WO2004103289A2 - Driverse thyroid hormone receptor antagonists and uses thereof - Google Patents

Driverse thyroid hormone receptor antagonists and uses thereof Download PDF

Info

Publication number
WO2004103289A2
WO2004103289A2 PCT/US2004/015048 US2004015048W WO2004103289A2 WO 2004103289 A2 WO2004103289 A2 WO 2004103289A2 US 2004015048 W US2004015048 W US 2004015048W WO 2004103289 A2 WO2004103289 A2 WO 2004103289A2
Authority
WO
WIPO (PCT)
Prior art keywords
thyroid hormone
compound
och
thyroid
effective amount
Prior art date
Application number
PCT/US2004/015048
Other languages
French (fr)
Other versions
WO2004103289A3 (en
Inventor
Herbert H. Samuels
Ruben Abagyan
Matthieu Schapira
Maxim Totrov
Bruce M. Raaka
Stephen R. Wilson
Li Fan
Zhiguo Zhou
Original Assignee
New York University
Molsoft Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by New York University, Molsoft Llc filed Critical New York University
Publication of WO2004103289A2 publication Critical patent/WO2004103289A2/en
Publication of WO2004103289A3 publication Critical patent/WO2004103289A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/68Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D211/70Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/04Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to the ring carbon atoms
    • C07D215/08Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to the ring carbon atoms with acylated ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/135Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/52Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to compounds and pharmaceutical compositions, and to the uses thereof, that are effective for treating conditions characterized by overproduction of thyroid hormone.
  • thyroid hormone hyperthyroidism or thyrotoxicosis
  • thyrotoxicosis an extremely common clinical entity caused by a number of different pathological conditions of the thyroid gland.
  • Approximately 0.5% of women will experience some clinical manifestation of hyperthyroidism in their lifetime (a frequency 3 to 5 times higher than that occuring in men), with potentially life-threatening effects on the cardiovascular system, including cardiac arrhythmias, heart failure, angina and myocardial infarction, particularly in the elderly 1"3 .
  • the treatment of hyperthyroidism has essentially remained unchanged for the past thirty years, and includes the use of radioactive iodine, surgery, or the use of anti-thyroid drugs, such as propylthiouracil, that inhibit thyroid hormone synthesis by blocking the iodination of thyroglobulin 1"3 .
  • anti-thyroid drugs such as propylthiouracil
  • Propylthiouracil and related drugs which block thyroid hormone synthesis, act slowly and can take up to six to eight weeks to fully deplete the thyroid gland and intrathyroidal stores of iodinated thyroglobulin, during which time hyperthyroidism can have severe consequences in certain individuals.
  • Radiochemical destruction of thyroid tissues by iodine may require four to six months to be fully effective while surgical thyroidectomy must be preceded with anti-thyroid drugs to prevent life threatening complications such as thyroid storm.
  • TR thyroid hormone receptor
  • the present invention concerns the usage of ligands having the effect of antagonizing TR as pharmaceutical agents.
  • the compounds of interest are ligands capable of bonding to TR. These compounds and pharmaceutical compositions containing them are useful for the treatment of conditions such as hyperthyroidism which are characterized by an overproduction of TR by the thyroid gland. Additionally, the invention includes a method for the computer based screening, optimization, in vitro testing, and synthesis of novel compounds having TR antagonist activity using a library that may include commercially available starting compounds.
  • the invention provides pharmaceutical compositions that are capable of antagonizing TR, that have as an active ingredient a compound or compounds that have the structure of Formula I:
  • Ri is CH(CH 3 ) 2 , CH 2 CH 3 , CH 3 , or H ;
  • R 2 is CF 3 , CH 3 , F, or H;
  • R 3 is F, CH 3 , OCH 3 , CF 3 , or H;
  • the invention concerns a particular group of compounds according to Formula I that have been identified and synthesized herein, and that are ligands having TR antagonist activity that and may be defined by Formula I:
  • Ri is CH(CH 3 ) 2 , CH 2 CH 3 , CH 3 , or H ;
  • R 2 is CF 3 , CH 3 , F, or H;
  • R 3 is F, CH 3 , OCH 3j CF 3 , or H;
  • R 4 is CH 3 , OCH 3 , or H
  • R 2 s CF 3 , R-i, R 3 and R 4 are H; when R- ⁇ s CH(CH 3 ) 2 , R 2 and R 3 are H and R 4 is either CH 3 or H; when R- ⁇ s CH 2 CH 3 , R 2 , R 3 , and R 4 are H; when R 2 s CH 3 , R 1 and R 3 are H and R 4 is OCH 3 ; when R 2 s F, R 3 is also F and R and R 4 are H; when R 3 s F, R 2 is also F and R-i and R 4 are H; when R 1 s CH 3 , R 3 and R 4 are also CH 3 and R 2 is H; when R 3 s OCH 3 , R 1 f R 2 , and R 4 are H; when R 3 s CF 3 , R 1 , R 2 , and R 4 are H.
  • the invention concerns derivatives of a certain compound which is designated herein Compound F, and methods for the synthesis thereof, and the sythesis of its derivatives in turn, whereby said derivatives of Compound F comprise a class of compounds that may be generally represented by Formula II
  • R1 is F
  • R2 may be Cl, OCH 3 or F
  • R3 may be H or OCH 3 ;
  • R4 may be H or N0 3 ;
  • R5 may be H, OCH 3 or NO 2 ;
  • R6 may be H or OCH 3 .
  • the compounds prepared in accordance with Formula may be selected from the following:
  • R1 F
  • R2 OCH 3
  • R3 OCH 3
  • R4 H
  • R5 NO 2
  • R6 H
  • the invention provides a method for the identification, screening, optimization of selectivity of, and synthesis of compounds capable of antagonizing the effects of TR, wherein the method comprises the steps of i) selecting a compound, such as a compound selected from the group consisting of
  • ii) generating a virtual library of derivatives of the compound chosen in step i); iii) screening said library in silico; iv) chemically synthesizing at least one compound screened in iii); and v) testing in vitro at least one compound synthesized in iv).
  • the invention provides original ligands with TR antagonist activity in the DM range and sub-DM range.
  • the invention provides pharmaceutical compositions comprising one or more compounds of the invention, effective for the treatment of conditions such as hyperthyroidism and thyrotoxicosis characterized by overproduction of thyroid hormone wherein the compositions act by antagonizing TR.
  • the invention provides methods for modulation a process mediated by thyroid hormone nuclear receptors by administering to a human a compound or composition according to the invention that is capable of antagonizing TR.
  • Figure 1A is a predicted conformation of Compound A bound to the TR ⁇ ligand- binding pocket.
  • a hydrogen bond between His 435 and a carbonyl oxygen of Compound A and possibly between Arg 282 and a nitro-oxygen of Compound A constitute the only polar interactions. All other contacts are hydrophobic (not shown for clarity).
  • Figure 1 B shows a predicted conformation of Compound A superimposed with the crystal structure of T3 bound to active TR, and clashing with the active conformation of helix H12.
  • FIG. 2 is a graph showing inhibition of [ 125 I]T3 binding to TR by Compound A in intact cells.
  • the GH4C1 pituitary cell line which contains endogenous TRs (TR ⁇ , TR ⁇ 1 , and TR ⁇ 2), was incubated with 0.1 nM [ 125 I]T3 alone and with the indicated concentrations on unlabeled T3 and Compound A. After incubation for 60 min. at 37°C, the cells were chilled, washed, and the nuclei isolated. The results indicate the inhibition of binding of [ 125 I]T3 by T3 and Compound A.
  • Figure 3 is a comparison of the antagonist activity of Compound A and two of its derivative compounds (A-i and A 3 ) identified through in silico virtual library screening in accordance with the invention.
  • Figure 4 is a graph showing the inhibition of T3-mediated co-activator recruitment to TR by compounds A ⁇ A 3 , and A in vitro.
  • Approximately 2.5-5 x 10 4 cpm of 35 S-labeled TR ⁇ ⁇ (20 fmol) in 2 ⁇ l of lysate was incubated with 500 ng of GST fused to the receptor interaction region of the co-activator NRC (NRC15) immobilized on glutathione-agarose beads.
  • the samples were also incubated for 15 min at room temperature with 2 ⁇ M of Ai or A 3 or 5 ⁇ M of Compound A in binding buffer.
  • the samples were then chilled on ice and incubated with 1 nM T3 for an additional 60 min at 4°C.
  • Control samples contained no T3 or antagonists, or received only T3.
  • the beads were washed and the bound 35 S-TR ⁇ electrophoresed in a 10% SDS-gel followed by analysis and quantitation of the amount of 35 S-TR ⁇ bound using a Molecular Dynamics Phosphorimager and ImageQuant software.
  • the percent inhibition of T3-mediated binding of 35 S-TR ⁇ to GST-NRC15 by compounds A, Ai, and A 3 was determined after subtracting the amount of 35 S-TR ⁇ bound to GST-NRC15 in the absence of T3.
  • Figure 5 is a graph of the standard UV absorption-concentration correlation for compound A4 concentration in mice serum, as discussed in Example 4.
  • pharmaceutically acceptable means that the carrier, diluent, vehicle excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof;
  • salts of the compounds of this invention may be formed of the compound itself, prodrugs, e.g. esters, isomers and the like, and include all of the pharmaceutically acceptable salts which are most often used in pharmaceutical chemistry; for example, salts may be formed with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, carboxylic acids, sulfonic acids including such agents as naphthalenesulfonic, ethanesulfonic, hydroxyethanesulfonic, methanesulfonic ("mesylate"), benzenesulfonic ("besylate”) and toluenesulfonic acids, e.g., p-toluenesulfonic ("tosylate”), sulfuric acid, nitric acid, phosphoric acid, tartaric acid, pyrosulfuric acid, metaphosphoric acid, succinic acid, formic acid, phthalic
  • salts include salts of organic acids selected from formic, acetic, trifluoroacetic, propionic, benzoic, citric, maleic, tartaric, methanesulfonic, benzenesulfonic or toluenesulfonic, salts of inorganic acids selected from hydrochloric, hydrobromic, sulfuric or phosphoric, amino acids selected from aspartic and glutamic, and salts of sodium and potassium;
  • a “prodrug” is a drug precursor which, following administration, releases the drug in vivo via some chemical or physiological process (e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the desired drug form);
  • treating includes, inter alia, preventative (e.g., prophylactic), palliative and curative treatment.
  • the invention relates to the discovery of original ligands with TR antagonist activity in the DM range and sub- DM range, to pharmaceutical compositions containing such compounds, and to the uses thereof.
  • the activity of the thyroid hormones, L-thyroxin (T4) and L-triiodothyronine (T3), is mediated by the thyroid hormone receptor ("jR") 45 ' 50 ' 51 .
  • j ne TRs are members of the nuclear hormone receptor (NR) superfamily that also includes receptors for steroid hormones, retinoids, and 1 ,25-dihydroxy-vitamin D3 5"7 .
  • receptors are transcription factors that can regulate expression of specific genes in various tissues, and are targets for widely used drugs, such as tamoxifen, an estrogen receptor (ER) partial antagonist, flutamide, an anti-androgen, or rosiglitazone, a peroxisome proliferator activated receptor-D (PPARD) agonist (Dees 1998) (Olef sky 2000).
  • drugs such as tamoxifen, an estrogen receptor (ER) partial antagonist, flutamide, an anti-androgen, or rosiglitazone, a peroxisome proliferator activated receptor-D (PPARD) agonist (Dees 1998) (Olef sky 2000).
  • TR-D1 , TR-D1 and TR-D2 are differentially expressed in various tissues and have been described (Lazar 1993).
  • TSH thyroid stimulating hormone
  • T3 the negative feedback of thyroid stimulating hormone
  • TRD thyroid stimulating hormone
  • compounds according to the invention act to antagonize TR. Previous expertise in the structure/function of NRs facilitated the construction of a TR model in its antagonist-bound conformation (see Figs. 1A and 1 B).
  • antagonist candidate molecules are selected by in silico screening from a large library that may include known compounds.
  • Each ligand of the Available Chemicals Directory (“ACD”) of over 240,000 commercially available chemical structures was automatically docked into the model of the TR antagonist binding pocket.
  • ACD Available Chemicals Directory
  • the unexpected chemical diversity of active molecules identified underlines the power of the receptor-based rational lead drug-discovery approach.
  • the best scoring compounds were then further energy-minimized using a full atom representation of the receptor according to a double-scheme Monte-Carlo energy minimization procedure with both flexible ligand and flexible receptor side-chains.
  • Each compound was scored according to its fit with the TRD receptor model, taking into account continuum as well as discrete electrostatics, hydrophobicity, and entropy parameters.
  • Fourteen structurally diverse TR antagonists were identified in this way.
  • the 14 known molecules displaying TR antagonist activity are listed below in Table I in the order of apparent efficacy against an agonist concentration of 8 nM T3.
  • the unexpected chemical diversity of active molecules identified underlines the power of the receptor-based rational lead drug discovery approach.
  • Compound A chosen for its large (90%) inhibition at a concentration of 20 ⁇ M is further derivatized by the synthetic schemes outlined below.
  • Figure 1 A the predicted conformation of antagonist candidate Compound A bound to TRD ligand-binding pocket is shown.
  • a hydrogen bond between His435 and a carbonyl oxygen of Compound A and possibly between Arg 282 and a nitro-oxygen of Compound A would be the only polar interactions. All other contacts would be hydrophobic (not shown for clarity) highlighting the antagonistic properties of Compound A at the receptor site.
  • Figure 1 B shows that Compound A would superimpose with the crystal structure of T3 bound to active TR and would clash with the active conformation of helix H12.
  • a compound within the scope of Formula I shall at all times be understood to include all active forms of such compounds, including, for example, the free form thereof, e.g., the free acid or base form and also, all prodrugs, polymorphs, hydrates, solvates, and the like, and all pharmaceutically acceptable salts as described above. It will also be appreciated that suitable active metabolites of compounds within the scope of Formula I, in any suitable form, are also included herein.
  • certain compounds suitable for use in the present invention such as, for example, certain compounds of Formula I may have asymmetric centers and therefore exist in different enantiomeric forms. All suitable optical isomers and stereoisomers of such compounds, and mixtures thereof, are considered to be within the scope of the invention.
  • the present invention includes the use of a racemate, a single enantiomeric form, a single diastereomeric form, or mixtures thereof, as suitable.
  • such compounds may also exist as tautomers. Accordingly, the present invention relates to the use of all such suitable tautomers and mixtures thereof.
  • Compound F is further derivativized with commercially available building blocks to obtain a class of compounds represented in general form by Formula II below.
  • R1 F
  • R2 OCH 3
  • R3 OCH 3
  • R4 H
  • R5 NO 2
  • the invention comprises pharmaceutical compositions having synthesized compounds of Table 2, or other novel derivatives of the compounds of Table I, synthesized as above described, or prodrugs, isomers or pharmaceutically acceptable salts thereof, as their active ingredients.
  • Pharmaceutical compositions according to the invention preferably comprise a suitable amount of at least one compound, prodrug, isomer or pharmaceutically acceptable salt of this compound, (i.e. an amount sufficient to provide the desired dosage) along with a pharmaceutically acceptable vehicle, carrier or diluent.
  • the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention can be administered in a wide variety of different dosage forms, i.e., they may be combined with various pharmaceutically acceptable inert carriers in any suitable form.
  • Such carriers include solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents.
  • the pharmaceutical compositions can be formulated to contain a daily dose, or a convenient fraction of a daily dose, in a dosage unit, which may be a single tablet or a capsule or a convenient volume of a liquid.
  • any suitable route of administration may be used in the present invention. It is usually preferred to administer the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention orally for reasons of convenience; however, they may be administered, for example, percutaneously, or as suppositories for absorption by the rectum, as desired in a given instance. As described above, the administration may be carried out in single or multiple doses, as appropriate.
  • compositions may be used in the present invention, including tablets, lozenges, hard candies, chewable tablets, granules, powders, sprays, capsules, pills, microcapsules, solutions, parenteral solutions, troches, injections (e.g., intravenous, intraperitoneal, intramuscular or subcutaneous), suppositories, elixirs, syrups and suspensions.
  • tablets lozenges, hard candies, chewable tablets, granules, powders, sprays, capsules, pills, microcapsules, solutions, parenteral solutions, troches, injections (e.g., intravenous, intraperitoneal, intramuscular or subcutaneous), suppositories, elixirs, syrups and suspensions.
  • the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may be used as solutions in sesame or peanut oil, or as aqueous solutions (e.g., aqueous propyleneglycol), as the case may be, and they are best used in the form of a sterile aqueous solution which may contain other substances; for example, enough salts or glucose to make the solution isotonic, the pH of the solution being suitably adjusted and buffered, where necessary, and surfactants such as, for example, hydroxypropylcellulose.
  • aqueous solutions are suitable for intra-articular, intramuscular and subcutaneous injection purposes.
  • aqueous solutions are suitable for intravenous injection purposes.
  • the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may also be administered topically and this may be done by way of, e.g., creams, jellies, salves, lotions, gels, pastes, ointments, and the like, in accordance with standard pharmaceutical practice.
  • the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention of the present invention may also be administered transdermally (e.g., through the use of a patch).
  • any suitable formulation for transdermal application comprising a compound of the present invention may be employed and such formulations would generally also contain a suitable transdermal carrier, e.g., an absorbable pharmacologically acceptable solvent to promote and assist passage of the compounds through the subject's skin.
  • suitable transdermal devices may comprise the form of a bandage having a backing member and a reservoir containing the subject compound.
  • bandage-type transdermal devices may further include suitable carriers, rate- controlling barriers, and means for securing the transdermal device to the subject's skin.
  • the pharmaceutical compositions are prepared according to methods usual in pharmaceutical chemistry.
  • the pharmaceutical compositions can be prepared by methods commonly employed using conventional, organic or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g., cellulose, methylcellulose, hydroxymethylcellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g., starch, carboxymethylcellulose, hydroxypropylstarch, low substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate, or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous silicic acid, talc or sodium
  • Capsules can be prepared by mixing a compound, prodrug, isomer or pharmaceutically acceptable salt of the invention with a suitable diluent and filling the proper amount of the mixture in capsules.
  • suitable diluents include inert powdered substances such as starch of many different kinds, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
  • Tablets can be prepared by direct compression, by wet granulation, or by dry granulation. Their formulations usually incorporate diluents, binders, lubricants and disintegrators as well as a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention.
  • Common diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives may also be used.
  • Common tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, glucose and the like. Natural and synthetic gums are also convenient, including acacia, alginates, methylcellulose, polyvinylpyrrolidine and the like. Polyethylene glycol, ethylcellulose and waxes can also serve as binders.
  • a lubricant is generally necessary in a tablet formulation to prevent the tablet and punches from sticking in the die.
  • the lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
  • Tablet disintegrators include substances which swell when wetted to break up the tablet and release a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention. They include starches, clays, celluloses, algins and gums. More particularly, corn and potato starches, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, cation-exchange resins, alginic acid, guar gum, citrus pulp and carboxymethylcellulose, for example, may be used as well as sodium lauryl sulfate. [0049] Tablets are often coated with sugar as a flavor and sealant, or with film- forming protecting agents to modify the dissolution properties of the tablet.
  • the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may also be formulated as chewable tablets, by using large amounts of pleasant-tasting substances such as mannitol in the formulation, as is now well- established in the art.
  • any suitable base can be used.
  • Cocoa butter is a traditional suppository base, which may be modified by the addition of waxes to raise its melting point.
  • Water-miscible suppository bases comprising, particularly, polyethylene glycols of various molecular weights are also in wide use.
  • a slowly soluble pellet of a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention may be prepared and incorporated in a tablet or capsule.
  • the technique may be improved by making pellets of several different dissolution rates and filling capsules with a mixture of the pellets. Tablets or capsules may be coated with a film which resists dissolution for a predictable period of time.
  • parenteral preparations may also be made long-acting by dissolving or suspending a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention, as the case may be, in oily or emulsified vehicles which allow it to disperse only slowly in the serum.
  • the compounds of this invention are administered in a pharmaceutically effective amount.
  • the amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like, but in general will be about 0.01 % to about 20% of the total weight of the composition.
  • compositions of this invention can be administered by any suitable routes including, by way of illustration, oral, topical, rectal, transdermal, subcutaneous, intravenous, intramuscular, intranasal, and the like.
  • the compounds of this invention are preferably formulated as either oral, topical or injectable compositions.
  • compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, such compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the nitrone compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like.
  • Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • Topical compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the active ingredients When formulated as an ointment, the active ingredients will typically be combined with either a paraffinic or a water- miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example, an oil-in-water cream base.
  • Such topical formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration or stability of the active ingredients or the formulation. All such known topical formulations and ingredients are included within the scope of this invention.
  • the compounds of this invention can also be administered by a transdermal device. Accordingly, topical administration can be accomplished using a patch either of the reservoir or porous membrane type or of a solid matrix variety.
  • Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art.
  • the alkyl nitrone compound in such compositions is typically a minor component, often being from about 0.05 to 2% by weight with the remainder being the injectable carrier and the like.
  • the compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in the incorporated materials in Remington's Pharmaceutical Sciences.
  • EXAMPLE 1 In order to study the inhibition of binding of [ 125 i]T3 to TRs in intact cells by the antagonist molecule Compound A, GH4 rat pituitary cells containing endogenous TRs (TR ⁇ 1 , TR ⁇ 1 , and TR ⁇ 2) were grown in monolayer culture in DMEM medium containing 10% calf serum. Cells were dispersed by incubation in a buffered solution of EDTA and incubated at 37 ° C for 60 min in serum-free DMEM to lower endogenous levels of thyroid hormones.
  • the samples were washed twice by re-suspension and vortexing with 1 ml of 50 mM Tris-HCI, pH 7.85, containing 1 mM MgCI 2 and 0.5% Triton X-100 and centrifugation at 1 ,000 x g for 10 min to isolate the nuclear fraction of the cells.
  • the amount of [ 125 I]T3 retained in the resulting pellet of washed nuclei was determined using a Packard gamma spectrometer.
  • the results are presented in Figure 2 as a percent of radioactivity retained in washed nuclei from cells incubated with 0.1 nM [ 125 I]T3 in the absence of unlabeled T3 or antagonist candidates. Each data point represents the average of duplicates, which generally varied by less than 5%.
  • EXAMPLE 2 Functional CAT assays were performed to compare the extent of inhibition of the T3 stimulation of CAT activity observed in the presence of the antagonist candidates.
  • HeLa cells were innoculated at 50,000 cells per well in 24 well plates in DMEM containing 10% calf serum. The cells were transfected 5 hours later by calcium phosphate precipitation using 450ng of the T3 responsive ⁇ MTV-IR-CAT reporter and 250 ng of a vector expressing TR ⁇ . At the time of transfection, the cells also received 6nM T3 and the different concentrations of the antagonist candidates. Cells were harvested 40h after transfection and assayed for protein content and CAT activity. Results, shown in Figure 3, are expressed as the extent of inhibition of the T3 stimulation of CAT activity observed in the presence of the antagonist candidates. Each data point reflects the average of triplicate samples which showed less than 10% variation.
  • EXAMPLE 3 A study to compare the T3-mediated co-activator recruitment to TR by A 1 ; A 3 , and Compound A was conducted in vitro. Approximately 2.5-5 x 10 4 cpm of 35 S- labeled TR ⁇ (20 fmol) in 2 ⁇ l of lysate was incubated with 500 ng of GST fused to the receptor interaction region of the co-activator NRC (NRC15) immobilized on a glutathione-agarose beads. The samples were also incubated for 15 min at room temperature with of A- ⁇ or A 3 or 5 ⁇ M of Compound A in binding buffer. The samples were then chilled on ice and incubated with 1 nM T3 for an additional 60 min at 4 ° C.
  • Control samples contained no T3 or antagonists, or received only T3.
  • the beads were washed and the bound 35 S-TR ⁇ electrphoresed in a 10% SDS gel followed by analysis and quantitation of that amount of 35 S-TR ⁇ bound using a Molecular Dynamics Phosphorimager and ImageQuant software.
  • the percent inhibition of T3- mediated binding of 35 S-TR ⁇ to GST-NRC15 by Compounds A, A 1 and A 3 was determined after subtracting the amount of 35 S-TR ⁇ bound to GST-NRC15 in the absence of T3.
  • the results are shown in Figure 4 and show the inhibitory effect of the compounds as A 3 >A ⁇ >A, confirming the increased efficacy of compounds, synthesized from antagonists candidates chosen according to the Invention, whose selectivity is optimized as described herein.
  • a 3 compound has Rf value of 0.3 when eluent is ethyl acetate in TLC study.
  • a 3 compound has water solubility approximately of 10 ⁇ M and ethyl acetate can be used to extract A 3 out of aqueous phase.
  • a 3 compound shows non-linear optical property.
  • Macchia PE Takeuchi Y, Kawai T, Cua K, Gauthier K, Chassande O, Seo H, Hayashi Y, Samarut J, Murata Y, Weiss RE, Refetoff S. (2001) Increased sensitivity to thyroid hormone in mice with complete deficiency of thyroid hormone receptor alpha. Proc Natl Acad Sci U S A; 98(1 ):349-54.
  • TLS Translocated-in-Liposarcoma

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Provided are compounds, pharmaceutical compositions, and methods for the synthesis and use thereof that are effective for the modulation or treatment of conditions characterized by overproduction of thyroid hormone, wherein the effective compounds act by antagonizing the effect of thyroid hormone at the receptor level.

Description

DIVERSE THYROID HORMONE RECEPTOR ANTAGONISTS AND USES THEREOF
CROSS REFERENCE TO RELATED APPLICATION [0001] This application claims benefit of priority to provisional patent application Serial No. 60/470,749, filed May 14, 2003, which is incorporated herein by reference in its entirety.
STATEMENT OF GOVERNMENTAL SUPPORT [0002] The research leading to the present invention was supported, at least in part, by National Institute of Health (NIH) Grant Nos. DK16636 and DK059041. Accordingly, the United States Government may have certain rights in the invention.
FIELD OF THE INVENTION [0003] The invention relates to compounds and pharmaceutical compositions, and to the uses thereof, that are effective for treating conditions characterized by overproduction of thyroid hormone.
BACKGROUND OF THE INVENTION
[0004] Overproduction of thyroid hormone (hyperthyroidism or thyrotoxicosis) is an extremely common clinical entity caused by a number of different pathological conditions of the thyroid gland. Approximately 0.5% of women will experience some clinical manifestation of hyperthyroidism in their lifetime (a frequency 3 to 5 times higher than that occuring in men), with potentially life-threatening effects on the cardiovascular system, including cardiac arrhythmias, heart failure, angina and myocardial infarction, particularly in the elderly1"3.
[0005] The treatment of hyperthyroidism has essentially remained unchanged for the past thirty years, and includes the use of radioactive iodine, surgery, or the use of anti-thyroid drugs, such as propylthiouracil, that inhibit thyroid hormone synthesis by blocking the iodination of thyroglobulin1"3. Each approach has its own intrinsic limitations and/or side effects. Propylthiouracil and related drugs, which block thyroid hormone synthesis, act slowly and can take up to six to eight weeks to fully deplete the thyroid gland and intrathyroidal stores of iodinated thyroglobulin, during which time hyperthyroidism can have severe consequences in certain individuals. Radiochemical destruction of thyroid tissues by iodine may require four to six months to be fully effective while surgical thyroidectomy must be preceded with anti-thyroid drugs to prevent life threatening complications such as thyroid storm.
[0006] The identification of thyroid hormone receptor ("TR") antagonists could play an important role in the future treatment of hyperthyroidism. Such molecules would act rapidly by directly antagonizing the effect of thyroid hormone at the receptor level, a significant improvement for individuals with hyperthyroidism who require surgery, have cardiac disease, or life threatening thyrotoxic storm.
SUMMARY OF THE INVENTION
[0007] The present invention concerns the usage of ligands having the effect of antagonizing TR as pharmaceutical agents. The compounds of interest are ligands capable of bonding to TR. These compounds and pharmaceutical compositions containing them are useful for the treatment of conditions such as hyperthyroidism which are characterized by an overproduction of TR by the thyroid gland. Additionally, the invention includes a method for the computer based screening, optimization, in vitro testing, and synthesis of novel compounds having TR antagonist activity using a library that may include commercially available starting compounds.
[0008] In a first aspect, the invention provides pharmaceutical compositions that are capable of antagonizing TR, that have as an active ingredient a compound or compounds that have the structure of Formula I:
Figure imgf000004_0001
In Formula I:
Ri is CH(CH3)2, CH2 CH3, CH3, or H ; R2 is CF3, CH3, F, or H; R3 is F, CH3, OCH3, CF3, or H; and
Figure imgf000004_0002
[0009] In a second aspect, the invention concerns a particular group of compounds according to Formula I that have been identified and synthesized herein, and that are ligands having TR antagonist activity that and may be defined by Formula I:
Figure imgf000005_0001
wherein
Ri is CH(CH3)2, CH2 CH3, CH3, or H ; R2 is CF3, CH3, F, or H; R3 is F, CH3, OCH3j CF3, or H; and R4 is CH3, OCH3, or H
provided that when R2 s CF3, R-i, R3 and R4are H; when R-\ s CH(CH3) 2, R2 and R3 are H and R4 is either CH3 or H; when R-\ s CH2CH3, R2, R3, and R4 are H; when R2 s CH3, R1 and R3 are H and R4 is OCH3; when R2 s F, R3 is also F and R and R4 are H; when R3 s F, R2 is also F and R-i and R4 are H; when R1 s CH3, R3 and R4 are also CH3 and R2 is H; when R3 s OCH3, R1 f R2, and R4 are H; when R3 s CF3, R1, R2, and R4 are H.
[0010] In a third aspect, the invention concerns derivatives of a certain compound which is designated herein Compound F, and methods for the synthesis thereof, and the sythesis of its derivatives in turn, whereby said derivatives of Compound F comprise a class of compounds that may be generally represented by Formula II
Figure imgf000005_0002
Formula II wherein
R1 is F;
R2 may be Cl, OCH3 or F;
R3 may be H or OCH3;
R4 may be H or N03;
R5 may be H, OCH3 or NO2; and
R6 may be H or OCH3.
In a preferred embodiment, the compounds prepared in accordance with Formula may be selected from the following:
R1 = F, R2 = Cl, R3 = H, R4=H, R5=NO2, R6=H
R1 = F, R2 = Cl, R3 = H, R4=H, R5=H, R6=OCH3
R1 = F, R2 = OCH3, R3 = OCH3, R4=H, R5=NO2, R6=H
R1 = F, R2 = Cl, R3 = H, R4=NO2, R5=H, R6=H
R1 = CH3, R2 = F, R3 = H, R4=H, R5= OCH3, R6=H
[001 1 ] In a fourth aspect, the invention provides a method for the identification, screening, optimization of selectivity of, and synthesis of compounds capable of antagonizing the effects of TR, wherein the method comprises the steps of i) selecting a compound, such as a compound selected from the group consisting of
Figure imgf000006_0001
Figure imgf000007_0001
ii) generating a virtual library of derivatives of the compound chosen in step i); iii) screening said library in silico; iv) chemically synthesizing at least one compound screened in iii); and v) testing in vitro at least one compound synthesized in iv).
[0012] In a fifth aspect, the invention provides original ligands with TR antagonist activity in the DM range and sub-DM range. [0013] In a sixth aspect, the invention provides pharmaceutical compositions comprising one or more compounds of the invention, effective for the treatment of conditions such as hyperthyroidism and thyrotoxicosis characterized by overproduction of thyroid hormone wherein the compositions act by antagonizing TR.
[0014] In a seventh aspect, the invention provides methods for modulation a process mediated by thyroid hormone nuclear receptors by administering to a human a compound or composition according to the invention that is capable of antagonizing TR.
BRIEF DESCRIPTION OF THE FIGURES
[0015] Figure 1A is a predicted conformation of Compound A bound to the TRβ ligand- binding pocket. A hydrogen bond between His 435 and a carbonyl oxygen of Compound A and possibly between Arg 282 and a nitro-oxygen of Compound A constitute the only polar interactions. All other contacts are hydrophobic (not shown for clarity).
[0016] Figure 1 B shows a predicted conformation of Compound A superimposed with the crystal structure of T3 bound to active TR, and clashing with the active conformation of helix H12.
[0017] Figure 2 is a graph showing inhibition of [125I]T3 binding to TR by Compound A in intact cells. The GH4C1 pituitary cell line, which contains endogenous TRs (TRα, TRβ1 , and TRβ2), was incubated with 0.1 nM [125I]T3 alone and with the indicated concentrations on unlabeled T3 and Compound A. After incubation for 60 min. at 37°C, the cells were chilled, washed, and the nuclei isolated. The results indicate the inhibition of binding of [125I]T3 by T3 and Compound A.
[0018] Figure 3 is a comparison of the antagonist activity of Compound A and two of its derivative compounds (A-i and A3) identified through in silico virtual library screening in accordance with the invention.
[0019] Figure 4 is a graph showing the inhibition of T3-mediated co-activator recruitment to TR by compounds A^ A3, and A in vitro. Approximately 2.5-5 x 104 cpm of 35S-labeled TRα α(20 fmol) in 2 μl of lysate was incubated with 500 ng of GST fused to the receptor interaction region of the co-activator NRC (NRC15) immobilized on glutathione-agarose beads. The samples were also incubated for 15 min at room temperature with 2 μM of Ai or A3 or 5 μM of Compound A in binding buffer. The samples were then chilled on ice and incubated with 1 nM T3 for an additional 60 min at 4°C. Control samples contained no T3 or antagonists, or received only T3. The beads were washed and the bound 35S-TRα electrophoresed in a 10% SDS-gel followed by analysis and quantitation of the amount of 35S-TRα bound using a Molecular Dynamics Phosphorimager and ImageQuant software. The percent inhibition of T3-mediated binding of 35S-TRα to GST-NRC15 by compounds A, Ai, and A3 was determined after subtracting the amount of 35S-TRα bound to GST-NRC15 in the absence of T3.
[0020] Figure 5 is a graph of the standard UV absorption-concentration correlation for compound A4 concentration in mice serum, as discussed in Example 4.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT [0021] Definitions. Unless otherwise provided herein:
[0022] "pharmaceutically acceptable" means that the carrier, diluent, vehicle excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof;
[0023] "pharmaceutically acceptable salts" of the compounds of this invention may be formed of the compound itself, prodrugs, e.g. esters, isomers and the like, and include all of the pharmaceutically acceptable salts which are most often used in pharmaceutical chemistry; for example, salts may be formed with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, carboxylic acids, sulfonic acids including such agents as naphthalenesulfonic, ethanesulfonic, hydroxyethanesulfonic, methanesulfonic ("mesylate"), benzenesulfonic ("besylate") and toluenesulfonic acids, e.g., p-toluenesulfonic ("tosylate"), sulfuric acid, nitric acid, phosphoric acid, tartaric acid, pyrosulfuric acid, metaphosphoric acid, succinic acid, formic acid, phthalic acid, malic acid, maleic acid, lactic acid, ascorbic acid, glycollic acid, gluconic acid, mandelic acid, glutamic acid, aspartic acid, fumaric acid, pyruvic acid, phenylacetic acid, pamoic acid, nicotinic acid, and the like; suitable pharmaceutically acceptable salts also include alkali metal salts (e.g. sodium, potassium salts), alkaline earth metal salts (e.g. magnesium, calcium salts), amine salts (e.g. ammonium, alkylammonium, dialkylammonium, trialkylammonium, tetraalkylammonium, diethanolaminium, tri-ethanolaminium and guanidinium salts); preferred salts include salts of organic acids selected from formic, acetic, trifluoroacetic, propionic, benzoic, citric, maleic, tartaric, methanesulfonic, benzenesulfonic or toluenesulfonic, salts of inorganic acids selected from hydrochloric, hydrobromic, sulfuric or phosphoric, amino acids selected from aspartic and glutamic, and salts of sodium and potassium;
[0024] a "prodrug" is a drug precursor which, following administration, releases the drug in vivo via some chemical or physiological process (e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the desired drug form);
[0025] "treating," "treat" or "treatment" includes, inter alia, preventative (e.g., prophylactic), palliative and curative treatment.
[0026] The invention relates to the discovery of original ligands with TR antagonist activity in the DM range and sub- DM range, to pharmaceutical compositions containing such compounds, and to the uses thereof. The activity of the thyroid hormones, L-thyroxin (T4) and L-triiodothyronine (T3), is mediated by the thyroid hormone receptor ("jR") 45'50'51. jne TRs are members of the nuclear hormone receptor (NR) superfamily that also includes receptors for steroid hormones, retinoids, and 1 ,25-dihydroxy-vitamin D35"7. These receptors are transcription factors that can regulate expression of specific genes in various tissues, and are targets for widely used drugs, such as tamoxifen, an estrogen receptor (ER) partial antagonist, flutamide, an anti-androgen, or rosiglitazone, a peroxisome proliferator activated receptor-D (PPARD) agonist (Dees 1998) (Olef sky 2000).
[0027] Several different isoforms of TR (TR-D1 , TR-D1 and TR-D2) are differentially expressed in various tissues and have been described (Lazar 1993). Gene knockout studies in mice indicate that the TRD isoforms plays a role in the development of the auditory system and in the negative feedback of thyroid stimulating hormone ("TSH") by T3 in the pituitary (Forrest 1996, Weiss 1997), while TRD modulates the effect of thyroid hormone on calorigenesis and on the cardiovascular system (Wikstrom 1998). [0028] In a preferred embodiment, compounds according to the invention act to antagonize TR. Previous expertise in the structure/function of NRs facilitated the construction of a TR model in its antagonist-bound conformation (see Figs. 1A and 1 B).
[0029] In a preferred embodiment, antagonist candidate molecules are selected by in silico screening from a large library that may include known compounds. Each ligand of the Available Chemicals Directory ("ACD") of over 240,000 commercially available chemical structures was automatically docked into the model of the TR antagonist binding pocket. The unexpected chemical diversity of active molecules identified underlines the power of the receptor-based rational lead drug-discovery approach. The best scoring compounds were then further energy-minimized using a full atom representation of the receptor according to a double-scheme Monte-Carlo energy minimization procedure with both flexible ligand and flexible receptor side-chains. Previous achievements demonstrate that this strategy can successfully identify receptor-specific ligands with appropriate biological systems and robust modeling tools (Schapira 2000, Filikov 2000, Schapira 2001 , Abagyan 2002). As a non-limiting example, the C-terminal H12 helix of TR was docked onto the hydrophobic co- activator recruitment site of the receptor, and the energy of the system was minimized in the internal coordinates space by an extensive Monte Carlo simulation, which may be (and this illustration was) performed with Molsoft's ICM technology (ICM 2.8 Manual).
[0030] Each compound was scored according to its fit with the TRD receptor model, taking into account continuum as well as discrete electrostatics, hydrophobicity, and entropy parameters. Fourteen structurally diverse TR antagonists were identified in this way. One optimization cycle, based on one of the 14 active structures, allowed improvement of the affinity for the receptor. The 14 known molecules displaying TR antagonist activity are listed below in Table I in the order of apparent efficacy against an agonist concentration of 8 nM T3. The unexpected chemical diversity of active molecules identified underlines the power of the receptor-based rational lead drug discovery approach.
TABLE 1
Concentration (DM) Inhibition (%)
Figure imgf000012_0001
Figure imgf000013_0002
[0031] In a preferred embodiment, Compound A, chosen for its large (90%) inhibition at a concentration of 20μM is further derivatized by the synthetic schemes outlined below. Referring to Figure 1 A, the predicted conformation of antagonist candidate Compound A bound to TRD ligand-binding pocket is shown. A hydrogen bond between His435 and a carbonyl oxygen of Compound A and possibly between Arg 282 and a nitro-oxygen of Compound A would be the only polar interactions. All other contacts would be hydrophobic (not shown for clarity) highlighting the antagonistic properties of Compound A at the receptor site. Figure 1 B shows that Compound A would superimpose with the crystal structure of T3 bound to active TR and would clash with the active conformation of helix H12.
[0032] Eight compounds, Compounds A^As, all derivatives of Compound A, were actually synthesized and tested in vitro. The calculated score, corresponding rank, structure and activity for each compound, represented as a species of a genus represented by Formula I, are listed below in Table 2. The best two inhibitors were among the top 4 scoring compounds (Ai and A3 respectively), while the two less active molecules were the worst scoring ones (A7 and A8 respectively). One derivative (A3) reached IC-50 in the nanomolar range (0.75 DM)
Formula I
Figure imgf000013_0001
TABLE 2
Figure imgf000013_0003
Figure imgf000014_0001
[0033] As disclosed herein, a compound within the scope of Formula I shall at all times be understood to include all active forms of such compounds, including, for example, the free form thereof, e.g., the free acid or base form and also, all prodrugs, polymorphs, hydrates, solvates, and the like, and all pharmaceutically acceptable salts as described above. It will also be appreciated that suitable active metabolites of compounds within the scope of Formula I, in any suitable form, are also included herein.
[0034] Moreover, certain compounds suitable for use in the present invention such as, for example, certain compounds of Formula I may have asymmetric centers and therefore exist in different enantiomeric forms. All suitable optical isomers and stereoisomers of such compounds, and mixtures thereof, are considered to be within the scope of the invention. With respect to such compounds, the present invention includes the use of a racemate, a single enantiomeric form, a single diastereomeric form, or mixtures thereof, as suitable. Moreover, such compounds may also exist as tautomers. Accordingly, the present invention relates to the use of all such suitable tautomers and mixtures thereof.
[0035] In a preferred embodiment of the invention, the scheme shown below is employed to generate a virtual library focused on Compound A, in which Compound A is divided into three structural units that can be derivatized independently with commercially available building blocks".
Figure imgf000015_0001
[0036] In another embodiment, Compound F is further derivativized with commercially available building blocks to obtain a class of compounds represented in general form by Formula II below.
Figure imgf000015_0002
Formula II
Where
R1 = F, R2 = Cl, R3 = H, R4=H, R5=NO2, R6=H =10μM
R1 = F, R2 = Cl, R3 = H, R4=H, R5=H, R6=OCH3 =10μM
R1 = F, R2 = OCH3, R3 = OCH3, R4=H, R5=NO2, R6=H =5μM
R1 = F, R2 = Cl, R3 = H, R4=NO2, R5=H, R6=H =10μM
R1 = CH3, R2 = F, R3 = H, R4=H, R5= OCH3, R6=H Ki=10μM
[0037] In a preferred embodiment, the invention comprises pharmaceutical compositions having synthesized compounds of Table 2, or other novel derivatives of the compounds of Table I, synthesized as above described, or prodrugs, isomers or pharmaceutically acceptable salts thereof, as their active ingredients. Pharmaceutical compositions according to the invention preferably comprise a suitable amount of at least one compound, prodrug, isomer or pharmaceutically acceptable salt of this compound, (i.e. an amount sufficient to provide the desired dosage) along with a pharmaceutically acceptable vehicle, carrier or diluent.
[0038] The compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention can be administered in a wide variety of different dosage forms, i.e., they may be combined with various pharmaceutically acceptable inert carriers in any suitable form. Such carriers include solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents. The pharmaceutical compositions can be formulated to contain a daily dose, or a convenient fraction of a daily dose, in a dosage unit, which may be a single tablet or a capsule or a convenient volume of a liquid.
[0039] Any suitable route of administration may be used in the present invention. It is usually preferred to administer the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention orally for reasons of convenience; however, they may be administered, for example, percutaneously, or as suppositories for absorption by the rectum, as desired in a given instance. As described above, the administration may be carried out in single or multiple doses, as appropriate.
[0040] All of the usual types of pharmaceutical compositions may be used in the present invention, including tablets, lozenges, hard candies, chewable tablets, granules, powders, sprays, capsules, pills, microcapsules, solutions, parenteral solutions, troches, injections (e.g., intravenous, intraperitoneal, intramuscular or subcutaneous), suppositories, elixirs, syrups and suspensions.
[0041] For parenteral administration, the compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may be used as solutions in sesame or peanut oil, or as aqueous solutions (e.g., aqueous propyleneglycol), as the case may be, and they are best used in the form of a sterile aqueous solution which may contain other substances; for example, enough salts or glucose to make the solution isotonic, the pH of the solution being suitably adjusted and buffered, where necessary, and surfactants such as, for example, hydroxypropylcellulose. Such oily solutions are suitable for intra-articular, intramuscular and subcutaneous injection purposes. Such aqueous solutions are suitable for intravenous injection purposes.
[0042] The compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may also be administered topically and this may be done by way of, e.g., creams, jellies, salves, lotions, gels, pastes, ointments, and the like, in accordance with standard pharmaceutical practice. The compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention of the present invention may also be administered transdermally (e.g., through the use of a patch). Any suitable formulation for transdermal application comprising a compound of the present invention may be employed and such formulations would generally also contain a suitable transdermal carrier, e.g., an absorbable pharmacologically acceptable solvent to promote and assist passage of the compounds through the subject's skin. For example, suitable transdermal devices may comprise the form of a bandage having a backing member and a reservoir containing the subject compound. Such bandage-type transdermal devices may further include suitable carriers, rate- controlling barriers, and means for securing the transdermal device to the subject's skin.
[0043] In general, all of the pharmaceutical compositions are prepared according to methods usual in pharmaceutical chemistry. As will be described in detail hereinbelow, the pharmaceutical compositions can be prepared by methods commonly employed using conventional, organic or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g., cellulose, methylcellulose, hydroxymethylcellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g., starch, carboxymethylcellulose, hydroxypropylstarch, low substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate, or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous silicic acid, talc or sodium lauryl sulfate), a flavoring agent (e.g., citric acid, menthol, glycine or orange powder), a preservative (e.g., sodium benzoate, sodium bisulfite, methylparaben or propylparaben), a stabilizer (e.g., citric acid, sodium citrate or acetic acid), a suspending agent (e.g., methylcellulose, polyvinylpyrrolidone, or aluminum stearate), a dispersing agent (e.g., hydroxypropylmethylcellulose), a diluent (e.g., water), a coloring agent, an emulsifying agent, and a base wax (e.g., cocoa butter, white petrolatum or polyethylene glycol). [0044] Any of the compounds, prodrugs, isomers or pharmaceutically acceptable salts of this invention may be readily formulated as tablets, capsules, and the like. It is preferable to prepare solutions from water-soluble salts, such as the hydrochloride salt.
[0045] Capsules can be prepared by mixing a compound, prodrug, isomer or pharmaceutically acceptable salt of the invention with a suitable diluent and filling the proper amount of the mixture in capsules. The usual diluents include inert powdered substances such as starch of many different kinds, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
[0046] Tablets can be prepared by direct compression, by wet granulation, or by dry granulation. Their formulations usually incorporate diluents, binders, lubricants and disintegrators as well as a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention. Common diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives may also be used. Common tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, glucose and the like. Natural and synthetic gums are also convenient, including acacia, alginates, methylcellulose, polyvinylpyrrolidine and the like. Polyethylene glycol, ethylcellulose and waxes can also serve as binders.
[0047] A lubricant is generally necessary in a tablet formulation to prevent the tablet and punches from sticking in the die. The lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
[0048] Tablet disintegrators include substances which swell when wetted to break up the tablet and release a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention. They include starches, clays, celluloses, algins and gums. More particularly, corn and potato starches, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, cation-exchange resins, alginic acid, guar gum, citrus pulp and carboxymethylcellulose, for example, may be used as well as sodium lauryl sulfate. [0049] Tablets are often coated with sugar as a flavor and sealant, or with film- forming protecting agents to modify the dissolution properties of the tablet. The compounds, prodrugs, isomers and pharmaceutically acceptable salts of this invention may also be formulated as chewable tablets, by using large amounts of pleasant-tasting substances such as mannitol in the formulation, as is now well- established in the art.
[0050] Where it is desired to administer a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention as a suppository, any suitable base can be used. Cocoa butter is a traditional suppository base, which may be modified by the addition of waxes to raise its melting point. Water-miscible suppository bases comprising, particularly, polyethylene glycols of various molecular weights are also in wide use.
[0051] As discussed above, the effect of a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention may be delayed or prolonged by proper formulation. For example, a slowly soluble pellet of a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention may be prepared and incorporated in a tablet or capsule. The technique may be improved by making pellets of several different dissolution rates and filling capsules with a mixture of the pellets. Tablets or capsules may be coated with a film which resists dissolution for a predictable period of time. The parenteral preparations may also be made long-acting by dissolving or suspending a compound, prodrug, isomer or pharmaceutically acceptable salt of this invention, as the case may be, in oily or emulsified vehicles which allow it to disperse only slowly in the serum.
[0052] Generally, the compounds of this invention are administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like, but in general will be about 0.01 % to about 20% of the total weight of the composition.
[0053] The pharmaceutical compositions of this invention can be administered by any suitable routes including, by way of illustration, oral, topical, rectal, transdermal, subcutaneous, intravenous, intramuscular, intranasal, and the like. Depending on the intended route of delivery, the compounds of this invention are preferably formulated as either oral, topical or injectable compositions.
[0054] Pharmaceutical compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, such compositions are presented in unit dosage forms to facilitate accurate dosing. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the nitrone compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
[0055] Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like. Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
[0056] Topical compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight. When formulated as an ointment, the active ingredients will typically be combined with either a paraffinic or a water- miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example, an oil-in-water cream base. Such topical formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration or stability of the active ingredients or the formulation. All such known topical formulations and ingredients are included within the scope of this invention.
[0057] The compounds of this invention can also be administered by a transdermal device. Accordingly, topical administration can be accomplished using a patch either of the reservoir or porous membrane type or of a solid matrix variety.
[0058] Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art. As before, the alkyl nitrone compound in such compositions is typically a minor component, often being from about 0.05 to 2% by weight with the remainder being the injectable carrier and the like.
[0059] The above-described components for orally and topically administrable or injectable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington's Pharmaceutical Sciences, 18th edition, 1990, Mack Publishing Company, Easton, Pennsylvania, 18042, which is incorporated herein by reference.
[0060] The compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in the incorporated materials in Remington's Pharmaceutical Sciences.
[0061] The following synthetic schemes and protocols may be used to synthesize compounds according to the invention.
Figure imgf000022_0001
Figure imgf000022_0002
R!-R5 = H in this example
Figure imgf000022_0003
Figure
[0062] The synthesis was carried out in 5 steps, as depicted the Figure above. In the first step (1 ), 2.78 g (20 mMol) of K2C03 was added to a solution of 1 ,4-Dioxa-8- azaspiro[4.5]decane (2.86 g, 20 mMol) and 1 -f luoro-4-nitrobenzene (1.41 g, lOmMol) in 10ml acetonitrile. The resulting suspension was ref luxed under nitrogen for 3 days, cooled to room temperature, and 50ml water was added and extracted with 300ml CH2CI2 twice. The organic layers were combined, washed with brine, and dried over Na2SO4. Trituration with cold ethyl ether afforded 2.53g (96%) of 1 as a white solid. This compound was pure enough and no further purification was needed. 1H NMR (CDCI3) δ 1.93 (triplet, 4H), 3.65 (triplet, 4H), 4.02 (singlet, 4H), 7.15 (doublet, 2H), 8.19 (doublet, 2H).
[0063] 1.69g (or 6.4 mMol) of 1 was dissolved in 20ml THF and 10ml 10% H2SO4 was added (1 ). This mixture was stirred at room temperature for 4 days, diluted with 30ml H2O and extracted with 300 ml CH2CI2. The organic part was washed with brine and dried. The solvent was removed to produce 1.32 g (94%) of 2, a slightly yellow solid. 1H NMR ( CDCI3) δ 2.68 (triplet, 4H), 3.91 (triplet, 4H), 6.98 (doublet, 2H), 8.22 (doublet 2H).
[0064] 0.66 g of 2 (3 mMol) and 2.1 g of Ph3P=CHCOOC2H5 (6 mMol) were mixed and heated to 160°C overnight under N2 (2). The reaction mixture was cooled to room temperature and purified by flash chromatography [hexane/ethyl acetate 7:3 ( v/v ), R, 0.26] to give 0.65 g (76%) of 3. 1 H NMR ( CDCI3) δ 1.34 (triplet, 3H), 2.34 (broad, 2H), 3.11 (broad, 2H), 3.59 (broad, 2H), 3.89 (broad, 2H), 4.18 (multiplet, 2H), 5.71 (br, 1 H), 6.81 ( triplet, 2H), 8.18 (triplet, 2H).
[0065] The mixture of 0.29 g of 3 (1 mMol) and 0.48 g anhydrous NH2NH2 (15 mMol) in 40 ml absolute ethanol was refluxed for 3 hours (3). After cooling to ambient temperature, the solution was concentrated in vacuo. The solid residue was crystallized from ethanol, affording compound 2.52 g (91 %) of 4. 1H NMR (DMSO) δ 2.22 (broad, 2H), 2.83 (broad, 2H), 3.60 (broad, 2H), 3.91 (broad, 2H), 5.68 (singlet, 1 H), 7.02 (doublet, 2H), 8.05 (doublet, 2H)
[0066] In the final step, 0.5 mMol of the commercially available phenylisocyanate was added to a solution of 0.13 g 4 (O.δmmol) in 1 ml dry CH2CI2 (4), and stirred at room temperature for 2 hours. The final product was separated by filtration.
A1 1H NMR 1.10 (doublet, 6H), 2.16 (singlet, 3H), 2.26 (broad, 2H), 2.96 (broad, 2H), 3.34 (multiplet, 1 H), 3.36 (broad, 2H), 3.92 (broad, 2H), 5.69 (singlet, 1H), 7.03 (multiplet, 3H), 7.13 (doublet, 2H), 7.87 (singlet, 1 H), 8.05 (doublet, 2H), 9.78 (singlet, 1 H). MS
A3 1H NMR 1.13 (doublet, 6H), 2.26 (broad, 2H), 2.97 (broad, 2H), 3.32 (multiplet, 1H), 3.64 (broad, 2H), 3.94 (broad, 2H), 5.71 (singlet, 1H), 6.96 (doublet, 2H), 7.12 (triplet, 2H), 7.26 (multiplet, 1 H), 7.44 (multiplet, 1 H), 8.11 (doublet, 2H), 8.28 (singlet, 1 H), 9.82 (singlet, 1 H).
[0067] Combinatorial chemistry (Figure below) is used to design and synthesize four new classes of compounds (Schemes 1-4).
Figure imgf000024_0001
Evolve (change) Evolve (change)
Figure imgf000024_0003
Figure imgf000024_0002
Figure imgf000024_0004
Figure: Combinatorial evolution of new compounds
-N o° NH2NH2
Figure imgf000024_0005
Figure imgf000024_0006
Scheme 1
Figure imgf000025_0001
Figure imgf000025_0002
Scheme 3
Figure imgf000026_0001
Scheme 4
[0068] While in no way intending to be bound by theory, in order to further illustrate and characterize the physiochemical and bioactive properties of the compounds and compositions of the invention, inventors provide the following non-limiting examples.
EXAMPLE 1 [0069] In order to study the inhibition of binding of [125i]T3 to TRs in intact cells by the antagonist molecule Compound A, GH4 rat pituitary cells containing endogenous TRs (TRα1 , TRβ1 , and TRβ2) were grown in monolayer culture in DMEM medium containing 10% calf serum. Cells were dispersed by incubation in a buffered solution of EDTA and incubated at 37°C for 60 min in serum-free DMEM to lower endogenous levels of thyroid hormones. Aliquots containing approximately 1.5 million cells were collected by centrif ugation at 1000 x g for 10 min and then suspended in 1 mL of serum free medium containing 0.1 nM [125I]T3 and the indicated concentrations (see Figure 2) of unlabeled T3 or the antagonist candidate, Compound A. Following incubation at 37°C for 60 min, the cells were chilled in ice and then centrifuged at 4°C at 1000x g for 10 min. The samples were washed twice by re-suspension and vortexing with 1 ml of 50 mM Tris-HCI, pH 7.85, containing 1 mM MgCI2 and 0.5% Triton X-100 and centrifugation at 1 ,000 x g for 10 min to isolate the nuclear fraction of the cells. The amount of [125I]T3 retained in the resulting pellet of washed nuclei was determined using a Packard gamma spectrometer. The results are presented in Figure 2 as a percent of radioactivity retained in washed nuclei from cells incubated with 0.1 nM [125I]T3 in the absence of unlabeled T3 or antagonist candidates. Each data point represents the average of duplicates, which generally varied by less than 5%.
EXAMPLE 2 [0070] Functional CAT assays were performed to compare the extent of inhibition of the T3 stimulation of CAT activity observed in the presence of the antagonist candidates. HeLa cells were innoculated at 50,000 cells per well in 24 well plates in DMEM containing 10% calf serum. The cells were transfected 5 hours later by calcium phosphate precipitation using 450ng of the T3 responsive ΔMTV-IR-CAT reporter and 250 ng of a vector expressing TRα. At the time of transfection, the cells also received 6nM T3 and the different concentrations of the antagonist candidates. Cells were harvested 40h after transfection and assayed for protein content and CAT activity. Results, shown in Figure 3, are expressed as the extent of inhibition of the T3 stimulation of CAT activity observed in the presence of the antagonist candidates. Each data point reflects the average of triplicate samples which showed less than 10% variation.
EXAMPLE 3 [0071] A study to compare the T3-mediated co-activator recruitment to TR by A1 ; A3, and Compound A was conducted in vitro. Approximately 2.5-5 x 104 cpm of 35S- labeled TRα (20 fmol) in 2 μl of lysate was incubated with 500 ng of GST fused to the receptor interaction region of the co-activator NRC (NRC15) immobilized on a glutathione-agarose beads. The samples were also incubated for 15 min at room temperature with of A-\ or A3 or 5 μM of Compound A in binding buffer. The samples were then chilled on ice and incubated with 1 nM T3 for an additional 60 min at 4°C. Control samples contained no T3 or antagonists, or received only T3. The beads were washed and the bound 35S-TRα electrphoresed in a 10% SDS gel followed by analysis and quantitation of that amount of 35S-TRα bound using a Molecular Dynamics Phosphorimager and ImageQuant software. The percent inhibition of T3- mediated binding of 35S-TRα to GST-NRC15 by Compounds A, A1 and A3 was determined after subtracting the amount of 35S-TRα bound to GST-NRC15 in the absence of T3. The results are shown in Figure 4 and show the inhibitory effect of the compounds as A3>Aι>A, confirming the increased efficacy of compounds, synthesized from antagonists candidates chosen according to the Invention, whose selectivity is optimized as described herein.
EXAMPLE 4 Part one: UV-vis detection of A3 in buffer
[0072] In order to detect the A3 concentration in mice serum after the above study, standard UV absorption-concentration correlation was studied. Standard A3 solution in buffer ranging from 100 μM to 1 nM with 10 time difference was prepared and their UV-vis absorption was measured. UV-vis can be used to detect 10 nM A3 solution, and the detection limit can be enhanced by fluorescence. A3 compound has Rf value of 0.3 when eluent is ethyl acetate in TLC study. A3 compound has water solubility approximately of 10 μM and ethyl acetate can be used to extract A3 out of aqueous phase. In addition, A3 compound shows non-linear optical property.
[0073] The following references contain material associated with the field of the invention disclosed herein; however, no determination has been made with regard to the relevance or lack thereof of any of said references; moreover, no assertion that any of said references is or is not relevant to the invention is intended. In the event that any of said references is actually found to contain material relevant to the present invention, such reference should be considered incorporated in its entirety into this specification.
1. DeGroot Ed. (1995), Saunders Endocrinology, 3rd ed., L.J.
2. Werner and Ingbar (1996) The Thyroid: a Fundamental and Clinical Test, Lippincott-Raven, 7th ed., L.E. Braverman and R.D. Utiger, eds.
3. Wilson J.D. Ed. (1998), Saunders Williams Textbook of Endocrinology
4. Yen, P.M. (2001 ) Physiol. Rev. 81 , 1097-1142.
5. Evans RM. (1988) The steroid and thyroid hormone receptor superfamily. Science 240:889-95
6. Carson-Jurica MA, Schrader WT, O'Malley BW. (1990) Steroid receptor family: structure and functions. Endocr Rev. 11 :201-20.
7. Chambon P. (1993) The molecular and genetic dissection of the retinoid signaling pathway. Gene.135:223-8.
8. Dees, E.C., Kennedy, M.J. (1998) Curr. Opin. Oncol. 10, 517-522.
9. Labrie F. (1993) Mechanism of action and pure antiandrogenic properties of flutamide. Cancer. 72:3816-27.
10. Olefsky JM, Saltiel AR (2000) PPAR gamma and the treatment of insulin resistance. Trends Endocrinol Metab. 11 (9):362-8.
11. Forrest D, Erway LC, Ng L, Altschuler R, Curran T. (1996) Thyroid hormone receptor beta is essential for development of auditory function. Nat Genet. 13:354-7.
12. Weiss RE, Forrest D, Pohlenz J, Cua K, Curran T, Refetoff S. (1997) Thyrotropin regulation by thyroid hormone in thyroid hormone receptor beta-deficient mice. Endocrinology. 138:3624-9.
13. Wikstrom L, Johansson C, Salto C, Barlow C, Campos Barros A, Baas F, Forrest D, Thoren P, Vennstrom B. (1998) Abnormal heart rate and body temperature in mice lacking thyroid hormone receptor alpha 1. EMBO J. 17:455-61.
14. Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T, Engstrom O, Ohman L, Greene GL, Gustafsson JA, Carlquist M. (1997) Molecular basis of agonism and antagonism in the oestrogen receptor. Nature. 389:753-8.
15. Moras D, Gronemeyer H. (1998) The nuclear receptor ligand-binding domain: structure and function. Curr Opin Cell Biol., 10:384-91.
16. Weatherman RV, Fletterick RJ, Scanlan TS. (1999) Nuclear-receptor ligands and ligand-binding domains. Annu Rev Biochem. 68:559-81. 17. Bourguet W, Germain P, Gronemeyer H. (2000) Nuclear receptor ligand- binding domains: three-dimensional structures, molecular interactions and pharmacological implications. Trends Pharmacol Sci. 21 :381 -8
18. Pike AC, Brzozowski AM, Walton J, Hubbard RE, Thorsell AG, Li YL, Gustafsson JA, Carlquist M (2001 ) Structural insights into the mode of action of a pure antiestrogen. Structure (Camb) 9:145-53.
19. Xu HE, Stanley TB, Montana VG, Lambert MH, Shearer BG, Cobb JE, McKee DD, Galardi CM, Plunket KD, Nolte RT, Parks DJ, Moore JT, Kliewer SA, Willson TM, Stimmel JB. (2002) Structural basis for antagonist-mediated recruitment of nuclear co-repressors by PPARalpha. Nature. 415:813-7.
20. Shiau, A. K., Barstad, D., Radek, J.T. Meyers, M.J. Nettles, K.W., Katzellenbogen, B.S., Katzellenbogen, J.A., Agard, D.A., Greene, G.L. (2002) Nat. Struc. Biol. 9, 359-364.
21. Shiau, A. K., Barstad, D., Loria, P. M., Cheng, L, Kushner, P. J., Agard, D. ' A., Greene, G. L. (1998) The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 95, 927-937.
22. Pike, A.C., Brzozowski, A.M., Hubbard, R.E., Bonn, T., Thorsell, A.G., Engstrom, O., Ljunggren, J., Gustafsson, J.A., Carlquist, M. (1999) EMBO J. 18, 4608-18.
23. Bourguet, W., Vivat, V., Wurtz, J.M., Chambon, P., Gronemeyer, H., Moras, D. (2000) Mol. Cell 5, 289-98.
24. Schapira M, Raaka BM, Samuels HH, Abagyan R. (2000) Rational discovery of novel nuclear hormone receptor antagonists Proc Natl Acad Sci U S A. 97(3):1008-13.
25. Baxter JD, Goede P, Apriletti JW, West BL, Feng W, Mellstrom K, Fletterick RJ, Wagner RL, Kushner PJ, Ribeiro RC, Webb P, Scanlan TS, Nilsson S. (2002), Structure-based design and synthesis of a thyroid hormone receptor (TR) antagonist. Endocrinology; 143(2):517-24.
26. Darimont, B.D., Wagner, R.L., Apriletti, J.W., Stallcup, M.R., Kushner, P.J., Baxter, J.D., Fletterick, R.J., Yamamoto, K.R. (1998) Genes Dev. 12, 3343-56.
27. Molsoft LLC, ICM 2.8 manual, freely available online at www.molsoft.com (Molsoft, San Diego, CA).
28. Abagyan, R., Totrov, M. (1994) J. Mol. Biol. 235, 983-1002.
29. Totrov, M., Abagyan, R. (1997) Proteins, Suppl. 1 , 215-20.
30. Totrov, M., Abagyan, R., (2001) in Drug-Receptor Thermodynamics: Introduction and Applications, ed., Raffa, R.B. (J. Wiley & Sons, Ltd.), pp. 603-624.
31. Abagyan, R., Totrov, M. (2001) Curr. Opin. Chem. Biol. 5, 375-82. 32. Raaka, B.M., Samuels, H.H. (1983) J. Biol. Chem. 258, 417-425.
33. Mahajan MA and Samuels HH. (2000). A new family of nuclear receptor coregulators that integrate nuclear receptor signaling through CREB-binding protein. Mol. Cell. Biol. 20; 5048-5063.
34. Li, D., Desai-Yajnik, V., Lo, E., Schapira, M., Abagyan, R., Samuels, H.H. (1999) Mol. Cell. Biol. 19, 7191-202.
35. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev.; 46(1 -3):3-26.
36. Bourguet W., Ruff M., Chambon P., Gronemeyer H., Moras D. (1995) Nature 375, 377-82.
37. Strynadka N.C., Eisenstein M., Katchalski-Katzir E., Shoichet B.K., Kuntz I.D., Abagyan R., Totrov M., Janin J., Cherfils J., Zimmerman F., Olson A., Duncan B., Rao M., Jackson R., Stemberg M., James M.N. (1996) Nat Struct Biol. 3, 233-9.
38. Abagyan R., Batalov S., Cardozo T., Totrov M., Webber J., Zhou Y. (1997) Proteins., Suppl 1 , 29-37.
39. Ajay A, Walters WP, Murcko MA. (1998) Can we learn to distinguish between "drug-like" and "nondrug-like" molecules? J Med Chem.; 41 (18):3314-24.
40. Barratt MD, Rodford RA. (2001 ) The computational prediction of toxicity. Curr Opin Chem Biol. 5(4):383-8.
41. Casanova J, Horowitz ZD, Copp RP, Mclntyre WR, Pascual A, Samuels HH.(1984) Photoaffinity labeling of thyroid hormone nuclear receptors. Influence of n- butyrate and analysis of the half-lives of the 57,000 and 47,000 molecular weight receptor forms J Biol Chem.; 259(19):12084-91
42. Egea PF, Mitschler A, Rochel N, Ruff M, Chambon P, Moras D. (2000) Crystal structure of the human RXRalpha ligand-binding domain bound to its natural ligand: 9-cis retinoic acid. EMBO J. Jun 1 ;19(11):2592-601.
43. Filikov AV, Mohan V, Vickers TA, Griffey RH, Cook PD, Abagyan RA, James TL. (2000) Identification of ligands for RNA targets via structure-based virtual screening: HIV-1 TAR J Comput Aided Mol Des. 14(6):593-610.
44. Greene N. (2002) Computer systems for the prediction of toxicity: an update. Adv Drug Deliv Rev.;54(3):417-31
45. Lazar MA (1993) Thyroid hormone receptors: multiple forms, multiple possibilities. Endocr Rev. 14 (2): 184-93
46. Macchia PE, Takeuchi Y, Kawai T, Cua K, Gauthier K, Chassande O, Seo H, Hayashi Y, Samarut J, Murata Y, Weiss RE, Refetoff S. (2001) Increased sensitivity to thyroid hormone in mice with complete deficiency of thyroid hormone receptor alpha. Proc Natl Acad Sci U S A; 98(1 ):349-54.
47. McGovern SL, Caselli E, Grigorieff N, Shoichet BK. (2002) A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screening. J Med Chem. 45(8):1712-22.
48. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH, Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Milburn MV.(1998) Ligand binding and co- activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature 395(6698):137-43.
49. Onate SA, Tsai SY, Tsai M-J, and O'Malley BW. (1995) Sequence and characterization of a coactivator of the steroid hormone receptor superfamily. Science 270; 1354-1357.
50. Oppenheimer JH, Koerner D, Schwartz HL, Surks Ml (1972) J. in. Endocrinol. Metab. 35:330-33
51. Oppenheimer, J.H. and H.H. Samuels. 1983. Molecular Basis of Thyroid Hormone Action. Academic Press, New York
52. Pohlenz J, Maqueem A, Cua K, Weiss RE, Van Sande J, Refetoff S. (1999) Improved radioimmunoassay for measurement of mouse thyrotropin in serum: strain differences in thyrotropin concentration and thyrotroph sensitivity to thyroid hormone. Thyroid.; 9(12):1265-71.
53. Powers CA, Mathur M, Raaka BM, Ron D, and Samuels HH. (1998) TLS (Translocated-in-Liposarcoma) is a high-affinity interactor for steroid, thyroid hormone, and retinoid receptors. Mol. Endocrinol. 12; 4-18.
54. Refetoff S. (1997) Resistance to thyroid hormone. Curr Ther Endocrinol Metab.; 6:132-4.
55. Sadowski J, Kubinyi H. (1998) A scoring scheme for discriminating between drugs and nondrugs. J Med Chem. 41 (18):3325-9.
56. Samuels HH, Tsai JS. (1973) Thyroid hormone action in cell culture: demonstration of nuclear receptors in intact cells and isolated nuclei. Proc Natl Acad Sci U S A. 70:3488-92
57. Schapira M, Raaka BM, Samuels HH, Abagyan R. (2001 ) In silico discovery of novel Retinoic Acid Receptor agonist structures. BMC Struct Biol. 2001 ;1 (1 ):1.
58. Steinmetz AC, Renaud JP, Moras D. (2001) Binding of ligands and activation of transcription by nuclear receptors. Annu Rev Biophys Biomol Struct. 30:329-59.
59. Takeuchi Y, Murata Y, Sadow P, Hayashi Y, Seo H, Xu J, O'Malley BW, Weiss RE, and Refetoff S. (2002) Steroid receptor coactivator-1 deficiency causes variable alterations in the modulation of T(3)-regulated transcription of genes in vivo. Endocrinology 143; 1346-52.
60. Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW, Kopple KD. (2002) Molecular properties that influence the oral bioavailability of drug candidates J. Med. Chem., 45 (12), 2615 -2623.
61. Viswanadhan VN, Balan C, Hulme C, Cheetham JC, Sun Y. (2002) Knowledge-based approaches in the design and selection of compound libraries for drug discovery. Curr Opin Drug Discov Devel.; 5(3):400-6.
62. Wagner, R. L., Darimont, B. D., Apriletti, J. W., Stallcup, M. R., Kushner, P. J., Baxter, J. D., Fletterick, R. J., Yamamoto, K. R.(1998) Structure and Specificity of Nuclear Receptor: Coactivator Interactions Genes Dev. 12, 3343.
63. Walters WP, Ajay, Murcko MA. (1999) Recognizing molecules with drug-like properties. Curr Opin Chem Biol. Aug;3(4):384-7.
64. Weiss RE, Refetoff S. (2000) Resistance to thyroid hormone. Rev Endocr Metab Disord. 1 (1 -2):97-108
65. Weiss RE, Chassande O, Koo EK, Macchia PE, Cua K, Samarut J, Refetoff S, Refetoff S. (2002) Thyroid function and effect of aging in combined hetero/homozygous mice deficient in thyroid hormone receptors alpha and beta genes. J Endocrinol.; 172(1): 177-85.
66. Weiss RE, Xu J, Ning G, Pohlenz J, O'Malley BW, and Refetoff S. (1999). Mice deficient in the steroid receptor co-activator 1 (SRC-1) are resistant to thyroid hormone. Embo J.18; 1900-4.
67. Taylor, E. C; Skotnicki, J. S. Synthesis. (1981), 8, 606.
68. Hu, Y.; Zorumski, C. F.; Covey, D. F. J. Org. Chem. (1995), 60, 3619.
69. Wang G.; Hollingsworth, R. I. Tetrahedron: Asymmetry. (1999), 10, 1895.
70. Plenat F.; Cassagne, M.; Cristau, H. J. Tetrahedron. (1995), 35, 9551
[0074] From the foregoing description, various modifications and changes in the compositions and methods of this invention will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein.
[0075] All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.

Claims

WHAT IS CLAIMED IS:
1. A compound capable of antagonizing the effects of thyroid hormone having the structure
Figure imgf000034_0001
wherein:
Rn is CH(CH3)2, CH2 CH3, CH3, or H ; R2 is CF3, CH3, F, or H; R3 is F, CH3, OCH3, CF3, or H; and
Figure imgf000034_0002
provided that when R2 s CF3, R-, and R3 are H; when R-i s CH(CH3)2, R2 and R3 are H and R4 is either CH3 or H; when R-i s CH2CH3, R2, R3, and R4 are H; when R2 s CH3, R and R3 are H and R4 is OCH3; when R2 s F, R3 is also F and Ri and R4 are H; when R3 s F, R2 is also F and R^ and R4 are H; when R-i s CH3, R3 and R4 are also CH3 and R2 is H; when R3 s OCH3, RL R2, and R4 are H; when R3 s CF3, R R2, and R4 are H.
2. A compound according to claim 1 wherein R-\ is CH(CH3)2 and R2, R3, and R4 are H.
3. A compound according to claim 1 wherein Ri is CH2CH3 and R2, R3, and R4 are H.
4. A compound according to claim 1 wherein Ri is CH(CH3)2, R is CH3, and R2 and R3 are H.
5. A compound according to claim 1 wherein R2 is CH3, R is OCH3, and R^ and R3 are Hi
6. A compound according to claim 1 wherein R2 is F, R3 is F, and Ri and R4 are H.
7. A compound according to claim 1 wherein Ri, R3, and R4 are CH3, and R2 is H.
8. A compound according to claim 1 wherein R1 ( R3, and R are CH3, and R2 is H.
9. A compound according to claim 1 wherein R3 is OCH3, and R-i, R2, and R4are H.
10. A compound according to claim 1 wherein R3 is CF3, and R1 ; R2, and R4are H.
11. A compound capable of antagonizing the effects of thyroid hormone having the structure
Figure imgf000035_0001
wherein
R1 = F, R2 = Cl, R3 = H, R4=H, R5=NO2, R6=H
R1 = F, R2 = Cl, R3 = H, R4=H, R5=H, R6=OCH3
R1 = F, R2 = OCH3, R3 = OCH3, R4=H, R5=NO2, R6=H
R1 = F, R2 = Cl, R3 = H, R4=NO2, R5=H, R6=H
R1 = CH3, R2 = F, R3 = H, R4=H, R5= OCH3, R6=H
12. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound according either of claims 1 or 11 and a pharmaceutically acceptable carrier.
13. A pharmaceutical composition comprising a pharmaceutically effective amount of a compiund having the structure
Figure imgf000036_0001
wherein
Ri is CH(CH3)2, CH2 CH3, CH3, or H ; R2 is CF3, CH3, F, or H; R3 is F, CH3, OCH3, CF3, or H; and R4 is CH3, OCH3, or H,
and a pharmaceutically acceptable carrier.
14. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound having the structure
Figure imgf000036_0002
wherein:
R1 is F;
R2 may be Cl, OCH3 or F;
R3 may be H or OCH3;
R4 may be H or NO3;
R5 may be H, OCH3 or NO2; and
R6 may be H or OCH3. and a pharmaceutically acceptable carrier.
15. A pharmaceutical composition according to any of claims 12, 13, or 14, wherein the composition is effective as an antagonist of thyroid hormone for modulation of a condition in a human characterized by overproduction of thyroid hormone to a degree detrimental to health.
16. A method of controlling thyroid hormone receptor activity comprising the in vivo administration of a compound according to either of claims 1 or 1 1.
17. A method of controlling thyroid hormone receptor activity comprising the in vivo administration of a therapeutically effective amount of a composition according to any of claims 12, 13, or 14.
18. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising administering a therapeutically effective amount of a composition according to any of claims 12, 13, or 14.
19. The method of any of claims 16-18 wherein the condition is selected from the group consisting of hyperthyroidism and thyrotoxicosis.
20. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising administering a pharmaceutically effective amount of a compound according to either of claims 1 or 11.
21. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising i) administering a pharmaceutically effective amount of a compound according to either of claims 1 or 11 ; and ii) administering a treatment selected from the group consisting of administering a pharmaceutical agent capable of inhibiting the synthesis of thyroid hormone, destruction of thyroid tissue by radioactive iodine, and surgical removal of thyroid tissue.
22. The method of either of claims 20 or 21 , wherein the condition is selected from the group consisting of hyperthyroidism and thyrotoxicosis.
23. The method of claim 22 wherein the condition is selected from the group consisting of hyperthyroidism and thyrotoxicosis.
24. A method for preparing compounds useful for the treatment of a human having a condition characterized by overproduction of thyroid hormone, said method comprising the steps of: i) selecting a compound selected from the group consisting of
Name Structure
Figure imgf000038_0001
Figure imgf000039_0001
ii) generating a virtual library of derivatives of the compound chosen in step i); iii) screening said library in silico; iv) chemically synthesizing at least one compound screened in iii); and v) testing in vitro at least one compound synthesized in iv).
25. The method of claim 24 further comprising the additional step of testing in vivo at least one compound chemically synthesized in iv).
26. A pharmaceutical composition comprising a pharmaceutically effective amount of one or more compounds according to either of claims 1 or 11 and a pharmaceutically acceptable carrier.
27. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising administering a pharmaceutically effective amount of at least one compound selected from the group consisting of:
Figure imgf000039_0002
Figure imgf000040_0001
28. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising administering a pharmaceutically effective amount of a pharmaceutical composition containing, as an active ingredient, a pharmaceutically effective amount of at least one compound selected from the group consisting of:
Figure imgf000040_0002
Figure imgf000041_0001
29. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising administering a pharmaceutically effective amount of at least one compound according to claim 27.
30. A method of treating a human for a condition characterized by overproduction of thyroid hormone comprising: i) administering a pharmaceutically effective amount of one or more compounds according to either of claims 1 or 11 ; and ii) administering a treatment selected from the group consisting of administering a pharmaceutical agent capable of inhibiting the synthesis of thyroid hormone, destruction of thyroid tissue by radioactive iodine, and surgical removal of thyroid tissue.
31. A method of controlling thyroid hormone receptor activity comprising the in vivo administration of one or more compounds according to claim 1.
32. A method of controlling thyroid hormone receptor activity comprising the in vivo administration of one or more compounds according to claim 11.
33. Use of a compound according to either of claims 1 or 11 for the preparation of a composition for the treatment of aberrant thyroid activity in a mammal.
34. Use of a composition according to any of claims 12-14 for the treatment of aberrant thyroid activity in a mammal.
35. Use according to either of claims 33 or 34 wherein the mammal is a human.
36. Use according to any of claims 33-35 wherein said aberrant thyroid activity comprises overproduction of thyroid hormone.
37. Use according to claim 36 wherein said overproduction of thyroid hormone is selected from the group consisting of hyperthyroidism and thyrotoxicosis.
PCT/US2004/015048 2003-05-14 2004-05-14 Driverse thyroid hormone receptor antagonists and uses thereof WO2004103289A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47074903P 2003-05-14 2003-05-14
US60/470,749 2003-05-14

Publications (2)

Publication Number Publication Date
WO2004103289A2 true WO2004103289A2 (en) 2004-12-02
WO2004103289A3 WO2004103289A3 (en) 2005-10-20

Family

ID=33476746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/015048 WO2004103289A2 (en) 2003-05-14 2004-05-14 Driverse thyroid hormone receptor antagonists and uses thereof

Country Status (1)

Country Link
WO (1) WO2004103289A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010122980A1 (en) * 2009-04-20 2010-10-28 田辺三菱製薬株式会社 Novel thyroid hormone β receptor agonist
US7829552B2 (en) 2003-11-19 2010-11-09 Metabasis Therapeutics, Inc. Phosphorus-containing thyromimetics
JP2012106996A (en) * 2010-10-19 2012-06-07 Mitsubishi Tanabe Pharma Corp NOVEL THYROID HORMONE β-RECEPTOR AGONIST
US10130643B2 (en) 2005-05-26 2018-11-20 Metabasis Therapeutics, Inc. Thyromimetics for the treatment of fatty liver diseases
US11202789B2 (en) 2016-11-21 2021-12-21 Viking Therapeutics, Inc. Method of treating glycogen storage disease
US11707472B2 (en) 2017-06-05 2023-07-25 Viking Therapeutics, Inc. Compositions for the treatment of fibrosis
US11787828B2 (en) 2018-03-22 2023-10-17 Viking Therapeutics, Inc. Crystalline forms and methods of producing crystalline forms of a compound

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE CAPLUS [Online] 2003 SCHAPIRA ET AL: 'Discovery of diverse thyroid hormone receptor antagonists by high docking.' Database accession no. (139:477528) & PNAS vol. 100, no. 12, 2003, pages 7354 - 7359 *
DATABASE CAPLUS [Online] August 1996 IIDA ET AL: 'Reversible thermal recording material containing color disappearance promoter.' Database accession no. (125:234450) *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7829552B2 (en) 2003-11-19 2010-11-09 Metabasis Therapeutics, Inc. Phosphorus-containing thyromimetics
US10130643B2 (en) 2005-05-26 2018-11-20 Metabasis Therapeutics, Inc. Thyromimetics for the treatment of fatty liver diseases
US10925885B2 (en) 2005-05-26 2021-02-23 Metabasis Therapeutics, Inc. Thyromimetics for the treatment of fatty liver diseases
WO2010122980A1 (en) * 2009-04-20 2010-10-28 田辺三菱製薬株式会社 Novel thyroid hormone β receptor agonist
JP5487202B2 (en) * 2009-04-20 2014-05-07 田辺三菱製薬株式会社 Novel thyroid hormone beta receptor agonist
US8791266B2 (en) 2009-04-20 2014-07-29 Mitsubishi Tanabe Pharma Corporation Thyroid hormone β receptor agonist
JP2012106996A (en) * 2010-10-19 2012-06-07 Mitsubishi Tanabe Pharma Corp NOVEL THYROID HORMONE β-RECEPTOR AGONIST
US11202789B2 (en) 2016-11-21 2021-12-21 Viking Therapeutics, Inc. Method of treating glycogen storage disease
US11707472B2 (en) 2017-06-05 2023-07-25 Viking Therapeutics, Inc. Compositions for the treatment of fibrosis
US11787828B2 (en) 2018-03-22 2023-10-17 Viking Therapeutics, Inc. Crystalline forms and methods of producing crystalline forms of a compound

Also Published As

Publication number Publication date
WO2004103289A3 (en) 2005-10-20

Similar Documents

Publication Publication Date Title
US6140354A (en) N-substituted aminotetralins as ligands for the neuropeptide Y Y5 receptor useful in the treatment of obesity and other disorders
DE60015927T2 (en) PHENYLENE AND PHENYLTHIUM DERIVATIVES
EP1723105B1 (en) Bicyclic substituted indole-derivative steroid hormone nuclear receptor modulators
US20060293292A1 (en) METHODS OF USING ACYL HYDRAZONES AS sEH INHIBITORS
JP2002536445A (en) Phenylurea and phenylthiourea derivatives as orexin receptor antagonists
CA2904985A1 (en) Pyrazole-amide compound and medicinal uses therefor
EP2997015B1 (en) Phenoxyethyl dihydro-1h-isoquinoline compounds
TW200424198A (en) Imiidazopyridine compounds as 5-HT4 receptor agonists
US10106523B2 (en) Amide compound
Scanio et al. Discovery and biological evaluation of potent, selective, orally bioavailable, pyrazine-based blockers of the Nav1. 8 sodium channel with efficacy in a model of neuropathic pain
TW201443004A (en) Phenoxyethoxy compounds
CA2957898A1 (en) Pyrrolopyrimidine derivatives as nr2b nmda receptor antagonists
WO2004103289A2 (en) Driverse thyroid hormone receptor antagonists and uses thereof
WO2020257261A1 (en) SUBSTITUTED HETEROCYCLES AS c-MYC TARGETING AGENTS
JP2006520780A (en) 6-sulfonamide derivatives of quinoline and chromene as androgen receptor antagonists
US10017521B2 (en) Chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors
EP3402780A1 (en) Mast-cell modulators and uses thereof
US8394811B2 (en) Hydrazide compounds as thyroid hormone receptor modulators and uses thereof
JP2022511287A (en) Substituted heterocyclic compound as a c-MYC targeting agent
JP2001525398A (en) Selective β3 adrenergic agonist
WO2019088057A1 (en) Anilide derivative and medicinal use thereof
US5658923A (en) Azepine derivatives and use thereof
US20070088016A1 (en) Tricyclic steroid hormone nuclear receptor modulators
WO2023138599A1 (en) Aromatic fused ring nav1.8 inhibitor, and use thereof
Rowbottom et al. Synthesis and structure–activity relationships of retro bis-aminopyrrolidine urea (rAPU) derived small-molecule antagonists of the melanin-concentrating hormone receptor-1 (MCH-R1). Part 1

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase