WO2012078519A2 - 3-acylidene-2-oxoindole derivatives for inhibition of transglutaminase 2 - Google Patents

3-acylidene-2-oxoindole derivatives for inhibition of transglutaminase 2 Download PDF

Info

Publication number
WO2012078519A2
WO2012078519A2 PCT/US2011/063312 US2011063312W WO2012078519A2 WO 2012078519 A2 WO2012078519 A2 WO 2012078519A2 US 2011063312 W US2011063312 W US 2011063312W WO 2012078519 A2 WO2012078519 A2 WO 2012078519A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
substituted
alkyl
another embodiment
unsubstituted
Prior art date
Application number
PCT/US2011/063312
Other languages
French (fr)
Other versions
WO2012078519A3 (en
Inventor
John Griffin
Brian Raimundo
Peter Madrid
Chaitan Khosla
Xi JIN
Cornelius Kloeck
Original Assignee
Numerate, Inc.
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Numerate, Inc., The Board Of Trustees Of The Leland Stanford Junior University filed Critical Numerate, Inc.
Publication of WO2012078519A2 publication Critical patent/WO2012078519A2/en
Publication of WO2012078519A3 publication Critical patent/WO2012078519A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Landscapes

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

Abstract

Provided herein are acylidene oxoindole derivatives, compositions including them, and methods of inhibiting transglutaminase 2 and treating diseases mediated by transglutaminase 2 by administering such derivatives and compositions.

Description

3-ACYLIDENE-2-OXOINDOLE DERIVATIVES FOR INHIBITION OF TRANSGLUTAMINASE 2
CROSS REFERENCE TO RELATED APPLICATION
[0001] This application claims priority under 35 U.S.C. § 1 19(e) of U.S. provisional application number 61/420,216 filed on December 6, 2010, which is incorporated herein in its entirety by reference.
FIELD OF THE INVENTION
[0002] The present technology provides compositions and methods for inhibiting transglutaminase 2 (TG2), and generally relates to the fields of medicinal chemistry, medicine, pharmacology, molecular biology, and biology.
BACKGROUND OF THE INVENTION
[0003] Transglutaminases (TGases) are a family of Ca2+-dependent enzymes that catalyze the formation of isopeptide bonds between the carboxamide group of protein/peptide-bound glutamine residues and the epsilon-amino group of protein/peptide- bound lysine residues to form NE-(y-glutamyl)lysine isopeptide bonds with loss of ammonia. Currently, eight TGase isoforms have been identified. The TGase 2 (i.e., tissue transglutaminase) isozyme (or TG2) is reportedly involved in several general biological functions, including apoptosis, cell adhesion and signal transduction. In addition, this particular isozyme has reportedly been linked to celiac disease, cancer and
neurodegenerative diseases. Therefore, TG2 inhibitors are needed in order to further elucidate its role in various patho-physiologies and to provide compounds for therapeutic development.
SUMMARY OF THE INVENTION
[0004] Provided herein are 3-acylidene-2-oxoindole derivatives and compositions including them. For Example, and without limitation, such 3-acylidene-2-oxoindole derivatives have generic and specific formulas provided hereinbelow. Also provided herein are methods of inhibiting transglutaminase 2 and treating diseases mediated by transglutaminase 2 by administering such derivatives and compositions. DETAILED DESCRIPTION OF THE INVENTION
Definitions
[0005] All technical and patent publications cited herein are incorporated herein by reference in their entirety.
[0006) All numerical designations, e.g., pH, temperature, time, concentration, and molecular weight, including ranges, are approximations which are varied ( + ) or ( - ) by increments of 0.1 or 1.0, as appropriate. It is to be understood, although not always explicitly stated that all numerical designations are preceded by the term "about". It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.
[0007] As used in the specification and claims, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise.
[0008] "Administering" or "administration of a compound or composition drug to a patient (and grammatical equivalents of this phrase) refers to direct administration, which may be administration to a patient by a medical professional or may be self- administration, and/or indirect administration, which may be the act of prescribing a drug. For example, a physician who instructs a patient to self-administer a drug and/or provides a patient with a prescription for a drug is administering the drug to the patient.
[0009] "Alkoxy" refers to an alkyl group covalently bonded to an oxygen atom. In other words, an alkoxy group has the general structure -O- alkyl. C -Ce alkoxy groups include, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n- pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy.
[0010] "Alkyl" refers to a straight (or linear) or branched chain alkyl group. C1-C6 alkyl groups include, for example, methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert- butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl and 3-methylpentyl.
[0011] "Alkylene" refers to a linear saturated divalent hydrocarbon radical or a branched saturated divalent hydrocarbon radical. Q-Ce alkylene groups include, for example, without limitation, methylene, ethylene, propylene, butylene, 2- methylpropylene, and pentylene.
[0012] "Amino" refers to a monovalent radical -NRaRb wherein Ra and Rb are independently hydrogen, alkyl, aryl or heteroaryl. The term "alkylamino" refers to the group -NRaRb where Ra is alkyl and Rb is H or alkyl. For dialkylamino groups, the alkyl portions can be the same or different and can also be combined to form a 3- to 8- membered ring with the nitrogen atom to which each is attached. Accordingly, a group represented as -NRaRb is meant to include piperidinyl, pyrrolidinyl, morpholinyl, azetidinyl and the like.
[0013] "Aryl" refers to a cyclic moiety that includes one or more monocyclic or fused ring aromatic systems. Such moieties include any moiety that has one or more monocyclic or bicyclic fused ring aromatic systems, including but not limited to phenyl and naphthyl.
[0014] "Arylene" refers to a divalent aryl group. Examples of arylene include, without
Figure imgf000004_0001
limitation, the phenylenes: , and various naphthylenes.
[0015] (Cm-Cn), Cm-Cn, or Cm-n refer to the number of carbon atoms in a certain group before which one of these symbols are placed. For example, C 1 -C6 alkyl refers to an alkyl group containing from 1 to 6 carbon atoms.
[0016] "Carrier" refers to a solid or liquid substance such as a polymer, solvent, suspending agent, absorbing agent, or adsorbing agent for the pre-delivery or capture of a compound of this technology delivery for subsequent delivery. The carrier may be liquid or solid and is selected with the planned manner of administration in mind.
[0017] "Carboxamide or carboxamido" refers to a monovalent radical -CO-NRaRb.
[0018] "Comprising" refers to compounds, compositions and methods including the recited elements, but not exclude others. "Consisting essentially of," when used to define compounds, compositions or methods, shall mean excluding other elements that would materially affect the basic and novel characteristics of the claimed technology.
"Consisting of," shall mean excluding any element, step, or ingredient not specified in the claim. Embodiments defined by each of these transition terms are within the scope of this technology.
[00191 "Cycloalkyl" refers to, unless otherwise stated, cyclic versions of "alkyl", "alkenyl" and "alkynyl" in which all ring atoms are carbon. "Cycloalkyl" refers to a mono- or polycyclic group. "Cycloalkyl" may form a bridged ring or a spiro ring. The cycloalkyl group may have one or more double or triple bond(s). Typical cycloalkyl groups have from 3 to 8 ring atoms. Examples of cycloalkyl include cyclopentyl, cyclohexyl, 1 -cyclohexenyl, 3-cyclohexenyl, and cycloheptyl.
[0020] "Halogen" or halo" refers to by themselves or as part of another substituent, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
[0021] "Heteroaryl" refers to a monocyclic aromatic system having 5 or 6 ring atoms, or a fused ring bicyclic aromatic system having 8-20 atoms, in which the ring atoms are C, O, S, SO, S02, or N, and at least one of the ring atoms is a heteroatom, i.e., O, S, SO, SO2, or N. Heteroaryl groups include, for example, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothio-furanyl, benzothiophenyl, benzoxazolyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, NH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, dithiazinyl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, isobenzofiiranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, quinuclidinyl,
tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, thiadiazinyl, thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl and xanthenyl. Unless indicated otherwise, the arrangement of the heteroatoms within the ring may be any arrangement allowed by the bonding
characteristics of the constituent ring atoms. [0022] "Heterocyclyl" or heterocyclic refers to a monocyclic or fused ring multicyclic cycloalkyl group at least a portion of which is not aromatic and in which one or more of the carbon atoms in the ring system is replaced by a heteroatom selected from O, S, SO, SO2, P, or N. Examples of heterocyclyl groups include but are not limited to imidazolinyl, morpholinyl, piperidinyl, piperidin-2-onyl, piperazinyl, pyrrolidinyl, pyrrolidine-2-onyl, tetrahydrofuranyl, and tetrahydroimidazo [4,5-c] pyridinyl.
[0023] "Leaving group" refers to an atom or a group that can be replaced by a nucleophile. Examples of leaving groups include, but are not limited to, halo and sulfonate (-OSOiRs, wherein Rs is Q-Ce alkyl, C1-C6 alkyl substituted with 1 -3 fluoro atoms, or is phenyl or phenyl substituted with 1 -2 C1-C6 alkyl or halo atoms.
[0024] "Pharmaceutically acceptable salts" refers to salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular acidic or basic nature of the compounds described herein. When compounds of the present technology contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of salts derived from pharmaceutically-acceptable inorganic bases include aluminum, ammonium, calcium, lithium, magnesium, potassium, sodium, and the like. Salts derived from
pharmaceutically-acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, diethylamine, 2- diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N- ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethyl amine, trimethylamine, tripropylamine, tromethamine and the like. When compounds of the present technology contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p- tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginine and the like, and salts of organic acids like glucuronic or galactunoric acids. Certain specific compounds of the present technology may contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
[0025] "Pharmaceutically acceptable excipient, carrier, or diluent" refers to an excipient, carrier, or diluent that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes an excipient, carrier, or diluent that is acceptable for human pharmaceutical use as well as veterinary use. A "pharmaceutically acceptable excipient, carrier, or diluent" includes both one and more than one such excipient, carrier, or diluent.
[0026] A "subject," "individual" or "patient" is used interchangeably herein, and refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, rats, rabbits, simians, bovines, ovines, porcines, canines, felines, farm animals, sport animals, pets, equines, and primates, particularly humans.
[0027] "Substituted" refers to a group as defined herein in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atom "substituents" include, but are not limited to, a halogen atom; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy, and acyloxy groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as nitro, amino (-NH2), alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, alkoxyamino, hydroxyamino, acylamino, sulfonylamino, N-oxides, imides, and enamines; and other heteroatoms in various other groups. "Substituents" also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom is replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, acyl, amido,
alkoxycarbonyl, aminocarbonyl, carboxyl, and ester groups; nitrogen in groups such as imines, oximes, hydrazones, and nitriles. "Substituents" further include groups in which one or more bonds to a carbon(s) or hydrogen(s) atoms is replaced by a bond to a cycloalkyl, heterocyclyl, aryl, and heteroaryl groups. For cycloalkyl, heterocyclyl, aryl, and heteroaryl groups, "substituents" still further include, substituted and unsubstituted alkyl groups. For alkylene groups, certain substituents such as an -O- , -S- or -NRa- moiety may be inserted between two carbon atoms. Other substituents include ethynyl, vinyl, carboxyl and its esters and amides, hydroxymethyl, and methyl. Another
"substituent" is the trifluoromethyl group and other groups that contain the
trifluoromethyl group. Two substituents on same or adjacent carbon atoms may together with the carbon atoms to which they are bonded form a heterocyclic or cycloalkyl group. Typically, a particular group may have 0, 1 , 2 or 3 substituents.
[0028] "Sulfonate" refers to a group of formula -OS02Rx wherein Rx is alkyl, trifluoromethyl, or substituted or unsubstituted aryl.
[0029] "Therapeutically effective amount" is an amount administered to a patient with a disease mediated by TG2 that is sufficient to effect beneficial or desired results. A therapeutically effective amount can be administered in one or more administrations, applications, or dosages.
[0030] "Treating" or "treatment of a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results such as the reduction of symptoms. For purposes of this technology, beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms of diseases mediated by TG2; diminishment of extent of such diseases; delay or slowing of such disease progression; amelioration, palliation, or stabilization of such diseases; or other beneficial results.
[0031] "Reduction" or "inhibition" of a symptom or symptoms (and grammatical equivalents of this phrase) of a pathological condition or disease refers to decreasing the severity or frequency of the symptom(s), or elimination of the symptom(s).
MODES FOR CARRYING OUT THE INVENTION
[0032] In one aspect, the present technology provides compounds of formula (I)
Figure imgf000009_0001
(I) wherein R\ is H or substituted or unsubstituted C1-C6 alkyl; R2 is substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted heterocyclyl, aryl or heteroaryl group, or -NR6R7 wherein R6 and R7 are independently H or substituted or unsubstituted C] -C6 alkyl or aryl group; R31 , R32, R33, and R34 independently are H, halo, -NO2, -O-Re, or -SO2 R26R27 wherein Rg is substituted or unsubstituted C) -C6 alkyl or aryl group and R26 and R27 are independently H, C1-C6 alkyl, or aryl; R4 is OH and R5 is hydrogen, or R4 and R5 together with the carbon atoms to which they are bonded form a carbon-carbon double bond; or a pharmaceutically acceptable salt thereof.
[0033] In one embodiment, the present technology provides compounds of formula (II), formula (IIA), and formula (IIB)
Figure imgf000009_0002
(II) (IIA) (IIB) and pharmaceutically acceptable salts thereof, wherein the bond indicates that formula IIA or IIB represents compounds having cis or trans (or E or Z) stereochemistry around the carbon-carbon double bond, m is 1 , 2, 3, or 4, and R3 is defined as R31-R34 above. In one embodiment, the stereochemistry is E. In another embodiment, the stereochemistry is Z. [0034J In certain embodiments, compounds of formula (II) and (I) wherein R4 is OH are useful intermediates in the synthesis of TG2 inhibitory compounds of the present technology.
[0035] In another embodiment, the present technology provides compounds of formula (III) and (IIIA) and pharmaceutically acceptable salts thereof:
Figure imgf000010_0001
(IIIB)
wherein is H, Ci-Ce alkyl, phenyl, or -(CH2)n-Rio;
n is 1 , 2, or 3;
Rio is C3-C8 cycloalkyl, substituted or unsubstituted phenyl, -CO2R11 or -CONRnRn; R11 and R)2 are independently H or C1-C6 alkyl, or Rn and R)2 together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring;
R2 is C1 -C3 alkyl, substituted or unsubstituted pyridyl, or substituted or unsubstituted phenyl; and
R3 is halo, -NO2, substituted or unsubstituted C1-C3 alkoxy, or C1-C3 alkyl. [0037] As used herein, stereoisomers refer to E and Z isomers around the exocyclic carbon-carbon double bond shown in IIIB above. In one embodiment, the stereoisomer is an E stereoisomer. In another embodiment, the stereoisomer is a Z stereoisomer.
[0038] In other embodiments, for the compound of formula (IIIB) or stereoisomers thereof, Ri is H or -(CH2)n-Rio- In another embodiment,
Figure imgf000011_0001
is H. In another
embodiment, Ri is -(CH2)n-Rio- In another embodiment, n is 1. In another embodiment, n is 2. In another embodiment, n is 3. In another embodiment, R\ is C C6 alkyl. In another embodiment, Ri is methyl, isopropyl, propyl, isobutyl, butyl, or tertiary butyl. In another embodiment, R| is phenyl.
[0039] In another embodiment, n is 1. In another embodiment, n is 2. In another embodiment, n is 3.
[0040] In another embodiment, R)0 is C3-C8 cycloalkyl. In another embodiment, Rio is cyclohexyl. In another embodiment, Rio is substituted or unsubstituted phenyl. In another embodiment, Ri0 is phenyl. In another embodiment, R!0 is substituted or unsubstituted phenyl. In another embodiment, Rio is a monosubstituted phenyl. In another embodiment, the monosubstituted phenyl is substituted at the meta or para position. Suitable phenyl substituents include without limitation, -CO2H and C1-C6 alkyl esters thereof. In another embodiment, Rio is -CO2R11. In another embodiment, Rio is - CONR1 1R12. In another embodiment, Rn is C1-C3 alkyl. In another embodiment, R]2 is C1-C3 alkyl. In another embodiment, Rn and R12 together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring
[0041] In another embodiment, R2 is C1-C3 alkyl. In another embodiment, R2 is methyl. In another embodiment, R2 is pyridyl. In another embodiment, R2 is 2-, 3-, or 4-pyridyl. In another embodiment, R2 is 2-pyridyl. In another embodiment, R2 is 3-pyridyl. In another embodiment, R2 is 4-pyridyl. In another embodiment, R2 is substituted pyridyl substituted with 1 -2, or 1 substituent. Suitable pyridyl substituents include without limitation halo, C1-C6 alkoxy, and C1-C6 alkoxy substituted with 1 -3 fluoro atoms. In another embodiment, R2 is substituted or unsubstituted phenyl. In another embodiment, R2 is substituted phenyl substituted with 1 -3, 1 -2, or 1 substituent. In another
embodiment, the substituted phenyl is monosubstituted phenyl. In another embodiment, the monosubstituted phenyl is substituted with a halo, C 1 -C3 alkoxy, nitro, or amino group. In another embodiment, the monosubstituted phenyl is substituted with a halo group. In another embodiment, the monosubstituted phenyl is substituted with an C1-C3 alkoxy. In another embodiment, the monosubstituted phenyl is substituted with an amino group. Other suitable substituents for the phenyl group include without limitation Q-C6 alkoxy substituted with 1 -3 fluoro atoms.
[0042] In another embodiment, R3 is hydrogen. In another embodiment, R3 is halo. In another embodiment, R3 is chloro. In another embodiment, R3 is bromo. In another embodiment, R3 is C1-C3 alkyl. In another embodiment, R3 is substituted or unsubstituted C 1-C3 alkoxy. In another embodiment, R3 is C 1-C3 alkoxy. In another embodiment, R3 is substituted C1-C3 alkoxy. Nonlimiting alkoxy substituents include halo, such as 1 -3 fluoro groups. In another embodiment, R3 is N02. In another embodiment, R3 is a»non- hydrogen substituent, substituted at the 4 position. In another embodiment, R3 is a non- hydrogen substituent, substituted at the 5 position. In another embodiment, R3 is a non- hydrogen substituent, substituted at the 6 position. In another embodiment, R3 is a non- hydrogen substituent, substituted at the 7 position.
[0043] In another embodiment, for the compound of formula (IIIB) or stereoisomers thereof, R2 is pyridyl, substituted pyridyl, phenyl, or phenyl substituted with a halo, methoxy, or amino group, and R3 is 4-chloro. In another embodiment, R2 is pyridyl and R3 is 4-chloro.
[0044] Various compounds of the present technology, and of formula (IIIB) include those illustrated in Table 1 hereinbelow and stereoisomers thereof.
[0045] In another embodiment, the present technology provides compounds of formula (IV) and pharmaceutically acceptable salts thereof:
Figure imgf000012_0001
(IV)
Within this embodiment, in one embodiment, Ri is H, methyl, or -Rg-Rio wherein R9 is substituted or unsubstituted methylene and Ri0 is -CONR1 1 R12 or substituted or unsubstituted aryl, cycloalkyl, or heterocyclyl group wherein Rn and R12 independently are H or C\-Ce alkyl or R) i and R12 together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring, and the heterocyclyl group contains at least one basic nitrogen atom. In another embodiment, R9 is -CH2-. In another embodiment, Rio is cycloalkyl. In another embodiment, Rio is aryl. In another embodiment, Rio is heteroaryl. In another embodiment, Rio is heterocyclyl. In another embodiment, R2 is
Figure imgf000013_0001
[0046] In another embodiment, R2 is
Figure imgf000013_0002
wherein R13 is halo or another leaving group, C1-C6 alkyl, or aryl. In another embodiment, Ri is H. In another embodiment, the present technology provides following compounds and pharmaceutically acceptable salts thereof:
Figure imgf000013_0003
[0047] In another embodiment, the present technology provides compounds of formula (IV), wherein R2 is substituted or unsubstituted pyridyl. In another embodiment, the substituted pyridyl is of formula:
Figure imgf000013_0004
wherein R14 is a bond, -O- or -NR16- and R15 is halo, substituted or unsubstituted C1-C6 alkyl or aryl, or is CONR18R19, or R15 and Ri6 together with the nitrogen atom to which they are bonded form a heterocyclic ring containing up to 2 nitrogen atoms, each Rig and R19 are independently C1-C6 alkyl, or Rig and R19 together with the nitrogen atom to which they are bonded form a heterocyclic ring containing up to 2 nitrogen atoms, and wherein Ri6 is substituted or unsubstituted C1-C6 alkyl, provided that when R] 5 is halo, R)4 is a bond. In another embodiment, RM is a bond. In another embodiment, R14 is -0-. In another embodiment, RH is -NRi6-. In another embodiment, R\5 is halo. In another embodiment, R15 is C\-Ce alkyl. In another embodiment, R15 is substituted C1-C6 alkyl. In another embodiment, R15 is aryl. In another embodiment, R) 5 substituted aryl. In another embodiment, R15 is CONRi8Ri . In another embodiment, R15 and Ri6 together with the nitrogen atom to which they are bonded form a heterocyclic ring containing up to 2 nitrogen atoms.
[0048] In another embodiment, the substituted pyridyl is of formula:
Figure imgf000014_0001
In another embodiment, R15 is substituted or unsubstituted aryl. In another embodiment, R15 is aryl. In another embodiment, R15 is unsubstituted aryl. In another embodiment, Ri5 is substituted or unsubstituted heteroaryl. In another embodiment, R15 is heteroaryl. In another embodiment, R15 is substituted heteroaryl. In another embodiment, R15 is halo. In another embodiment, 15 is substituted or unsubstituted benzyl. In another embodiment, R15 is benzyl. In another embodiment, R $ is substituted benzyl substituted at the methylene group. In another embodiment, R15 is substituted benzyl substituted at the phenyl ring. In another embodiment, R15 is CONRi8Ri9.
[0049] In another embodiment, the substituted pyridyl is of formula:
Figure imgf000014_0002
In another embodiment, R15 is C1 -C4 alkyl. In another embodiment, R15 is C1-C2 alkyl substituted with a carboxyl, carboxamido, or aryl group.
[0050] In another embodiment, the substituted pyridyl is of formula:
Figure imgf000015_0001
In another embodiment, R15 and Ri6 are independently C 1 -C3 alkyl. In another embodiment, R15 and Ri6 together with the carbon atom to which they are bonded form a 5-6 membered hetercyclic ring containing up to 2 ring nitrogen atoms. [0051] In other embodiments within these embodiments, the compounds provided are
Figure imgf000015_0002
Figure imgf000016_0001
and pharmaceutically acceptable salts thereof, wherein the -R within the phenyl ring denotes a phenyl ring that is unsubstituted or substituted (i.e., R is hydrogen or another substituent). In another embodiment, R14 is -O- and Ri6 is -CH2-Rn wherein R17 is aryl, carboxamide, or a carboxylic acid group.
In another embodiment, the present technology provides compounds of formula
Figure imgf000016_0002
(V) and pharmaceutically acceptable salts thereof, wherein Rio is -CONR11R12 or substituted or unsubstituted aryl, cycloalkyl, or heterocyclyl group wherein Rn and R12
independently are H or C1-C6 alkyl or Rn and Rn together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring, and the heterocyclyl group (referring to Rio) contains at least one basic nitrogen atom. In another embodiment, Rio is - CONR1 1R12. In another embodiment, Rio is substituted or unsubstituted aryl. In another embodiment, Rio is aryl. In another embodiment, Rio is substituted aryl. In another embodiment, aryl is phenyl. In another embodiment, Rio is substituted or unsubstituted 5- 6 membered heterocyclic ring containing up to 2 ring nitrogen atoms. In another embodiment, R]0 is an unsubstituted 5-6 membered heterocyclic ring. In another embodiment, Rio is a substituted 5-6 membered heterocyclic ring. In another embodiment, R)0 is substituted or unsubstituted C3-Q cycloalkyl group. In another embodiment, Rio is a C3-Q cycloalkyl group. In another embodiment, Ri0 is a substituted C3-Q cycloalkyl group. In another embodiment, the present technology provides a compound:
Figure imgf000017_0001
(VA) and pharmaceutically acceptable salts thereof, wherein R13 is defined as above. [0053] In another embodiment, the present technology provides the compound:
Figure imgf000017_0002
and pharmaceutically acceptable salts thereof, wherein Rn-Ri2 are defined as [0054] In another embodiment, the present technology provides compound:
Figure imgf000018_0001
(VC) and pharmaceutically acceptable salts thereof, wherein R19 is hydrogen or substituted or unsubstiruted C1-C6 alkyl. In another embodiment, R19 is hydrogen. In another embodiment, R1 is C\-Ce alkyl. In another embodiment, R19 is substituted C1 -C6 alkyl.
[0055] In another embodiment, the present technology provides compounds of formula
(VI)
Figure imgf000018_0002
(VI) and pharmaceutically acceptable salts thereof, wherein R6 and R7 are independently H, unsubstituted or substituted C1-C4 alkyl, or aryl. In another embodiment, R6 is H. In another embodiment, R6 is C1-C4 alkyl. In another embodiment, R6 is substituted C 1-C4 alkyl. In another embodiment, R6 is aryl. In another embodiment, -NRsR7 is -NH2, - NHMe, -NHPh, -NHCH2Ph, or -N(CH2Ph)2.
[0056] In another embodiment, the present technology provides compounds of formula
(VII)
Figure imgf000019_0001
(VII) and pharmaceutically acceptable salts thereof, wherein R32 is H, -0-R8, or -SO2NR26R27 wherein, wherein Ri, R26 and R27 are defined as in any aspect or embodiment above, and Rg is
Figure imgf000019_0002
or substituted or unsubstituted phenyl or benzyl, and R33 is H or -0-R8 wherein Rs is substituted or unsubstituted phenyl, provided that one of R32 and R33 is H. In another embodiment within this embodiment, Ri is H and R2 is
Figure imgf000019_0003
[0057] In another embodiment the present technology provides the following compound and pharmaceutically acceptable salts thereof:
Figure imgf000019_0004
[0058] In another embodiment within this embodiment, the present technology provides the compound:
Figure imgf000020_0001
and pharmaceutically acceptable salts thereof, wherein R13 is defined as above. In another embodiment within this embodiment, the present technology provides the compound:
Figure imgf000020_0002
wherein R13 is defined as above. In another embodiment within this embodiment, the present technology provides the following compound and pharmaceutically acceptable salts thereof:
Figure imgf000020_0003
[0059] In another embodiment within this embodiment, the present technology provides the following compound and pharmaceutically acceptable salts thereof:
Figure imgf000020_0004
wherein R26 and R27 are defined as above. [0060] In another embodiment, the present technology provides compounds of formula (VIIIA) and (VIIIB)
Figure imgf000021_0001
(VIIIA) (VIIIB) and pharmaceutically acceptable salts thereof, wherein R\ is H, substituted or unsubstituted CpC6 alkyl; R2 is C1-C4 alkyl or substituted or unsusbtituted aryl or heteroaryl group; and R31, R32, R33, and R34 are independently H, halo, or substituted or unsubstituted C]-C4 alkoxy. In one embodiment, the compound provided is of formula (VIIIA). In another embodiment, the present technology provides compounds of formula (VIIIA) wherein R2 is selected from the group consisting of methyl, unsubstituted phenyl, phenyl substituted with a C1-C4 alkyl or an aryl group, 3-pyridyl, or 3-pyridyl substituted with a C1-C4 alkoxy group. In another embodiment, the 3-pyridyl is substituted with a C1-C4 alkoxy group at the 4 position. In another embodiment, Ri is unsubstituted C1-C4 alkyl or C1 -C4 alkyl substituted with substituted or unsubstituted piperidinyl, cyclohexyl and phenyl groups.
[0061 ] In another embodiment, the present technology provides compounds of formula (IX) and steroisomers thereof, and pharmaceutically acceptable salts of each thereof:
Figure imgf000021_0002
(IX) wherein Ri, R31 , and R2 are defined as in formula (VIIIA) above. In another embodiment, Ri is H or C1-C4 alkyl; R31 is CI or H, and R2 is methyl, phenyl, phenyl substituted with a C1-C4 alkyl group, or
Figure imgf000022_0001
wherein R25 is H or C1-C4 alkoxy. In another embodiment, Ri is H. In another embodiment, Ri is methyl. In another embodiment, R2 is
Figure imgf000022_0002
wherein R25 is defined as in formula (IX). In another embodiment, R2 is methyl. In another embodiment, R2 is phenyl. In another embodiment, R2 is phenyl substituted with a C1-C4 alkyl group.
[0062] In another embodiment, the present technology provides compounds of formula (X) and steroisomers thereof, and pharmaceutically acceptable salts of each thereof:
Figure imgf000022_0003
(X) wherein Ri, R32, and R2 are defined as in formula (VIIIA) above. In another embodiment, R32 is H or chloro. In another embodiment, Ri is H. In another embodiment, R] is methyl. In another embodiment, R2 is
Figure imgf000023_0001
wherein R25 is defined as in formula (IX). In another embodiment, R2 is methyl. In another embodiment, R2 is phenyl. In another embodiment, R2 is phenyl substituted with a C1 -C4 alkyl group.
[0063] In another embodiment, the present technology provides compounds of formula (XI) and steroisomers thereof, and pharmaceutically acceptable salts of each thereof:
Figure imgf000023_0002
(XI) wherein Ri , R33, and R2 are defined as in formula (VIIIA) above. In another embodiment, R33 is H, fluoro, trifluoromethoxy-, or C1-C4 alkoxy. In another embodiment, Ri is H. In another embodiment, Ri is methyl. In another embodiment, R2 is
Figure imgf000023_0003
wherein R25 is defined as in formula (IX). In another embodiment, R2 is methyl. In another embodiment, R2 is phenyl. In another embodiment, R2 is phenyl substituted with a C1 -C4 alkyl group.
[0064] In another embodiment, the present technology provides compounds of formula (XII) and stereoisomers thereof, and pharmaceutically acceptable salts of each thereof:
Figure imgf000024_0001
(XII) wherein Ri, R34, and R2 are defined as in formula (VIIIA) above. In another embodiment, R34 is H or CI. In another embodiment, R\ is H. In another embodiment, R\ is methyl. In another embodiment, R2 is:
Figure imgf000024_0002
wherein R25 is defined as in formula (IX). In another embodiment, R2 is methyl. In another embodiment, R2 is phenyl. In another embodiment, R2 is phenyl substituted with a C1-C4 alkyl group.
[0065] In another embodiment, the present technology provides compounds wherein Ri is hydrogen, R2 is methyl, and the oxindole part of the molecule is unsubstituted or substituted with one R31 , R32, R33 , or R34 group selected from chloro, methoxy, trifluoromethoxy, or fluoro. In another embodiment, the present technology provides the following compounds and pharmaceutically acceptable salts thereof:
Figure imgf000024_0003
wherein R3 is H, 6-OMe, 6-F, 5-Cl, 4-Cl, 7-Cl, or 6-O-CF3, wherein the number before a substituent refers to the position of the substituent in the structure. [0066] In another embodiment, the present technology provides compounds wherein Ri is hydrogen or methyl, R2 is phenyl, phenyl substituted with a methyl group, 3-pyridyl, or 3-pyridyl substituted with a methoxy group, and the oxindole part of the molecule is unsubstituted or substituted with only R3 ] which is chloro. In another embodiment, the present technology provides the following compounds and pharmaceutically acceptable salts thereof:
Figure imgf000025_0001
wherein R| is H, R2 is 3-pyridyl, R3 is CI; Ri is H, R2 is 3-pyridyl, R3 is H; Ri is H, R2 is phenyl, R3 is CI; R\ is Me, R2 is 3-pyridyl, R3 is CI; R\ is H, R2 is phenyl, R3 is H; Ri is H, R2 is 4-methoxy-3-pyridyl, R3 is CI; and R| is H, R2 is 2-methylphenyl, R3 is CI or stereoisomers thereof.
[0067] In other embodiments, compounds useful in accordance with the present technology include those illustrated in the Examples section below.
[0068] In another embodiment, the present technology provides compounds that demonstrate IC50 values of about 0.01 micro molar to about 50 micro molar as determined in the Example section entitled "Enzyme Kinetic Measurements." In other embodiments, compounds demonstrate ICso values of about 0.1 micromolar to about 5 micromolar, 0.5 micromolar to about 1 micromolar. In another embodiment, the present technology provides compounds that demonstrate low cell permeation. Without being bound by mechanism, since TG2 is substantially found in the outer cell membrane, a compound of the present technology useful in inhibiting TG2 does not necessarily need to enter into the cell for such inhibition. A variety of methods to determine cell permeation is well known in the art using, e.g., octanol water partitioning or Caco-2 cell based permeation testing.
[0069] In one embodiment, the present technology excludes the compound:
Figure imgf000026_0001
[0070] In another embodiment, the present technology provides the compounds tabulated below and pharmaceutically acceptable salts thereof:
Table 1
Figure imgf000026_0002
Figure imgf000027_0001
Figure imgf000028_0001
43 5-Br 4.8
44 5-Me 6.3
45 5-N02 7.9
46 6-Cl 2.1 (70%)
47 6-Br 1.5 (35%)
48 7-Cl 7.3
49 7-Br 2.9
[0071] One of ordinary skill in the art will appreciate that many of the compounds of the present technology and prodrugs thereof, may exhibit the phenomena of tautomerism, conformational isomerism, geometric isomerism, and/or optical isomerism. For example, the compounds and prodrugs of the present technology may include one or more chiral centers and/or double bonds and as a consequence may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers, diasteromers, and mixtures thereof, such as racemic mixtures. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present technology.
[0072] As another example, the compounds and prodrugs of the present technology may exist in several tautomeric forms, including the enol form, the keto form, and mixtures thereof. As the various compound names, formulae and compound drawings within the specification and claims can represent only one of the possible tautomeric, conformational isomeric, optical isomeric, or geometric isomeric forms, it should be understood that the present technology encompasses any tautomeric, conformational isomeric, optical isomeric, and/or geometric isomeric forms of the compounds or prodrugs having one or more of the utilities described herein, as well as mixtures of these various different isomeric forms.
[0073] Depending upon the nature of the various substituents, the compounds and prodrugs of the present technology can be in the form of salts. Such salts include pharmaceutically acceptable salts, salts suitable for veterinary uses, etc. Such salts can be derived from acids or bases, as is well-known in the art. In one embodiment, the salt is a pharmaceutically acceptable salt.
[0074] The various compounds of the technology are synthesized according to the procedures described here, and following methods known in the art upon reading this disclosure. A variety of substituted isatins are useful as starting materials for the synthesis of the compounds of the technology and intermediates to them. Certain general synthetic methods are schematically shown below.
Figure imgf000030_0001
[0075] A variety of bases may be used including without limitation dialkyl amines. Suitable dialkyl amines include, without limitation, diethyl amine. A variety of acids may be used for the dehydration, including without limitation mineral acids such as HCl, organic acids such as acetic acid, and their mixtures.
Figure imgf000030_0002
Ri is substitued or unsubstitued alkyl
[0076] The substituted or unsubstituted isatin may be alkylated before reacting with a methyl ketone. When Ri is an aryl group, the alkylation is conveniently carried out by reacting the starting compound with a aryl boronic acid or a aryl boronic acid ester in presence of suitable palladium catalysts and bases well known to the skilled artisan.
Figure imgf000031_0001
Ri3 = X (a leaving group) Ri3 = substituted or unsubstituted aryl, heteroaryl, or benzyl
[0077] In certain instances, compounds of the present technology wherein R2 is a
substituted aryl group, may be synthesized as shown above.
Figure imgf000031_0002
Ri4 is a bond and
R15 is substituted or unsubstituted alkyl or aryl, or CONR, 8R19
[0078J In certain instances, compounds of the present technology wherein R2 is a
substituted pyridyl group, may be synthesized as shown above. [0079J The reactions are performed for a period of time sufficient to provide a substantial amount of a product, which is readily determined by a skilled artisan using well known techniques such as thin layer chromatography, Ή-NMR, and the likes. A product may be used in subsequent steps without purification or may be separated using methods well known to the skilled artisan such as column chromatography,
crystallization, precipitation, and the likes.
[0080] In another aspect, the present technology provides compositions comprising or consisting essentially of a compound of the present technology and a carrier, diluent, or excipient. Compounds of the present technology include without limitation, those of formulas (I)-(XI). And subformulas included therein. In another embodiment, the carrier, diluent, or excipient is pharmaceutically acceptable. In another embodiment, the compounds employed in the compositions are of formulas (III) - (XI). In another embodiment, the compounds employed are of formula (IIB). In another embodiment, the compounds employed are of formula (IIIB). In another embodiment, the carrier is a pharmaceutically acceptable polymer. A variety of carrier, diluent, or excipient, pharmaceutically acceptable or not, are well known to one of skill in the art. See for example, pages 24-25 of PCT Pub. No. WO 2007/050795 incorporated herein by reference. Polymeric pharmaceutically acceptable carriers can form tubes, micelles, or other structures wherein the active ingredient is retained for eventual release into the patient. See, for example, Biodegradable Polymers as Drug Carriers, Peter Markland, Encyclopedia of Pharmaceutical Technology 02 October 2006.
[0081 ) In another aspect, the present technology provides the use of a compound or the pharmaceutically acceptable composition of the present technology, in the preparation of a medicament. In another aspect, the present technology provides methods of inhibiting transglutaminase-2 (TG2) comprising contacting an effective amount of a compound or a composition of of the present technology with the TG2. As used herein, an amount that provides a concentration in the subject, tissue or organ system of interest sufficient to inhibit TG2, for example an amount which provides a concentration in the subject, tissue or organ system of interest greater than the IC50 value for TG2 inhibition as determined. In another embodiment, the contacting takes place in vitro or in vivo. In another aspect, the present technology provides a complex comprising, or consisting essentially of a compound of the present technology and a TG2. [0082J In another aspect, the present technology provides methods of inhibiting TG2 in a tissue comprising contacting the tissue with a compound or a composition of the present technology. In another embodiment, the contacting is performed in vitro or in vivo.
[0083] In another aspect, the present technology provides methods of treating diseases relating to the pathological presence of a TG2 in a tissue, comprising administering a therapeutically effective amount of a compound or a composition of the present technology to a patient in need of such treatment, thereby treating the disease. In another embodiment, the tissue is one or more of the small intestine, the brain, and the lungs. In another embodiment, the disease is Alzheimer's disease, atherosclerosis, celiac sprue, cystic fibrosis, dermatitis herpetiformis, glioblastoma, carcinoma, and other cancers, Huntington's disease, keratosis, Parkinson's disease, or a wound to be healed. A variety of routes of administration may be employed to administer the compounds and compositions of the present technology in accordance with the methods of the present technology. In another embodiment, the compound or composition is administered by one or more of by ingestion, by inhalation, intranasal administration, or by injection.
[0084] The compounds or isomers, prodrug, tautomer, or pharmaceutically acceptable salts thereof, of the present technology can be formulated in the pharmaceutically acceptable compositions per se, or in the form of a hydrate, solvate, N-oxide, or pharmaceutically acceptable salt, as described herein. Typically, such salts are more soluble in aqueous solutions than the corresponding free acids and bases, but salts having lower solubility than the corresponding free acids and bases may also be formed. The present technology includes within its scope solvates of the compounds and salts thereof, for example, hydrates.
[0085] In one embodiment, the present technology provides a pharmaceutically acceptable composition (formulation) comprising a compound selected from the compounds of the present technology or isomers, hydrates, tautomers, or
pharmaceutically acceptable salts thereof and at least one pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof.
[0086] In one embodiment, the methods can be practiced as a therapeutic approach towards the treatment of the conditions described herein. Thus, in a specific embodiment, the compounds of the present technology can be used to treat the conditions described herein in animal subjects, including humans. The methods generally comprise administering to the subject an amount of a compound of the present technology, or a salt, prodrug, hydrate, or N-oxide thereof, effective to treat the condition.
[0087] In some embodiments, the subject is a non-human mammal, including, but not limited to, bovine, horse, feline, canine, rodent, or primate. In another embodiment, the subject is a human.
[0088] The compounds of the present technology can be provided in a variety of formulations and dosages. It is to be understood that reference to the compound of the present technology, or "active" in discussions of formulations is also intended to include, where appropriate as known to those of skill in the art, formulation of the prodrugs of the compounds.
[0089] In one embodiment, the compounds are provided as non-toxic pharmaceutically acceptable salts. In certain embodiments, suitable pharmaceutically acceptable salts of the compounds of the present teclinology include acid addition salts such as those formed with hydrochloric acid, fumaric acid, p-toluenesulphonic acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, or phosphoric acid. Salts of amine groups may also comprise quaternary ammonium salts in which the amino nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl, or substituted alkyl moiety. In certain other embodiments, where the compounds of the present technology contain an acidic moiety, suitable pharmaceutically acceptable salts thereof may include metal salts such as alkali metal salts, e.g., sodium or potassium salts; and alkaline earth metal salts, e.g., calcium or magnesium salts.
[0090] The pharmaceutically acceptable salts of the present technology can be formed by conventional means, such as by reacting the free base form of the product with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is insoluble or in a solvent such as water which is removed in vacuo, by freeze drying, or by exchanging the anions of an existing salt for another anion on a suitable ion exchange resin.
[0091] Pharmaceutically acceptable compositions comprising the compounds described herein (or prodrugs thereof) can be manufactured by means of conventional mixing, dissolving, granulating, dragee-making levigating, emulsifying, encapsulating, entrapping, or lyophilization processes. The compositions can be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients, or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
[0092] The compounds of the present technology can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray nasal, vaginal, rectal, sublingual, urethral (e.g., urethral suppository) or topical routes of administration (e.g., gel, ointment, cream, aerosol, etc.) and can be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles appropriate for each route of administration.
[0093] The pharmaceutically acceptable compositions for the administration of the compounds can be conveniently presented in unit dosage form and can be prepared by any of the methods well known in the art. The pharmaceutically acceptable compositions can be, for example, prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier, a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired therapeutic effect. For example, pharmaceutically acceptable compositions of the present technology may take a form suitable for virtually any mode of administration, including, for example, topical, ocular, oral, buccal, systemic, nasal, injection, transdermal, rectal, and vaginal, or a form suitable for administration by inhalation or insufflation.
[0094] For topical administration, the compound(s) or prodrug(s) can be formulated as solutions, gels, ointments, creams, suspensions, etc., as is well-known in the art.
[0095] Systemic pharmaceutically acceptable compositions include those designed for administration by injection (e.g., subcutaneous, intravenous, intramuscular, intrathecal, or intraperitoneal injection) as well as those designed for transdermal, transmucosal, oral, or pulmonary administration.
[0096] Useful injectable pharmaceutically acceptable compositions include sterile suspensions, solutions, or emulsions of the active compound(s) in aqueous or oily vehicles. The pharmaceutically acceptable compositions may also contain formulating agents, such as suspending, stabilizing, and/or dispersing agents. The formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, and may contain added preservatives.
[0097J Alternatively, the injectable pharmaceutically acceptable compositions can be provided in powder form for reconstitution with a suitable vehicle, including but not limited to sterile pyrogen free water, buffer, and dextrose solution, before use. To this end, the active compound(s) can be dried by any art-known technique, such as lyophilization, and reconstituted prior to use.
[0098] For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the pharmaceutically acceptable compositions. Such penetrants are known in the art.
[0099] For oral administration, the pharmaceutically acceptable compositions may take the form of, for example, lozenges, tablets, or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone, or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose, or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulfate). The tablets can be coated by methods well known in the art with, for example, sugars, films, or enteric coatings.
Additionally, the pharmaceutically acceptable compositions containing the compounds of the present technology or prodrug thereof in a form suitable for oral use may also include, for example, troches, lozenges, aqueous, or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
[0100] Pharmaceutically acceptable compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutically acceptable compositions, and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents, and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient (including drug and/or prodrug) in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents (e.g., corn starch or alginic acid); binding agents (e.g. starch, gelatin, or acacia); and lubricating agents (e.g., magnesium stearate, stearic acid, or talc). The tablets can be left uncoated or they can be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. They may also be coated by the techniques described in the U.S. Pat. Nos. 4,256,108; 4, 166,452; and 4,265,874 to form osmotic therapeutic tablets for control release. The pharmaceutically acceptable compositions of the present technology may also be in the form of oil-in-water emulsions.
[0101] Liquid pharmaceutically acceptable compositions (or liquid preprarations) for oral administration may take the form of, for example, elixirs, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats); emulsifying agents (e.g., lecithin, or acacia); nonaqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, cremophore™, or fractionated vegetable oils); and preservatives (e.g., methyl or propylphydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, preservatives, flavoring, coloring, and sweetening agents as appropriate.
[0102] Preparations for oral administration can be suitably formulated to give controlled release or sustained release of the active compound, as is well known. The sustained release formulations (or sustained release pharmaceutically acceptable compositions) of the present technology are preferably in the form of a compressed tablet comprising an intimate mixture of compound of the present technology and a partially neutralized pH- dependent binder that controls the rate of compound dissolution in aqueous media across the range of pH in the stomach (typically approximately 2) and in the intestine (typically approximately about 5.5).
[0103] To provide for a sustained release of compounds of the present technology, one or more pH-dependent binders can be chosen to control the dissolution profile of the sustained release pharmaceutically acceptable compositions so that such pharmaceutically acceptable compositions release compound slowly and continuously as the
pharmaceutically acceptable compositions are passed through the stomach and gastrointestinal tract. Accordingly, the pH-dependent binders suitable for use in the present technology are those which inhibit rapid release of drug from a tablet during its residence in the stomach (where the pH is-below about 4.5), and which promotes the release of a therapeutic amount of the compound of the present technology from the dosage form in the lower gastrointestinal tract (where the pH is generally greater than about 4.5). Many materials known in the pharmaceutical art as "enteric" binders and coating agents have a desired pH dissolution properties. The examples include phthalic acid derivatives such as the phthalic acid derivatives of vinyl polymers and copolymers, hydroxyalkylcelluloses, alkylcelluloses, cellulose acetates, hydroxyalkylcellulose acetates, cellulose ethers, alkylcellulose acetates, and the partial esters thereof, and polymers and copolymers of lower alkyl acrylic acids and lower alkyl acrylates, and the partial esters thereof. One or more pH-dependent binders present in the sustained release formulation of the present technology are in an amount ranging from about 1 to about 20 wt %, more preferably from about 5 to about 12 wt % and most preferably about 10 wt %.
[0104] One or more pH-independent binders may be in used in oral sustained release pharmaceutically acceptable compositions of the present technology. The pH- independent binders can be present in the pharmaceutically acceptable compositions of the present technology in an amount ranging from about 1 to about 10 wt %, and preferably in amount ranging from about 1 to about 3 wt % and most preferably about 2 wt %.
[0105] The sustained release pharmaceutically acceptable compositions of the present technology may also contain pharmaceutically acceptable excipients intimately admixed with the compound and the pH-dependent binder. Pharmaceutically acceptable excipients may include, for example, pH-independent binders or film-forming agents such as hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose,
polyvinylpyrrolidone, neutral poly(meth)acrylate esters, starch, gelatin, sugars, carboxymethylcellulose, and the like. Other useful pharmaceutical excipients include diluents such as lactose, mannitol, dry starch, microcrystalline cellulose and the like; surface active agents such as polyoxyethylene sorbitan esters, sorbitan esters and the like; and coloring agents and flavoring agents. Lubricants (such as talc and magnesium stearate) and other tableting aids can also be optionally present.
[0106] The sustained release pharmaceutically acceptable compositions of the present technology have a compound of the present technology in the range of about 50% by weight to about 95% or more by weight, and preferably between about 70% to about 90% by weight; a pH-dependent binder content of between 5% and 40%, preferably between 5% and 25%, and more preferably between 5% and 15%; with the remainder of the dosage form comprising pH-independent binders, fillers, and other optional excipients.
[0107] For buccal administration, the pharmaceutically acceptable compositions may take the form of tablets or lozenges formulated in the conventional manner.
[0108] For rectal and vaginal routes of administration, the active compound(s) can be formulated as solutions (for retention enemas), suppositories, or ointments containing conventional suppository bases such as cocoa butter or other glycerides.
[0109] For nasal administration or administration by inhalation or insufflation, the active compound(s) or prodrug(s) can be conveniently delivered in the form of an aerosol spray from pressurized packs or a nebulizer with the use of a suitable propellant (e.g., dichlorodifiuoromethane, trichlorofluoromethane, dichlorotetrafluoroethane,
fluorocarbons, carbon dioxide, or other suitable gas). In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges for use in an inhaler or insufflator (for example, capsules and cartridges comprised of gelatin) can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0110] The pharmaceutically acceptable compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution, and isotonic sodium chloride solution. The compounds may also be administered in the form of suppositories for rectal or urethral administration of the drug. [0111] For topical use, creams, ointments, jellies, gels, solutions, suspensions, etc., containing the compounds of the present technology, can be employed. In some embodiments, the compounds of the present technology can be formulated for topical administration with polyethylene glycol (PEG). These formulations may optionally comprise additional pharmaceutically acceptable ingredients such as diluents, stabilizers, and/or adjuvants.
[0112] Included among the devices which can be used to administer compounds of the present technology, are those well-known in the art, such as metered dose inhalers, liquid nebulizers, dry powder inhalers, sprayers, thermal vaporizers, and the like. Other suitable technology for administration of particular compounds of the present technology includes electrohydrodynamic aerosolizers. As those skilled in the art will recognize, the formulation of compounds, the quantity of the formulation delivered, and the duration of administration of a single dose depend on the type of inhalation device employed as well as other factors. For some aerosol delivery systems, such as nebulizers, the frequency of administration and length of time for which the system is activated will depend mainly on the concentration of compounds in the aerosol. For example, shorter periods of administration can be used at higher concentrations of compounds in the nebulizer solution. Devices such as metered dose inhalers can produce higher aerosol
concentrations and can be operated for shorter periods to deliver the desired amount of compounds in some embodiments. Devices such as dry powder inhalers deliver active agent until a given charge of agent is expelled from the device. In this type of inhaler, the amount of compounds in a given quantity of the powder determines the dose delivered in a single administration.
[0113] Pharmaceutically acceptable compositions of compounds of the present technology for administration from a dry powder inhaler may typically include a finely divided dry powder containing compounds, but the powder can also include a bulking agent, buffer, carrier, excipient, another additive, or the like. Additives can be included in such a dry powder composition of compounds of the present technology, for example, to dilute the powder as required for delivery from the particular powder inhaler, to facilitate processing of the formulation, to provide advantageous powder properties to the formulation, to facilitate dispersion of the powder from the inhalation device, to stabilize the formulation (e.g., antioxidants or buffers), to provide taste to the formulation, or the like. Typical additives include mono-, di-, and polysaccharides; sugar alcohols and other polyols, such as, for example, lactose, glucose, raffinose, melezitose, lactitol, maltitol, trehalose, sucrose, mannitol, starch, or combinations thereof; surfactants, such as sorbitols, diphosphatidyl choline, or lecithin; and the like.
[0114] For prolonged delivery, the compound(s) or prodrug(s) of the present technology can be formulated as a depot preparation for administration by implantation or intramuscular injection. The active ingredient can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt). Alternatively, transdermal delivery systems manufactured as an adhesive disc or patch which slowly releases the active compound(s) for percutaneous absorption can be used. To this end, permeation enhancers can be used to facilitate transdermal penetration of the active compound(s). Suitable transdermal patches are described in, for example, U.S. Patent No. 5,407,713.; U.S. Patent No. 5,352,456; U.S. Patent No. 5,332,213; U.S. Patent No.
5,336, 168; U.S. Patent No. 5,290,561 ; U.S. Patent No. 5,254,346; U.S. Patent No.
5, 164,189; U.S. Patent No. 5,163,899; U.S. Patent No. 5,088,977; U.S. Patent No.
5,087,240; U.S. Patent No. 5,008, 1 10; and U.S. Patent No. 4,921 ,475.
[0115] Alternatively, other pharmaceutical delivery systems can be employed.
Liposomes and emulsions are well-known examples of delivery vehicles that can be used to deliver active compound(s) or prodrug(s). Certain organic solvents such as
dimethylsulfoxide (DMSO) may also be employed, for example for topical
administration, although usually at the cost of greater toxicity.
[0116] The pharmaceutical compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active compound(s). The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device can be accompanied by instructions for
administration.
[0117] The compound(s) or prodrug(s) described herein, or compositions thereof, will generally be used in an amount effective to achieve the intended result, for example, in an amount effective to treat or prevent the particular condition being treated. The compound(s) can be administered therapeutically to achieve therapeutic benefit or prophylactically to achieve prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated and/or eradication or amelioration of one or more of the symptoms associated with the underlying disorder such that the patient reports an improvement in feeling or condition, notwithstanding that the patient may still be afflicted with the underlying disorder. For example,
administration of a compound to a patient suffering from Celiac Sprue provides therapeutic benefit not only when Celiac Sprue is eradicated or ameliorated, but also when the patient reports a decrease in the severity or duration of the symptoms associated with Celiac Sprue. Therapeutic benefit also includes halting or slowing the progression of the disease, regardless of whether improvement is realized.
[0118] The amount of compound administered will depend upon a variety of factors, including, for example, the particular condition being treated, the mode of administration, the severity of the condition being treated, the age and weight of the patient, the bioavailability of the particular active compound. Determination of an effective dosage is well within the capabilities of those skilled in the art. As known by those of skill in the art, the preferred dosage of compounds of the present technology will also depend on the age, weight, general health, and severity of the condition of the individual being treated. Dosage may also need to be tailored to the sex of the individual and/or the lung capacity of the individual, where administered by inhalation. Dosage, and frequency of administration of the compounds or prodrugs thereof, will also depend on whether the compounds are formulated for treatment of acute episodes of a condition or for the prophylactic treatment of a disorder. A skilled practitioner will be able to determine the optimal dose for a particular individual.
[0119] For prophylactic administration, the compound can be administered to a patient at risk of developing one of the previously described conditions. Alternatively, prophylactic administration can be applied to avoid the onset of symptoms in a patient diagnosed with the underlying disorder.
[0120] Effective dosages can be estimated initially from in vitro assays. For example, an initial dosage for use in animals can be formulated to achieve a circulating blood or serum concentration of active compound that is at or above an IC50 of the particular compound as measured in as in vitro assay. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular compound is well within the capabilities of skilled artisans. For guidance, the reader is referred to Fingl & Woodbury, "General Principles," GOODMAN AND GILMAN'S THE PHARMACEUTICAL BASIS OF THERAPEUTICS, Chapter 1 , pp. 1 - 46, latest edition, Pergamon Press, and the references cited therein.
[0121] Initial dosages can also be estimated from in vivo data, such as animal models. Certain animal models useful for testing the efficacy of compounds to treat or prevent the various diseases described above are well-known in the art. Ordinarily skilled artisans can routinely adapt such information to determine dosages suitable for human administration.
[0122] Dosage amounts will typically be in the range of from about 0.0001 or 0.001 or 0.01 mg/kg/day to about 100 mg/kg/day, but can be higher or lower, depending upon, among other factors, the activity of the compound, its bioavailability, the mode of administration, and various factors discussed above. Dosage amount and interval can be adjusted individually to provide levels in the organ system of interest of the compound(s) which are sufficient to maintain therapeutic or prophylactic effect. For example, the compounds can be administered once per week, several times per week (e.g., every other day), once per day, or multiple times per day, depending upon, among other things, the mode of administration, the specific indication being treated, and the judgment of the prescribing physician. In cases of local administration or selective uptake, such as local topical administration, the effective local concentration of active compound(s) may not be related to plasma concentration. Skilled artisans will be able to optimize effective local dosages without undue experimentation.
[0123] The compound(s) useful in the treatment methods of the present technology will provide therapeutic or prophylactic benefit without causing substantial toxicity. Toxicity of the compound(s) can be determined using standard pharmaceutical procedures. The dose ratio between toxic and therapeutic (or prophylactic) effect is the therapeutic index. In certain embodiments, the compounds(s) exhibit high therapeutic indices as pertinent to the disease treated.
[0124] The foregoing disclosure pertaining to the dosage requirements for the compounds of the present technology is pertinent to dosages required for prodrugs, with the realization, apparent to the skilled artisan, that the amount of prodrug(s) administered will also depend upon a variety of factors, including, for example, the bioavailability of the particular prodrug(s) and the conversation rate and efficiency into active drug compound under the selected route of administration. Determination of an effective dosage of prodrug(s) for a particular use and mode of administration is well within the capabilities of those skilled in the art.
[0125] Also provided are kits for administration of the compounds of the present technology, prodrug thereof, or pharmaceutical formulations comprising the compound that may include a dosage amount of at least one compound or a composition comprising at least one compound, as disclosed herein. Kits may further comprise suitable packaging and/or instructions for use of the compound. Kits may also comprise a means for the delivery of the at least one compound or compositions comprising at least one compound of the present technology, such as an inhaler, spray dispenser (e.g., nasal spray), syringe for injection, or pressure pack for capsules, tablets, suppositories, or other device as described herein.
[0126] Other types of kits provide the compound and reagents to prepare a composition of the present technology for administration. The composition can be in a dry or lyophilized form or in a solution, particularly a sterile solution. When the composition is in a dry form, the reagent may comprise a pharmaceutically acceptable diluent for preparing a liquid formulation. The kit may contain a device for administration or for dispensing the compositions, including, but not limited to, syringe, pipette, transdermal patch, or inhalant.
[0127] The kits may include other therapeutic compounds for use in conjunction with the compounds of the present technology described herein. These compounds can be provided in a separate form or mixed with the compounds of the present technology. The kits will include appropriate instructions for preparation and administration of the composition, side effects of the compositions, and any other relevant information. The instructions can be in any suitable format, including, but not limited to, printed matter, videotape, computer readable disk, or optical disc.
[0128] In one embodiment, the present technology provides a kit comprising a compound selected from the compounds of the present technology or a prodrug thereof, packaging, and instructions for use. [0129] In another embodiment, the present technology provides a kit comprising the pharmaceutically acceptable composition comprising a compound selected from the compounds of the present technology or a prodrug thereof and at least one
pharmaceutically acceptable excipient, diluent, preservative, stabilizer, or mixture thereof, packaging, and instructions for use. In another embodiment, kits for treating an individual who suffers from or is susceptible to the conditions described herein are provided, comprising a container comprising a dosage amount of a compound or composition of the present technology, as disclosed herein, and instructions for use. The container can be any of those known in the art and appropriate for storage and delivery of oral, intravenous, topical, rectal, urethral, or inhaled formulations.
[0130] Kits may also be provided that contain sufficient dosages of the compounds or composition to provide effective treatment for an individual for an extended period, such as a week, 2 weeks, 3, weeks, 4 weeks, 6 weeks, or 8 weeks or more.
[0131] The technology having been described in summary and in detail is illustrated and not limited by the examples below.
EXAMPLES
[0132] The following abbreviations are used in the examples section.
Ac Acetyl
DCM Dichloromethane
DMF Dimethylformamide
dppf Diphenylphosphinoferrocene
E E (from the German entgegen) stereochemistry around a double bond
based on the Cahn-Ingold-Prelog priority rules.
EDTA Ethylene diamine tetraacetic acid
Et Ethyl
[Etotai] Total enzyme concentration
Km Michaelis constant
Me Methyl
mL Milliliter
μΐ- Microliter
μΜ Micromolar mmol(e) Millimole
Ms Methanesulfonyl
NADH Nicotinamide adenine dinucleotide reduced form
NMR Nuclear magnetic resonance
OCT Glycol-based freezing medium for histology
PBS Phosphate buffered saline
[S] Substrate concentration
TLC Thin layer chromatography
V Reaction velocity
z Z (from the German zusammen) stereochemistry around a double bond based on the Cahn-Ingold-Prelog priority rules.
Z-Gln-Gly ZQG
Synthesis of 3-Acvlidene-2-Oxoindole Derivatives
Example 1 : Synthesis of (Z)-3-(2-(5-bromopyridin-3-yl)-2-oxoethylidene)-4- chloroindolin-2-one
Figure imgf000046_0001
[0133] This example demonstrates the synthesis of a compound of the technology wherein R2 is substituted pyridine, wherein the substituted pyridine group may further be reacted to provide other substituted pyridine compounds of the present technology. 4- chloroindoline-2,3-dione (1.0 g, 5.1 mmol) was suspended in 20 mL anhydrous ethanol and l -(5-bromopyridin-3-yl)ethanone (1.21 g, 6.06 mmol) was added, followed by diethylamine (0.63 mL, 6.06 mmol) which was slowly dropped into the solution while the solid dissolved and the solution turned dark red/brown. Within a few minutes, a large amount of solid precipitated, leaving a yellow solution behind. After 2 hours of additional stirring, the mixture was triturated with 50 mL of n-pentane and all precipitate was filtered off, washed with diethyl ether (2 X 15 mL) and dried under vacuum, furnishing 3- (2-(5-bromopyridin-3-yl)-2-oxoethyl)-4-chloro-3-hydroxyindolin-2-one (1 .660 g, 4.35 mmol, 79 % yield) as a light grey solid that was used without further purification.
[0134] A sample of 3-(2-(5-bromopyridin-3-yl)-2-oxoethyl)-4-chloro-3-hydroxyindolin- 2-one ( 1.4 g, 3.67 mmol) was dissolved in 10 mL glacial acetic acid and 5 mL cone, hydrochloric acid. The solution was heated to boiling with a heatgun for 15 minutes when TLC (DCM:MeOH = 9: 1 ) showed complete conversion of the starting material. The reaction mixture was diluted and carefully neutralized with saturated sodium bicarbonate, precipitating the crude product. The solid was filtered, washed with deionized water (2 X 40 mL) and cold diethyl ether ( 1 X 20 mL). The filter cake was dissolved in acetone (50 mL) and the solution triturated with n-pentane (80 mL) and DCM (50 mL), until a dark brown solid precipitated. Anhydrous sodium sulfate was added to dry the organic solution. Then, all solids were filtered off and the volatiles were removed from the filtrate under reduced pressure, yielding (Z)-3-(2-(5-bromopyridin-3-yl)-2-oxoethylidene)-4- chloroindolin-2-one ( 1.12 g, 3.08 mmol, 84 % yield) as yellow-orange, fluffy needles of analytical purity of the bromo-derivative.
10135] Ή (400 MHz, DMSO-d6): δ 10.93 (s, 1 H), 9.02 (d, J = 1 .6 Hz, 1 H), 8.95 (d, J = 2.2 Hz, 1 H), 8.46 (dd, J = 2.2, 1 .6 Hz, 1 H), 7.86 (s, 1 H), 7.33 (dd, J = 8.2, 7.7 Hz, 1 H), 7.09 (d, J = 8.2 Hz, 1 H), 6.86 (d, J = 7.7 Hz, 1 H). 13C (100 MHz, DMSO-de): δ 193.6, 165.9, 154.2, 147.9, 145: 1 , 137.7, 133.9, 132.8, 132.1 , 131 .8, 129.6, 122.9, 120.6, 1 17.8, and 109.3. Low-resolution ES+ MS: 364.77 (M+H)
Example 2: Synthesis of (Z)-4-chloro-3-(2-oxo-2-(5-phenylpyridin-3- yl)ethylidene)indolin-2-one
Figure imgf000047_0001
[0136] This example demonstrated the synthesis of a compound wherein R.2 is an aryl substituted pyridine. A sample of (Z)-3-(2-(5-bromopyridin-3-yl)-2-oxoethylidene)-4- chloroindolin-2-one (20 mg, 0.055 mmol), phenylboronic acid (10 mg, 0.083 mmol), cesium carbonate (53.8 mg, 0. 1 7 mmol) and PdCi2(dppf)-CH2Cl2Adduct (4.49 mg, 5.50 μηιοΐ) were placed in a glass vial which was sealed with a septum and subsequently degassed and backfilled with nitrogen three times. Then, 1.5 mL of toluene and 0.5 mL of deionized water were added and the mixture was heated to 80 °C under vigorous stirring for 16 hours. After cooling to room temperature, the mixture was passed through a pad of silica gel topped by anhydrous sodium sulfate. The silica was washed well with ethyl acetate. All volatiles were removed from the filtrate under reduced pressure and the crude product subjected to purification by column chromatography, furnishing the (Z)-4-chloro- 3-(2-oxo-2-(5-phenylpyridin-3-yl)ethylidene)indolin-2-one in 50 % yield. LC-MS (low resolution ES+): 360.98 (35C1-M + H), 362.97 (37C1-M + H).
Example 3: A general procedure for making N-substituted 4-chloro-isatins (procedure A)
[0137] 4-Chloro-isatin (1 eq.) was dissolved in DMF and potassium carbonate (3 eq.) added, followed by 2 eq. of the desired alkylating agent (alkyl bromides or iodides). The mixture was stirred for 24 hours and the conversion monitored by TLC. If necessary, additional alkylating agent was added and stirring continued until all 4-chloro-isatin reactant had disappeared. The product could be precipitated by diluting the reaction mixture tenfold with water and isolated by filtration. The products were sufficiently pure by 1 H NMR and used without.any further purification.
General two-step procedure for synthesis of 3-acylidene-2-oxoindole derivatives (procedure B)
[0138] An isatin (1 eq.) with the desired substitution pattern or N-alkylation (see general procedure above) was condensed with the desired methyl ketone (1 eq.) using diethylamine (1 eq.) in anhydrous ethanol for 2-48 hours (most reactions were complete after 2-3 h). The condensation product could be precipitated in quantitative yield using pentane and isolated by filtration. Without further purification, it was subjected to a 4: 1 mixture of glacial acetic acid with concentrated hydrochloric acid and carefully heated to boiling with a heatgun for 20-30 min. The acidic mixture was diluted tenfold with water and neutralized with sodium hydroxide and sodium bicarbonate, whereby the highly colored 3-acylidene-2-oxoindole products precipitated. After filtration and drying, the crude products were dissolved in a minimal amount of acetone and DCM. The solution was gradually triturated with pentane and any precipitating dark impurities filtered off. The final product either crystallized from this solution or could be obtained by removing the volatiles. In most cases, the product was sufficiently pure without any further effort. If necessary, a small quantity of product could be purified by preparative TLC on 1 mm glass-backed silica gel plates, typically using 3:2 ethyl acetate - pentane as developing solvent and 100% ethyl acetate as eluent.
(Z)-[2,3'-biindolinylidene]-2',3-dione (7)
[01391 Isatin (0.30 g, 2.0 mmol), l H-indol-3-yl acetate (0.36 g, 2.0 mmol) and sodium carbonate (0.54 g, 5.1 mmol) were stirred in 15 mL anhydrous methanol. After 48 hours, a dark purple precipitate had formed which was collected by filtration and extensively washed with methanol and water to remove any unreacted materials, yielding (Ε)-[2,3'- biindolinylidene]-2',3-dione (0.50 g, 1.91 mmol, 94 % yield) in analytically pure form. Ή NMR (400 MHz, DMSO- ) δ 1 1.02 (br s, 2H), 8.77 (d, J = 7.2 Hz, 1 H), 7.65 (d, J = 7.6 Hz, 1H), 7.58 (t, J = 7.7 Hz, 1H), 7.42 (d, J = 8.1 Hz, 1H), 7.25 (t, J = 7.6 Hz, 1 H), 7.02 (t, J = 7.5 Hz, 2H), 6.90 (d, J = 7.7 Hz, 1 H). 13C NMR (101 MHz, DMSO-</6) δ 188.76, 171.12, 152.63, 141.25, 138.39, 137.15, 129.32, 124.69, 124.40, 121.58, 121.26, 121.23, 1 19.07, 1 13.50, 109.67, 106.71. ESI-MS (ES-) m/z calcd for C]6HioN202, 262.0742, found, 261.2781 (M-H)- (E)-[3,3'-biindolinylidene]-2,2'-dione (8)
[0140] Isatin (1.5 g, 10 mmol) and indolin-2-one (1 ,35 g, 10 mmol) were placed in a round-bottomed flask and suspended in 30 mL glacial acetic acid and 0.5 mL
concentrated hydrochloric acid. The mixture was heated to reflux, while turning into a dark red solution. After 3 hours, the solution was allowed to cool, while a solid precipitated. This solid was filtered off and washed thoroughly with methanol, water, sodium bicarbonate solution and eventually pentane to yield (E)-[3,3'-biindolinylidene]- 2,2'-dione (2.4 g, 9.0 mmol, 88 % yield) in analytically pure form. Ή NMR (400 MHz, DMSO-e ) δ 10.90 (s, 1H), 9.05 (d, J = 7.9 Hz, 1H), 7.34 (t, J = 7.6, 1H), 6.96 (d, J = 7.7 Hz, 1 H), 6.84 (d, J = 7.7 Hz, 1H). , 3C NMR (101 MHz, DMSO-cfe) δ 169.65, 144.75, 134.02, 133.30, 129.99, 122.36, 121.82, 1 10.21. ESI-MS (ES+) m/z calcd for C16HioN202, 262.0742, found, 285.1973 (M+Na)+
(E)-4-chloro-3-(2-oxopropylidene)indolin-2-one ( 10)
[0141] To a solution of 4-chloro isatin (500 mg, 2.75 mmol) in acetone (5 mL) was added diethylamine (314 μί, 3.02 mmol) at room temperature. The mixture was heated to 50 °C for 14 hours with magnetic stirring and then concentrated under reduced pressure. The resulting alcohol intermediate was triturated in diethyl ether with sonication, filtered and the resulting white solids were used without further purification. To a solution of the alcohol intermediate dissolved in ethanol (3 mL) was added concentrated hydrochloric acid (1 mL). The mixture was stirred at room temperature for 14 hours and then quenched with a solution of saturated sodium bicarbonate (5 mL). The mixture was extracted with dichloromethane (20 mL), concentrated and purified by silica gel flash chromatography using a gradient of 30 - 80% ethyl acetate in hexanes to afford the target compound 10 as a yellow solid. Ή NMR (500 MHz, DMSO-cfe) δ 10.97 (s, 1H), 7.57 (s, 1H), 7.30 (t, J = 8.0 Hz, 1H), 7.06 (d, J= 8.2 Hz, 1H), 6.85 (d, J= 7.3 Hz, 1H), 2.40 (s, 3H).13C NMR (126 MHz, DMSO-</6) δ 203.08, 166.07, 144.55, 137.99, 131.80, 129.06, 128.13, 122.82, 117.63, 109.15, 30.16. ESI-MS (ES+) m/z calcd for CnH8ClN02, 221.0244, found, 222.13 (M+H)+
(E)-5-chloro-3-(2-oxopropylidene)indolin-2-one (11)
[0142] Compound 11 was prepared according to the method used to prepare compound 10. Ή NMR (400 MHz, DMSO-i6) δ 10.89 (s, 1H), 8.31 (d,J=2.2 Hz, 1H), 7.42 (dd,J = 8.4,2.3 Hz, 1H), 7.12 (s, 1H), 6.88 (d,J=8.3Hz, 1H),2.47 (s, 3H). ,3CNMR(101 MHz, DMSO-i/e) δ 199.45, 168.25, 143.74, 133.80, 132.47, 129.47, 126.67, 125.53, 121.44, 111.70, 32.10. ESI-MS (ES-) m/z calcd for CnH8ClN02, 221.0244, found, 220.2041 (M-H)"
(E)-6-fluoro-3 -(2-oxopropylidene)indolin-2-one ( 12)
[0143] Compound 12 was prepared according to the method used to prepare compound 10. Ή NMR (400 MHz, DMSO-</6) δ 10.78 (s, 1H), 8.09 (dd, J= 9.7, 2.7 Hz, 1H), 7.23 (td,J= 9.0, 2.8 Hz, 1H), 7.10 (s, 1H), 6.85 (dd, J = 8.6, 4.5 Hz, 1H), 2.47 (s, 3H).13C NMR (101 MHz, DMSO-<6) δ 199.41, 168.50, 157.36 (d,J= 235.5 Hz), 141.38, 134.44, 129.22, 120.85 (d,J=9.9 Hz), 119.45 (d, J= 23.9 Hz), 114.03 (d, J= 26.5 Hz), 111.04 (d,J= 8.2 Hz), 32.11. ESI-MS (ES+) m/z calcd for C,,H8FN02, 205.0539, found, 228.1752 (M+Na)+
(E)-3-(2-oxopropylidene)-6-(trifluoromethoxy)indolin-2-one (13)
[0144] Compound 13 was prepared according to the method used to prepare compound 10. Ή NMR (400 MHz, DMSO-efe) δ 10.95 (s, 1H), 8.32 (s, 1H), 7.38 (dd, J= 8.5, 2.0 Hz, 1H), 7.13 (s, 1H), 6.94 (d,J=8.5 Hz, 1H), 2.47 (s, 3H).13CNMR(126 MHz, DMSO-c6)0199.45, 168.52, 144.07, 142.74, 134.06, 129.54, 126.12, 120.90, 120.32, 120.27 (q, J =255.5 Hz), 111.16, 32.17. ESI-MS (ES-) m/z calcd for Ci2H8F3N03, 271.0456, found, 270.22 (M-H)- (E)-7-chloro-3-(2-oxopropylidene)indolin-2-one (14) [0145] Compound 14 was prepared according to the method used to prepare compound 10. Ή NMR (400 MHz, DMSO-i/6) δ 1 1.18 (s, 1H), 8.22 (d, J = 1.1 Hz, 1 H), 7.42 (dd, J = 8.2, 0.7 Hz, 1 H), 7.12 (s, 1 H), 7.00 (t, J = 8.0 Hz, 1 H), 2.46 (s, 3H). 13C NMR (101 MHz, DMSO-ί ή) δ 199.19, 168.46, 142.28, 133.90, 132.48, 129.69, 125.62, 122.90, 121.80, 1 14.46, 32.07. ESI-MS (ES+) m/z calcd for CnHgClN02, 221.0244, found, 244.1 177(M+Na)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-2-yl)ethylidene)indolin-2-one (15)
[0146] Compound 15 was prepared by general procedure B (1.1 g, 47 % for two steps). Ή NMR (500 MHz, DMSO-c/6) δ 10.86 (s, 1H), 8.65 (dt, J = 4.7, 1.3 Hz, 1H), 8.06 (d, J = 1.3 Hz, 1 H), 8.05 (dd, J = 2.7, 1.3 Hz, 1H), 8.02 (s, 1 H), 7.66 (ddd, J = 5.9, 4.8, 3.1 Hz, 1H), 7.32 (t, J = 8.0 Hz, 1 H), 7.10 (dd, J = 8.2, 0.8 Hz, 1H), 6.85 (dd, J = 7.8, 0.8 Hz, 1 H). I3C NMR (101 MHz, DMSO- 6) δ 195.49, 165.97, 153.06, 149.21 , 144.65, 138.05, 137.61 , 131.81 , 130.22, 129.12, 127.94, 122.83, 121.59, 1 17.71 , 109.20. ESI-MS (ES+) m/z calcd for C5H9CIN2O2, 284.0353, found, 307.1844 (M+Na)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (16)
[0147] Compound 16 was prepared by general procedure B (3.2 g, 67 % for two steps). Ή NMR (400 MHz, DMSO-i 6) δ 10.92 (s, 1H), 9.04 (dd, J = 2.2, 0.9 Hz, 1H), 8.80 (dd, J = 4.8, 1.7 Hz, 1 H), 8.26 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 7.92 (s, 1 H), 7.57 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.34 (t, 8.0 Hz, 1 H), 7.1 1 (dd, J = 8.2, 0.8 Hz, 1H), 6.86 (dd, J = 7.8, 0.8 Hz, 1 H). 13C NMR ( 101 MHz, DMSO-cfe) δ 194.51 , 165.80, 153.65, 149.50, 144.95,
135.83, 134.81 , 132.06, 131.25, 131.21 , 129.46, 124.00, 122.85, 1 17.65, 109.21. ESI-MS (ES+) m/z calcd for Ci5H9ClN202, 284.0353, found, 307.1476 (M+Na)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-4-yl)ethylidene)indolin-2-one ( 17)
[0148] Compound 17 was prepared by general procedure B (1.83 g, 58 % for two steps). Ή NMR (400 MHz, DMSO-c 6) δ 10.96 (s, 1H), 8.79 (dd, J = 4.4, 1.7 Hz, 2H), 7.90 (s, 1 H), 7.79 (dd, J = 4.4, 1.7 Hz, 2H), 7.34 (t, J = 8.0 Hz, 1 H), 7.10 (dd, J = 8.2, 0.8 Hz, 1 H), 6.86 (dd, J = 7.8, 0.8 Hz, 1 H). I 3C NMR (101 MHz, DMSO-ek) δ 195.21,
165.84, 150.82, 145.07, 141.53, 134.65, 132.24, 131.70, 129.56, 122.92, 121.39, 1 17.56, 109.33. ESI-MS (ES+) m/z calcd for C15H9CIN2O2, 284.0353, found, 307.1735 (M+Na)+ (E)-3-(2-(5-bromopyridin-3-yl)-2-oxoethylidene)-4-chloroindolin-2-one (18)
[0149] Compound 18 was prepared by general procedure B (3.81 g, 66 % for two steps). Ή NMR (400 MHz, DMSO-cfe) δ 10.93 (s, 1 H), 9.02 (d, J = 1.6 Hz, 1 H), 8.95 (d, J = 2.2 Hz, 1 H), 8.46 (t, J = 1.9 Hz, 1 H), 7.86 (s, 1 H), 7.33 (t, J = 8.0 Hz, 1 H), 7.09 (d, J = 8.2 Hz, 1H), 6.86 (d, J = 7.7 Hz, 1H). 13C NMR (101 MHz, DMSO-i/6) δ 193.57, 165.91 , 154.22, 147.93, 145.09, 137.73, 133.89, 132.80, 132.13, 131.77, 129.57, 122.85, 120.63, 1 17.75, 109.25. ESI-MS (ES+) m/z calcd for C15H8BrClN202, 361.9458, found, 385.0456 (M+Na)+
(E)-4-chloro-3-(2-(6-methoxypyridin-3-yl)-2-oxoethylidene)indolin-2-one (19)
[0150] To a solution of isatin (100 mg, 0.68 mmol) in ethanol (2 mL) was added l -(6- methoxypyridin-3-yl)efhanone (1 12 mg, 0.74 mmol), followed by diethylamine (77 μί, 0.74 mmol). The mixture was stirred at room temperature for 2 hours then concentrated under reduced pressure. The resulting alcohol intermediate was triturated in diethyl ether with sonication, filtered and the resulting white solid was used without further purification. To a solution of the alcohol intermediate dissolved in ethanol (5 mL) was added concentrated hydrochloric acid (1 mL). The mixture was stirred at room
temperature for 14 hours and then quenched with a solution of saturated sodium bicarbonate (5 mL). The mixture was extracted with dichloromethane (20 mL), concentrated and purified by silica gel chromatography using a gradient of 30 - 80% ethyl acetate in hexanes to afford the target compound as a yellow. Ή NMR (400 MHz, DMSO-i¾ 5 10.90 (s, 1H), 8.71 (d, 7 = 1.8 Hz, 1H), 8.17 (dd, 7 = 8.7, 2.5 Hz, 1 H), 7.86 (s, 1 H), 7.33 (t, 7 = 8.0 Hz, 1H), 7.09 (d, 7= 8.2 Hz, 1H), 6.95 (d, 7= 8.7 Hz, 1H), 6.85 (d, 7 = 7.1 Hz, 1H), 3.94 (s, 3H). 13C NMR (101 MHz, DMSO-i¾ δ 192.83, 166.36, 165.75, 149.59, 144.81 , 138.70, 135.06, 131.94, 130.78, 129.36, 126.17, 122.82, 1 17.75, 1 10.96, 109.13, 54.03. ESI-MS (ES+) m/z calcd for Ci6Hi iClN203, 314.0458, found, 337.1444 (M+Na)+
(E)-4-chloro-3-(2-oxo-2-phenylethylidene)indolin-2-one (20)
[0151 ] Compound 20 was prepared according to the method used to prepare compound 19. Ή NMR (400 MHz, DMSO-i/6) δ 10.89 (s, 1H), 7.94 (s, 1H), 7.92 (dd, 7 = 8.3, 1.2 Hz, 2H), 7.66 (t, 7 = 7.4 Hz, 1H), 7.53 (t, 7 = 7.7 Hz, 2H), 7.33 (t, 7 = 8.0 Hz, 1 H), 7.10 (dd, 7 = 8.2, 0.7 Hz, 1 H), 6.85 (dd, 7= 7.8, 0.7 Hz, 1 H). 13C NMR (101 MHz, DMSO^) δ 194.84, 165.69, 144.76, 136.30, 135.67, 133.67, 131.90, 130.50, 129.27, 128.82, 128.50, 122.80, 1 17.68, 109.13. ESI-MS (ES+) m/z calcd for C,6H10ClNO2, 283.04, found, 306.1557 (M+Na)+
(E)-4-chloro-3-(2-(3-chlorophenyl)-2-oxoethylidene)indolin-2-one (21 )
[0152] Compound 21 was prepared by general procedure B (1.1 g, 36 % for two steps).
Ή NMR (400 MHz, DMSO-i 6) δ 10.92 (s, 1 H), 7.91 (s, 1H), 7.90 - 7.85 (m, 2H), 7.73 (ddd, J = 8.0, 2.2, 1.1 Hz, 1 H), 7.61 - 7.52 (m, 1 H), 7.38 - 7.29 (t, J = 8.0 Hz, 1H), 7.10 (dd, J = 8.2, 0.8 Hz, 1H), 6.86 (dd, J = 7.8, 0.8 Hz, 1H). 13C NMR (101 MHz, DMSO-i/5) δ 193.95, 165.77, 144.90, 137.60, 135.24, 133.70, 133.32, 132.02, 131.10, 130.88, 129.43, 127.64, 127.31 , 122.84, 1 17.67, 109.21. ESI-MS (ES+) m/z calcd for
C16H9CI2NO2, 317.0010, found, 340.1020 (M+Na)+
(E)-4-chloro-3-(2-(4-chlorophenyl)-2-oxoethylidene)indolin-2-one (22)
[0153] Compound 22 was prepared by general procedure B (2.6 g, 67 % for two steps). Ή NMR (400 MHz, DMSO-<4) δ 10.91 (s, 1H), 7.95 - 7.91 (m, 2H), 7.90 (s, 1 H), 7.62 - 7.55 (m, 2H), 7.37 - 7.28 (t, J= 8.0 Hz, 1H), 7.09 (dd, J = 8.2, 0.8 Hz, 1H), 6.85 (dd, J = 7.8, 0.8 Hz, 1H). 13C NMR (101 MHz, DMSO-i 6) δ 193.95, 165.73, 144.84, 138.50, 135.50, 134.48, 132.00, 130.87, 130.32, 129.37, 128.95, 122.83, 1 17.64, 109.18. ESI-MS (ES+) m/z calcd for C16H9C12N02, 317.0010, found, 340.1019 (M+Na)+
(E)-4-chloro-3-(2-(2-methoxyphenyl)-2-oxoethylidene)indolin-2-one (23)
[0154] Compound 23 was prepared by general procedure B (3.0 g, 96 % for two steps). Ή NMR (400 MHz, DMSO-c/6) δ 10.78 (s, 1H), 7.94 (d, J = 0.6 Hz, 1 H), 7.82 (dd, J = 7.8, 1.8 Hz, 1H), 7.60 (ddd, J = 8.4, 7.3, 1.8 Hz, 1H), 7.28 (t, J = 8 Hz, 1 H), 7.16 (d, J = 7.8 Hz, 1 H), 7.13 - 7.08 (m, 1H), 7.07 (dd, J = 8.2, 0.8 Hz, 1 H), 6.82 (dd, J = 7.8, 0.8 Hz, 1 H), 3.77 (s, 3H). 13C NMR (101 MHz, DMSO-i 6) δ 193.29, 165.96, 159.48, 144.29, 140.20, 135.02, 131.27, 130.27, 128.84, 126.03, 126.01, 122.72, 120.79, 1 18.19, 1 12.98, 109.00, 56.05. ESI-MS (ES-) m/z calcd for C17H12CINO3, 313.0506, found, 312.2631 (M- H)-
(E)-4-chloro-3-(2-(3-methoxyphenyl)-2-oxoethylidene)indolin-2-one (24)
[0155] Compound 24 was prepared by general procedure B (1.7 g, 82 % for two steps). 'H NMR (500 MHz, DMSO-</6) δ 10.90 (s, 1H), 7.92 (s, 1 H), 7.48 (dt, J = 7.6, 1.2 Hz, 1 H), 7.44 (t, J = 7.8 Hz, 1 H), 7.40 (dd, J = 2.4, 1.4 Hz, 1 H), 7.33 (t, J = 8.0 Hz, 1 H), 7.24 (ddd, J = 8.0, 2.6, 1.0 Hz, 1 H), 7.10 (dd, J = 8.2, 0.6 Hz, 1H), 6.85 (dd, J = 7.8, 0.6 Hz, 1 H), 3.82 (s, 3H). I 3C NMR ( 101 MHz, DMSO-i/6) δ 194.56, 165.71 , 159.49, 144.79, 137.07, 136.18, 131.92, 130.62, 130.09, 129.32, 122.82, 121.61 , 1 19.71 , 1 17.69, 1 12.30, 109.16, 55.40. ESI-MS (ES+) m/z calcd for C17H12CINO3, 313.0506, found, 312.2634 (ES-) (M-H)"
(E)-4-chloro-3-(2-(4-methoxyphenyl)-2-oxoethylidene)indolin-2-one (25)
[0156] Compound 25 was prepared by general procedure B (2.0 g, 77 % for two steps).
Ή NMR (400 MHz, DMSO-i/6) δ 10.87 (s, 1 H), 7.89 (s, 1 H), 7.89 - 7.85 (m, 2H), 7.32 (t, J = 8.0 Hz, 1H), 7.09 (dd, J = 8.2, 0.8 Hz, 1H), 7.07 - 7.02 (m, 2H), 6.85 (dd, J = 7.8, 0.8 Hz, 1 H), 3.84 (s, 3H). 13C NMR (101 MHz, DMSO-</6) δ 193.10, 165.66, 163.58, 144.65, 136.50, 131.79, 130.89, 130.12, 129.20, 128.81 , 122.79, 1 17.77, 1 14.09, 109.07, 55.63. ESI-MS (ES+) m/z calcd for C, 7H12C1N03> 313.0506, found, 314.1921 (M+H)+
(E)-3-(2-(3-aminophenyl)-2-oxoethylidene)-4-chloroindolin-2-one (26)
[0157] Compound 26 was prepared by general procedure B (1.3 g, 79 % for two steps). Ή NMR (400 MHz, DMSO-</6) δ 10.89 (s, 1 H), 7.88 (d, J = 0.6 Hz, 1H), 7.32 (t, J = 8.0 Hz, 1H), 7.15 (t, J = 7.9 Hz, 1 H), 7.13 - 7.1 1 (m, 1H), 7.09 (dd, J = 8.2, 0.8 Hz, 1H), 7.05 (ddd, J = 7.6, 1.6, 1.1 Hz, 1H), 6.85 (dd, J = 7.8, 0.8 Hz, 1H), 6.81 (ddd, J = 7.9, 2.4, 1.1 Hz, 1H), 5.42 (br s, 2H). 13C NMR (101 MHz, DMSO- /6) δ 195.63, 166.29, 146.07, 145.30, 137.68, 137.04, 132.45, 130.66, 129.90, 129.83, 123.42, 1 19.88, 1 18.37, 1 16.89,
1 14.35, 109.75. ESI-MS (ES+) m/z calcd for Ci6Hi iClN202) 298.0509, found, 321.1.956 (M+Na)+
(E)-3-(2-(4-aminophenyl)-2-oxoethylidene)-4-chloroindolin-2-one (27)
[0158] Compound 27 was prepared by general procedure B (1.1 g, 67 % for two steps). Ή NMR (400 MHz, DMSO-ok) δ 10.84 (s, 1H), 7.82 (d, J = 0.6 Hz, 1H), 7.66 - 7.54 (m, 2H), 7.30 (t, J = 8.0 Hz, 1 H), 7.07 (dd, J = 8.2, 0.8 Hz, 1 H), 6.83 (dd, J = 7.8, 0.8 Hz, 1 H), 6.64 - 6.53 (m, 2H), 4.57 (br s, 2H). I3C NMR (101 MHz, DMSO-i/6) δ 191.27, 165.60,
154.36, 144.39, 137.56, 131.45, 131.14, 129.24, 128.97, 123.52, 122.69, 1 17.94, 1 12.65, 108.91. ESI-MS (ES+) m/z calcd for C^HnClNzOz, 298.0509, found, 321.1579 (M+Na)+ (E)-4-chloro- 1 -methyl-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (28)
[0159] To a stirring solution of 4-chloroisatin (255 mg, 1.41 mmol) in DMF (4 mL) was added methyl iodide (175 μΐ-, 2.82 mmol), potassium carbonate (390 mg, 2.82 mmol) and catalytic potassium iodide (5 mg). The suspension was allowed to stir at room
temperature for 16 hours or until no more starting material was present as determined by TLC analysis (10% ethyl acetate/90% hexanes). The product was extracted into ethyl acetate (20 mL) from three washes with water (20 mL). After evaporating the solvent, the product was triturated in ethanol (5 mL), filtered and used without further purification.
[0160] To a suspension of 4-chloro-l -methylisatin (90 mg, 0.46 mmol) in ethanol (2 mL) was added 3-acetylpyridine (56.7 uL, 0.51 mmol), then diethylamine (53 μί, 0.51 mmol). The reaction was stirred two hours at room temperature, and then the reaction mixture was concentrated under reduced pressure. The crude product was re-dissolved in pyridine (0.5 mL) and treated with methanesulfonyl chloride (100 μί, 0.59 mmol). After stirring 1 hour at room temperature, pyridine was removed under reduced pressure at 60 °C. The crude product was then purified by silica gel chromatography using a gradient of 0 - 10% methanol in dichloromethane. Ή NMR (400 MHz, DMSO-i/6) δ 9.04 (s, 1H), 8.81 (dd, 7= 4.8, 1.6 Hz, 1 H), 8.27 (dd, J = 8.0, 1.7 Hz, 1 H), 8.00 (s, 1 H), 7.57 (dd, J = 7.9, 4.9 Hz, 1H), 7.44 (t, J = 8.1 Hz, 1H), 7.18 (d, J = 8.3 Hz, 1H), 7.07 (d, J= 7.9 Hz, 1 H), 3.08 (s, 3H). 13C NMR (126 MHz, DMSO-cfe) δ 194.39, 164.36, 153.73, 149.59, 146.05, 135.88, 135.34, 132.00, 131.26, 130.12, 129.26, 124.06, 123.45, 1 16.89, 108.22, 26.14. ESI-MS (ES+) m/z calcd for Ci6H, ,ClN202! 298.0509, found, 299.1481 (M+H)+ (E)-4-chloro-l -isopropyl-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (29)
[0161] Compound 29 was prepared by alkylating 4-chloro-isatin with isopropyl bromide using general procedure A and subjecting the product to general procedure B (200 mg, 23 % for three steps). Ή NMR (400 MHz, DMSO-i 6) δ 9.05 (dd, J = 2.2, 0.9 Hz, 1H), 8.81 (dd, J = 4.8, 1.7 Hz, 1H), 8.27 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.00 (s, 1 H), 7.58 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.40 (t, J = 8.1 Hz, 1H), 7.25 (dd, J = 8.1 , 0.7 Hz, 1 H), 7.17 (dd, J = 8.2, 0.7 Hz, 1H), 4.50 - 4.33 (m, 1H), 1.33 (d, J = 7.0 Hz, 6H). 13C NMR (101 MHz, DMSO-oW δ 194.53, 164.04, 153.72, 149.55, 144.67, 135.82, 135.35, 131.85, 131.24, 130.31 , 129.65, 124.07, 123.12, 1 17.15, 109.23, 43.80, 18.98. ESI-MS (ES+) m/z calcd for CigH,5ClN202, 326.0822, found, 349.1992 (M+Na)+
(E)-4-chloro-l -isobutyl-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (30)
[0162) Compound 30 was prepared by alkylating 4-chloro-isatin with isobutyl bromide using general procedure A and subjecting the product to general procedure B (330 mg, 30 % for three steps). Ή NMR (400 MHz, DMSO- 6) δ 9.04 (dd, J = 2.3, 0.9 Hz, 1 H), 8.79 (dd, J = 4.8, 1.7 Hz, 1H), 8.27 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.00 (s, 1 H), 7.56 (ddd, J = 8.0, 4.8, 0.9 Hz, 1 H), 7.41 (t, J = 8.1 Hz, 1H), 7.15 (ddd, J = 13.5, 8.1 , 0.7 Hz, 2H), 3.41 (d, J = 7.4 Hz, 2H), 2.08 - 1.80 (m, 1 H), 0.80 (d, J = 6.7 Hz, 6H). 13C NMR (101 MHz, DMSO-oy a 194.38, 164.58, 153.70, 149.54, 145.69, 135.81 , 135.39, 131.95, 131.20, 130.07, 129.45, 124.02, 123.37, 1 16.85, 108.62, 46.75, 26.65, 19.83. ESI-MS (ES+) m/z calcd for CsHnCl^C^, 340.0979, found, 341.2502 (M+H)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)- 1 -phenylindolin-2-one (31 )
[0163| Compound 31 was prepared by a Chan-Lam arylation of compound 16. For this, (Z)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (200 mg, 0.703 mmol), phenylboronic acid (90 mg, 0.738 mmol) and copper(II) acetate (134 mg, 0.738 mmol) were placed in a vial and suspended in 10 mL anhydrous DCM to which pyridine (1 14 μΐ, 1.405 mmol) was added. The mixture was stirred for 20 h before it was diluted with 40 mL ethyl acetate and washed five times 15 mL aliquots of aqueous sodium chloride (5 x 15 mL). The organic layer phase was dried over sodium sulfate and the volatiles removed under reduced pressure. The crude product was spotted on a 1 mm glass backed silica gel plate which was developed in 3:2 ethyl acetate - pentane. The bright yellow pure product (E)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)-l -phenylindolin-2-one (25 mg, 0.069 mmol, 9.9 % yield) could be isolated with ethyl acetate at Rf = 0.4. Ή NMR (400 MHz, CDCl3) 5 9.19 (d, J = 1.7 Hz, 1 H), 8.78 (dd, J = 4.8, 1.5 Hz, 1H), 8.39 - 8.31 (m, 1 H), 7.97 (s, 1 H), 7.50 - 7.42 (m, 3H), 7.37 (t, J = 7.4 Hz, 1H), 7.32 (dd, J = 8.2, 0.9 Hz, 2H), 7.24 (t, J = 8.0 Hz, 1H), 7.13 (dd, J = 8.2, 0.7 Hz, 1H), 6.77 (d, J = 7.8 Hz, 1H). 13C NMR (101 MHz, C,DCl3) 5 194.1 1 , 164.41 , 153.75, 150.24, 146.06, 136.00, 135.12, 133.31 , 131.86, 131.58, 131.55, 131.32, 129.83, 128.69, 126.54, 124.73, 124.13, 1 17.54, 108.62. ESI-MS (ES+) m/z calcd for C21H,3C1N202, 360.0666, found, 383.1792 (M+Na)+ (E)-4-chloro-l -(cyclohexylmethyl)-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (32)
[0164] Compound 32 was prepared according to the method used to prepare compound 28. Ή NMR (400 MHz, DMSO-cfe) δ 9.04 (dd, / = 2.2, 0.8 Hz, 1 H), 8.80 (dd, J = 4.8, 1.7 Hz, 1H), 8.37 - 8.17 (m, 1H), 8.00 (s, 1 H), 7.56 (ddd, J = 8.0, 4.8, 0.8 Hz, 1 H), 7.41 (t, J = 8.1 Hz, 1H), 7.14 (dd, J= 14.9, 7.8 Hz, 2H), 3.43 (d, J = 7.2 Hz, 2H), 1.90 - 1.25 (m, 6H), 1.26 - 0.98 (m, 3H), 0.98 - 0.73 (m, 2H). 13C NMR (101 MHz, DMSO-i 6) δ 194.37,
164.54, 153.70, 149.53, 145.75, 135.80, 135.37, 131.95, 131.22, 130.08, 129.43, 124.00, 123.32, 1 16.83, 108.61 , 45.53, 35.76, 30.12, 25.76, 25.15. ESI-MS (ES+) m/z calcd for C22H21C1N202, 380.1292, found, 381.2503 (M+H)+
(E)- l-benzyl-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (33)
[0165] Compound 33 was prepared according to the method used to prepare compound 28. Ή NMR (400 MHz, DMSO-< ) δ 9.09 (dd, J = 2.2, 0.9 Hz, 1 H), 8.82 (dd, J= 4.8, 1.7 Hz, 1 Η),·8.31 (ddd, J = 8.0, 2.2, 1.7 Hz, 1 H), 8.07 (s, 1 H), 7.58 (ddd, J = 8.0, 4.8, 0.9 Hz, 1 H), 7.36 (t, J= 8.1 Hz, 1 H), 7.33 - 7.21 (m, 5H), 7.16 (dd, J= 8.3, 0.8 Hz, 1 H), 6.99 (dd, J= 7.9, 0.8 Hz, 1H), 4.85 (s, 2H). I 3C NMR (101 MHz, DMSO-cfe) δ 194.26, 164.48, 153.77, 149.61 , 144.93, 135.89, 135.88, 135.79, 131.91 , 131.20, 129.92, 129.51 , 128.69,
127.55, 127.19, 124.05, 123.65, 1 17.06, 108.69, 42.81. ESI-MS (ES+) m/z calcd for C22H15C1N202, 374.0822, found, 375.2066 (M+H)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)-l -phenethylindolin-2-one (34) [0166] Compound 34 was prepared by alkylating 4-chloro-isatin with (2- bromoethyl)benzene using general procedure A and subjecting the product to general procedure B (400 mg, 50 % for three steps). Ή NMR (400 MHz, DMSO-i 6) δ 9.02 (d, J = 1.7 Hz, 1 H), 8.82 (dd, J = 4.8, 1.6 Hz, 1H), 8.21 (dt, J = 8.0, 1.9 Hz, 1 H), 7.99 (s, 1H), 7.58 (dd, J = 7.8, 5.0 Hz, 1H), 7.40 (t, J = 8.1 Hz, 1 H), 7.29 - 7.09 (m, 7H), 3.83 (t, J = 7.4 Hz, 2H), 2.82 (t, J = 7.4 Hz, 2H). 13C NMR (101 MHz, DMSO-c 6) δ 194.28, 164.14, 153.62, 149.43, 145.1 1 , 137.99, 135.93, 135.45, 131.95, 131.18, 129.91 , 129.39, 128.76, 128.35, 126.45, 124.09, 123.39, 1 16.79, 108.50, 40.96, 32.75. ESI-MS (ES+) m/z calcd for C23H,7C1N202, 388.0979, found, 41 1.21 (M+Na)+
(E)-4-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)-l-(3-phenylpropyl)indolin-2-one (35)
[0167] Compound 35 was prepared by alkylating 4-chloro-isatin with (3- bromopropyl)benzene using general procedure A and subjecting the product to general procedure B (500 mg, 74 % for three steps). Ή NMR (400 MHz, DMSO-cfe) δ 9.06 (dd, J = 2.2, 0.8 Hz, 1H), 8.80 (dd, J = 4.8, 1.7 Hz, 1 H), 8.28 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 7.99 (s, 1 H), 7.57 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.42 (t, J = 8.1 Hz, 1H), 7.25 - 7.08 (m, 7H), 3.63 (t, J = 7.2 Hz, 2H), 2.53 (t, J = 7.3 Hz, 2H), 1.86 - 1.77 (m, 2H). I3C NMR (101 MHz, DMSO- 6) 6 194.34, 164.31, 153.61 , 149.49, 145.28, 141.07, 135.97, 135.31 , 131.98, 131.27, 130.16, 129.46, 128.27, 128.17, 125.84, 124.09, 123.37, 1 17.00, 108.32, 39.52, 32.28, 28.53. ESI-MS (ES+) m/z calcd for C24H19C1N202, 402.1 135, found, 403.30 (M+H)+
(E)-2-(4-chloro-2-oxo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-l -yl)-N,N- dimethylacetamide (36)
[0168] Compound 36 was prepared according to the method used to prepare compound 28. Ή NMR (400 MHz, DMSO-i/6) δ 9.04 (dd, J = 2.2, 0.8 Hz, 1H), 8.81 (dd, J = 4.8, 1.7 Hz, 1 H), 8.27 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.06 (s, 1H), 7.59 (ddd, J = 8.0, 4.9, 0.9 Hz, 1 H), 7.39 (t, J = 8.1 Hz, 1H), 7.17 (dd, 7 = 8.2, 0.7 Hz, 1 H), 7.00 (dd, J = 8.0, 0.7 Hz, 1H), 4.57 (s, 2H), 3.01 (s, 3H), 2.79 (s, 3H). 13C NMR (101 MHz, DMSO-i/6) δ 194.24, 165.28, 164.65, 153.53, 149.31 , 145.89, 135.94, 135.59, 131.85, 131.41 , 129.93, 129.33, 124.13, 123.37, 1 16.74, 108.84, 41.33, 35.77, 35.19. ESI-MS (ES+) m/z calcd for C9H16CIN3O3, 369.088, found, 392.18 (M+Na)+
(E)-ethyl 2-(4-chloro-2-oxo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-l -yl)acetate (37)
[0169] Compound 37 was prepared by alkylating 4-chloro-isatin with ethyl iodoacetate using general procedure A and subjecting the product to general procedure B (140 mg, 19 % for three steps). Ή NMR (400 MHz, DMSO- e) δ 9.04 (dd, J = 2.3, 0.8 Hz, 1 H), 8.81 (dd, J = 4.8, 1.7 Hz, 1H), 8.26 (ddd, J = 8.0, 2.2, 1.8 Hz, 1H), 8.1 1 (s, 1 H), 7.58 (ddd, J = 8.1 , 4.8, 0.8 Hz, 1 H), 7.43 (t, J = 8.1 Hz, 1 H), 7.22 (dd, J = 8.2, 0.5 Hz, 1 H), 7.12 (dd, J = 7.9, 0.7 Hz, 1 H); 4.54 (s, 2H), 4.10 (q, J = 7.2 Hz, 2H), 1.16 (t, J = 7.1 Hz, 3H). 13C NMR (101 MHz, OMSO- 6) 8 194.12, 167.51 , 164.42, 153.83, 149.55, 144.91, 136.32, 135.79, 132.01 , 129.50, 129.40, 124.08, 123.83, 1 16.83, 1 16.79, 108.59, 61.29, 41.16, 13.97. ESI- MS (ES+) m/z calcd for Ci9H15ClN204, 370.072, found, 393.2094 (M+Na)+
(E)-methyl 3-((4-chloro-2-oxo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-l- yl)methyl)benzoate (38)
[0170] Compound 38 was prepared by alkylating 4-chloro-isatin with methyl 3- (bromomethyl)benzoate using general procedure A and subjecting the product to general procedure B. Ή NMR (500 MHz, CDC13) δ 9.20 (s, 1H), 8.82 (s, 1H), 8.36 (dt, J = 8.0, 1.9 Hz, 1 H), 7.97 - 7.89 (m, 3H), 7.48 (dd, J = 7.9, 4.8 Hz, 1H), 7.45 - 7.33 (m, 2H), 7.19 (t, J = 8.1 Hz, 1H), 7.05 (dd, J = 8.3, 0.8 Hz, 1H), 6.62 (dd, J = 7.9, 0.8 Hz, 1H), 4.86 (s, 2H), 3.90 (s, 3H) 13C NMR (101 MHz, CDC13) δ 194.04, 166.69, 165.07, 154.15, 150.53, 144.95, 135.77, 135.57, 134.92, 131.87, 131.69, 131.40, 131.22, 130.87, 129.34, 129.31 , 128.49, 124.49, 124.05, 1 17.64, 108.09, 52.41 , 43.69. ESI-MS (ES+) m/z calcd for C24H17CIN2O4, 432.0877, found, 433.2022 (M+H)+
(E)-methyl 4-((4-chloro-2-oxo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin- 1 - yl)methyl)benzoate (39)
[0171] Compound 39 was prepared by alkylating 4-chloro-isatin with methyl 3- (bromomethyl)benzoate using general procedure A and subjecting the product to general procedure B. 'H NMR (400 MHz, CDC13) δ 9.18' (s, 1H), 8.81 (d, J = 3.8 Hz, 1 H), 8.39 - 8.31 (m, 1 H), 7.98 - 7.94 (m, 2H), 7.93 (s, 1 H), 7.47 (ddd, J = 7.9, 4.8, 0.6 Hz, 1 H), 7.28 - 7.24 (m, 2H), 7.17 (t, J = 8.1 Hz, 1 H), 7.06 (dd; J = 8.3, 0.8 Hz, 1 H), 6.58 (dd, J = 7.9, 0.8 Hz, 1 H), 4.86 (s, 2H), 3.88 (s, 3H). I 3C NMR (101 MHz, CDC13) δ 193.99, 166.64, 165.03, 154.18, 150.54, 144.88, 140.09, 135.72, 135.03, 131.70, 131.66, 131.32, 131.23, 130.32, 129.97, 127.28, 124.52, 124.04, 1 17.62, 108.05, 52.32, 43.68. ESI-MS (ES+) m/z calcd for C24Hi7ClN204, 432.0877, found, 433.2022 (M+H)+
(E)-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (40)
[0172} Compound 40 was prepared according to the method used to prepare compound 19. Ή NMR (400 MHz, DMSO- d6) δ 10.84 (s, 1 H), 9.20 (dd, J = 2.3, 0.8 Hz, 1 H), 8.85 (dd, J = 4.8, 1.7 Hz, 1 H), 8.42 (ddd, J= 8.0, 2.3, 1.7 Hz, 1 H), 8.16 (d, J = 7.3 Hz, 1 H), 7.71 (s, 1 H), 7.63 (ddd, 7 = 8.0, 4.8, 0.9 Hz, 1 H), 7.37 (td, 7 = 7.7, 1.2 Hz, 1 H), 6.97 (td, J = 7.7, 1.1 Hz, 1 H), 6.89 (d, J = 7.8 Hz, 1 H). 13C NMR (101 MHz, DMSO- d6) δ 190.34, 168.17, 153.92, 149.57, 145.28, 137.34, 136.1 1 , 133.44, 132.70, 127.13, 124.79, 124.18,
121.85, 1 19.91 , 1 10.46. ESI-MS (ES+) m/z calcd for Ci5H,oN202, 250.0742, found, 251.1892 (M+H)+
(E)-4-bromo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (41)
(0173] Compound 41 was prepared by general procedure B. Ή NMR (400 MHz, DMSO-</6) δ 10.92 (s, 1 H), 9.04 (dd, J = 2.3, 0.9 Hz, 1H), 8.80 (dd, J = 4.8, 1.7 Hz, 1H), 8.26 (ddd, J = 8.0, 2.2, 1.7 Hz, 1H), 8.05 (d, J = 0.7 Hz, 1H), 7.57 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.29 - 7.23 (m, 2H), 6.90 (dd, J = 5.8, 2.9 Hz, 1H). I3C NMR ( 101 MHz, DMSO-i/6) 5 194.41 , 165.83, 153.64, 149.47, 145.32, 135.84, 134.16, 132.21 , 132.05, 131.25, 126.09, 124.03, 1 19.1 1 , 1 18.07, 109.70. ESI-MS (ES+) m/z calcd for
Ci5H9BrN202, 327.9847, found, 329.1079 (M+H)+
(E)-5-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (42)
[0174] Compound 42 was prepared by general procedure B (2.9 g, 77 % for two steps). Ή NMR (400 MHz, DMSO-rfe) δ 1 1.01 (s, 1H), 9.23 (d, J = 2.1 Hz, 1H), 8.86 (dd, J = 4.8, 1.6 Hz, 1H), 8.45 (dt, J = 7.9, 1.9 Hz, 1 H), 8.26 (d, J = 2.2 Hz, 1H), 7.81 (s, 1H), 7.64 (dd, J = 8.0, 4.8 Hz, 1H), 7.45 (dd, J = 8.4, 2.3 Hz, 1H), 6.92 (d, J = 8.4 Hz, 1H). 13C NMR (126 MHz, DMSO-</6) δ 190.04, 167.89, 154.00, 149.67, 144.16, 136.67, 136.21 ,
132.86, 132.66, 126.72, 125.99, 125.60, 124.15, 121.35, 1 1 1.90. ESI-MS (ES+) m/z calcd for C[ 5H9C1N202, 284.0353, found, 285.1973 (M+H)+
(E)-5-bromo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (43)
[0175] Compound 43 was prepared by general procedure B. Ή NMR (400 MHz, DMSO-fife) δ 10.99 (s, 1 H), 9.22 (d, J = 2.0 Hz, 1H), 8.85 (dd, J = 4.8, 1.4 Hz, 1H), 8.43 (m, 1H), 8.39 (d, J = 1.9 Hz, 1H), 7.78 (s, 1H), 7.63 (dd, J = 8.0, 4.9 Hz, 1 H), 7.55 (dd, J = 8.3, 2.0 Hz, 1 H), 6.86 (d, J = 8.3 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 190.06, 167.89, 154.07, 149.73, 144.57, 136.68, 136.31 , 135.77, 132.76, 129.62, 125.96, 124.27, 121.89, 1 13.41 , 1 12.48. ESI-MS (ES+) m/z calcd for C,5H9BrN202, 327.9847, found, 329.1079 (M+H)+
(E)-5-methyl-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (44)
[0176] Compound 44 was prepared by general procedure B (0.5 g, 23.4 % for two steps). Ή NMR (400 MHz, DMSO-^s) δ 10.74 (s, 1 H), 9.21 (d, J = 1.9 Hz, 1 H), 8.85 (dd, J = 4.8, 1.5 Hz, 1 H), 8.42 (dt, J = 8.0, 1.9 Hz, 1 H), 8.02 (s, 1 H), 7.69 (s, 1 H), 7.63 (dd, J = 8.0, 4.8 Hz, 1H), 7.19 (d, J = 7.3 Hz, 1H), 6.78 (d, J = 7.9 Hz, 1H), 2.24 (s, 3H).1JC NMR (101 MHz, DMSO-c4)5190.21, 168.24, 153.89, 149.56, 143.09, 137.73, 136.12, 133.91, 132.79, 130.56, 127.60, 124.41, 124.18, 120.00, 110.20, 20.75. ESI-MS (ES+) m/z calcd for C,6Hi2N202) 264.0899, found, 287.1902 (M+Na)+
(E)-5-nitro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (45)
[0177] Compound 45 was prepared by general procedure B (1.8 g, 59 % for two steps). Ή NMR (400 MHz, DMSO-i/6) δ 11.58 (s, 1H), 9.27 (dd, J = 2.3, 0.8 Hz, 1H), 9.15 (d, J = 2.4 Hz, 1H), 8.88 (dd, J = 4.8, 1.6 Hz, 1H), 8.49 (ddd, J = 8.0, 2.3, 1.7 Hz, 1H), 8.33 (dd, J = 8.7, 2.4 Hz, 1H), 7.92 (s, 1H), 7.65 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.10 (d, J = 8.7 Hz, 1H).13C NMR (101 MHz, DMSO- 6) δ 189.96, 168.51, 154.14, 150.85, 149.78, 142.04, 136.33, 135.81, 132.59, 129.47, 127.26, 124.21, 122.67, 120.07, 110.67. ESI-MS (ES+) m/z calcd for C15H9N3O4, 295.0593, found, 296.1903 (M+H)+
(E)-6-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (46)
[0178] Compound 46 was prepared by general procedure B (0.8 g, 57 % for two steps). Ή NMR (400 MHz, DMSO-i/6) δ 11.02 (s, 1H), 9.21 (dd, J = 2.3, 0.7 Hz, 1H), 8.86 (dd, J = 4.8, 1.6 Hz, 1H), 8.43 (ddd, J = 8.0, 2.2, 1.8 Hz, 1H), 8.22 (d, J = 8.4 Hz, 1H), 7.76 (s, 1H), 7.63 (ddd, J = 8.0, 4.8, 0.8 Hz, 1H), 7.07 (dd, J = 8.4, 2.0 Hz, 1H), 6.92 (d, J = 2.0 Hz, 1H).13CNMR(101 MHz, DMSO-c¾ δ 190.08, 168.21, 153.97, 149.59, 146.66, 137.48, 136.35, 136.15, 132.67, 128.63, 125.12, 124.18, 121.70, 118.85, 110.48. ESI-MS (ES+) m/z calcd for Ci5H9ClN202, 284.0353, found, 285.1973 (M+H)+
(E)-6-bromo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (47)
[0179] Compound 47 was prepared by general procedure B. Ή NMR (400 MHz, DMSO-</6)611.00 (s, 1H), 9.21 (dd, J = 2.3, 0.8 Hz, 1H), 8.86 (dd, J = 4.8, 1.7 Hz, 1H), 8.43 (ddd, J = 8.0, 2.3, 1.7 Hz, 1H), 8.14 (d, J = 8.3 Hz, 1H), 7.78 (s, 1H), 7.63 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.22 (dd, J = 8.3, 1.9 Hz, 1H), 7.05 (d, J = 1.6 Hz, 1H). I3C NMR (101 MHz/DMSO-cfe) δ 190.16, 168.08, 153.98, 149.59, 146.64, 136.43, 136.16, 132.66, 128.72, 126.37, 125.37, 124.64, 124.19, 119.17, 113.29, 39.52. ESI-MS (ES+) mz calcd for Ci5H9BrN202, 327.9847, found, 329.1079 (M+H)+
(E)-7-chloro-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (48)
[0180] Compound 48 was prepared by general procedure B (400 mg, 51 % for two steps). Ή NMR (400 MHz, DMSO-c6) δ 11.27 (s, 1 H), 9.19 (dd, J = 2.3, 0.7 Hz, 1 H), 8.84 (dd, J = 4.8, 1.7 Hz, 1H),8.41 (ddd, J = 8.0, 2.3, 1.7 Hz, 1H), 8.03 (d, J = 7.8 Hz, 1H), 7.77 (s, 1H), 7.61 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.44 (dd, J = 8.2, 1.0 Hz, 1H), 6.99 (t, 1H). 13C NMR (101 MHz, DMSO-efe) δ 190.38, 168.07, 154.08, 149.68, 142.55, 136.42, 136.23, 132.72, 132.47, 126.86, 125.41 , 124.22, 123.00, 121.71 , 1 14.69. ESI-MS (ES+) m/z calcd for C5H9CIN2O2, 284.0353, found, 285.1973 (M+H)+
(E)-7-bromo-3-(2-oxo-2-(pyridin-3-yl)ethylidene)indolin-2-one (49)
[0181] Compound 49 was prepared by general procedure B. Ή NMR (500 MHz, DMSO-i/6) 5 1 1.15 (s, 1 H), 9.21 (dd, J = 2.3, 0.9 Hz, 1H), 8.86 (dd, J = 4.8, 1.7 Hz, 1H), 8.43 (ddd, J = 8.0, 2.3, 1.7 Hz, 1H), 8.09 (dd, J = 7.7, 0.7 Hz, 1H), 7.78 (s, 1H), 7.64 (ddd, J = 8.0, 4.8, 0.9 Hz, 1H), 7.58 (dd, J = 8.2, 0.8 Hz, 1H), 6.95 (t, J = 8.1 Hz, 1 H). I3C NMR (126 MHz, DMSO < ) 5 190.42, 168.14, 154.08, 149.68, 144.53, 136.77, 136.23, 135.69, 132.49, 126.82, 125.82, 124.22, 123.30, 121.77, 102.91. ESI-MS (ES+) m/z calcd for Ci5H9BrN202, 327.9847, found, 329.1078 (M+H)+
(2-oxo-2H-chromen-7-yl-4-(2-(((benzyloxy)carbonyl)amino)acetamido)butanoate (50)
[0182] Cbz-Gly-OH (3.0 g, 14 mmol), methyl γ-aminobutyrate hydrochloride (2.2 g, 14 mmol), EDCI (2.8 g, 14 mmol) and hydroxybenzotriazole hydrate (2.2 g, 14 mmol) were dissolved in DMF (30 mL) and N-methyl-morpholine (1.6 mL, 14 mmol) added. After stirring the mixture for 16 hours, methyl 4-(2-
(((benzyloxy)carbonyl)amino)acetamido)butanoate (4.3 g, 97 %) was precipitated by diluting the reaction mixture tenfold with water and isolated by filtration. The white solid was washed extensively with water and then dissolved in THF (40 mL). Aqueous 2M LiOH (7 mL) was added followed by about 15 mL methanol to dissolve the resulting precipitate. The mixture was monitored for full conversion by TLC and subsequently diluted with ethyl acetate (150 mL). The organic phase was washed three times with 0.1 M HC1 (50 mL each) and once with brine (30 mL) and finally dried over sodium sulfate. The volatiles were removed under reduced pressure and the resulting fluffy white 4-(2- (((benzyloxy)carbonyl)amino)acetamido)butanoic acid (3.5 g, 85 %) directly subjected to the final coupling step without any further purification. An aliquot of the acid (1.5 g, 5.1 mmol) and EDCI (0.98 g, 5.1 mmol) were dissolved in DMF (15 mL), followed by 7- hydroxycoumarin (1.7 g, 10.2 mmol) and N-methyl-morpholine (560 μί, 5.1 mmol). The mixture was stirred for 24 hours before diluting it tenfold with water. The crude solid was filtered, washed extensively with sodium bicarbonate and water and dried under vacuum. 2-oxo-2H-chromen-7-yl-4-(2-(((benzyloxy)carbonyl)amino)acetamido)butanoate (1.45 g, 65 %) was recrystallized from ethyl acetate twice to remove residual 7-hydroxycoumarin to below 0.1 % as judged by fluorescence. Ή NMR (300 MHz, DMSO-c/6) δ 8.08 (d, 7 = 9.6 Hz, 1 H), 7.97 (t, J= 5.5 Hz, 1H), 7.77 (d, J= 8.5 Hz, 1 H), 7.47 (t, J = 6.1 Hz, 1H), 7.43 - 7.22 (m, 6H), 7.17 (dd, J = 8.4, 2.1 Hz, 1H), 6.48 (d, J = 9.6 Hz, 1H), 5.03 (s, 2H), 3.60 (d, J= 6.1 Hz, 2H), 3.18 (q, J = 6.5 Hz, 2H), 2.63 (t, J = 7.3 Hz, 2H), 1.77 (p, J= 6.9 Hz, 2H). 13C NMR (75 MHz, DMSO-</6) δ 171.22, 169.20, 159.75, 156.50, 154.1 1 , 152.94, 143.88, 137.04, 129.37, 128.36, 127.81 , 127.76, 1 18.73, 1 16.68, 1 15.56, 1 10.16, 65.51 , 43.63, 37.49, 30.76, 24.35. ESI-MS (ES+) m/z calcd for (^^NaOy, 438.1427, found, 439.2463 (M+H)+
Biological and Pharmacological Testing
Example 4: Expression and purification of recombinant human tg2
[0183] Recombinant human TG2 was expressed and purified as described previously (Piper et al, Biochemistry 2002, 41 (1), 386-393, incorporated herein by reference). Protein preparations were frozen at concentrations of 50 μΜ to 100 μΜ, and each aliquot was thawed 1 -2 hours before kinetic analysis.
Example 5: Enzyme kinetic measurements and data analysis
[0184] Steady state kinetic parameters for TG2-catalyzed deamidation and inhibition were measured using a coupled enzyme assay, as described previously with minor modifications (Pinkas et al, Plos Biology 2007, 5 (12), 2788-2796, Piper et al,
Biochemistry 2002, 41 (1 ), 386-393, each of which is incorporated herein by reference). Briefly, 120 uL reaction buffer consisting of 200 mM MOPS (pH=7.2), 10 mM a- ketoglutarate, I mM EDTA, 5 mM Ca2+, varying quantities of Z-Gln-Gly (3 to 30 mM), 36 unit/mL glutamate dehydrogenase, 0.3-0.4 mM NADH and a defined concentration of inhibitor was pre-incubated at room temperature for 20 minutes until the UV absorbance at 340 nm was stable. To start the reaction, 1.2 uL of TG2 was added to each cuvette, and the absorbances were recorded up to 120 minutes. Slopes were measured from linear region of each trace, usually 20 to 60 minutes after initiating reaction.
[0185] To measure IC50 values of reversible inhibitors, a series of assays were performed with inhibition concentrations of 1 , 3, 10, 30, and 100 uM, and constant ZQG concentration at 20 mM. Enzyme turnover rates were plotted against inhibition concentrations, and fitted into a four-parameter logarithm equation using BioDataFit 1.02 from Chang Bioscience Inc. [0186] To study inhibition effect of low IC5o compounds, a 4x4 assay was performed with inhibitor concentrations of 0.5xIC50, IC50, 2xIC50 and 4xIC5o, and ZQG substrate concentrations of 6, 9, 12, and 30 mM. Turnover rates were fitted into a modified
V [S]
Michaelis-Menten equation for reversible inhibitors ( = — — — )
[E,„lal ] ( 1 + m)K iii + (l + ii)[S] to calculate competitive (Ki) and non-competitive (Kj') inhibition constants.
[0187] Irreversible inhibitors were assayed in a manner similar to the above described steady state kinetics, with only 20 or 30 mM ZQG, and various inhibitor concentrations from 3 to 100 μΜ. The reaction was started by addition of TG2 and inhibitor, and the absorbance at 340 nm was monitored up to one hour. The progress curves were fitted to the following equation: Λ"3-Λ"!, k" . Substrate dependence of vo and k' yielded the kinetic parameters Kj and kjnh.
[0188] The results are tabulated below (Table 1). Under testing conditions, certain compounds may not have shown an IC50 low enough to be considered, under these conditions, to be efficacious in vitro; however these compounds may be useful in other test conditions as described here and known to one of skill in the art.
Example 6: Steady state kinetic analysis
[0189] TG2 activity and inhibition were assayed in the previously described GDH- coupled assay and a direct fluorescent assay. Piper, J. L. et al., Biochemistry 2002, 41, 386-93 and Gillet, S. M. et al., Anal. Biochem. 2005, 347, 221-6, each of which is incorporated herein by reference. Briefly, in the GDH coupled assay, TG2 (0.5 μΜ) enzymatic deamidation of 5-50 mM ZQG in 200 mM MOPS buffer (pH = 7.2) containing 4 mM CaC and 1 % DMSO was monitored at 340 nm through the consumption of NADH (0.35 mM) by the GDH (36 units/mL) mediated incorporation of nascent ammonia into a-ketoglutarate (10 mM). Absorbance measurements were recorded in a Perkin-Elmer Lambda 25 spectrophotometer for 50-80 min and steady state slopes measured after full equilibration of the samples, typically at 30-60 min timepoints. The raw slopes were converted to rates using the extinction coefficient of NADH (ε = 6220 L mor' cm'1) For IC50 measurements, the substrate concentration was typically about 10 mM (ca. Km) and for Kj measurements it varied from 5 to 50 mM. [0190] In the fluorescent method, TG2 was assayed in a buffer consisting of 200 mM MOPS, 4 mM CaCl2, 0.05 % PEG-20000, 1-2 % DMSO, 5-30 μΜ ZGBC substrate (2- oxo-2H-chromen-7-yl-4-(2-(((benzyloxy)carbonyl)amino)acetamido)butanoate) (50) and 0 to 100 μΜ inhibitor in deionized water at pH 7.2. The reactions were initiated by the addition of TG2. All measurements were carried out in triplicate on black Corning NBS 96-well plates and recorded in a Berthold Mithras LB940 microplate reader with excitation at 340 nm and emission at 460 nm. Raw fluorescence traces were evaluated through their slopes after the samples have fully equilibrated, typically at 30-60 min timepoints. The slopes were converted to rates using a standard curve and corrected for background hydrolysis of the substrate. For IC50 measurements, the substrate concentration was typically about 10 μΜ (ca. 2 * Km) and for K, measurements it varied from 5 to 30 μΜ. To determine IC50 values, the rates were fitted to a 4-parameter Hill- Model and for Kj determination, the rates were fitted to a non-linear mixed inhibition model using GraphPad Prism 5:0. The results are tabulated in Tables 1 , 2, 3, and 4.
[0191] Enzyme inhibition was measured using the coupled GDH assay ([TG2] = 0.5 μΜ). For IC50 values, the substrate was used at its Km = 10 mM.
Table 2
Figure imgf000064_0001
[0192] The inhibitory characteristics various 3-acylidene-2-oxoindole inhibitors against TG2 were measured in the GDH assay ([TG2] = 0.5 μΜ) and tabulated in Table 1 above. For IC50 values, the substrate was used at its Km = 10 mM. If partial inhibition was observed, the maximum inhibition effect is shown in parentheses.
[0193] In Table 3, IC50 concentrations determined by coupled GDH assay and the coumarin-based fluorescence assay with a better dynamic range for submicromolar values are compared. For IC50 values in the GDH-coupled assay, the substrate was used at its m = 10 mM with 500 nM TG2. In the fluorescent assay, IC50 values were measured at 10 μΜ substrate concentration (= 2 * Km) with 15 nM TG2.
Table 3
Figure imgf000066_0001
|0194] Table 4 below illustrates inhibition constants of selected 3-acylidene-2- oxoindoles in the GDH assay ([TG2] = 0.5 μΜ) and the new fluorescent assay ([TG2] = 15 nM), a is the ratio Kj' Kj in a mixed inhibition model. The larger the a, the more competitive the inhibition is; when a = 1 , the inhibition is non-competitive and when a < 1 , the inhibition is increasingly un-competitive.
Table 4
Figure imgf000067_0001
Example 7: Inhibition of tg2 in a wi-38 fibroblast wound model
[0195] The WI-38 fibroblast scratch assay is performed based upon modification of previously published protocols [Upchurch, H.F., Conway, E., Patterson, M.K. Jr, Maxwell, M.D. (1991) Localization of cellular transglutaminase on the extracellular matrix after wounding: characteristics of the matrix bound enzyme. J Cell Physiol 149(3), 375-382; Siegel, M.; Strnad, P.; Watts, R.E., Choi, K., Jabri, B., Omary, M.B., Khosla, C. (2008) Extracellular transglutaminase 2 is catalytically inactive but is transiently activated upon tissue injury. PLoS One 3(3), el 861 , each of which is incorporated herein by reference]. WI-38 fibroblasts are plated in an 8-well chamber glass slide at 105 cells/well and grown for 7-10 days with a media change every other day. Inhibitor is added at a range of concentrations (50, 25, 12.5, 10, 8, 6.25, 3.1 , 1.5 and 0 μΜ). Small scratches are made in the monolayer using a 0.1-10 μΙ_ pipette tip, and the cells are incubated at 37 ° C for 10 min. 5-biotinamido pentylamine (5BP) is added to a final concentration of 300 μΜ and the cells are incubated for 1 hour at 370 C with anti-fibronectin Ab. The cells are washed 3 times with PBS then fixed with -20°C methanol for 10 minutes. The cells are washed twice with PBS for 5 minutes, blocked with 1% BSA in PBS for 5 minutes at room temperature, and then washed twice using PBS. The cells are incubated with Streptavidin Alexa fluor 555 conjugate (to visualize TG2 activity) and anti-rabbit goat Ab 488 (to visualize fibronectin protein), and examined by fluorescence microscopy.
Example 8: Inhibition of tg2 in small intestines following poly(i:c)-induced small intestinal damage in mice
[0196] This in vivo assay is adapted from a previously described protocol [Siegel, M.; Stmad, P.; Watts, R.E., Choi, K., Jabri, B., Omary, M.B., Khosla, C. (2008) Extracellular transglutaminase 2 is catalytically inactive but is transiently activated upon tissue injury. PLoS One 3(3), el 861 , supra]. C57BL/6 female mice (8 weeks old) are IP injected with 30-90 mg/kg poly(I:C) (Sigma, PI 530) dissolved in PBS to injure the small intestine as described [Zhou, R., Wei, H., Sun, R., Tian, Z. (2007) Recognition of double-stranded RNA by TLR3 induces severe small intestinal injury in mice. J Immunol 178(7), 4548- 4556, incorporated herein by reference]. Controls are injected with PBS vehicle only. 5- BP is dissolved to 27.8 mg/mL in water and diluted to 25 mg/mL using lOx PBS. Mice are ΓΡ (intraperitoneally) injected twice with either 5-BP at 100 mg/kg or PBS at three hour intervals. For mice sacrificed after 6 hours, 5-BP or PBS is injected at 0 and 3 hours. For mice sacrificed after 1 1 hours, 5-BP or PBS is injected at 6 and 9 hours. The lumens of isolated small intestines are rinsed with PBS using a needle and syringe, and small pieces of tissue are formalin-fixed for hematoxylin and eosin (H&E) staining or frozen in OCT for subsequent immunofluorescence staining. The intestine is cut longitudinally, and the intestinal mucosa is scraped using microscope slides, wrapped in parafilm, and frozen on dry ice. Upon thawing, tissue is homogenized with a Wheaton dounce homogenizer in 50 mM mannitol, 2 mM tris, 10 mM EDTA, pH = 7.4, sonicated 2 times, and diluted in Laemmli sample buffer for SDS-PAGE and Western blotting. HRP conjugated neutravidin (Invitrogen, A-2664) and ECL plus substrate (GE Biosciences) are used to visualize biotinylated proteins. In order to study the effects of TG2 inhibitors, inhibitors are administered by oral gavage at doses ranging from 0 - 300 mg kg prior to and/or at one or more intervals after the first injection of poly(I:C).
[0197] The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein. [0198] While certain embodiments have been illustrated and described, it will be understood that changes and modifications can be made therein in accordance with ordinary skill in the art without departing from the present technology in its broader aspects as defined in the following claims.
[0199] In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group.
[0200] All publications, patent applications, patents, and other references mentioned herein are expressly incorporated by reference in their entirety, to the same extent as if each were incorporated by reference individually.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula (I)
Figure imgf000070_0001
(I) wherein Ri is H or substituted or unsubstituted Ci-C6 alkyl;
R2 is C1-C4 alkyl, substituted or unsusbtituted aryl or heteroaryl group, or NR6R7 wherein R6 and R7 are independently H or substituted or unsubstituted Ci-C6 alkyl or aryl group;
R31, R32, R33, and R34 independently are H, halo, N02, -0-R8, or -S02NR26R27 wherein R8 is substituted or unsubstituted Ci-C6 alkyl or aryl group and Ri6 and R17 are independently H, Ci-C6 alkyl, or aryl;
R4 is OH and R5 is hydrogen, or R4 and R5 together with the carbon atoms to which they are bonded form a carbon-carbon double bond; or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1 of formula (II) or formula (IIA)
Figure imgf000070_0002
(II) (IIA)
3. The compound of claim 1 of formula (IIIA)
Figure imgf000071_0001
(IV)
5. The compound of claim 4, wherein Ri is H, methyl, or -R9-R10 wherein R9 is substituted or unsubstituted methylene and Rio is -CONR11R12 or substituted or unsubstitutedaryl, cycloalkyl, or heterocyclyl group containing at least one basic nitrogen atom wherein Rn and R12 independently are H or Ci-C6 alkyl or Rn and R12 together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring.
6. The compound of claim 5, wherein R2 is
Figure imgf000071_0002
7. The compound of claim 4, wherein R2 is
Figure imgf000072_0001
wherein R13 is halo or another leaving group, Ci-C6 alkyl, or aryl.
8. The compound of claim 7, wherein Ri is H.
9. The compound of claim 4, wherein R2 is substituted or unsubstituted pyridyl.
10. The compound of claim 4, wherein R2 is
Figure imgf000072_0002
wherein R14 is a bond, -O- or -NR16- and R15 is halo, substituted or unsubstituted Ci-C6 alkyl, aryl, or CONRi8Ri9, or Ri5 and Ri6 together with the nitrogen atom to which they are bonded form a heterocyclic ring containing up to 2 nitrogen atoms, or R18 and R19 together with the nitrogen atom to which they are bonded form a heterocyclic ring containing up to 2 nitrogen atoms and wherein Ri6 is substituted or unsubstituted Ci-C6 alkyl, provided that when Ri5 is halo, R14 is a bond.
11. The compound of claim 10, wherein R2 is
Figure imgf000072_0003
12. The compound of claim 11, wherein Ri is H.
13. The compound of claim 10, wherein R14 is -O- and R½ is -CH2-R17 wherein Rn is aryl, carboxamide, or a carboxylic acid group. The compound of claim 4 of formula (V):
Figure imgf000073_0001
(V) wherein R10 is -CONR11R12 or substituted or unsubstituted aryl, cycloalkyl, or heterocyclyl group wherein Rn and R12 independently are H or Ci-C6 alkyl.
The compound of claim 1 of formula (VI)
Figure imgf000073_0002
(VI) wherein R6 and R7 are H, unsubstitutedor substituted C1-C4 alkyl, or aryl.
The compound of claim 1 of formula (VII)
Figure imgf000073_0003
(VII) wherein R32 is H, -0-R8, or -S02NR26R27 wherein R8 is
Figure imgf000074_0001
or substituted or unsubstituted phenyl or benzyl, and R33 is H or -0-R8 wherein R8 is substituted or unsubstituted phenyl, provided that one of R32 and R33 is H.
17. The compound of claim 16, wherein Ri is H and R2 is
Figure imgf000074_0002
18. The compound of claim 1 of formula (VIIIA)
Figure imgf000074_0003
(VIIIA) wherein Ri is H, substituted or unsubstituted Ci-C6 alkyl;
R2 is C1-C4 alkyl or substituted or unsusbtituted aryl or heteroaryl group;
R31, R32, and R33 are independently H, halo, or substituted or unsubstituted C1-C4 alkoxy.
19. The compound of claim 18 wherein R2 is selected from the group consisting of methyl, unsubstituted phenyl, phenyl substituted with a C1-C4 alkyl or an aryl group, 3-pyridyl, or 3-pyridyl substituted with a C1-C4 alkoxy group.
20. The compound of claim 18, wherein Ri is unsubstituted C1-C4 alkyl or C\- C4 alkyl substituted with substituted or unsubstituted piperidinyl, cyclohexyl and phenyl groups. The compound of claim 18 of formula (IX)
Figure imgf000075_0001
(IX)
The compound of claim 21, wherein Ri is H or C1-C4 alkyl;
R31 is CI or H; and
R2 is methyl, phenyl, phenyl substituted with a C1-C4 alkyl group, or
Figure imgf000075_0002
wherein R25 is H or C1-C4 alkoxy.
The compound of claim 18 of formula (X)
Figure imgf000075_0003
(X)
The compound of claim 23, wherein R32 is H or chloro. The compound of claim 18 of formula (XI) or (XII)
Figure imgf000076_0001
(XI) (XII)
26. The compound of claim 25 of formula (XI), wherein R33 is H, fluoro or C\- C4 alkoxy or the compound of formula (XII) wherein R34 is H or chloro.
27. A compound of formula IIIB)
Figure imgf000076_0002
(OB)
wherein Ri is H, Ci-C6 alkyl, phenyl, or -(CH2)n-Rio;
n is 1 , 2, or 3;
Rio is C3-C8 cycloalkyl, substituted or unsubstituted phenyl, -C02Rn or -CONRnRi2; R11 and R12 are independently Ci-C6 alkyl or Rn and R12 together with the carbon atom they are bonded to form a 5-6 membered heterocyclic ring;
R2 is C1-C3 alkyl, pyridyl, or substituted or unsubstituted phenyl; and
R3 is halo, -NO2, substituted or unsubstituted C1-C3 alkoxy, or C1-C3 alkyl.
28. The compound of claim 1 of formula:
Figure imgf000077_0001
29. A composition comprising a compound of any one of claims 1-28 and a carrier, diluent, or excipient.
30. The composition of claim 29, wherein the carrier, diluent, or excipient is pharmaceutically acceptable.
31. The composition of claim 30, wherein the carrier is a pharmaceutically acceptable polymer.
32. Use of a compound of any one of claims 1 to 28 or the composition of claim 30, in the preparation of a medicament.
33. A method of inhibiting a transglutaminase-2 (TG2) comprising contacting an effective amount of a compound of one of claims 1-28 or the composition of claim 30 with the TG2.
34. The method of claim 33, wherein the contacting takes place in vitro or in vivo.
35. A complex comprising a compound of any one of claims 1-28 and a TG2.
36. A method of inhibiting a TG2 in a tissue comprising contacting the tissue with a compound of any one of claims 1-28 or a composition of claim 30.
37. The method of claim 36, wherein the contacting is performed in vitro or in vivo.
38. A method of treating a disease relating to the pathological presence of a TG2 in a tissue, comprising administering a therapeutically effective amount of a compound of one of claims 1-28 or the composition of claim 30 to a patient in need of such treatment, thereby treating the disease.
39. The method of claim 38, wherein the tissue is one or more of the small intestine, the brain, the lungs. 40. The method of claim 38, wherein the disease is Alzheimer's disease, atherosclerosis, celiac sprue, cystic fibrosis, dermatitis herpetoformis, glioblastoma and other cancers, Huntington's disease, keratosis, Parkinson's disease, or a wound to be healed.
41. The method of claim 38, wherein the compound or composition is administered by one or more of by ingestion, by inhalation, intranasal administration or by injection.
PCT/US2011/063312 2010-12-06 2011-12-05 3-acylidene-2-oxoindole derivatives for inhibition of transglutaminase 2 WO2012078519A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42021610P 2010-12-06 2010-12-06
US61/420,216 2010-12-06

Publications (2)

Publication Number Publication Date
WO2012078519A2 true WO2012078519A2 (en) 2012-06-14
WO2012078519A3 WO2012078519A3 (en) 2012-08-02

Family

ID=46207668

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/063312 WO2012078519A2 (en) 2010-12-06 2011-12-05 3-acylidene-2-oxoindole derivatives for inhibition of transglutaminase 2

Country Status (1)

Country Link
WO (1) WO2012078519A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104496885A (en) * 2015-01-12 2015-04-08 中国药科大学 N-methyl-4-amino-5-nitroisatin preparation method
WO2016057698A1 (en) * 2014-10-09 2016-04-14 Tokalas, Inc. Indolinone compounds and uses thereof
US9714222B2 (en) 2012-04-12 2017-07-25 Georgetown University Methods and compositions for treating Ewings sarcoma family of tumors
US9822122B2 (en) 2016-03-31 2017-11-21 Oncternal Therapeutics, Inc. Indoline analogs and uses thereof
US10159660B2 (en) 2016-07-29 2018-12-25 Oncternal Therapeutics, Inc. Uses of indolinone compounds
WO2020033784A1 (en) 2018-08-10 2020-02-13 Sitari Pharma, Inc. Transglutaminase 2 (tg2) inhibitors
RU2781370C2 (en) * 2018-08-10 2022-10-11 Ситари Фарма, Инк. Transglutaminase 2 (tg2) inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056769A2 (en) * 2002-12-20 2004-07-08 Ciba Specialty Chemicals Holding Inc. Synthesis of amines and intermediates for the synthesis thereof
US20060052308A1 (en) * 2003-11-18 2006-03-09 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
WO2009123080A1 (en) * 2008-04-01 2009-10-08 アステラス製薬株式会社 Indolinone compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056769A2 (en) * 2002-12-20 2004-07-08 Ciba Specialty Chemicals Holding Inc. Synthesis of amines and intermediates for the synthesis thereof
US20060052308A1 (en) * 2003-11-18 2006-03-09 The Board Of Trustees Of The Leland Stanford Junior University Transglutaminase inhibitors and methods of use thereof
WO2009123080A1 (en) * 2008-04-01 2009-10-08 アステラス製薬株式会社 Indolinone compound

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CASSABDRA L. WOODARD ET AL.: 'Oxindole-based compounds are selective inhibitors of Plasmodium falciparum cyclin dependent protein kinase' JOURNAL OF MEDICINAL CHEMISTRY vol. 46, no. 18, 2003, ISSN 0022-2623 pages 3877 - 3883 *
CORNELIUS KLOECK ET AL.: 'Acylideneoxoindoles: A new class of reversible inhibitors of human transglutaminase 2' BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, 2011 vol. 21, no. 9, 16 December 2010, ISSN 0960-894X pages 2692 - 2696 *
LINDSEY J. MACPHERSON ET AL.: 'Noxious compounds activate TRPA1 ion channels through covalent modification of cysteines' NATURE vol. 445, 2007, ISSN 0028-0836 pages 541 - 545 *
STANLEY REHN ET AL.: 'The reaction between 3-aminocrotonates and oxindole- 3-ylidene derivatives: synthesis of highly substituted pyrroles' TETRAHEDRON vol. 61, no. 12, 2005, ISSN 0040-4020 pages 3115 - 3123 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9714222B2 (en) 2012-04-12 2017-07-25 Georgetown University Methods and compositions for treating Ewings sarcoma family of tumors
EA032644B1 (en) * 2014-10-09 2019-06-28 Онктернал Терапьютикс, Инк. Indolinone compounds and uses thereof
WO2016057698A1 (en) * 2014-10-09 2016-04-14 Tokalas, Inc. Indolinone compounds and uses thereof
US9604927B2 (en) 2014-10-09 2017-03-28 Oncternal Therapeutics, Inc. Indolinone compounds and uses thereof
US9895352B2 (en) 2014-10-09 2018-02-20 Oncternal Therapeutics, Inc. Indolinone compounds and uses thereof
US9987251B2 (en) 2014-10-09 2018-06-05 Oncternal Therapeutics, Inc. Indolinone compounds and uses thereof
AU2015328121B2 (en) * 2014-10-09 2019-11-21 Oncternal Therapeutics, Inc. Indolinone compounds and uses thereof
CN104496885A (en) * 2015-01-12 2015-04-08 中国药科大学 N-methyl-4-amino-5-nitroisatin preparation method
US9822122B2 (en) 2016-03-31 2017-11-21 Oncternal Therapeutics, Inc. Indoline analogs and uses thereof
US10351569B2 (en) 2016-03-31 2019-07-16 Oncternal Therapeutics, Inc. Indoline analogs and uses thereof
US10159660B2 (en) 2016-07-29 2018-12-25 Oncternal Therapeutics, Inc. Uses of indolinone compounds
US10646470B2 (en) 2016-07-29 2020-05-12 Oncternal Therapeutics, Inc. Uses of indolinone compounds
US11285132B2 (en) 2016-07-29 2022-03-29 Oncternal Therapeutics, Inc. Uses of indolinone compounds
WO2020033784A1 (en) 2018-08-10 2020-02-13 Sitari Pharma, Inc. Transglutaminase 2 (tg2) inhibitors
CN112789040A (en) * 2018-08-10 2021-05-11 思达利医药公司 Transglutaminase 2(TG2) inhibitors
EP3833348A4 (en) * 2018-08-10 2021-06-30 Sitari Pharma, Inc Transglutaminase 2 (tg2) inhibitors
RU2781370C2 (en) * 2018-08-10 2022-10-11 Ситари Фарма, Инк. Transglutaminase 2 (tg2) inhibitors
US11548892B1 (en) 2018-08-10 2023-01-10 Sitari Pharma, Inc. Transglutaminase 2 (TG2) inhibitors
EP4219460A1 (en) * 2018-08-10 2023-08-02 Sitari Pharma, Inc. Transglutaminase 2 (tg2) inhibitors

Also Published As

Publication number Publication date
WO2012078519A3 (en) 2012-08-02

Similar Documents

Publication Publication Date Title
ES2461268T3 (en) Indole-3-carboxylic acid amide, ester, thioamide and thiol ester compounds bearing aryl or heteroaryl groups having sphingosine-1-phosphate receptor antagonist biological activity (S1P)
ES2927954T3 (en) N-substituted 2,5-dioxo-azoline compounds for use in the treatment of cancer
CA2774133C (en) Compounds effective as xanthine oxidase inhibitors, method for preparing the same, and pharmaceutical composition containing the same
JP2021521200A (en) Compounds for the treatment of cancer
AU2017204598B2 (en) Meglumine salt formulations of 1-(5,6-dichloro-1H-benzo[d]imidazol-2-yl)-1H-pyrazole-4-carboxylic acid
WO2012078519A2 (en) 3-acylidene-2-oxoindole derivatives for inhibition of transglutaminase 2
BRPI0714840A2 (en) compound, process for the preparation and use thereof, pharmaceutical composition, process for the preparation thereof, and, product
PT832066E (en) N-SUBSTITUTED- (INDOLE-2-CARBONYL) -AMIDES AND DERIVATIVES AS GLYCOGENE-PHOSPHORYLASE INHIBITORS
MX2010008376A (en) Ampk modulators.
RU2507203C1 (en) Compound, certain novel forms thereof, pharmaceutical compositions based thereon and methods of production and use
KR20210097100A (en) 2-(1-acyloxy-n-pentyl)benzoic acid and a salt formed by a basic amino acid or aminoguanidine, preparation method and use thereof
BR112016005606B1 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, AND USES OF A COMPOUND
ES2927959T3 (en) Heterocyclic imidazole compounds, pharmaceutical compositions thereof, method of preparation thereof and use thereof
WO2018157801A1 (en) Cyano group-substituted fused bicyclic derivative, preparation method therefor, and application thereof
EP4105206A1 (en) Novel pyrazole derivative
JP2005535576A (en) Indol-2-ones as selective inhibitors of cyclooxygenase-2
BR112020022220A2 (en) new compound and pharmaceutical composition comprising the same
CN112047931B (en) FXIa coagulation factor inhibitor, pharmaceutical composition and application thereof
CN116496269A (en) Prolyl hydroxylase inhibitor and application thereof
WO2019001307A1 (en) Amide compound, composition containing same, and use thereof
WO2024071371A1 (en) Heterocyclic compound
TW202412778A (en) A prolyl hydroxylase inhibitor and use thereof
TW202328073A (en) Ep4 antagonis, salt and polymorphs thereof, method and application thereof
WO2023193733A1 (en) At2r agonist
RU2562773C2 (en) Glutarimide derivatives, application thereof, pharmaceutical composition based thereon, methods of obtaining thereof

Legal Events

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

Ref document number: 11846904

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11846904

Country of ref document: EP

Kind code of ref document: A2