THF (3 mL) was then added dropwise. After stirring for 30 minutes, a solution of boc-L- prolinal (500 mg, 2.51 mmol) was added dropwise. The reaction mixture was gradually wanned to room temperature and stirred for 20 h. The reaction mixture was then cooled to 0 °C and a saturated aqueous solution of ammonium chloride (5 mL) was added dropwise, then the solution was stirred at 0 °C for 1 h. The crude reaction mixture was adsorbed directly onto celite and purified by silica gel chromatography (0-5% EtOAc/hexanes gradient) to afford the desired product as a yellow oil (659 mg, 81% yield), (m/z): [M+Na]+ calcd for C17H30BNO4Na 346.22, found 346.20.
[0249] Step B: Preparation of tert-butyl (2S)-2-((E)-2-(2-(6-(2-ethyl-4-hydroxyphenyl)-7-fluoro-1- (tetrahydro-2H -pyran-2-yl)-1H -indazol-3-yl)-1H -imidazol-4-yl)vinyl)pyrrolidine-l-carboxylate (4-3). To a vial was added 2-10 (100 mg, 0.206 mmol) and 4-2 (100 mg, 0.309 mmol) in dioxane (3 mL), followed by a solution of sodium carbonate (65 mg, 0.618 mmol) in water (1 mL). The reaction mixture was sparged with nitrogen for 10 minutes. XPhos Pd G4 (9 mg, 10.3 pmol) was added and the reaction mixture was sparged again with nitrogen for an additional 10 minutes. The reaction mixture was heated to 110 °C and stirred for 2 h. The reaction mixture was cooled to room temperature and the volatiles were removed under reduced pressure. The residue was dissolved in EtOAc (10 mL) and extracted with a solution of saturated aqueous sodium bicarbonate (20 mL). The aqueous solution was then extracted with EtOAc (3x10 mL), and the combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was used directly in the following reaction without further purification, (m/z): [M+H]+ calcd for C34H40FN5O4602.31 , found 602.45.
[0250] Step C: Preparation of (S,E )-3-ethyl-4-(7-fluoro-3-(4-(2-(pyrrolidin-2-yl)vinyl)-1H -imidazol-2-yl)- 1H -indazol-6-yl)phenol (4-4). To a solution of 4-3 (124 mg, 0.206 mmol) in DCM (2 mL) was added trifluoroacetic acid (2 mL). The reaction mixture was stirred at 50 °C for 1 h, after which the reaction mixture was concentrated under reduced pressure. The crude product was purified by preparative HPLC chromatography to afford the TFA salt of the title compound as a white solid, (m/z): [M+H]+ calcd for C24H24FN5O 418.20, found 418.25.
[0251] Step D: Preparation of (S,E)-3-ethyL4-(7-fluoro-3-(4-(2-(l-methylpyrrolidin-2-yl)vinyl )-1H - imidazol-2-yl)-1H -indazol-6-yl)phenol (40). To a solution of 4-4 (10 mg, 0.024 mmol) in methanol (2 mL) was added formaldehyde (37 wt% in water, 100 μL, 0.077 mmol), followed by sodium cyanoborohydride (7.5 mg, 0.120 mmol). The reaction mixture was stirred for 90 minutes at room temperature, after which ethylenediamine (30 μL) and sodium borohydride (20 mg) were added. The reaction mixture was stirred for an additional 10 minutes, then concentrated under reduced pressure. The crude product was purified by preparative HPLC chromatography to afford the TFA salt of the title compound as a white solid (2.7 mg, 20.8% yield), (m/z): [M+H]+ calcd for C25H26FN5O 432.21, found 432.25.
[0252] Example 5: Synthesis of 3-ethyl-4-(7-fluoro-3-(4-(piperidin-4-ylidenemethyl)-lH-imidazol-2-yl)- lH-indazol-6-yl)phenol (47).
Figure imgf000106_0001
[0253] Step A: Preparation of tert-butyl 4-((4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)methylene)piperidine-1 -carboxylate (5-2). To an oven dried round bottom flask cooled under nitrogen was added 2,2,6,6-tetramethylpiperidine (508 μL, 3.01 mmol) in THF (3 mL) and the solution was cooled to -78 °C. n-Butyllithium (2.5 M in hexanes, 1.205 mL, 3.01 mmol) was added dropwise over 10 minutes, and the reaction mixture was stirred at -78 °C for 30 minutes. A solution of bis[(pinacolato)boryl]methane (670 mg, 2.51 mmol) in THF (3 mL) was added dropwise. After stirring for 30 minutes, a solution of 1-boc-4- piperidone (500 mg, 2.51 mmol) was added dropwise. The reaction mixture was allowed to slowly warm to room temperature and stirred for 24 h. The reaction mixture was then cooled to 0 °C and a saturated aqueous solution of ammonium chloride (5 mL) was added dropwise, then the solution was stirred at 0 °C for 1 h. The crude reaction mixture was adsorbed directly onto celite and purified by silica gel chromatography (0-5% EtOAc/hexanes gradient) to afford the desired product as a white solid (354 mg, 43.6% yield), (m/z): [M+H- Boc]+ calcd for C12H22BNO2224.17, found 224.10.
[0254] Step B: Preparation of tert-butyl 4-((2-(6-(2-ethyl-4-hydroxy phenyl)- 7-fluoro-1 -(tetrahydro -2H - pyran-2-yl)-1H -indazol-3-yl)-1H -imidazol-4-yl)methylene)piperidine-l -carboxylate (5-3). To a vial was added 2-10 (230 mg, 0.474 mmol) and 5-2 (153 mg, 0.474 mmol) in dioxane (3 mL), followed by a solution of sodium carbonate (151 mg, 1.422 mmol) in water (1 mL). The reaction mixture was then sparged with nitrogen for 10 minutes. XPhos Pd G4 (20.4 mg, 0.024 mmol) was added and the reaction mixture was sparged again with nitrogen for 10 minutes. The reaction mixture was heated to 1 10 °C and stirred for 2 h. The reaction mixture was cooled to room temperature, then concentrated under reduced pressure. The residue was dissolved in ethyl acetate (10 mL) and extracted with a solution of a saturated aqueous sodium bicarbonate (20 mL). The aqueous layer was extracted with ethyl acetate (3x10 mL), and the combined organic layers were washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The crude product was used directly in the following reaction without further purification, (m/z): [M+H]+ calcd for C34H40FN5O4602.31 , found 602.45.
[0255] Step C: Preparation of 3-ethyl-4-(741uoro-3-(4-(piperidin-4-ylidenemethyl)4#-imidazol-2-yi)-1H - indazol-6-yl)phenol (47). To a solution of 5-3 (285 mg, 0.474 mmol) in DCM (2 mL) was added trifluoroacetic acid (2 mL). The reaction mixture was stirred at 50 °C for 1 h, then the reaction mixture was concentrated under reduced pressure. The crude product was purified by preparative HPLC chromatography to afford a TFA salt of the title compound as a white solid (6.2 mg, 2.5% yield), (m/z): [M+H]+ calcd for C24H24FN5O 418.20, found 418.25.
[0256] Example 6: Synthesis of 3-ethyl-4-(3-(4-(3-(4-methylpiperazin-l-yl)prop-l -yn-l-yl)-1H -imidazol-2- y1)-1H-indazol-6-yl)phenol (66).
Figure imgf000107_0001
[0257] Step A : Preparation of 3-(4,5-diiodo-1H -imidazol-2-yl)-6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy)phenyr)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazole (6-1). A solution of 11-1 (1.0 g, 1.93 mmol) was dissolved in DCM (30 mL) and the solution was cooled to 0 °C, N-iodosuccinamide (0.95 g, 4.24 mmol) in DCM (6 mL) and THF (4.5 mL) was then added dropwise, and the reaction mixture was stirred at 0 °C for 3 h. Once the reaction was judged complete by LCMS, the reaction was quenched by addition of saturated aqueous sodium sulfite (80 mL) and the mixture was stirred at 0 °C for 10 minutes. The mixture was then extracted with DCM (80 mL), and the organic layer was washed with brine (60 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was used without further purification.
[0258] Step B: Preparation of 4-(3-(4,5-diiodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H-indazol- 6-yl)-3 -ethylphenol (6-2). Compound 6-1 (1.95 g, 2.53 mmol) was dissolved in a mixture of THF (30 mL), methanol (10 mL), and water (10 mL), then trifluoro acetic acid (3.90 mL, 50,6 mmol) was added and the reaction mixture was stirred at 50 °C for 6 h. The reaction mixture was then diluted with ethyl acetate (60 mL) and a solution of sodium bicarbonate (10.6 g, 127 mmol) in water (50 mL) was added slowly. The organic layer was then separated, washed with brine (50 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was then purified by silica gel chromatography (10-30% EtOAc/hexanes gradient) to afford the desired product as a white solid (1.23 g, 76% yield), (m/z): [M+H]+ calcd for C23H22I2N4O2 640.98, found 640.7.
[0259] Step C: Preparation of 3-ethyl-4-(3-(4-iodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran~2-yl)-1H- indazol-6-yl)phenol (6-3). Sodium sulfite (3.63 g, 28.8 mmol) was added to a solution of 6-2 (1 .23 g, 1.92 mmol) in ethanol (26 mL) and water (13 mL), then the reaction vessel was sealed and heated with stirring at 110 °C for 2 days. The reaction was then cooled, water (20 mL) was added, and the mixture was extracted with ethyl acetate (2x60 mL). The combined organic layers were washed with brine (60 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (10-40% EtOAc/hexanes gradient) to afford the desired product as a white solid (0.62 g, 62.3% yield), (m/z): [M+H]+ calcd for C23H231N4O2 515,09, found 515.0.
[0260] Step D: Preparation of 3-ethyl-4-(3 -(4-(3 -(4-methylpiperazin- 1 -y l)prop-1 -yn- 1 -y 1)- 1H-imidazol-2- yl)-1H-indazol~6-yl)phenol (66). 1-methyl-4~(prop-2-yn~l-yl)piperazine (18.8 mg, 0.136 mmol), 6-3 (35 mg, 0.068 mmol), and triethylamine (28 uL, 0.204 mmol) were dissolved in DMF (1.2 mL), then the reaction was degassed with nitrogen for 10 minutes. Cuprous iodide (2.6 mg, 0,014 mmol) and bis(triphenylphosphine)- palladium(II)chloride (4.8 mg, 0.0068 mmol) were added, the reaction was again degassed with nitrogen for 5 minutes, then the reaction was sealed and stirred at 80 °C for 1 h. The reaction mixture was concentrated and the residue was dissolved in DCM (1 mL), then trifluoroacetic acid (0.5 mL) was added and the reaction mixture was stirred at 45 °C for 40 minutes. The reaction mixture was concentrated and the crude product w’as purified by preparative HPLC (5-45% ACN/water gradient with 0.05% TFA) to afford the TEA salt of the title compound (14.6 mg, 38.7% yield), (m/z): [M+H]+ calcd for
Figure imgf000108_0001
441.23, found 441.2.
[0261] Example 7: Synthesis of 4-(3-(4-(azetidm-3-ylethynyl)-1H-imidazol-2-yl)-1H-indazol-6-yl)-3- ethylphenol (120).
Figure imgf000109_0002
[0262] Tert-butyl 3 -ethynylazetidrne-1 -carboxylate (21.1 mg, 0.117 mmol), 6-3 (30 mg, 0.058 mmol), and triethylamine (24 uL, 0.175 mmol) were dissolved in DMF (1.2 mL), then the reaction was degassed with nitrogen for 10 minutes. Cuprous iodide (2.2 mg, 0.012 mmol) and bis(triphenylphosphine)- palladium(II)chloride (4.1 mg, 0.0058 mmol) were then added, the reaction was again degassed with nitrogen for 5 minutes, then the reaction was sealed and stirred at 50 °C for 2 h. The reaction mixture was concentrated, the residue was dissolved in DCM (1 mL), then trifluoroacetic acid (0.5 mL) was added and the reaction mixture was stirred at 45 °C for 1 h. The reaction mixture was concentrated and the crude product was purified by preparative HPLC to afford the TFA salt of the title compound (3.1 mg, 10.3% yield), (m/z): [M+H]+ calcd for Cr,H2iN5O 384.17, found 384,2,
[0263] Example 8: Synthesis of (R)-3-ethyl-4-(7-fluoro-3-(4-(morpholin-3-ylethynyl)-1H-imidazol-2-yl)- 1H-indazol-6-yl)phenol (22).
Figure imgf000109_0001
[0264] Step A : Preparation of tert-butyl (R)-3-ethynylmorpholine-4-carboxylate (8-2). (S)-N-Boc-3- morpholmecarbaldehyde (30 mg, 0.139 mmol) was dissolved in methanol (2.0 mL), then potassium carbonate (38,5 mg, 0.279 mmol) was added, followed by dimethyl (2-oxopropanimidoy1)phosphonate (32.3 mg, 0,167 mmol), and the reaction mixture was stirred at room temperature. After judged complete by TLC (24 h), the reaction mixture was concentrated, then the residue was partitioned between ethyl acetate (3 mL) and water (3 mL). The organic layer was then dried over sodium sulfate and concentrated. The crude product was used without further purification.
Figure imgf000110_0001
NMR (600 MHz, Chloroform-<7) 84.76 (s, 1H), 3.94 (m, 2H), 3.73 (d, J = 13.1 Hz, 1H), 3.62 (d, 8.1 Hz, 1H), 3.49 (t, .7 = 11.8 Hz, 1 H), 3.34 (in, 1H), 2.33 (s, 1H), 1.50 (s, 9H).
[0265] Step B: Preparation of 3-(4,5-diiodo-1H-imidazol-2-yl)-6-(2-ethyl-4-((2- (trimethylsilyl)ethoxy)methoxy)phenyl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yI)-1H-indazole (8-3). A solution of 2-7 (1 .0 g, 1.86 mmol) was dissolved in DCM (23 ml.) and the solution was cooled to 0 °C. N- iodosuccinamide (0.92 g, 4.10 mmol) in DCM (4 mL) and THF (3 mL) was then added dropwise, and the reaction mixture was stirred at 0 °C for 3 h, then at ambient temperature for 16 h. The reaction was quenched by addition of saturated aqueous sodium sulfite (40 mL). The mixture was then extracted with DCM (40 mL), and the organic layer was washed with brine (40 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was used without further purification.
[0266] Step C: Preparation of 4-(3-(4,5-diiodo-1H-imidazol-2-yl)-7-fluoro-l-(tetrahydro-2H-pyran-2-yl)-1H- indazol-6-yl)-3 -ethylphenol (8-4). Compound 8-3 (1.56 g, 1.98 mmol) was dissolved in a mixture of THF (22.5 mL), methanol (7.5 mL), and water (7.5 mL), then trifluoroacetic acid (3.05 mL, 39.6 mmol) was added and the reaction mixture was stirred at 50 °C for 6 h. The reaction mixture was then diluted with ethyl acetate (40 mL) and a solution of sodium bicarbonate (10.48 g, 99 mmol) in water (40 mL) was added slowly. The organic layer was then separated, washed with brine (40 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was then purified by silica gel chromatography (10-35% EtOAc/hexanes gradient) to afford the desired product as a white solid (0.93 g, 72% yield), (m/z): [M+H] + calcd for C23H21FT2N4O2 658.97, found 659.1.
[0267] Step D: Preparation of 3-ethyl-4-(7-fluoro-3-(4-iodo-1H-imidazol-2-yl)-l-(tetrahydro-2H-pyran-2- yl)-1H-indazol-6-yl)phenol (8-5). Sodium sulfite (3.58 g, 28.4 mmol) was added to a solution of 8-4 (0.93 g, 1.42 mmol) in ethanol (19 mL) and water (9.5 mL), then the reaction vessel was sealed and heated with stirring at 110 °C for 2 days. The reaction was then cooled, water (20 mL) was added, and the mixture was extracted with ethyl acetate (2x60 mL). The combined organic layers were washed with brine (60 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (10-45% EtOAc/hexanes gradient) to afford the desired product as a white solid (0.56 g, 74.4% yield), (m/z): [M+H] + calcd for C23H22FIN4O2 533.08, found 533.2.
[0268] Step E: (R)-3-ethyl-4-(7-fluoro-3-(4-(morpholin-3-ylethynyl)-1H -imidazol-2-yl)-1H-indazol-6- yl)phenol (22). Compound 8-5 (25 mg, 0.047 mmol), tert-butyl (R)-3-ethynylmorpholine-4-carboxyiate (8-2) (24,80 mg, 0.117 mmol), and triethylamine (0.020 ml, 0. 141 mmol) were dissolved in DMF (1.0 ml) and the solution was sparged with nitrogen for 10 minutes. Bis(triphenylphosphine)palladium(II)chloride (3.30 mg, 4.70 pmol) and copper (I) iodide (1.789 mg, 9.39 pmol) were then added and the reaction mixture was stirred at 60 °C for 4 h. The reaction mixture was concentrated, then 1 mL of trifluoroacetic acid was added and the reaction mixture was stirred at 40 °C for 30 minutes, after which LCMS showed the reaction was complete.
The reaction mixture was concentrated and the crude product -was purified by preparative HPLC (5-70% ACN/water) to afford the TFA salt of the title compound (7.3 mg, 28.5% yield), (m/z): [M+H] calcd for
C24H22FN5O2 432.18, found 432.2.
[0269] Example 9: Synthesis of 3-etliyl-4-(7-fluoro-3-(4-(((2S,4S)-4-methoxypyiToIidin-2-yl)ethynyI)-1H- imidazol-2-; rl)-1H-indazoi-6-yl)phenol (49).
Figure imgf000111_0001
[0270] Step A: Preparation of tert-butyl (2S,4S)-2-ethynyl-4-hydroxypyrrolidine-l -carboxylate (9-2). To a stirred solution of 9-1 (2 g, 8.15 mmol) in DCM (25 mL) at -78 °C was added DIBAL-H (IM in toluene, 20.4 mL, 20.4 mmol) dropwise with stirring. The resulting solution was stirred at -78 CC for 2.5 h until disappearance of starting material was observed by TLC. The reaction was then quenched by the slow addition of methanol (20 mL), then the solution was wanned to 0 °C. Potassium carbonate (2.25 g, 16.3 mmol) was added, followed by the addition of dimethyl (l-diazo-2-oxopropyl)phosphonate (1.88 g, 9,78 mmol), and the resulting mixture w'as allowed to warm to room temperature and stir for 16 h. An aqueous solution of 1 M sodium potassium tartrate (20 mL) and diethyl ether (20 mL) were then added and the solution was stirred for 1 h. The organic layer was separated, washed with brine (30 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (15% EtOAc/heptane) to afford the desired product as an off-white solid (0.7 g, 36.6% yield). 1H NMR (400 MHz, DMSCM.) 5 4.97 (s. 1H), 4.36 (m, 1H), 4.20 (m, 1H), 3.43 (m, 1H), 3.09 (m, 1H), 2.28 (m, 1H), 1.83 (m, 1H), 1.40 (s, 9H).
[0271] Step B: Preparation of tert-butvl (2S,4S)-2-ethynyl-4-methoxypyrrolidine-l -carboxylate (9-3). To a stirred solution of 9-2 (0.75 g, 3.55 mmol) in DMF (5 mL) was added sodium hydride (60% dispersion in mineral oil, 156 mg, 3.91 mmol) at -20 °C and the resulting solution was stirred for 30 minutes. Methyl iodide (265 μL, 4.26 mmol) was then added and the reaction mixture wrns allowed to warm to room temperature and stir for 5 h. After the reaction was judged complete by TLC, the reaction mixture was quenched by addition of ice-cold water (10 mL), then was extracted with ethyl acetate (3x10 mL). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (12% EtOAc/heptane) to afford the desired product as a viscous oil (405 mg, 50.1% yield), (m/z): [M+H]+ calcd for CuH^NOs 226.14, found 226.32.
[0272] Step C: Preparation of 3-ethyl-4-(7-fluoro-3-(4-(((2S,4S)-4-methoxypyrrolidin-2-yl)ethynyl)-1H- itnidazol-2-yl)-TH-indazol-6-yl)phenol (49). Compound 8-5 (50 mg, 0.094 mmol), 9-3 (52.9 mg, 0.235 mmol), and triethylamine (0.039 ml, 0.282 mmol) were dissolved in DMF (1.0 mL) and the solution was sparged with nitrogen for 10 minutes. Bis(triphenylphosphine)palladium(ll)chloride (6.59 mg, 9.39 pmol) and copper (I) iodide (3.58 mg, 0.019 mmol) were then added and the reaction mixture was stirred at 60 °C for 16 h. The reaction mixture was concentrated, then 1 mL of trifluoroacetic acid was added and the reaction mixture was stirred at 40 °C for 30 minutes, after which LCMS showed the reaction was complete. The reaction mixture was concentrated and the crude product was purified by preparative HPLC (5-70% ACN, 'water) to afford a TFA salt of the title compound, (m/z): [M+H]+ calcd for C25H24FN5O2 446.19, found 446.3.
[0273] Example 10: Biochemical JAK Kinase Assays to Measure pKi.
[0274] A panel of four LanthaScreen JAK biochemical assays (JAK1, 2, 3 and Tyk2) were carried in a common kinase reaction buffer (50 mM HEPES, pH 7.5, 0.01% Brij-35, 10 mM MgCL, and 1 mM EGTA). Recombinant GST-tagged JAK enzymes and a GFP-tagged ST ATI peptide substrate were obtained from Life Technologies.
[0275] Serially diluted compounds were pre-incubated with each of the four JAK enzymes and the substrate in white 384-well microplates (Coming) at ambient temperature for Ih. ATP was subsequently added to initiate the kinase reactions in 10 μL total volume, with 1% DMSO. The final enzyme concentrations for JAK1, 2, 3 and Tyk2 are 4.2 nM, 0.1 nM, 1 nM, and 0.25 nM respectively; the corresponding Km ATP concentrations used are 25 uM, 3 uM, 1.6 μM , and 10 pM; while the substrate concentration is 200 nM for all four assays. Kinase reactions were allowed to proceed for 1 hour at ambient temperature before a 10 μL preparation of EDTA (lOmM final concentration) and Tb-anti-pSTATl (pTyr701) antibody (Life Technologies, 2 nM final concentration) in TR-FRET dilution buffer (Life Technologies) was added. The plates were allowed to incubate at ambient temperature for Ih before being read on the EnVision reader (Perkin Elmer). Emission ratio signals (520nm/495nm) were recorded and utilized to calculate the percent inhibition values based on DMSO and background controls.
[0276] For dose-response analyses, percent inhibition data were plotted vs. compound concentrations, and IC50 values were determined from a 4-parameter robust fit model with the Prism software (GraphPad Software). Results were expressed as pICso (negative logarithm of IC50) and subsequently converted to pKi (negative logarithm of dissociation constant. Kt) using the Cheng-Prusoff equation. The higher the value of pKi (lower value of Ki), the greater the inhibition of JAK activity. Certain compounds disclosed herein exhibited pKi values of greater than 8 or greater than 9 when tested in the biochemical JAK assay.
[0277] Table 2 shows biological activities of selected compounds in a biochemical JAK assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
Table 2
Figure imgf000113_0001
[0278] Example 11: Cellular JAKI Potency Assay to Measure pICso-
[0279] The JAKI cellular potency assay was carried out by measuring inhibition of interleukin- 13 (IL-13, R&D Systems) induced STAT6 phosphorylation in BEAS-2B human lung epithelial ceils (ATCC), BEAS-2B cells were grown at 37°C in a 5% CO2 humidified incubator in 50% DMEM/50% F-12 medium (Life Technologies) supplemented with 10% FBS (Hyclone), 100 U/mL penicillin, 100 μg/mL streptomycin (Life Technologies), and 2 mM GlutaMAX (Life Technologies). On day 1 of the assay, ceils were seeded at a 7,500 cells/well density in white poly-D-lysine-coated 384-weil plates (Coming) with 25μL medium and were allowed to adhere overnight in the incubator. On day 2 of the assay, the medium was removed and replaced with 12 μL of assay buffer (Hank's Balanced Salt Solution/HBSS, 25mM HEPES, and 1 mg/ml bovine serum album in/BSA) containing dose-responses of test compounds. Compounds were serially diluted in DMSO and then diluted another 1000-fold in media to bring the final DMSO concentration to 0.1 %. Cells were incubated with test compounds at 37 °C for 1 h and followed by the addition of 12 μl of pre-wanned IL-13 (80 ng/mL in assay buffer) for stimulation. After incubating at 37 °C for 30 min, the assay buffer (containing compound and IL- 13) was removed, and 10 μL of cell lysis buffer added (25 mM HEPES, 0.1 % SDS, 1 % NP-40, 5 mM MgCh, 1.3 mM EDTA, 1 mM EGTA, supplemented with Complete Ultra mini protease inhibitors and PhosSTOP from Roche Diagnostics). The plates were shaken at ambient temperature for 30min before the addition of detection reagents. Levels of pSTAT6 were measured using the AlphaLISA SureFire Ultra pSTAT6 (Tyr641) assay kit from PerkinElmer. For dose-response analy sis, percent inhibition data were plotted vs. compound concentrations, and ICso values were determined from a 4-parameter robust fit model with the Graphpad Prism software.
[0280] Data are expressed as pICLu (negative decadic logarithm IC50) values. Certain compounds disclosed herein exhibited pICso values of greater than 6 or greater than 7 when tested in BEAS-2B cells. Test compounds having a lower IC50 value or higher pICso value in this assay show greater inhibition of IL-13 induced STAT6 phosphorylation.
[0281] Table 3 shows biological activities of selected compounds in a cellular JAK potency assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
Table 3
Figure imgf000114_0001
[0282] Example 12: Cytotoxicity Measured by Premature Chromosome Condensation [15] (pCC’15).
[0283] The impact of a compound of the present disclosure on cellular adenosine triphosphate (ATP) levels was measured in Beas2B cells, a human lung epithelial cell line. Levels of ATP are correlated with the viability of cells and are often measured to determine the potential cytotoxicity of compounds. CellTiter-Glo, which lyses the cells and produces a luminescent signal proportional to the amount of ATP present, was used to determine the effect of test compound on cell viability.
[0284] Beas2B cells were grown in 50% DMEM (Life Technologies) and 50% F-12 (Life Technologies) media, supplemented with 10% Fetal Bovine Serum (ATCC), 25 mM HEPES (Life Technologies), and lx Pen-Strep (Life Technologies). Cells were cultured in a humidified incubator set at 37 °C, 5% CO2, and trypsinized using 0.25% Trypsin with 0.5% polyvinylpyrrolidone (PVP). [0285] For the assay, Beas2B cells were seeded at 500 cells/well (25 |iL/well) in a 384-well plate and cultured overnight. Compounds were serially diluted in DMSO, then further diluted with growth media (40 pL/well) to create a compound plate 6x of the final assay concentration, at 0.6% DMSO. The diluted compounds were then added to the cells (5 μL/well) and incubated at 37 °C, 5% CO2 for 48 hours. After the compound incubation, CellTiter-Glo (Promega) was added directly to the cells (30 μL/mL). The assay plate was sealed and shaken at 700 rpm for 15 minutes in a darkened environment, then centrifuged for 2 minutes at 1500 rpm to settle the lysate at the bottom of the well. The effect of the compound on cell viability was determined through analysis of dose-dependent quantified changes in ATP from baseline (non-compound treated cells) and wells treated with 60 pM AT9283, a well-characterized cytotoxic compound. Data are expressed as pCCi5 (negative decadic logarithm CC15) values. Certain compounds disclosed herein exhibited pCCi5 values of less than 6, or less than 5.7, or less than 5.5 when tested in Beas2B cells.
[0286] Table 4 shows cytotoxicities of selected compounds in a premature chromosome condensation assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
Table 4
Figure imgf000115_0001
[0287] Example 13: Murine model of IL-13 induced pSTAT6 induction in lung tissue.
[0288] IL-13 binds to cell surface receptors activating members of the Janus family of kinases (JAK) which then phosphorylate STAT6 and subsequently activate further transcription pathways. In the described model, a dose of IL-13 was delivered locally into the lungs of mice to induce the phosphorylation of STAT6 (pSTAT6) which is then measured as the endpoint.
[0289] Adult balb/c mice from Harlan were used in the assay. On the day of study, animals were lightly anesthetized with isoflurane and administered either vehicle or test compound (0.5 mg/mL, 50 μL total volume over several breaths) via oral aspiration.
[0290] Animals were placed in lateral recumbency post dose and monitored for full recovery from anesthesia before being returned to their home cage. Eight hours later, animals were once again briefly anesthetized and challenged with either vehicle or EL-13 (0.03 pg total dose delivered, 50 μL total volume) via oral aspiration before being monitored for recovery from anesthesia and returned to their home cage. One hour after vehicle or IL-13 administration, lungs were collected for both pSTAT6 detection using an AlphaLISA Immunoassay (PerkinElmer) and analyzed for total drug concentration.
[0291] Selected compounds of the present disclosure were tested in the assay. Activity in the model is evidenced by a decrease in the level of pSTAT6 present in the lungs of treated animals at 9 hours compared to the vehicle treated, IL-13 challenged control animals. The difference between the control animals which were vehicle treated, IL-13 challenged and the control animals which were vehicle treated, vehicle challenged dictated the 0% and 100% inhibitory effect, respectively, in any given experiment. Exemplary’ compounds were tested in the assay and exhibited inhibition of STAT6 phosphoiylation at 9 hours after IL- 13 challenge as documented below.
[0292] Table 5 shows inhibition of STAT6 phosphorylation of selected compounds in the murine model. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
Table 5
Figure imgf000116_0001
[0293] Example 14: Pharmacokinetics in Plasma and Lung in Mouse After Oral Aspiration of Test Compounds.
[0294] Plasma and lung concentrations of test compounds were quantified and pharmacokinetic parameters were calculated in the following manner. Male CD1 mice from Charles River Laboratories were used in the pharmacokinetic studies. Test compounds were individually formulated in 20% propylene glycol in pH 4 citrate buffer at a concentration of 0.2 mg/mL. Test compounds were administered in two, 25 gL increments introduced into the trachea of each mouse by oral aspiration using a calibrated pipette once the animal was anesthetized using isoflurane. Blood samples were collected as terminal collections via cardiac puncture at 0.167, 1, 4, 8, and 24 hr post-dosing. Following inhalation with CCL, a direct cardiac puncture was performed while avoiding puncturing the lung, and blood was immediately transferred into K2EDTA tubes and placed on wet ice. Blood samples were centrifuged (Eppendorf centrifuge, 5804R) for 4 minutes at approximately 12,000 rpm at 4 °C to collect plasma. Intact lungs were also excised from these mice using the same timepoints (0. 167, 1, 4, 8, and 24 hr). Lungs were washed with sterile water to remove any blood residue and were patted dry, weighed, and homogenized in 0, 1% formic acid in water at a dilution of 1:3 (lung:water, weight/volume). Plasma and lung concentrations of test compounds were determined by LC-MS/MS analysis against analytical standards constructed into a standard curee in the test matrix. The pharmacokinetic parameters of test compounds were determined by non -com partmental analysis. For concentrations below the limit of quantification, zero was used for mean calculations. Mean values were not reported if more than 50% of the samples were below the limit of quantification at a timepoint, or if more than 50% of a calcuiated pharmacokinetic parameter was not reportable. The area under the concentration-time curve extrapolated to infinity (AUQo-inf)) was calculated as follows: AUQo-inf) = AUCH + Ciw / k, where AUCc&p is the area under the concentration-time curve from the time of dosing to the last measurable concentration calculated by the linear trapezoidal rule, Ciast is the last measurable concentration, and k is the first order rate constant associated with the terminal elimination phase, estimated by linear regression of time versus log concentration. The lung-to-plasma AUC ratio was determined as the ratio of the lung AUC(o-inf) in μg*hr/g to the plasma AUC(o-inf) in μg*hr/mL. Certain compounds disclosed herein exhibited lung-to-plasma AUC ratios of greater than 100, such as greater than 200, in male CD1 mice.
[0295] Table 6 shows plasma AUC(o-24) values, lung tissue AUC(o-24) values and the lung-to-plasma AUC ratios of selected compounds as assessed in accordance with this Example. For Plasma AUC(0-24), H- H- denotes a value < 0.5, +++ denotes a value from 0.6 to 1 .0, and ++ denotes a value from 1 .1 to 1 .5. For Lung Tissue AUCaw*), ++++ denotes a value from 101 to 200, +++ denotes a value from 51 to 100, and ++ denotes a value from 10 to 50. For the Lung:Plasma AUC Ratio, ++++ denotes a value > 300, +++ denotes a value from 201 to 300, ++ denotes a value from 101 to 200, and + denotes a value from 50 to 100. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-9.
Table 6
Figure imgf000117_0001

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula (I):
Figure imgf000118_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is independently selected at each occurrence from halogen, -OH, -O(C1-4 alkyl), -O(C1-4 haloalkyl), and C1-4 alkyl;
R2 is selected from hydrogen, halogen, and C1-4 alkyl;
R3 is independently selected at each occurrence from halogen and C1-4 alkyl; m is an integer from 0 to 5 ; n is an integer from 0 to 2; sss represents a double bond or triple bond; when is a double bond, then A is CR4R5 when as- is a triple bond, then A is CR6;
R4 and R5 are each independently selected from hydrogen and RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from hydrogen and RA;
RA is independently selected at each occurrence from: halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10);
Ci-10 alkyl, C2-10 alkenyl, and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)R10, -S(=O)2R10, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10,
-NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -C(O)R10, -C(O)OR10, -OC(O)R10, -OC(O)OR10, -OC(O)N(R10)2, -OC(O)NR11R12, -NR10C(O)R10, -NR10C(O)OR10, -NR10C(O)N(R10)2, -NR10C(O)NR11R12, -C(O)N(R10)2, -C(O)NR11R12, -P(O)(OR10)2, -P(O)(R10)2, =O, =S, =N(R10), R10, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, Co-3 alkyl-(C3-12 carbocycle), and Co-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH20H, -CH2CH2OH, -OCH3, -OCH2CH3, -OCH2CH2-OCH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R11 and R12 are taken together with the nitrogen atom to which they are attached to form a 3- to 12- membered heterocycle, optionally substituted with one or more R10; and
R7 is hydrogen or C1-6 alkyl.
2. The compound or salt of claim 1, wherein m is an integer from 1 to 3.
3. The compound or salt of claim 2, wherein m is 2.
4. The compound or salt of any one of claims 1 to 3, wherein n is 0.
5. The compound or salt of any one of claims 1 to 4, wherein the compound is a compound of
Formula (I-A):
Figure imgf000119_0001
6. The compound or salt of any one of claims 1 to 5, wherein R1 is independently selected at each occurrence from halogen, -OH, and -CH2CH3.
7. The compound or salt of any one of claims 1 to 6, wherein the compound is a compound of
Formula (I-D):
Figure imgf000120_0001
8. The compound or salt of any one of claims 1 to 7, wherein:
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA; and
R6 is selected from RA;
RA is independently selected at each occurrence from: halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12;
Ci-10 alkyl, optionally substituted at each occurrence with one or more substituents selected from halogen, -OR10, -SR10, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, C3-12 carbocycle, and 3- to 12-membered heterocycle; and
C3.i2 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -SR10, -CH2N(R10)2, -N(R10)2, -NR11R12, -S(=O)2N(R10)2, -S(=O)2NR11R12, -NR10S(=O)2R10, -NR10S(=O)2N(R10)2, -NR10S(=O)2NR11R12, -NR10C(O)R10, -C(O)N(R10)2, -C(O)NR11R12, R10, C1-6 alkyl, and C1-6 haloalkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, Co-3 alkyl-(C3-12 carbocycle), and Co-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-6 carbocycle, and 3- to 6-membered heterocycle.
9. The compound or salt of claim 8, wherein:
RA is independently selected at each occurrence from:
-N(R10)2;
CI-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 3- to 12-membered heterocycle; and C3-12 carbocycle and 3- to 12-membered heterocycle, wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-6 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C0-3 alkyl-(C3-i2 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
10. The compound or salt of any one of claims 1 to 9, wherein === is a double bond and A is CR4R5.
11. The compound or salt of claim 10, wherein the compound is a compound of Formula (I-E):
Figure imgf000121_0001
12. The compound or salt of claim 10 or 11, wherein:
R4 is selected from RA and R5 is selected from hydrogen and CH3;
R4 is selected from hydrogen and CH3 and R5 is selected from RA; or
R4 and R5 are taken together with the carbon atom to which they are attached to form a C4-10 carbocycle or 4- to 10-membered heterocycle, each of which is optionally substituted with one or more RA; and
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NHS(=O)2R10, -NHC(O)R10, and 4- to 10-membered heterocycle; and
4- to 10-membered heterocycle, wherein each 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from -OCH3, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-6 carbocycle), and C0-3 alkyl-(3- to 6-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -N(CH3)z, =O, -OH, and -CH3.
Figure imgf000122_0001
Figure imgf000123_0001
14. The compound or salt of any one of claims 1 to 9, wherein when === is a triple bond and A is
15. The compound or salt of claim 14, wherein the compound is a compound of Formula (I-F):
Figure imgf000123_0002
16. The compound or salt of claim 14 or 15, wherein R6 is selected from: C1-4 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, C3 -IO carbocycle, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in R6 is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen and C1-4 alkyl.
Figure imgf000123_0003
Figure imgf000124_0001
18. The compound or salt of any one of claims 1 to 17, wherein R2 is selected from hydrogen and halogen.
19. The compound or salt of claim 18, wherein R2 is hydrogen.
20. The compound or salt of claim 18, wherein R2 is F.
21. The compound or salt of claim 7, wherein:
R2 is selected from hydrogen and F;
R4 is selected from RA and R5 is selected from hydrogen and CH3: R4 is selected from hydrogen and CH3 and R5 is selected from RA; or R4 and R5 are taken together with the carbon atom to which they are attached to form a C3-12 carbocycle or 3- to 12-membered heterocycle, each of which is optionally substituted with one or more RA;
R6 is selected from RA;
RA is independently selected at each occurrence from:
-N(R10)2;
C1-3 alkyl, optionally substituted at each occurrence with one or more substituents selected from -N(R10)2, -NR11R12, -NR10S(=O)2R10, -NR10C(O)R10, and 4- to 10-membered heterocycle; and C3 -10 carbocycle and 4- to 10-membered heterocycle, wherein each C3 -10 carbocycle and 4- to 10-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR10, -N(R10)2, and C1-4 alkyl; and
R10 is independently selected at each occurrence from hydrogen; and C1-4 alkyl, C0-3 alkyl-(C3-io carbocycle), and C0-3 alkyl-(4- to 10-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2, -N(CH3)2, =O, -OH, and -CH3.
22. The compound or salt of claim 7, wherein:
R2 is selected from hydrogen and F; and
Figure imgf000125_0001
23. The compound or salt of claim 7, wherein:
R2 is selected from hydrogen and F; and
Figure imgf000125_0002
Figure imgf000126_0001
24. The compound or salt of claim 7, wherein:
R2 is selected from hydrogen and F; and
Figure imgf000126_0002
25. The compound or salt of claim 7, wherein:
R2 is selected from hydrogen and F; and
Figure imgf000126_0003
26. A substantially pure stereoisomer of the compound or salt of any one of claims 1 to 25.
27. The compound or salt of claim 26, wherein the stereoisomer is provided in at least 90% enantiomeric excess.
28. A compound selected from Table 1, or a pharmaceutically acceptable salt thereof.
29. A pharmaceutical composition comprising the compound or salt of any one of claims 1 to 28 and a pharmaceutically acceptable carrier.
30. The pharmaceutical composition of claim 29, further comprising one or more additional therapeutic agents.
31. The pharmaceutical composition of claim 29 or 30, wherein the pharmaceutical composition is formulated for inhalation.
32. A method of inhibiting JAK, comprising contacting JAK with an effective amount of the compound or salt of any one of claims 1 to 28.
33. A method of treating a JAK-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1 to 28.
34. The method of claim 33, wherein the disease or condition is a respiratory disease.
35. The method of claim 34, wherein the respiratory disease is selected from asthma, chronic obstructive pulmonary disease, cystic fibrosis, pneumonitis, idiopathic pulmonary fibrosis, acute lung injusy, acute respiratory distress syndrome, bronchitis, emphysema, bronchiolitis obliterans, sarcoidosis, an eosinophilic disease, a helminthic infection, pulmonary arterial hypertension, lymphangioleiomyomatosis, bronchiectasis, an infiltrative pulmonary disease, drug-induced pneumonitis, fungal induced pneumonitis, allergic bronchopulmonary’ aspergillosis, hypersen sitivity pneumonitis, eosinophilic granulomatosis with polyangiitis, idiopathic acute eosinophilic pneumonia, idiopathic chronic eosinophilic pneumonia, hypereosinophilic syndrome, Loftier syndrome, bronchiolitis obliterans organizing pneumonia, lung graft- versus-host disease, COVID-19, SARS, MERS, chronic rhino sinusitis with or without nasal polyps, nasal polyposis, sinusitis with nasal polyps, rhinitis, and immune-checkpoint-inhibitor induced pneumonitis.
36. The method of claim 34, wherein the respiratory disease is selected from asthma and chronic obstructive pulmonary disease.
37. The method of claim 36, wherein the asthma is selected from T2-dominant (eosinophilic) asthma and non-T2-dominant (non-eosinophilic) asthma.
38. A method of treating lung transplant rejection in a subject, comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1 to 28.
39. The method of claim 38, wherein the lung transplant rejection is selected from primary graft dysfunction, organizing pneumonia, acute rejection, lymphocytic bronchiolitis, and chronic lung allograft dysfunction.
40. The method of claim 38, wherein the lung transplant rejection is acute lung transplant rejection.
41. The method of claim 38, wherein the lung transplant rejection is chronic lung allograft dysfunction.
42. The method of claim 38, wherein the lung transplant rejection is selected from bronchiolitis obliterans, restrictive chronic lung allograft dysfunction, and neutrophilic allograft dysfunction.
43. The method of any one of claims 32 to 42, comprising administering a second therapeutic agent.
44. The method of any one of claims 32 to 43, wherein the compound or salt is administered by inhalation.
45. A compound or salt of any one of claims 1 to 28 for use in treating a respiratory disease.
46. A compound or salt of any one of claims 1 to 28 for use in treating lung transplant rejection.
47. The use of a compound or salt of any one of claims 1 to 28 for the manufacture of a medicament for treating a respiratory disease.
48. The use of a compound or salt of any one of claims 1 to 28 for the manufacture of a medicament for treating lung transplant rejection.
PCT/US2022/050466 2021-11-19 2022-11-18 Bicyclic inhibitors of jak and methods of use WO2023091707A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163281218P 2021-11-19 2021-11-19
US63/281,218 2021-11-19

Publications (1)

Publication Number Publication Date
WO2023091707A1 true WO2023091707A1 (en) 2023-05-25

Family

ID=84602349

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/050466 WO2023091707A1 (en) 2021-11-19 2022-11-18 Bicyclic inhibitors of jak and methods of use

Country Status (1)

Country Link
WO (1) WO2023091707A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
WO2021136345A1 (en) * 2019-12-30 2021-07-08 路良 Jak inhibitor compound and use thereof
WO2022272020A1 (en) * 2021-06-25 2022-12-29 Theravance Biopharma R&D Ip, Llc Imidazolo indazole compounds as jak inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
WO2021136345A1 (en) * 2019-12-30 2021-07-08 路良 Jak inhibitor compound and use thereof
EP4071145A1 (en) * 2019-12-30 2022-10-12 Liang Lu Jak inhibitor compound and use thereof
WO2022272020A1 (en) * 2021-06-25 2022-12-29 Theravance Biopharma R&D Ip, Llc Imidazolo indazole compounds as jak inhibitors

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WHITE
ASSIRI ET AL., N ENGL J MED., vol. 369, 2013, pages 407 - 1
BERASTEGUI ET AL., CLIN. TRANSPLANT., vol. 31, 2017, pages e12898
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
COTTIN ET AL., CLIN. CHEST. MED., vol. 37, no. 3, 2016, pages 535 - 56
EL-HASHEMITE ET AL., AM. J. RESPIR. CELL. MOL. BIOL., vol. 33, 2005, pages 227 - 230
EL-HASHEMITE ET AL., CANCER RES., vol. 64, 2004, pages 3436 - 3443
GAUTHIER ET AL., CURR. TRANSPLANT. REP., vol. 3, no. 3, 2016, pages 185 - 191
GEORAS STEVE N ET AL: "JAK inhibitors for asthma", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 148, no. 4, 1 October 2021 (2021-10-01), pages 953 - 963, XP086812861, ISSN: 0091-6749, [retrieved on 20211005], DOI: 10.1016/J.JACI.2021.08.013 *
GUAN ET AL., N ENGL J MΕ·D., vol. 382, 2020, pages 1708 - 1720
H.C. ANSEL ET AL.: "Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WHITE
HIGUCHI, T. ET AL.: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14, 1987
HUANG ET AL., THE LANCET, vol. 395, no. 10223, 2020, pages 507 - 506
JOO ET AL.: "Therapeutic Effects of Intranasal Tofacitinib on Chronic Rhinosinusitis with Nasal Polyps in Mice", THE LARYNGOSCOPE, vol. 202A, Retrieved from the Internet <URL:https://doi.org/10.1002/lary.29129>
KUDLACZ ET AL., EUR. J. PHARMACOL, vol. 582, 2008, pages 154 - 161
KURUVILLA ET AL., CLIN REV ALLERGY IMMUNOL, vol. 56, no. 2, 2019, pages 219 - 233
MALAVIYA ET AL., INT. IMMUNOPHARMACOL., vol. 70, 2010, pages 829 - 836
MATSUNAGA ET AL., BIOCHEM. AND BIOPHYS. RES. COMMUN., 2011, pages 261 - 267
PETER JONES ET AL: "Design and Synthesis of a Pan-Janus Kinase Inhibitor Clinical Candidate (PF-06263276) Suitable for Inhaled and Topical Delivery for the Treatment of Inflammatory Diseases of the Lungs and Skin", JOURNAL OF MEDICINAL CHEMISTRY, vol. 60, no. 2, 4 January 2017 (2017-01-04), US, pages 767 - 786, XP055468539, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.6b01634 *
SHINO ET AL., PL(7S ONE,, vol. 12, no. 7, 2017, pages eO 180281
VICENTI ET AL., AMERICAN JOURNAL OF TRANSPLANTATION, vol. 12, 2012, pages 2446 - 56
WANG ET AL., JAMA, vol. 323, no. 11, 2020, pages 1061 - 1069
XU XHAN MLI TSUN WWANG DFU B ET AL.: "Effective Treatment of Severe COVID-19 Patients with T<x:ilizumab", PNAS, 2020, Retrieved from the Internet <URL:https://doi.org/10.1073/pnas.2005615117>
ZEISER ET AL., LEUKEMIA, vol. 29, no. 10, 2015, pages 2062 - 68

Similar Documents

Publication Publication Date Title
US11878977B2 (en) JAK inhibitors containing a 4-membered heterocyclic amide
EP3837258B1 (en) Dimethyl amino azetidine amides as jak inhibitors
US11713315B2 (en) 5 to 7 membered heterocyclic amides as JAK inhibitors
US11702415B2 (en) Crystalline hydrate of a JAK inhibitor compound
WO2022272020A1 (en) Imidazolo indazole compounds as jak inhibitors
WO2022178215A1 (en) Amino amide tetrahydro imidazo pyridines as jak inhibitors
WO2023091707A1 (en) Bicyclic inhibitors of jak and methods of use
NZ756823B2 (en) Jak inhibitors containing a 4-membered heterocyclic amide

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22830034

Country of ref document: EP

Kind code of ref document: A1