HETEROBICYCLIC CARBOXAMIDES AND USES THEREOF

Information

  • Patent Application
  • 20230002404
  • Publication Number
    20230002404
  • Date Filed
    September 11, 2020
    3 years ago
  • Date Published
    January 05, 2023
    a year ago
Abstract
Provided herein are heterobicyclic cyclopropanecarboxamide and heterobicyclic carboxamide compounds. In particular, provided herein are compounds that affect the function of kinases in a cell, and that are useful as therapeutic agents or with therapeutic agents. The compounds provided herein are useful in the treatment of a variety of diseases and conditions including eye diseases such as glaucoma, cardiovascular diseases, inflammatory diseases, and diseases characterized by abnormal growth, such as cancers. Also provided herein are compositions comprising heterobicyclic cyclopropanecarboxamide or heterobicyclic carboxamide compounds.
Description
FIELD OF THE INVENTION

The present disclosure relates to heterobicyclic cyclopropanecarboxamide compounds that affect the function of kinases and other proteins in a cell and that are useful as therapeutic agents or with therapeutic agents. In particular, these compounds are useful in the treatment of eye diseases such as glaucoma and retinal diseases, as anti-inflammatory agents, for the treatment of cardiovascular diseases, and for diseases characterized by abnormal growth, such as cancers.


BACKGROUND

A variety of hormones, neurotransmitters and biologically active substances control, regulate or adjust the functions of living bodies via specific receptors located in cell membranes. Many of these receptors mediate the transmission of intracellular signals by activating guanine nucleotide-binding proteins (G proteins) to which the receptor is coupled. Such receptors are generically referred to as G-protein coupled receptors (GPCRs) and include, among others, α-adrenergic receptors, β-adrenergic receptors, opioid receptors, cannabinoid receptors and prostaglandin receptors. The biological effects of activating or inhibiting these receptors is not direct but is mediated by a host of intracellular proteins. The importance of these secondary proteins has been recognized and modulation of this class is now being investigated as intervention points in disease states. One of the most important classes of these downstream effectors is the “kinase” class.


The various kinases play important roles in the regulation of various physiological functions. For example, kinases have been implicated in a number of disease states, including, but not limited to: cardiac indications such as angina pectoris, essential hypertension, myocardial infarction, supraventricular and ventricular arrhythmias, congestive heart failure, atherosclerosis, renal failure, diabetes, respiratory indications such as asthma, chronic bronchitis, bronchospasm, emphysema, airway obstruction, upper respiratory indications such as rhinitis, seasonal allergies, inflammatory disease, inflammation in response to injury, rheumatoid arthritis. The importance of p38 MAPK inhibitors in particular as new drugs for rheumatoid arthritis is reflected by the large number of compounds that has been developed over the last years (J. Westra and P. C. Limburg Mini-Reviews in Medicinal Chemistry Volume 6, Number 8, August 2006). Other conditions include chronic inflammatory bowel disease, glaucoma, hypergastrinemia, gastrointestinal indications such as acid/peptic disorder, erosive esophagitis, gastrointestinal hypersecretion, mastocytosis, gastrointestinal reflux, peptic ulcer, Zollinger-Ellison syndrome, pain, obesity, bulimia nervosa, depression, obsessive-compulsive disorder, organ malformations (e.g., cardiac malformations), neurodegenerative diseases such as Parkinson's Disease and Alzheimer's Disease, multiple sclerosis, Epstein-Barr infection and cancer (Nature Reviews Drug Discovery 2002, 1: 493-502). In other disease states, the role of kinases is only now becoming clear. The retina is a complex tissue composed of multiple interconnected cell layers, highly specialized for transforming light and color into electrical signals that are perceived by the brain. Damage or death of the primary light-sensing cells, the photoreceptors, results in devastating effects on vision. Despite the identification of numerous mutations that cause inherited retinal degenerations, the cellular and molecular mechanisms leading from the primary mutations to photoreceptor apoptosis are not well understood but may involve the wnt pathway (A S Hackam “The Wnt Signaling Pathway in Retinal Degeneration” IUBMB Life Volume 57, Number 6/June 2005).


The success of the tyrosine-kinase inhibitor ST1571 (Gleevec) in the treatment of chronic myelogenous leukemia (Nature Reviews Drug Discovery 2003, 2: 296-313) has spurred considerable efforts to develop other kinase inhibitors for the treatment of a wide range of other cancers (Nature Reviews Cancer 2003, 3: 650-665). The balance between the initiation and the inactivation of intracellular signals determines the intensity and duration of the response of the receptors to stimuli such as agonists. When desensitization occurs, the mediation or regulation of the physiological function mediated or regulated by the G proteins to which the receptors are coupled is reduced or prevented. For example, when agonists are administered to treat a disease or condition by activation of certain receptors, the receptors relatively quickly become desensitized from the action of the GRKs such that agonist administration may no longer result in therapeutic activation of the appropriate receptors. At that point, administration of the agonist no longer enables sufficient or effective control of or influence on the disease or condition intended to be treated.


Janus Kinases (or JAK) are a family of cytoplasmic protein tyrosine kinases. The JAK family plays a role in the cytokine-dependent regulation of proliferation and function of cells involved in immune response. Currently, there are four JAK family members are known JAK1, JAK2, JAK3, and TYK2. 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 inflammation. 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. Phosphorylated 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. There is recent evidence that the kinases ROCK1 and ROCK2 may also participate in similar interactions with the STAT enzymes. Inhibition of ROCK kinases has been noted to have an anti-inflammatory effect as well.


In view of the role that kinases play in many disease states, there is an urgent and continuing need for small molecule ligands which inhibit or modulate the activity of kinases. Without wishing to be bound by theory, it is thought that modulation of the activity of kinases, in particular ROCK and JAK kinases, by the compounds of the present disclosure is, at least in part, responsible for their beneficial effects.


SUMMARY

In one aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H or halogen;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is CH2, phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S;


R9 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 alkyl-CN, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl; and


n is 0 or 1; or




embedded image


is H, and R1-6 and n are as defined above.


In another aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R13 is CH2, CH2NH,




embedded image


R14 is H, OH, NH2, CN, C1-3 alkyl, phenyl, phenyl-CH3, CH2-phenyl, phenyl-(O—C1-6 alkyl), SO2—(C1-6 alkyl), CHCH2, C1-3 alkylene-CN, C1-6 alkyl-OH, C1-6 alkyl-NH2, a monocyclic C3-5 heterocycloalkyl having 1-3 (e.g., 1 or 2, e.g., 1) heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S (e.g., N or O, e.g., N, e.g., O), and wherein each heteroaryl may be substituted with one or two halogen substituents; and


R15 is H, CN, OH, or NH2.


In another aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R16 is phenyl, CH2-phenyl, a CH2-monocyclic C3-5 heteroaryl having 1-3 heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S, and wherein each heteroaryl may be substituted with one or two halogen substituents;


R17 is H, OH, halogen, C1-6 alkyl-OH, C1-6 alkyl-O—C(O)-phenyl-(R18)z;


R18 is H, OH, halogen, or C1-6 alkyl;


R19 is H or C1-6 alkyl;


R20 is H or C1-6 alkyl;


z is 0, 1, 2, 3, 4, or 5; and


m is 0, 1, or 2.


In another aspect, the present disclosure provides a pharmaceutical composition comprising a compound according to the present disclosure and a pharmaceutically acceptable excipient.


In another aspect, the present disclosure provides a method of treating an ocular disorder in a subject in need of treatment, comprising administering to the subject a compound or composition according to the present disclosure.


In another aspect, the present disclosure provides a method of reducing intraocular pressure in a subject in need thereof, comprising administering to an eye of the subject a compound or composition according to the present disclosure.


In another aspect, the present disclosure provides a kit comprising a compound or composition according to the present disclosure and instructions for use.







DETAILED DESCRIPTION

Publications and patents are referred to throughout this disclosure. All U.S. Patents cited herein are hereby incorporated by reference. All percentages, ratios, and proportions used herein are percent by weight unless otherwise specified.


Heterobicyclic cyclopropanecarboxamides are provided herein.


As used herein, the term “alkyl” refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups. “Alkyl” may be exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl and the like. “C1-6 alkyl” refers to alkyl groups containing one to six carbon atoms.


As used herein, the term “contacting a cell” is used to mean contacting a cell in vitro or in vivo i.e. in a subject, such as a mammal, including humans, rabbits, cats and dogs.


As used herein, the term “controlling the disease or condition” is used to mean changing the activity of one or more kinases to affect the disease or condition.


As used herein, the term “disease or condition associated with kinase activity” is used to mean a disease or condition treatable, in whole or in part, by inhibition of one or more kinases.


As used herein, the term “excipient” includes physiologically compatible additives useful in preparation of a pharmaceutical composition. Examples of pharmaceutically acceptable carriers and excipients can for example be found in Remington Pharmaceutical Science, 16th Ed.


As used herein, the term “eye disease” includes, but is not limited to, glaucoma, allergy, cancers of the eye, neurodegenerative diseases of the eye, such as diabetic eye disease, macular degeneration (AMD), inflammation, and dry eye.


As used herein, the term “halogen” refers to fluoro, chloro, bromo or iodo moieties. In some embodiments, the halogen is fluoro, chloro, or bromo. In some embodiments, the halogen is fluoro.


As used herein, the term “haloalkyl” refers to an alkyl group substituted with one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) halogens, each independently selected from fluoro, chloro, bromo or iodo.


As used herein, the term “heteroaryl” refers to an aromatic carbocyclic radical having one or more heteroatoms in the carbocyclic ring.


As used herein, the term “pharmaceutically acceptable carrier” means a carrier that is useful for the preparation of a pharmaceutical composition that is: generally compatible with the other ingredients of the composition, not deleterious to the recipient, and neither biologically nor otherwise undesirable. “A pharmaceutically acceptable carrier” includes both one and more than one carrier. Embodiments include carriers for topical, ocular, parenteral, intravenous, intraperitoneal intramuscular, sublingual, nasal and oral administration. “Pharmaceutically acceptable carrier” also includes agents for preparation of aqueous dispersions and sterile powders for injection or dispersions.


As used herein, “pharmaceutically acceptable salts” refers to an ionizable therapeutic agent that has been combined with a counter-ion to form a neutral complex. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.


“Treatment” of an individual or a cell is any type of intervention provided as a means to alter the natural course of a disease or pathology in the individual or cell. Treatment includes, but is not limited to, administration of, e.g., a pharmaceutical composition, and may be performed either prophylactically, or subsequent to the initiation of a pathologic event or contact with an etiologic agent. Treatment includes any desirable effect on the symptoms or pathology of a disease or condition associated with inflammation, among others described herein.


Also included are “prophylactic” treatments, which can be directed to reducing the rate of progression of the disease or condition being treated, delaying the onset of that disease or condition, or reducing the severity of its onset. “Treatment” or “prophylaxis” does not necessarily indicate complete eradication, cure, or prevention of the disease or condition, or associated symptoms thereof.


As used herein, the term “therapeutically effective amount” as used herein refers to a dosage of the compounds or compositions effective for influencing, reducing or inhibiting the activity of or preventing activation of a kinase. This term as used herein may also refer to an amount effective at bringing about a desired in vivo effect in an animal, preferably, a human, such as reduction in intraocular pressure.


Compounds

In one aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H or halogen;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is CH2, phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S;


R9 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 alkyl-CN, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl; and


n is 0 or 1; or




embedded image


is H, and R1-6 and n are as defined above.


In one aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R13 is CH2, CH2NH,




embedded image


R14 is H, OH, NH2, CN, C1-3 alkyl, phenyl, phenyl-CH3, CH2-phenyl, phenyl-(O—C1-6 alkyl), SO2—(C1-6 alkyl), CHCH2, C1-3 alkylene-CN, C1-6 alkyl-OH, C1-6 alkyl-NH2, a monocyclic C3-5 heterocycloalkyl having 1-3 (e.g., 1 or 2, e.g., 1) heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S (e.g., N or O, e.g., N, e.g., O), and wherein each heteroaryl may be substituted with one or two halogen substituents; and


R15 is H, CN, OH, or NH2.


In some embodiments,




embedded image


comprises 1 chemical ring. In some embodiments,




embedded image


comprises 2 chemical rings.


In another aspect, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, halogen, CN, NH2, OH, C1-6 alkyl, C1-6 haloalkyl, O—(C1-6 alkyl), O—(C1-6 haloalkyl), or C3-6 cycloalkyl;


R2 is H, halogen, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R16 is phenyl, CH2-phenyl, a CH2-monocyclic C3-5 heteroaryl having 1-3 heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S, and wherein each heteroaryl may be substituted with one or two halogen substituents;


R17 is H, OH, halogen, C1-6 alkyl-OH, C1-6 alkyl-O—C(O)-phenyl-(R18)z;


R18 is H, OH, halogen, or C1-6 alkyl;


R19 is H or C1-6 alkyl;


R20 is H or C1-6 alkyl;


z is 0, 1, 2, 3, 4, or 5; and


m is 0, 1, or 2.


In some embodiments of the formulae provided herein, the compounds are of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, F, Cl, Br, I, CN, NH2, OH, OCF3, OCH2CF3, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl;


R2 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H, F, Cl, Br, or I;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S; when R7 is not H


R9 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl when R7 is not H; and


n is 0 or 1; or




embedded image


is H, and R1-6 and n are as defined above.


In some embodiments of the formulae provided herein, the compounds are of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, F, Cl, Br, I, CN, NH2, OH, OCF3, OCH2CF3, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl;


R2 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H, F, Cl, Br, or I;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S;


R9 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl; and


n is 0 or 1.


In some embodiments, provided herein are compounds of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R2 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H, F, Cl, Br, or I;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S;


R9 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl; and


n is 0 or 1; or




embedded image


is H, and R1-6 and n are as defined above.


In some embodiments of the formulae provided herein, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


R1 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R2 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;


R3 is




embedded image


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H, F, Cl, Br, or I;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S;


R9 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl; and


n is 0 or 1.


In some embodiments of the formulae provided herein, the compound is a compound of the formula selected from:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


wherein


n is 1;


R10 is




embedded image


R11 and R12 are connected by an amide or sulfonamide linkage;


R12 is




embedded image


and


R11 is




embedded image


or n is 0, R10 and R12 are as defined above, and R11 is




embedded image


In some embodiments, the compound is a compound of the formula:




embedded image


or a pharmaceutically acceptable salt thereof,


In some embodiments, the compound is a compound of the formula selected from:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound is of the formula:




embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments of the formulae provided herein:


R1 is H, F, Cl. Me, CN, or C1-3 alkyl;


R2 is H, F, or C1-3 alkyl; or


R9 is H, F, or C1-3 alkyl.


In some embodiments




embedded image


is H. In some embodiments




embedded image


is H, and n is 1.

In some embodiments R1 is H, Br, Cl, F, Me, Et, iPr, Pr, nBu, iBu, sBu, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CHF2, —CF3, —OCF3, —CH2CF3, —CN, —CH2CN, —NH2, —OH, —CH2OH, or —CH2NH2.


In some embodiments R4 is S.


In some embodiments R4 is —C(H)═C(H)—.


In some embodiments R4 is —C(H)═N—.


In some embodiments R6 is CO.


In some embodiments R6 is SO2.


In some embodiments R7 is a bond.


In some embodiments R7 is a CH2.


In some embodiments R1 is H, R5 is H, or R9 is H.


In some embodiments R8 is phenyl.


In some embodiments R8 is a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S.


In some embodiments R8 is a monocyclic C3-5 heteroaryl having 1-3 nitrogen atoms.


In some embodiments R8 is a pyridyl, pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, pyrimidinyl, triazinyl, thiazolyl, pyrazinyl, pyridazinyl, or oxazolyl.


In some embodiments R14 is a pyridyl, pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, pyrimidinyl, triazinyl, thiazolyl, pyrazinyl, pyridazinyl, or oxazolyl.


In some embodiments R16 is a pyridyl, pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, pyrimidinyl, triazinyl, thiazolyl, pyrazinyl, pyridazinyl, or oxazolyl.


In some embodiments the heteroaryl is a pyridyl, pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, pyrimidinyl, triazinyl, thiazolyl, pyrazinyl, pyridazinyl, or oxazolyl.


In some embodiments:




embedded image


R1 is H, Cl, or CH3;


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C4-5 heteroaryl having 1 or 2 nitrogen atoms;


R9 is H or F; and


n is 1.


In some embodiments:




embedded image


R1 is H, Br, Cl, F, Me, Et, iPr, Pr, nBu, iBu, sBu, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CHF2, —CF3, —OCF3, —CH2CF3, —CN, —CH2CN, —NH2, —OH, —CH2OH, or —CH2NH2;


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H;


R6 is CO or SO2;


R7 is a bond or CH2;


R8 is phenyl, or a monocyclic C4-5 heteroaryl having 1 or 2 nitrogen atoms;


R9 is H or F; and


n is 1.


In some embodiments:




embedded image


R1 is H, Br, Cl, F, Me, Et, iPr, Pr, nBu, iBu, sBu, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CHF2, —CF3, —OCF3, —CH2CF3, —CN, —CH2CN, —NH2, —OH, —CH2OH, or —CH2NH2;


R4 is S, —C(H)═C(H)—, or —C(H)═N—;


R5 is H;


R6 is CO or SO2;




embedded image


is H; and

n is 1.


In some embodiments:




embedded image


R1 is H, Cl, or CH3;


R4 is —C(H)═C(H)— or —C(H)═N—;


R5 is H; and


n is 0.


In some embodiments:




embedded image


R1 is H, Br, Cl, F, Me, Et, iPr, Pr, nBu, iBu, sBu, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —CHF2, —CF3, —OCF3, —CH2CF3, —CN, —CH2CN, —NH2, —OH, —CH2OH, or —CH2NH2;


R4 is —C(H)═C(H)— or —C(H)═N—;


R5 is H; and


n is 0.


In some embodiments R13 is CH2 or CH2NH.


In some embodiments, R13 is




embedded image


In some embodiments, R13 is




embedded image


In some embodiments R14 is H, OH, NH2, CN, C1-3 alkyl, SO2—(C1-6 alkyl), CHCH2, C1-3 alkylene-CN, C1-6 alkyl-OH, or C1-6 alkyl-NH2.


In some embodiments R14 is OH, NH2, CN, C1-3 alkyl, phenyl, phenyl-CH3, CH2-phenyl, phenyl-(O—C1-6 alkyl), SO2—(C1-6 alkyl), CHCH2, C1-3 alkylene-CN, C1-6 alkyl-OH, C1-6 alkyl-NH2, a monocyclic C3-5 heterocycloalkyl having 1-3 (e.g., 1 or 2, e.g., 1) heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S (e.g., N or O, e.g., N, e.g., O), and wherein each heteroaryl may be substituted with one or two halogen substituents.


In some embodiments R14 is phenyl, phenyl-CH3, CH2-phenyl, phenyl-(O—C1-6 alkyl), a monocyclic C3-5 heterocycloalkyl having 1-3 (e.g., 1 or 2, e.g., 1) heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S (e.g., N or O, e.g., N, e.g., O), and wherein each heteroaryl may be substituted with one or two halogen substituents.


In some embodiments R15 is CN, OH, or NH2. In some embodiments R15 is H.


In some embodiments R16 is phenyl, or CH2-phenyl.


In some embodiments R16 is a CH2-monocyclic C3-5 heteroaryl having 1-3 heteroatoms, or a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S, and wherein each heteroaryl may be substituted with one or two halogen substituents.


In some embodiments R17 is H, OH, halogen, or C1-6 alkyl-OH.


In some embodiments R17 is C1-6 alkyl-OH or C1-6 alkyl-O—C(O)-phenyl-(R18)z.


In some embodiments R17 is C1-6 alkyl-O—C(O)-phenyl-(R18)z.


In some embodiments z is 1 or 2. In some embodiments, z is 0.


In some embodiments m is 1 or 2. In some embodiments m is 0


In some embodiments the compound is a compound provided in a Table herein, or a pharmaceutically acceptable salt thereof.


In some embodiments the compound is:




embedded image


embedded image


embedded image


embedded image


or a pharmaceutically acceptable salt thereof.


In some embodiments of the formulae provided herein, the formula is trans racemic or trans-R,R with respect to the cyclopropane stereocenters.


General Scheme for the Coupling of 1,6-Naphthyridin-2-Amine to Carboxylic Acid:



embedded image


N-(1,6-naphthyridin-2-yl)-2-(pyridine-4-yl)cyclopropane-1-carboxamide (S1-2). To 2-(pyridine-4-yl)cyclopropane-1-carboxylic acid S1-1 (1.0 eq) was charged 1,6 naphthyridin-2-amine (1.0 eq), EDC (1.6 eq), HOBt (1.6 eq), DMAP (0.2 eq), and pyridine (1.0 mL). Stirred at 70 deg Celsius overnight. Poured into saturated aqueous sodium bicarbonate and extracted with ethyl acetate (3×). Washed combined organics with saturated sodium chloride. Dried combined organics over magnesium sulfate, filtered, and evaporated. The crude reaction mixture was purified by column chromatography (DCM/MeOH) followed by RPLC (ACN/water) to afford N-(1,6-naphthyridin-2-yl)-2-(pyridine-4-yl)cyclopropane-1-carboxamide (S1-2, 53% yield). Trans racemic or trans-R,R compounds were prepared according to a similar procedure using the appropriate starting materials.


Using the general scheme shown above, making adjustments as necessary for protecting groups, etc., the compounds of Table 1 were synthesized.











TABLE 1





Compound




Number
Structure
IC50

















1


embedded image


12 nM JAK2





2


embedded image


5.0 nM ROCK2





3


embedded image


6.0 nM ROCK2 7.0 nM ROCK1





4


embedded image


22 nM JAK2 6.0 nM ROCK2





5


embedded image


8.9 nM JAK2





6


embedded image


8.0 nM TYK2





7


embedded image


1.1 nM TYK2





8


embedded image








9


embedded image


12 nM JAK2





10


embedded image








11


embedded image


7.0 nM ROCK2





12


embedded image


12 nM ROCK2





13


embedded image


18 nM ROCK2





14


embedded image


230 nM ROCK2





15


embedded image


350 nM JAK1





16


embedded image


54 nM ROCK2





17


embedded image


30 nM ROCK2





18


embedded image


30 nM ROCK2





19


embedded image


43 nM ROCK1





20


embedded image


35 nM TYK2





21


embedded image


111 nM ROCK2





22


embedded image


22 nM JAK2










Using a substantially the same procedures as described above, with appropriate modifications, compounds E26-E61 (Table 2) can be prepared.










TABLE 2







E26


embedded image







E27


embedded image







E28


embedded image







E29


embedded image







E30


embedded image







E31


embedded image







E32


embedded image







E33


embedded image







E34


embedded image







E35


embedded image







E36


embedded image







E37


embedded image







E38


embedded image







E39


embedded image







E40


embedded image







E41


embedded image







E42


embedded image







E43


embedded image







E44


embedded image







E45


embedded image







E46


embedded image







E47


embedded image







E48


embedded image







E49


embedded image







E50


embedded image







E51


embedded image







E52


embedded image







E53


embedded image







E54


embedded image







E55


embedded image







E56


embedded image







E57


embedded image







E58


embedded image







E59


embedded image







E60


embedded image







E61











embedded image


(E)-5-(3-(tert-butoxy)-3-oxoprop-1-en-1-yl)thiophene-3-carboxylic acid (S2-2). To 5-formylthiophene-3-carboxylic acid in DME at 0° C. was added tert-butyl 2-(diethoxyphosphoryl)acetate and NaH and solution was warmed to room temperature then heated at 30° C. After 3 hours the reaction is poured into EtOAc and HCl (1N), extracted, dried (Na2SO4) filtered and evaporated to give crude 2. Column chromatography (0-20% EtOAc-Hexanes) gave pure (E)-5-(3-(tert-butoxy)-3-oxoprop-1-en-1-yl) thiophene-3-carboxylic acid (S2-2, 82%).


5-(2-(tert-butoxycarbonyl)cyclopropyl)thiophene-3-carboxylic acid (S2-3). To TMSOI in DMSO was added NaH and solution was stirred for 1 hour at room temperature. Then (E)-5-(3-(tert-butoxy)-3-oxoprop-1-en-1-yl)thiophene-3-carboxylic acid (S2-2) in DMSO was added and solution stirred at 30° C. for 1-3 hours. The mixture was cooled and poured into cold EtOAc-HCl(1N) and extracted, dried (Na2SO4) filtered and evaporated. Column chromatography 5% MeOH—CH2Cl2 gave pure 5-(2-(tert-butoxycarbonyl)cyclopropyl)thiophene-3-carboxylic acid (73%, S2-3).


tert-Butyl-2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl)cyclopropane-1-carboxylate (S2-4). To 5-(2-(tert-butoxycarbonyl)cyclopropyl)thiophene-3-carboxylic acid (S2-3) in pyridine was added EDC, DMAP and pyrimidin-2-ylmethanamine hydrochloride and solution was stirred at room temperature overnight. The mixture was poured into EtOAc-NaHCO3 (sat) and extracted, dried (Na2SO4), filtered and evaporated. Column chromatography (0-5%) MeOH—CH2Cl2 gave pure tert-butyl 2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl)cyclopropane-1-carboxylate (97%, S2-4)


2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl) cyclopropane-1-carboxylic acid (S2-5). To tert-butyl 2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl)cyclopropane-1-carboxylate (S2-4) in CH2Cl2 was added HCl (4N) and solution was stirred overnight. The CH2Cl2 was decanted off and the compound was washed with Et2O, dried to give 2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl)cyclopropane-1-carboxylic acid hydrochloride (S2-5). (>90%)


5-(2-((4-chlorothieno[2,3-c]pyridin-2-yl)carbamoyl)cyclopropyl)-N-(pyrimidin-2-ylmethyl)thiophene-3-carboxamide (S2-6). To 2-(4-((pyrimidin-2-ylmethyl)carbamoyl)thiophen-2-yl)cyclopropane-1-carboxylic acid hydrochloride (S2-5) in pyridine was added EDC, DMAP and 2-(λ2-azanyl)-4-chlorothieno[2,3-c]pyridine and the solution was stirred at room temperature overnight. The reaction was poured into EtOAc-NaHCO3(sat) extracted, dried (Na2SO4) filtered and evaporated. Column chromatography to give 5-(2-((4-chlorothieno[2,3-c]pyridin-2-yl)carbamoyl)cyclopropyl)-N-(pyrimidin-2-ylmethyl)thiophene-3-carboxamide (48%, S2-6).


Using procedures similar to those set forth above for Scheme 1 and substituting the appropriate starting materials, the compounds E107-119 (Table 3) were made and E120-146 (Table 4) could be synthesized.










TABLE 3









embedded image


E107





trans racemic








embedded image


E108





R,R enantiomer








embedded image


E109





trans racemic








embedded image


E110





R,R enantiomer








embedded image


E111





R,R enantiomer








embedded image


E112





R,R enantiomer








embedded image


E113





R,R enantiomer








embedded image


E114





R,R enantiomer








embedded image


E115





trans racemic








embedded image


E116





trans racemic








embedded image


E116





R,R enantiomer








embedded image


E117





trans racemic








embedded image


E118





R,R enantiomer








embedded image


E119





trans racemic

















TABLE 4









embedded image


E120







embedded image


E121







embedded image


E122







embedded image


E123







embedded image


E124







embedded image


E125







embedded image


E126







embedded image


E127







embedded image


E128







embedded image


E129







embedded image


E130







embedded image


E131







embedded image


E132







embedded image


E133







embedded image


E134







embedded image


E135







embedded image


E136







embedded image


E137







embedded image


E138







embedded image


E139







embedded image


E140







embedded image


E141







embedded image


E142







embedded image


E143







embedded image


E144







embedded image


E145







embedded image


E146











embedded image


Preparation of methyl 2-phenylcyclopropane-1-carboxylate (S3-2)

To a solution of 2-phenylcyclopropane-1-carboxylic acid (S3-1) in anhydrous DMF at 0° C. was added K2CO3, then methyl iodide. Reaction was stirred at room temperature for 3 hours or until complete by TLC. The mixture was poured over sat NaHCO3 and extracted with EtOAc. The organic layer was washed with brine and then dried over Na2SO4, filtered and concentrated under reduced vacuum. Column chromatography 0-10% hex/EtOAc gave methyl 2-phenylcyclopropane-1-carboxylate as a clear, colorless oil (S3-2, 82%).


Preparation of methyl 2-(4-(chlorosulfonyl)phenyl)cyclopropane-1-carboxylate (S3-3)

To chlorosulfonic acid at 0° C. was added neat methyl 2-phenylcyclopropane-1-carboxylate (S3-2) dropwise over 15 minutes. The reaction was stirred for 3 hours or until complete by TLC. The mixture was poured over ice water and extracted with EtOAc. The organic layer was washed with brine and then dried over Na2SO4, filtered and concentrated under reduced vacuum. Column chromatography 0-15% hex/EtOAc gave methyl 2-(4-(chlorosulfonyl)phenyl)cyclopropane-1-carboxylate as a white solid (S3-3, 79%).


Preparation of methyl 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylate (S3-4)

To a solution of methyl 2-(4-(chlorosulfonyl)phenyl)cyclopropane-1-carboxylate (3) in anhydrous pyridine at 0° C. was added pyridin-2-amine. The reaction was stirred for 1 hour or until complete by TLC. The mixture was poured over sat NaHCO3 and extracted with EtOAc. The organic layer was washed with brine and then dried over Na2SO4, filtered and concentrated under reduced vacuum. Column chromatography 0-5% MeOH/CH2Cl2 gave methyl 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylate as a white solid (S3-4, 74%).


Preparation of 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylic acid (S3-5)

To a solution of 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylate (S3-4) in THF/H2O was added LiOH—H2O and the solution was stirred for 6 hours. The pH was adjusted to 5 with HCl (1N). The aqueous layer was extracted with EtOAc, dried (Na2SO4), filtered and evaporated to give 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylic acid as a white solid (S3-5, 85%).


Preparation of 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)-N-(thiazolo[5,4-c]pyridin-2-yl)cyclopropane-1-carboxamide (S3-6)

To a solution of 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)cyclopropane-1-carboxylic acid (S3-5) in anhydrous pyridine was added EDC, DMAP and thiazolo[5,4-c]pyridin-2-amine and the solution was stirred under N2 overnight. The reaction mixture was concentrated. Column chromatography 0-5% MeOH/CH2Cl2 gave pure 2-(4-(N-(pyridin-2-yl)sulfamoyl)phenyl)-N-(thiazolo[5,4-c]pyridin-2-yl)cyclopropane-1-carboxamide (S3-6, 40%).


Using procedures similar to those set forth above for Scheme 3 and substituting the appropriate starting materials, the compounds E201-E203 (Table 5) were made and E204-218 (Table 6) could be synthesized.












TABLE 5









E201


embedded image









E202


embedded image









E203


embedded image




















TABLE 6









embedded image


E204







embedded image


E205







embedded image


E206







embedded image


E207







embedded image


E208







embedded image


E209







embedded image


E210







embedded image


E211







embedded image


E212







embedded image


E213







embedded image


E214







embedded image


E215







embedded image


E216







embedded image


E217







embedded image


E218







embedded image


E219







embedded image


E220







embedded image


E221







embedded image


E222







embedded image


E223







embedded image


E224









Isomers

Compounds described herein may exist in one or more particular geometric, optical, enantiomeric, diastereomeric, epimeric, atropic, stereoisomer, tautomeric, conformational, or anomeric forms, including but not limited to, cis- and trans-forms; E- and Z-forms; c-, t-, and r-forms; endo- and exo-forms; R-, S-, and meso-forms; D- and L-forms; d- and 1-forms; (+) and (−) forms; keto-, enol-, and enolate-forms; syn- and anti-forms; synclinal- and anticlinal-forms; α- and β-forms; axial and equatorial forms; boat-, chair-, twist-, envelope-, and half chair-forms; and combinations thereof, hereinafter collectively referred to as “isomers” (or “isomeric forms”).


In one embodiment, a compound described herein may be an enantiomerically enriched isomer of a stereoisomer described herein. For example, the compound may have an enantiomeric excess of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. Enantiomer, when used herein, refers to either of a pair of chemical compounds whose molecular structures have a mirror-image relationship to each other.


In one embodiment, a preparation of a compound disclosed herein is enriched for an isomer of the compound having a selected stereochemistry, e.g., R or S, corresponding to a selected stereocenter. For example, the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.


In one embodiment, a composition described herein includes a preparation of a compound disclosed herein that is enriched for a structure or structures having a selected stereochemistry, e.g., R or S, at a selected stereocenter. Exemplary R/S configurations can be those provided in an example described herein.


An “enriched preparation,” as used herein, is enriched for a selected stereoconfiguration of one, two, three or more selected stereocenters within the subject compound. Exemplary selected stereocenters and exemplary stereoconfigurations thereof can be selected from those provided herein, e.g., in an example described herein. By enriched is meant at least 60%, e.g., of the molecules of compound in the preparation have a selected stereochemistry of a selected stereocenter. In an embodiment it is at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. Enriched refers to the level of a subject molecule(s) and does not connote a process limitation unless specified.


Compounds may be prepared in racemic form or as individual enantiomers or diastereomers by either stereospecific synthesis or by resolution. The compounds may, for example, be resolved into their component enantiomers or diastereomers by standard techniques, such as the formation of stereoisomeric pairs by salt formation with an optically active base, followed by fractional crystallization and regeneration of the free acid. The compounds may also be resolved by formation of stereoisomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral chromatography column. The enantiomers also may be obtained from kinetic resolution of the racemate of corresponding esters using lipase enzymes.


Except as discussed below for tautomeric forms, specifically excluded from the term “isomers,” as used herein, are structural (or constitutional) isomers (i.e., isomers which differ in the connections between atoms rather than merely by the position of atoms in space). For example, a reference to a methoxy group, —OCH3, is not to be construed as a reference to its structural isomer, a hydroxymethyl group, —CH2OH. Similarly, a reference to ortho-chlorophenyl is not to be construed as a reference to its structural isomer, meta-chlorophenyl. However, a reference to a class of structures may well include structurally isomeric forms falling within that class (e.g., C3-alkyl or propyl includes n-propyl and iso-propyl; C4-alkyl or butyl includes n-, iso-, sec-, and tert-butyl; methoxyphenyl includes ortho-, meta-, and para-methoxyphenyl).


The above exclusion does not pertain to tautomeric forms, for example, keto-, enol-, and enolate-forms, as in, for example, the following tautomeric pairs: keto/enol, imine/enamine, amide/imino alcohol, amidine/amidine, nitroso/oxime, thioketone/enethiol, N-nitroso/hydroxyazo, and nitro/aci-nitro.


Note that specifically included in the term “isomer” are compounds with one or more isotopic substitutions. For example, H may be in any isotopic form, including 1H, 2H (D), and 3H (T); C may be in any isotopic form, including 12C, 13C, and 14C; O may be in any isotopic form, including 16O and 18O; and the like.


Salts

A compound described herein can be in the form of a salt, e.g., a pharmaceutically acceptable salt. The term “pharmaceutically acceptable salt” includes salts of the active compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. Neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of this disclosure. Examples of pharmaceutically acceptable salts are discussed in Berge et al, 1977, “Pharmaceutically Acceptable Salts.” J. Pharm. Sci. Vol. 66, pp. 1-19. In an embodiment, the compound is present in mono-salt form. In embodiments, the compound is present in di-salt form.


For example, if the compound is anionic, or has a functional group which may be anionic (e.g., —COOH may be —COO), then a salt may be formed with a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Na+ and K+, alkaline earth cations such as Ca2+ and Mg2+, and other cations. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e., NH4+) and substituted ammonium ions (e.g., NH3R1+, NH2R2+, NHR3+, NR4+). Examples of some suitable substituted ammonium ions are those derived from: ethylamine, diethylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as dibasic amino acids, such as lysine and arginine.


If the compound is cationic, or has a functional group that may be cationic (e.g., —NH2 may be —NH3+), then a salt may be formed with a suitable anion. Examples of suitable inorganic anions include, but are not limited to, those derived from the following inorganic acids: hydrochloric, hydrobromic, hydroiodic, sulfuric, sulfurous, nitric, nitrous, phosphoric, and phosphorous.


Examples of suitable organic anions include, but are not limited to, those derived from the following organic acids: 2-acetyoxybenzoic, acetic, ascorbic, aspartic, benzoic, camphorsulfonic, cinnamic, citric, edetic, ethanedisulfonic, ethanesulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, hydroxymaleic, hydroxynaphthalene carboxylic, isethionic, lactic, lactobionic, lauric, maleic, malic, methanesulfonic, mucic, oleic, oxalic, palmitic, pamoic, pantothenic, phenylacetic, phenylsulfonic, propionic, pyruvic, salicylic, stearic, succinic, sulfanilic, tartaric, p-toluenesulfonic, and valeric. Examples of suitable polymeric organic anions include, but are not limited to, those derived from the following polymeric acids: tannic acid, carboxymethyl cellulose.


Unless otherwise specified, a reference to a particular compound also includes salt forms thereof.


Chemically Protected Forms

It may be convenient or desirable to prepare, purify, and/or handle an active compound in a chemically protected form. The term “chemically protected form” is used herein in the conventional chemical sense and pertains to a compound in which one or more reactive functional groups are protected from undesirable chemical reactions under specified conditions (e.g., pH, temperature, radiation, solvent, and the like). In practice, well known chemical methods are employed to reversibly render unreactive a functional group, which otherwise would be reactive, under specified conditions. In a chemically protected form, one or more reactive functional groups are in the form of a protected or protecting group (also known as a masked or masking group or a blocked or blocking group). By protecting a reactive functional group, reactions involving other unprotected reactive functional groups can be performed, without affecting the protected group; the protecting group may be removed, usually in a subsequent step, without substantially affecting the remainder of the molecule. See, for example, Protective Groups in Organic Synthesis (T. Green and P. Wuts; 3rd Edition; John Wiley and Sons, 1999). Unless otherwise specified, a reference to a particular compound also includes chemically protected forms thereof.


A wide variety of such “protecting,” “blocking,” or “masking” methods are widely used and well known in organic synthesis. For example, a compound which has two nonequivalent reactive functional groups, both of which would be reactive under specified conditions, may be derivatized to render one of the functional groups “protected,” and therefore unreactive, under the specified conditions; so protected, the compound may be used as a reactant which has effectively only one reactive functional group. After the desired reaction (involving the other functional group) is complete, the protected group may be “deprotected” to return it to its original functionality.


A hydroxyl group may be protected as an ether (—OR) or an ester (—OC(O)R), for example, as: a t-butyl ether; a benzyl, benzhydryl (diphenylmethyl), or trityl (triphenylmethyl) ether; a trimethylsilyl or t-butyldimethylsilyl ether; or an acetyl ester (—OC(O)CH3, —OAc).


An aldehyde or ketone group may be protected as an acetal (RCH(OR)2) or ketal (R2C(OR)2), respectively, in which the carbonyl group (R2C═O) is converted to a diether (R2C(OR)2), by reaction with, for example, a primary alcohol. The aldehyde or ketone group is readily regenerated by hydrolysis using a large excess of water in the presence of acid.


An amine group may be protected, for example, as an amide (—NRC(O)R) or a urethane (—NRC(O)OR), for example, as: a methyl amide (—NHC(O)CH3); a benzyloxy amide (—NHC(O)OCH2C6H5, —NH-Cbz); as a tert-butoxy amide (—NHC(O)OC(CH3)3, —NH-Boc); a 2-biphenyl-2-propoxy amide (—NHCO(O)C(CH3)2C6H4C6H5, —NH-Bpoc), as a 9-fluorenylmethoxy amide (—NH-Fmoc), as a 6-nitroveratryloxy amide (—NH-Nvoc), as a 2-trimethylsilylethyloxy amide (—NH-Teoc), as a 2,2,2-trichloroethyloxy amide (—NH-Troc), as an allyloxy amide (—NH-Alloc), as a 2(-phenylsulphonyl)ethyloxy amide (—NH-Psec); or, in suitable cases (e.g., cyclic amines), as a nitroxide radical (>N—O«).


A carboxylic acid group may be protected as an ester, for example, as: an alkyl ester (e.g., a methyl ester; a t-butyl ester); a haloalkyl ester (e.g., a haloalkyl ester); a trialkylsilylalkyl ester; or an arylalkyl ester (e.g., a benzyl ester; a nitrobenzyl ester); or as an amide, for example, as a methyl amide.


A thiol group may be protected as a thioether (—SR), for example, as: a benzyl thioether; an acetamidomethyl ether (—S—CH2NHC(O)CH3)


Prodrugs and Other Modifications

In addition to salt forms, the present disclosure may also provide compounds that are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds described herein. Prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present disclosure when placed in a transdermal patch reservoir with or without a suitable enzyme or chemical reagent.


A compound described herein can also be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those that increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism, and/or alter rate of excretion. Examples of these modifications include, but are not limited to, esterification with polyethylene glycols, derivatization with pivolates or fatty acid substituents, conversion to carbamates, hydroxylation of aromatic rings, and heteroatom substitution in aromatic rings.


Synthesis

Methods of synthesizing the herein described compounds will be evident to those of ordinary skill in the art. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.


Methods of Use and Activity

The compounds as disclosed herein and compositions including them have kinase inhibitory activity and are thus useful in modulating the action of kinases, and in treatment and/or prevention of diseases or conditions influenced by kinases. The above compounds and compositions may be used to modulate (e.g., influence or inhibit) the action of kinases either in a cell in vitro or in a cell in a living body in vivo. Specifically, in one embodiment, a method is provided of inhibiting the action of a kinase comprising applying to a medium such as an assay medium or contacting with a cell either in a cell in vitro or in a cell in a living body in vivo an effective inhibitory amount of a compound as disclosed herein. In one embodiment, the kinase inhibited is a rho kinase (e.g., ROCK1 or ROCK2). In another embodiment, the kinase inhibited is a JAK (e.g., JAK2) kinase.


JAK inhibitors are useful in treating various JAK-associated diseases or disorders. Examples of JAK-associated diseases include diseases involving the immune system including, for example, organ transplant rejection (e.g., allograft rejection and graft versus host disease). Further examples of JAK-associated diseases include autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease (COPD), and the like. In some embodiments, the autoimmune disease is arthritis.


Further examples of JAK-associated diseases include allergic conditions such as asthma, food allergies, eczematous dermatitis, contact dermatitis, atopic dermatitis (atropic eczema), and rhinitis. Further examples of JAK-associated diseases include viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV, HTLV 1, Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).


Further examples of JAK-associated diseases or conditions include those characterized by solid tumors (e.g., prostate cancer, renal cancer, hepatic cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, Castleman's disease, uterine leiomyosarcoma, melanoma etc.), hematological cancers (e.g., lymphoma, leukemia such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and cutaneous B-cell lymphoma. Example CTCLs include Sezary syndrome and mycosis fungoides. Other examples of JAK-associated diseases or conditions include pulmonary arterial hypertension.


Other examples of JAK-associated diseases or conditions include inflammation-associated cancers. In some embodiments, the cancer is associated with inflammatory bowel disease. In some embodiments, the inflammatory bowel disease is ulcerative colitis. In some embodiments, the inflammatory bowel disease is Crohn's disease. In some embodiments, the inflammation-associated cancer is colitis-associated cancer. In some embodiments, the inflammation-associated cancer is colon cancer or colorectal cancer. In some embodiments, the cancer is gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small intestine cancer, or rectal cancer. All of these indications are also indications for ROCK inhibitiors.


The compounds of the present disclosure are used in methods of inhibiting kinases in a cell, a tissue or a subject such as a human comprising contacting the cell with an amount of one or more of the compounds of the present disclosure effective to inhibit the kinase. In one embodiment, the compounds are administered in a pharmaceutically acceptable composition, such as in or with a pharmaceutically acceptable carrier.


In another embodiment, the compounds of the present disclosure are used in methods for modulating the action of a kinase in a cell comprising contacting the cell with amount of one or more compounds of the present disclosure effective to modulate the action of a kinase in a cell. In one embodiment, the compounds of the present disclosure are administered in a pharmaceutically acceptable composition, such as in or with a pharmaceutically acceptable carrier.


Treatment or prevention of diseases or conditions for which the compounds of the present disclosure may be useful includes any of the diseases or conditions associated with kinase activity or diseases or conditions affected by kinases. Examples of these types of diseases include neurodegenerative diseases, such as Alzheimer's; ocular diseases, such as diabetic eye diseases, wet age-related macular degeneration, or dry age-related macular degeneration, inflammatory eye diseases, retinal degradation and glaucoma; cardiovascular diseases; and cancer. Additional examples include bone disorder, obesity, hepatic disease, renal disease, pancreatitis, gastric disturbance, hypertension, fertility control, disorders of hair growth, nasal congestion, neurogenic bladder disorder, gastrointestinal disorder, dermatological disorder, and respiratory indications.


In some embodiments, the compounds of the present disclosure will be administered in conjunction with one or more additional therapeutic agents. Suitable classes of additional therapeutic agents include, but are not limited to, beta blockers, alpha-agonists, carbonic anhydrase inhibitors, prostaglandin-like compounds, miotic or cholinergic agents, epinephrine compounds, or neuroprotective or anti-inflammatory compounds.


Beta blockers. These compounds are thought to lower intraocular pressure (IOP) by reducing the production of aqueous humor. Examples include levobunolol (BETAGAN™), timolol (BETIMOL™, TIMOPTIC™), betaxolol (BETOPTIC™) and metipranolol (OPTIPRANOLOL™).


Alpha-agonists. These compounds are thought to lower IOP by reducing the production of aqueous humor and increasing drainage. Examples include apraclonidine (IOPIDINE™) and brimonidine (ALPHAGAN™).


Carbonic anhydrase inhibitors. These compounds are thought to lower IOP by also reducing the production of aqueous humor. Examples include dorzolamide (TRUSOPT™) and brinzolamide (AZOPT™).


Prostaglandin-like compounds. These compounds are thought to lower IOP by increasing the outflow of aqueous humor by the uveoscleral pathway. Examples include AR-102, latanoprost (XALATAN™), bimatoprost (LUMIGAN™), tafluprost (ZIOPTAN™), and travoprost (TRAVATAN™).


Miotic or cholinergic agents. These agents are thought to function by causing the pupil to constrict, which opens drainage channels in the eye. Examples include pilocarpine (ISOPTO CARPINE™, PILOPINE™) and carbachol (ISOPTO CARBACHOL™).


Epinephrine compounds. These compounds, such as dipivefrin (PROPINE™), are thought to function by both decreasing the outflow of aqueous humor, as well as increasing fluid drainage.


Neuroprotective or anti-inflammatory compounds. These compounds, such as Aflibercept (EYLEA™) are treatments for conditions of the retina such as Macular Degeneration, and are designed as anti-VEGF treatments or have similar types of anti-growth or anti-inflammatory activity.


Thus, provided herein are methods of treating an ocular disorder in a subject in need thereof, comprising administering to the subject a compound, a composition, or a pharmaceutical composition provided herein.


Also provided herein are methods of reducing intraocular pressure in a subject in need thereof, comprising administering to the subject a compound, a composition, or a pharmaceutical composition provided herein.


In one aspect, provided herein are methods of treating an ocular disorder in a subject in need thereof, comprising administering to the subject a compound, or a pharmaceutically acceptable salt thereof, provided herein.


In some embodiments, the ocular disorder is glaucoma.


In another aspect, provided herein are methods of reducing intraocular pressure in a subject in need thereof, comprising administering to the subject a compound, or a pharmaceutically acceptable salt thereof, provided herein.


In some embodiments of these aspects, the compound is administered topically to an eye of the subject.


In some embodiments, provided herein are methods of treating an ocular disorder in a subject in need thereof, comprising administering to the subject a compound of one of the Formulae provided herein, or a pharmaceutically acceptable salt thereof.


In some embodiments, provided herein are methods of treating an ocular disorder in a subject in need thereof, comprising administering to the subject a compound provided in Table 1, or a pharmaceutically acceptable salt thereof.


In some embodiments, provided herein are methods of reducing intraocular pressure in a subject in need thereof, comprising administering to the subject a compound of one of the Formulae provided herein, or a pharmaceutically acceptable salt thereof.


In some embodiments, provided herein are methods of reducing intraocular pressure in a subject in need thereof, comprising administering to the subject a compound provided in Table 1, or a pharmaceutically acceptable salt thereof.


In some embodiments of these methods, the method further comprises administering one or more additional therapeutic agents. In some embodiments, the one or more additional therapeutic agents is a beta blocker, an alpha-agonist, a carbonic anhydrase inhibitor, a prostaglandin or a prostaglandin-like compound, a miotic or cholinergic agent, an epinephrine compound, or a neuroprotective or anti-inflammatory compound. In some embodiments, the one or more additional therapeutic agents is a prostaglandin or a prostaglandin-like compound. In some embodiment, the prostaglandin-like compound is AR-102, latanoprost, bimatoprost, tafluprost, or travoprost.


Also provided herein are methods of treating an autoimmune disease in a subject in need thereof, comprising administering to the subject a compound, a composition, or a pharmaceutical composition provided herein.


In some embodiments, provided herein are methods of treating an autoimmune disease in a subject in need thereof, comprising administering to the subject a compound of one of the Formulae provided herein, or a pharmaceutically acceptable salt thereof.


In some embodiments, provided herein are methods of treating an autoimmune disease in a subject in need thereof, comprising administering to the subject a compound provided in Table 1, or a pharmaceutically acceptable salt thereof.


In some embodiments, the autoimmune disease is multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, or chronic obstructive pulmonary disease.


Compositions and Administration

The additional therapeutic agent or agents can be administered simultaneously or sequentially with the compounds of the present disclosure. Sequential administration includes administration before or after the compounds of the present disclosure. In some embodiments, the additional therapeutic agent or agents can be administered in the same composition as the compounds of the present disclosure. In other embodiments, there can be an interval of time between administration of the additional therapeutic agent and the compounds of the present disclosure.


In some embodiments, the administration of an additional therapeutic agent with a compound of the present disclosure will enable lower doses of the other therapeutic agents to be administered for a longer period of time.


Also provided herein are compositions comprising a compound provided herein, or a pharmaceutically acceptable salt thereof. In one embodiment, the compositions provided herein are pharmaceutical compositions comprising a pharmaceutically acceptable carrier.


Pharmaceutical compositions for use in accordance with the present disclosure may be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. Thus, the compounds and their physiologically acceptable salts and solvates may be formulated for administration by, for example, solid dosing, eyedrop, in a topical oil-based formulation, injection (including injection of a drug-eluting device either into the body as a whole, or into specific tissues of the eye), inhalation (either through the mouth or the nose), implants, or oral, buccal, parenteral or rectal administration. Techniques and formulations may generally be found in “Remington's Pharmaceutical Sciences,” (Meade Publishing Co., Easton, Pa.).


The route by which the compounds of the present disclosure (component A) will be administered and the form of the composition will dictate the type of carrier (component B) to be used. The composition may be in a variety of forms, suitable, for example, for systemic administration (e.g., oral, rectal, nasal, sublingual, buccal, implants, or parenteral, or by ocular injection into one of the chambers of the eye, such as intravitreal injection, intracameral injection, or injection into the aqueous humour.) or topical administration (e.g., local application on the skin, ocular, liposome delivery systems, or iontophoresis).


Carriers for systemic administration typically comprise at least one of a) diluents, b) lubricants, c) binders, d) disintegrants, e) colorants, f) flavors, g) sweeteners, h) antioxidants, j) preservatives, k) glidants, m) solvents, n) suspending agents, o) wetting agents, p) surfactants, combinations thereof, and others. All carriers are optional in the systemic compositions.


Ingredient a) is a diluent. Suitable diluents for solid dosage forms include sugars such as glucose, lactose, dextrose, and sucrose; diols such as propylene glycol; calcium carbonate; sodium carbonate; sugar alcohols, such as glycerin; mannitol; and sorbitol. The amount of ingredient a) in the systemic or topical composition is typically about 50 to about 90%.


Ingredient b) is a lubricant. Suitable lubricants for solid dosage forms are exemplified by solid lubricants including silica, talc, stearic acid and its magnesium salts and calcium salts, calcium sulfate; and liquid lubricants such as polyethylene glycol and vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil of Theobroma. The amount of ingredient b) in the systemic or topical composition is typically about 5 to about 10%.


Ingredient c) is a binder. Suitable binders for solid dosage forms include polyvinyl pyrrolidone; magnesium aluminum silicate; starches such as corn starch and potato starch; gelatin; tragacanth; and cellulose and its derivatives, such as sodium carboxymethylcellulose, ethyl cellulose, methylcellulose, microcrystalline cellulose, and sodium carboxymethylcellulose. The amount of ingredient c) in the systemic composition is typically about 5 to about 50%, and in ocular solid dosing forms up to 99%.


Ingredient d) is a disintegrant. Suitable disintegrants for solid dosage forms include agar, alginic acid and the sodium salt thereof, effervescent mixtures, croscarmelose, crospovidone, sodium carboxymethyl starch, sodium starch glycolate, clays, and ion exchange resins. The amount of ingredient d) in the systemic or topical composition is typically about 0.1 to about 10%.


Ingredient e) for solid dosage forms is a colorant such as an FD&C dye. When used, the amount of ingredient e) in the systemic or topical composition is typically about 0.005 to about 0.1%.


Ingredient f) for solid dosage forms is a flavor such as menthol, peppermint, and fruit flavors. The amount of ingredient f), when used, in the systemic or topical composition is typically about 0.1 to about 1.0%.


Ingredient g) for solid dosage forms is a sweetener such as aspartame and saccharin. The amount of ingredient g) in the systemic or topical composition is typically about 0.001 to about 1%.


Ingredient h) is an antioxidant such as butylated hydroxyanisole (“BHA”), butylated hydroxytoluene (“BHT”), and vitamin E. The amount of ingredient h) in the systemic or topical composition is typically about 0.1 to about 5%.


Ingredient j) is a preservative such as benzalkonium chloride, methyl paraben and sodium benzoate. The amount of ingredient j) in the systemic or topical composition is typically about 0.01 to about 5%.


Ingredient k) for solid dosage forms is a glidant such as silicon dioxide. The amount of ingredient k) in the systemic or topical composition is typically about 1 to about 5%.


Ingredient m) is a solvent, such as water, isotonic saline, ethyl oleate, glycerine, hydroxylated castor oils, alcohols such as ethanol, and phosphate buffer solutions. The amount of ingredient m) in the systemic or topical composition is typically from about 0 to about 100%.


Ingredient n) is a suspending agent. Suitable suspending agents include Avicel® RC-591 (from FMC Corporation of Philadelphia, Pa.) and sodium alginate. The amount of ingredient n) in the systemic or topical composition is typically about 1 to about 8%.


Ingredient o) is a surfactant such as lecithin, Polysorbate 80, and sodium lauryl sulfate, and the TWEENS® from Atlas Powder Company of Wilmington, Del. Suitable surfactants include those disclosed in the C.T.F.A. Cosmetic Ingredient Handbook, 1992, pp. 587-592; Remington's Pharmaceutical Sciences, 15th Ed. 1975, pp. 335-337; and McCutcheon's Volume 1, Emulsifiers & Detergents, 1994, North American Edition, pp. 236-239. The amount of ingredient o) in the systemic or topical composition is typically about 0.1% to about 5%.


Although the amounts of components A and B in the systemic compositions will vary depending on the type of systemic composition prepared, the specific derivative selected for component A and the ingredients of component B, in general, system compositions comprise 0.01% to 50% of component A and 50 to 99.99% of component B.


Compositions for parenteral administration typically comprise A) 0.1 to 10% of the compounds of the present disclosure and B) 90 to 99.9% of a carrier comprising a) a diluent and m) a solvent. In one embodiment, component a) comprises propylene glycol and m) comprises ethanol or ethyl oleate.


Compositions for oral administration can have various dosage forms. For example, solid forms include tablets, capsules, granules, and bulk powders. These oral dosage forms comprise a safe and effective amount, usually at least about 5%, and more particularly from about 25% to about 50% of component A). The oral dosage compositions further comprise about 50 to about 95% of component B), and more particularly, from about 50 to about 75%.


Tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, or multiple-compressed. Tablets typically comprise component A, and component B a carrier comprising ingredients selected from the group consisting of a) diluents, b) lubricants, c) binders, d) disintegrants, e) colorants, f) flavors, g) sweeteners, k) glidants, and combinations thereof. Specific diluents include calcium carbonate, sodium carbonate, mannitol, lactose and cellulose. Specific binders include starch, gelatin, and sucrose. Specific disintegrants include alginic acid and croscarmelose. Specific lubricants include magnesium stearate, stearic acid, and talc. Specific colorants are the FD&C dyes, which can be added for appearance. Chewable tablets preferably contain g) sweeteners such as aspartame and saccharin, or f) flavors such as menthol, peppermint, fruit flavors, or a combination thereof.


Capsules (including implants, time release and sustained release formulations) typically comprise component A, and a carrier comprising one or more a) diluents disclosed above in a capsule comprising gelatin. Granules typically comprise component A, and preferably further comprise k) glidants such as silicon dioxide to improve flow characteristics. Implants can be of the biodegradable or the non-biodegradable type. Implants may be prepared using any known biocompatible formulation.


The selection of ingredients in the carrier for oral compositions depends on secondary considerations like taste, cost, and shelf stability, which are not critical for the purposes of this disclosure. One skilled in the art would know how to select appropriate ingredients without undue experimentation.


The solid compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that component A is released in the gastrointestinal tract in the vicinity of the desired application, or at various points and times to extend the desired action. The coatings typically comprise one or more components selected from the group consisting of cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropyl methyl cellulose phthalate, ethyl cellulose, EUDRAGIT® coatings (available from Rohm & Haas G.M.B.H. of Darmstadt, Germany), waxes and shellac.


Compositions for oral administration can also have liquid forms. For example, suitable liquid forms include aqueous solutions, emulsions, suspensions, solutions reconstituted from non-effervescent granules, suspensions reconstituted from non-effervescent granules, effervescent preparations reconstituted from effervescent granules, elixirs, tinctures, syrups, and the like. Liquid orally administered compositions typically comprise component A and component B, namely, a carrier comprising ingredients selected from the group consisting of a) diluents, e) colorants, f) flavors, g) sweeteners, j) preservatives, m) solvents, n) suspending agents, and o) surfactants. Peroral liquid compositions preferably comprise one or more ingredients selected from the group consisting of e) colorants, f) flavors, and g) sweeteners.


Other compositions useful for attaining systemic delivery of the subject compounds include injection, sublingual, buccal and nasal dosage forms. Such compositions typically comprise one or more of soluble filler substances such as a) diluents including sucrose, sorbitol and mannitol; and c) binders such as acacia, microcrystalline cellulose, carboxymethyl cellulose, and hydroxypropyl methylcellulose. Such compositions may further comprise b) lubricants, e) colorants, f) flavors, g) sweeteners, h) antioxidants, and k) glidants.


In one embodiment of the disclosure, the compounds of the present disclosure are topically administered. Topical compositions that can be applied locally to the eye may be in any form known in the art, non-limiting Examples of which include solids, gelable drops, sprays, ointments, or a sustained or non-sustained release unit placed in the conjunctival cul-du-sac of the eye or another appropriate location.


Topical compositions that can be applied locally to the skin may be in any form including solids, solutions, oils, creams, ointments, gels, lotions, shampoos, leave-on and rinse-out hair conditioners, milks, cleansers, moisturizers, sprays, skin patches, and the like. Topical compositions comprise: component A, the compounds described above, and component B, a carrier. The carrier of the topical composition preferably aids penetration of the compounds into the eye. Component B may further comprise one or more optional components.


An effective amount of a compound according to the present disclosure will vary with the particular condition being treated, the age and physical condition of the patient being treated, the severity of the condition, the duration of treatment, the nature of concurrent therapy, the route of administration, the particular pharmaceutically-acceptable carrier utilized, and like factors within the knowledge and expertise of the attending physician. For example, an effective amount of the compounds of the present disclosure for systemic administration is from about 0.01 to about 1000 μg/kg body weight, preferably from about 0.1 to about 100 μg/kg per body weight, most preferably form about 1 to about 50 μg/kg body weight per day. The transdermal dosages will be designed to attain similar serum or plasma levels, based upon techniques known to those skilled in the art of pharmacokinetics and transdermal formulations. Plasma levels for systemic administration are expected to be in the range of 0.01 to 100 ng/mL, more preferably from 0.05 to 50 ng/mL and most preferably from 0.1 to 10 ng/mL. While these dosages are based upon a daily administration rate, the compounds of the present disclosure may also be administered at other intervals, such as twice per day, twice weekly, once weekly, or once a month. One of ordinary skill in the art would be able to calculate suitable effective amounts for other intervals of administration.


The compounds of the present disclosure are useful in a method of reducing or decreasing intraocular pressure. The compounds of the present disclosure may be administered to a subject in need of treatment in an amount effective to reduce intraocular pressure. Thus, these compounds are useful in the treatment of glaucoma. The preferred route of administration for treating glaucoma is topically.


The exact amounts of each component in the topical composition depend on various factors. The amount of component A added to the topical composition is dependent on the IC50 of component A, typically expressed in nanomolar (nM) units. For example, if the IC50 of the medicament is 1 nM, the amount of component A will be from about 0.001 to about 0.3%. If the IC50 of the medicament is 10 nM, the amount of component A) will be from about 0.01 to about 1%. If the IC50 of the medicament is 100 nM, the amount of component A will be from about 0.1 to about 10%. If the amount of component A is outside the ranges specified above (i.e., lower), efficacy of the treatment may be reduced. One skilled in the art understands how to calculate and understand an IC50. The remainder of the composition, up to 100%, is component B.


The amount of the carrier employed in conjunction with component A is sufficient to provide a practical quantity of composition for administration per unit dose of the medicament. Techniques and compositions for making dosage forms useful in the methods of this disclosure are described in the following references: Modern Pharmaceutics, Chapters 9 and 10, Banker & Rhodes, eds. (1979); Lieberman et al., Pharmaceutical Dosage Forms: Tablets (1981); and Ansel, Introduction to Pharmaceutical Dosage Forms, 2nd Ed., (1976).


Component B may comprise a single ingredient or a combination of two or more ingredients. In the topical compositions, component B comprises a topical carrier. Suitable topical carriers comprise one or more ingredients selected from the group consisting of phosphate buffered saline, isotonic water, deionized water, monofunctional alcohols, symmetrical alcohols, aloe vera gel, allantoin, glycerin, vitamin A and E oils, mineral oil, propylene glycol, PPG-2 myristyl propionate, dimethyl isosorbide, castor oil, combinations thereof, and the like. More particularly, carriers for skin applications include propylene glycol, dimethyl isosorbide, and water, and even more particularly, phosphate buffered saline, isotonic water, deionized water, monofunctional alcohols and symmetrical alcohols.


The carrier of the topical composition may further comprise one or more ingredients selected from the group consisting of q) emollients, r) propellants, s) solvents, t) humectants, u) thickeners, v) powders, w) fragrances, x) pigments, and y) preservatives.


Ingredient q) is an emollient. The amount of ingredient q) in a skin-based topical composition is typically about 5 to about 95%. Suitable emollients include stearyl alcohol, glyceryl monoricinoleate, glyceryl monostearate, propane-1,2-diol, butane-1,3-diol, mink oil, cetyl alcohol, isopropyl isostearate, stearic acid, isobutyl palmitate, isocetyl stearate, oleyl alcohol, isopropyl laurate, hexyl laurate, decyl oleate, octadecan-2-ol, isocetyl alcohol, cetyl palmitate, di-n-butyl sebacate, isopropyl myristate, isopropyl palmitate, isopropyl stearate, butyl stearate, polyethylene glycol, triethylene glycol, lanolin, sesame oil, coconut oil, arachis oil, castor oil, acetylated lanolin alcohols, petroleum, mineral oil, butyl myristate, isostearic acid, palmitic acid, isopropyl linoleate, lauryl lactate, myristyl lactate, decyl oleate, myristyl myristate, and combinations thereof. Specific emollients for skin include stearyl alcohol and polydimethylsiloxane.


Ingredient r) is a propellant. The amount of ingredient r) in the topical composition is typically about 0 to about 95%. Suitable propellants include propane, butane, isobutane, dimethyl ether, carbon dioxide, nitrous oxide, and combinations thereof.


Ingredient s) is a solvent. The amount of ingredient s) in the topical composition is typically about 0 to about 95%. Suitable solvents include water, ethyl alcohol, methylene chloride, isopropanol, castor oil, ethylene glycol monoethyl ether, diethylene glycol monobutyl ether, diethylene glycol monoethyl ether, dimethylsulfoxide, dimethyl formamide, tetrahydrofuran, and combinations thereof. Specific solvents include ethyl alcohol and homotopic alcohols.


Ingredient t) is a humectant. The amount of ingredient t) in the topical composition is typically 0 to 95%. Suitable humectants include glycerin, sorbitol, sodium 2-pyrrolidone-5-carboxylate, soluble collagen, dibutyl phthalate, gelatin, and combinations thereof. Specific humectants include glycerin.


Ingredient u) is a thickener. The amount of ingredient u) in the topical composition is typically about 0 to about 95%.


Ingredient v) is a powder. The amount of ingredient v) in the topical composition is typically 0 to 95%. Suitable powders include beta-cyclodextrins, hydroxypropyl cyclodextrins, chalk, talc, fullers earth, kaolin, starch, gums, colloidal silicon dioxide, sodium polyacrylate, tetra alkyl ammonium smectites, trialkyl aryl ammonium smectites, chemically-modified magnesium aluminum silicate, organically-modified Montmorillonite clay, hydrated aluminum silicate, fumed silica, carboxyvinyl polymer, sodium carboxymethyl cellulose, ethylene glycol monostearate, and combinations thereof. For ocular applications, specific powders include beta-cyclodextrin, hydroxypropyl cyclodextrin, and sodium polyacrylate. For gel dosing ocular formulations, sodium polyacrylate may be used.


Ingredient w) is a fragrance. The amount of ingredient w) in the topical composition is typically about 0 to about 1.5%, particularly, about 0.001 to about 0.1%. For ocular applications a fragrance is not typically used.


Ingredient x) is a pigment. Suitable pigments for skin applications include inorganic pigments, organic lake pigments, pearlescent pigments, and mixtures thereof. Inorganic pigments useful in this disclosure include those selected from the group consisting of rutile or anatase titanium dioxide, coded in the Color Index under the reference CI 77,891; black, yellow, red and brown iron oxides, coded under references CI 77,499, 77,492 and, 77,491; manganese violet (CI 77,742); ultramarine blue (CI 77,007); chromium oxide (CI 77,288); chromium hydrate (CI 77,289); and ferric blue (CI 77,510) and mixtures thereof.


The organic pigments and lakes useful in this disclosure include those selected from the group consisting of D&C Red No. 19 (CI 45,170), D&C Red No. 9 (CI 15,585), D&C Red No. 21 (CI 45,380), D&C Orange No. 4 (CI 15,510), D&C Orange No. 5 (CI 45,370), D&C Red No. 27 (CI 45,410), D&C Red No. 13 (CI 15,630), D&C Red No. 7 (CI 15,850), D&C Red No. 6 (CI 15,850), D&C Yellow No. 5 (CI 19,140), D&C Red No. 36 (CI 12,085), D&C Orange No. 10 (CI 45,425), D&C Yellow No. 6 (CI 15,985), D&C Red No. 30 (CI 73,360), D&C Red No. 3 (CI 45,430), the dye or lakes based on Cochineal Carmine (CI 75,570) and mixtures thereof.


The pearlescent pigments useful in this disclosure include those selected from the group consisting of the white pearlescent pigments such as mica coated with titanium oxide, bismuth oxychloride, colored pearlescent pigments such as titanium mica with iron oxides, titanium mica with ferric blue, chromium oxide and the like, titanium mica with an organic pigment of the above-mentioned type as well as those based on bismuth oxychloride and mixtures thereof. The amount of pigment in the topical composition is typically about 0 to about 10%. For ocular applications a pigment is generally not used.


In a particularly preferred embodiment of the disclosure, topical pharmaceutical compositions for ocular administration are prepared typically comprising component A and B (a carrier), such as purified water, and one or more ingredients selected from the group consisting of y) sugars or sugar alcohols such as dextrans, particularly mannitol and dextran 70, z) cellulose or a derivative thereof, aa) a salt, bb) disodium EDTA (Edetate disodium), and cc) a pH adjusting additive.


Examples of z) cellulose derivatives suitable for use in the topical pharmaceutical composition for ocular administration include sodium carboxymethylcellulose, ethylcellulose, methylcellulose, and hydroxypropyl-methylcellulose, particularly, hydroxypropyl-methylcellulose.


Examples of aa) salts suitable for use in the topical pharmaceutical composition for ocular administration include mono-, di- and trisodium phosphate, sodium chloride, potassium chloride, and combinations thereof.


Examples of cc) pH adjusting additives include HCl or NaOH in amounts sufficient to adjust the pH of the topical pharmaceutical composition for ocular administration to the range of 4.5-7.5 pH units.


Component A may be included in kits comprising a compound as described herein, a systemic or topical composition described above, or both; and information, instructions, or both that use of the kit will provide treatment for cosmetic and medical conditions in mammals (particularly humans). The information and instructions may be in the form of words, pictures, or both, and the like. In addition or in the alternative, the kit may comprise the medicament, a composition, or both; and information, instructions, or both, regarding methods of application of medicament, or of composition, preferably with the benefit of treating or preventing cosmetic and medical conditions in mammals (e.g., humans).


The disclosure will be further explained by the following illustrative Examples that are to be considered to be non-limiting.


EXAMPLES
Example 1: ROCK and JAK Assays
ROCK Kinase Inhibition Assays.

All compounds were initially prepared as 10 mM stocks in anhydrous dimethylsulfoxide (DMSO). A 20 μL aliquot of the 10 mM solutions was transferred to individual wells in column 1 of a 96-well polypropylene microtiter plate (Corning #3363) and diluted with DMSO to give a final compound concentration of 4 mM. Test compounds were then serially diluted 1:5 in DMSO for an 11-point concentration response and further diluted in the assay buffer bringing all compound concentrations to a final range of 100 μM to 10 μM in 2.5% DMSO. The assay was performed in white 96-well, flat-bottom, half-area, non-binding assay plate (Corning #3642) in assay buffer consisting of 20 mM HEPES (pH 7.5), 10 mM MgCl2*6H2O, 100 μM sodium orthovanadate, 0.05% CHAPS and 0.1% bovine serum albumin. A 10 μL aliquot of compound from each well of the intermediate dilution plate and 20 μL of a 2X substrate/enzyme solution containing acceptor substrate (800 nM RSK2 peptide KRRRLSSLRA (SEQ ID NO: 1)), ROCK2 enzyme (10 nM), or ROCK1 enzyme, and 1,4-Dithiothreitol (DTT, 2 uM) were added to all wells. The reaction was initiated by the addition of 10 μL of 4x stock solution ATP (2 μM). Reactions were thoroughly mixed manually, covered and allowed to incubate at room temperature for 75 min. Protein kinase activity was quantitated using Promega's KINASE-GLO™ luminescent Kinase Assay Kit according to the manufacturer's directions. ATP concentrations remaining in Test wells following the termination of the enzymatic reaction were compared against control wells containing equivalent amounts of DMSO containing no inhibitor (CTRL). ATP concentrations in both Test wells and CTRL wells were normalized against background (BKG) ATP concentrations in wells containing concentrations of inhibitor that completely inhibited the protein kinase under investigation (i.e. a concentration that prevented any consumption of ATP over the course of the incubation). Percent of Control (POC) values were determined for each concentration of compound tested according to the equation:





POC=((Test well value−BKG)/(CTRL−BKG))*100


IC50 values were calculated using the following 4-parameter logistic curve-fitting algorithm:






f(x)=(A+((B−A)/(1+((x/C){circumflex over ( )}D))))


IC50 values were converted to Ki values using the following Cheng-Prusoff Equation:






K
i
=IC
50/(1+([ATP]/Km ATP])).


JAK Kinase Assays.

Compounds were prepared in the exact same manner as described in the ROCK Kinase Assay with the exception to the substrate and enzyme. The JAK 2X substrate/enzyme solution consisted of acceptor substrate (800 nM Abl peptide EAIYAAPFAKKK (SEQ ID NO:2)), JAK2 or JAK3 enzyme (10 nM) and DTT (2 uM). All other steps and solutions remain identical to the ROCK Kinase Assay above. Results are shown above in Table 1.


Example 2. PTM-HTM Assay

Porcine Trabecular Meshwork cells (PTM) were isolated from freshly obtained enucleated porcine eyes. Immortalized Human Trabecular Meshwork cells (TM-1) were obtained through a kind gift from Donna Peters in the Department of Ophthalmology and Visual Sciences at the University of Wisconsin. Cells were plated onto fibronectin coated glass-bottom 96-well plates and allowed to attach overnight. Media was removed and replaced with test compound in media with 1% fetal bovine serum and incubated for various times. After incubation, cells were formaldehyde fixed, triton solubilized, and stained. PTM cells were stained with Alexa Fluor® 488 phalloidin (F-actin) and Hoechst 33342 (nuclei). TM-1 cells were stained with anti-paxillin followed by Alexa Fluor® 488 goat-anti-mouse IgG (focal adhesions) and Hoechst 33342 (nuclei). All staining reagents were obtained through Invitrogen. Images were collected on an INCell 2200 imager with a 20X objective. The actin fiber length and total area of focal adhesions were analyzed using custom algorithms developed in the INCell Developer Toolbox, v1.9.3. Data collected were converted to percent of control (untreated cells). Curves were fit to data in GraphPad Prizm using sigmoidal dose-response and constraining top and bottom to 100% and 0%, respectively.


Example 3

Topical pharmaceutical compositions for lowering intraocular pressure are prepared by conventional methods and formulated as shown in Table 7.











TABLE 7






Ingredient
Amount (wt %)


















heterobicyclic cyclopropanecarboxamide
0.50



Dextran 70
0.1



Hydroxypropyl methylcellulose
0.3



Sodium Chloride
0.77



Potassium chloride
0.12



Disodium EDTA
0.05



Benzalkonium chloride
0.01



HCl and/or NaOH
pH 4.5-6.5



Purified water
q.s. to 100%









A compound provided herein is used as the heterobicyclic cyclopropanecarboxamide. When the composition is topically administered to the eyes once daily, the above composition decreases intraocular pressure in a subject suffering from glaucoma.


Example 4
Reference Example One. Pharmacological Activity for Glaucoma Assay

Pharmacological activity for glaucoma can also be demonstrated using assays designed to test the ability of the subject compounds to decrease intraocular pressure. Examples of such assays are described in the following reference, incorporated herein by reference: C. Liljebris, G. Selen, B. Resul, J. Sternschantz, and U. Hacksell, “Derivatives of 17-phenyl-18, 19, 20-trinorprostaglandin FIsopropyl Ester: Potential Anti-glaucoma Agents”, Journal of Medicinal Chemistry 1995, 38 (2): 289-304.


While the disclosure has been described in detail and with reference to specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made without departing from the spirit and scope of the disclosure.


Groupings of alternative elements or embodiments disclosed herein may be referred to and claimed individually or in any combination with other members of the group or other elements found herein.


The contents of all references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art.


Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims
  • 1. A compound of the formula:
  • 2. A compound as in claim 1, wherein: R1 is H, F, Cl, Br, CN, NH2, OH, OCF3, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl;R2 is H, F, or C1-3 alkyl; orR9 is H, F, or C1-3 alkyl.
  • 3. A compound as in claim 1 or 2, wherein the compound is of the formula:
  • 4. A compound as in claim 1 or 2, wherein the compound is of the formula:
  • 5. A compound as in claim 1 or 2, wherein the compound is of the formula:
  • 6. A compound as in one of claims 1-5, wherein R4 is S.
  • 7. A compound as in one of claims 1-5, wherein R4 is —C(H)═C(H)—.
  • 8. A compound as in one of claims 1-5, wherein R4 is —C(H)═N—.
  • 9. A compound as in one of claims 1-8, wherein R6 is CO.
  • 10. A compound as in one of claims 1-8, wherein R6 is SO2.
  • 11. A compound as in one of claims 1-10, wherein R7 is a bond.
  • 12. A compound as in one of claims 1-10, wherein R7 is a CH2.
  • 13. A compound as in one of claims 1-12, wherein R1 is H, Cl or Me, R5 is H, or R9 is H.
  • 14. A compound as in one of claims 1-13, wherein R8 is phenyl.
  • 15. A compound as in one of claims 1-13, wherein R8 is a monocyclic C3-5 heteroaryl having 1-3 heteroatoms, wherein each heteroatom is, independently, N, O, or S.
  • 16. A compound as in one of claims 1-13, wherein R8 is a monocyclic C3-5 heteroaryl having 1-3 nitrogen atoms.
  • 17. A compound as in claim 1, wherein:
  • 18. A compound as in claim 1, wherein:
  • 19. A compound as in claim 1, wherein the compound is:
  • 20. A compound as in claim 1, wherein the compound is of the formula:
  • 21. A compound as in any of claims 1, 6-8, or 11-16, wherein the compound is selected from:
  • 22. A compound as in claim 1, wherein: R1 is H, F, Cl, Br, I, CN, NH2, OH, OCF3, OCH2CF3, C1-6 alkyl, C3-6 cycloalkyl, or C1-6 haloalkyl;R2 is H, F, Cl, Br, I, C1-6 alkyl, or C1-6 haloalkyl;R3 is
  • 23. A compound of the formula:
  • 24. A compound of the formula:
  • 25. A composition, comprising the compound as in one of claims 1-24.
  • 26. A composition as in claim 25, wherein the composition is a pharmaceutical composition.
  • 27. A composition as in claim 26, wherein the composition further includes a pharmaceutically acceptable excipient.
  • 28. A method of treating an ocular disorder in a subject in need thereof, comprising administering to the subject a compound or composition as in one of claims 1-26.
  • 29. A method as in claim 28, wherein the ocular disorder is glaucoma.
  • 30. A method of reducing intraocular pressure in a subject in need thereof, comprising administering to the subject a compound or composition as in one of claims 1-26.
  • 31. A method of treating an autoimmune disease in a subject in need thereof, comprising administering to the subject a compound or composition as in one of claims 1-26.
  • 32. A method as in claim 31, wherein the autoimmune disease is multiple sclerosis, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis, autoimmune thyroid disorders, chronic obstructive pulmonary disease, or a combination thereof.
  • 33. A method of treating a disease or condition associated with kinase activity or diseases or conditions affected by kinases, comprising administering to the subject a compound or composition as in one of claims 1-26.
  • 34. A method as in claim 33, wherein the disease is a neurodegenerative disease, an ocular disease, a cardiovascular disease, or a cancer.
  • 35. A method as in one of claims 28-34, wherein the administering is topically to an eye of the subject.
  • 36. A kit, including a compound or composition as in one of claims 1-26, and instructions for use.
  • 37. A compound as described herein.
  • 38. A method as described herein.
SEQUENCE LISTING

This application contains a sequence listing having the filename 1959002-00266_ST25.txt, which is 655 bytes in size, and was created on Sep. 8, 2020. The entire content of this sequence listing is herein incorporated by reference. This application is a national stage entry under 35 USC § 371 of international application No. PCT/US2020/050554, filed Sep. 11, 2020, which claims priority of U.S. Provisional Patent Application No. 62/900,263, filed Sep. 13, 2019, the entire content of which is herein incorporated by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/050554 9/11/2020 WO
Provisional Applications (1)
Number Date Country
62900263 Sep 2019 US