AU2002229033A1 - 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone - Google Patents

7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone

Info

Publication number
AU2002229033A1
AU2002229033A1 AU2002229033A AU2002229033A AU2002229033A1 AU 2002229033 A1 AU2002229033 A1 AU 2002229033A1 AU 2002229033 A AU2002229033 A AU 2002229033A AU 2002229033 A AU2002229033 A AU 2002229033A AU 2002229033 A1 AU2002229033 A1 AU 2002229033A1
Authority
AU
Australia
Prior art keywords
group
alkyl
aryl
compound
aralkyl
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
AU2002229033A
Other versions
AU2002229033B2 (en
Inventor
James C. Lanter
Mark Macielag
Zhihua Sui
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceuticals Inc
Original Assignee
Ortho McNeil Pharmaceutical Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/992,785 external-priority patent/US6583153B2/en
Application filed by Ortho McNeil Pharmaceutical Inc filed Critical Ortho McNeil Pharmaceutical Inc
Publication of AU2002229033A1 publication Critical patent/AU2002229033A1/en
Application granted granted Critical
Publication of AU2002229033B2 publication Critical patent/AU2002229033B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

7-HETEROCYCLYL QUINOLINE AND THIENO[2,3-b]PYRIDINE DERIVATIVES USEFUL AS ANTAGONISTS OF GONADOTROPIN
RELEASING HORMONE
FIELD OF THE INVENTION
The present invention is directed to novel 7-heterocyclyl quinoline and thieno[2,3-/.]pyridine derivatives, pharmaceutical compositions containing them and their use in the treatment of disorders and conditions associated with gonadotropin releasing hormone (GnRH). The compounds of the invention are antagonists of GnRH, useful in the treatment of infertility, prostate cancer, benign prostate hyperplasia (BPH), and useful as contraceptives.
BACKGROUND OF THE INVENTION
Gonadotropin-releasing hormone (GnRH), also referred to as luteinizing hormone-releasing hormone (LHRH) is a linear decapeptide amide, pGlu-His- Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2, originally isolated from porcine (Matsuo, H., et. al., Biochem. Biophys. Res. Commun. 1972, 43, 1334-1339) and ovine
(Burgus, R., et. al., PNAS, USA, 1972, 69, 278-282) sources. GnRH plays a key role in the reproductive system. The hormone is released from the hypothalamus and acts on the pituitary gland to stimulate the biosynthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). LH released from the pituitary gland is primarily responsible for the regulation of gonadal steroid production in both males and females, whereas FSH regulates spermatogenesis in males and follicular development in females.
GnRH-based therapies using peptidic GnRH agonists and antagonists have been shown effective in the treatment of conditions associated with
LH/FSH release, such as endometriosis, uterine fibroids, polycystic ovarian disease, precocious puberty and some gonadal steroid-dependent neoplasia, particularly prostate cancer, breast cancer and ovarian cancer. GnRH agonists and antagonists are also useful in the treatment of fertility and as a contraceptive in both males and females.
Although the compounds of the present invention are useful primarily for the treatment of disorders and conditions associated with the reproductive system, they may also be useful for the treatment of other GnRH mediated disorders and conditions including pituitary gonadotrope adenomas, sleep disorders, benign prostate hyperplasia, and prostate cancer.
Peptide-like GnRH antagonists are known, for example, derivatives of straight-chain peptides (US Patent 5140009 and 517835), cyclic hexapeptide derivatives (Japanese Patent Application Laid-open No. 61 (1986)-191698), and bicyclic peptide derivatives (J. Med. Chem. 1993, 36, 3265). However, due to a lack of bioavailability, these compounds are limited to intravenous and subcutaneous administration.
Recently, small molecule, non-peptide GnRH antagonists have been disclosed. Kato, et al., in EP0679642 disclose isochroman derivatives which have gonadotropin releasing hormone receptor antagonizing activity, as well as calcium-antagonizing and monoamine-uptake inhibiting activities.
Ohkawa et al., in WO96/38438 disclose tricyclic diazepine derivatives which have gonadotropin releasing hormone receptor antagonist activity. Ohkawa et al., in WO95/29900 disclose condensed heterocyclic compounds which have GnRH receptor antagonistic action and / or an action of improving sleep disturbances.
Furuya et al., in WO97/14682 disclose quinolone derivatives as GnRH antagonists, useful as prophylactic or therapeutic agents for the prevention or treatment of sex hormone dependent disease. Goulet et al., in WO97/44037 and in WO97/44041 , Goulet et al., in W097/44321 and Goulet et al., in W097/44339 disclose non-peptide antagonists of GnRH useful for the treatment of a variety of sex-hormone related conditions in men and women. Goulet et al., in WO97/21703 and in W097/21707 disclose non-peptide antagonists of GnRH useful for the treatment of a variety of sex-hormone related conditions in men and women.
Furuya et al., in WO95/28405 disclose bicyclic thiophene derivatives with gonadotropin releasing hormone receptor antagonizing activity. Furuya et al., in W097/41126 disclose 4,7-dihydro-4-oxothieno[2,3-b]pyridine derivatives having GnRH antagonistic activity. Furuya, et al., in WO97/14697 disclose thieno[2,3-b]pyridine derivatives as GnRH antagonists.
SUMMARY OF THE INVENTION
The present invention is directed to a compound of formula (I) or (II):
wherein
L1 is selected from the group consisting of CH2, CH(CH3) and C(CH3)2;
R1 and R2are independently selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the aryl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, -C(O)-alkyl, -C(0)-aryl or -C(0)-cycloalkyl;
X is selected from the group consisting of O, S and NRA; where RA is selected from hydrogen, alkyl, aryl or aralkyl; R4 is selected from the group consisting of -C(0)-RB, -C(O)O-RB, -C(0)NH2, -C(0)-NHRB, -C(O)-N(RB)2, and -C(0)NHNH2; wherein RB is selected from the group consisting of alkyl, aryl, aralkyl and cycloalkyl; alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc; wherein each Rc is independently selected from the group consisting of oxo, alkyl, alkoxy, amino, alkylamino, dialkylamino, aryl, -O-aryl, aralkyl and -
O-aralkyl;
L2 is selected from the group consisting of alkyl;
R3 is selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido;
R5 is selected from the group consisting of halogen, cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido; provided that when X is O, then R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
In an aspect of the present invention is the compound 4,7-dihydro-2-(4- methoxyphenyl)-7-[(2-methoxyphenyl)methyl]-3-
[[methyl(phenylmethyl)amino]methyl]-4-oxo-thieno[2,3-i ]pyridine-5-carboxylic acid hydrazide, and pharmaceutically acceptable salts, esters and prodrugs thereof.
Illustrative of the invention is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and any of the compounds described above. An illustration of the invention is a pharmaceutical composition made by mixing any of the compounds described above and a pharmaceutically acceptable carrier. Illustrating the invention is a process for making a pharmaceutical composition comprising mixing any of the compounds described above and a pharmaceutically acceptable carrier.
Exemplifying the invention are methods of treating disorders or diseases which respond to antagonism of GnRH, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above.
An example of the invention is a method for treating infertility, prostate cancer or benign prostate hyperplasia (BPH), in a subject in need thereof comprising administering to the subject an effective amount of any of the compounds or pharmaceutical compositions described above.
A further example of the invention is a method of female or male contraception, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above.
Yet another example of the invention is the use of any of the compounds described herein in the preparation of a medicament for treating: (a) infertility, (b) prostate cancer, (c) benign prostate hyperplasia (BPH) or for (d) contraception, in a subject in need thereof. DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to a compound of formula (I) or (II):
wherein L1, R1, R2, X, R4, L2, R3 and R5 are as previously described, useful in the treatment of disorders or diseases which respond to antagonism of the GnRH such as infertility, prostate cancer, benign prostate hyperplasia (BPH), and the like. The compounds of the present invention are further useful as contraceptives.
In one embodiment of the present invention are compounds of the formula (I) wherein
L is CH2;
R1 and R2 are independently selected from the group consisting of lower alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the aryl, aralkyl, heteroaryl or heterocycloalkyl may be optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl) or N(lower alkyl)2;
X is O;
R4 is selected from the group consisting of -C(0)0-alkyl, -C(O)0-aryl and -C(0)NHNH2; alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, pyrazolinyl, dihydropyridyl and dihydropyrimidyl, wherein the ring structure is optionally substituted with one to two substituents independently selected from oxo, lower alkyl, lower alkoxy, aryl, -O-aryl, aralkyl or -O-aralkyl; L2 is selected from the group consisting of lower alkyl;
R3 is selected from the group consisting of aryl; wherein the aryl group is optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl), N(Iower alkyl)2, cyano or sulfonamido;
R5 is selected from the group consisting of halogen, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocycloalkyl wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl is optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl), N(lower alkyl)2, cyano or sulfonamido; provided that when X is O, then R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocyloalkyl group is optionally substituted with one to two substituents selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl), N(lower alkyl)2, cyano or sulfonamido; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
In another embodiment of the present invention are compounds of the formula (I) or (II) wherein X is S and R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido;
In a further embodiment of the present invention are compounds of the formula (I) or (II) wherein X is NRA or alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc, wherein Rc is as defined above.
In a particularly preferred embodiment of the present invention are compounds of the formula (I) and (II) as listed in Tables 1 and 2. TABLE
TABLE
As used herein, "halogen" shall mean chlorine, bromine, fluorine and iodine.
As used herein, the term "alkyl" whether used alone or as part of a substituent group, includes straight and branched chains comprising one to ten carbon atoms. For example, alkyl radicals include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, t-butyl, pentyl and the like. Unless otherwise noted, "lower" when used with alkyl means a carbon chain composition of 1-6 carbon atoms.
As used herein, unless otherwise noted, "alkoxy" shall denote an oxygen ether radical of the above described straight or branched chain alkyl groups. For example, methoxy, ethoxy, n-propoxy, sec-butoxy, t-butoxy, n-hexyloxy and the like. Unless otherwise noted, "lower" when used with alkoxy means an oxygen ether radical of the above described straight or branched carbon chain alkyl group wherein the alkyl is of 1-6 carbon atoms. As used herein, unless otherwise noted, "aryl" shall refer to carbocyclic aromatic groups such as phenyl, naphthyl, and the like.
As used herein, unless otherwise noted, "aralkyl" shall mean any lower alkyl group substituted with an aryl group such as phenyl, naphthyl and the like.
For example, benzyl, phenylethyl, phenylpropyl, naphthyl methyl, and the like.
As used herein, unless otherwise noted, "cycloalkyl" shall mean any three to eight membered, monocyclic, saturated, carbocyclic ring structure including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cylcooctyl.
As used herein, unless otherwise noted, "heteroaryl" shall denote any five or six membered monocyclic aromatic ring structure containing at least one heteroatom selected from the group consisting of O, N and S, optionally containing one to three additional heteroatoms independently selected from the group consisting of O, N and S; or a nine or ten membered bicyclic aromatic ring structure containing at least one heteroatom selected from the group consisting of O, N and S, optionally containing one to four additional heteroatoms independently selected from the group consisting of O, N and S. The heteroaryl group may be attached at any heteroatom or carbon atom of the ring such that the result is a stable structure.
Examples of suitable heteroaryl groups include, but are not limited to, pyrrolyl, furyl, thienyl, oxazolyl, imidazolyl, purazolyl, isoxazolyl, isothiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyranyl, furazanyl, indolizinyl, indolyl, isoindolinyl, indazolyl, benzofuryl, benzothienyl, benzimidazolyl, benzthiazolyl, purinyl, quinolizinyl, quinolinyl, isoquinolinyl, isothiazolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, and the like.
As used herein, the term "heterocycloalkyl" shall denote any five to seven membered monocyclic, saturated, partially unsaturated or partially aromatic ring structure containing at least one heteroatom selected from the group consisting of O, N and S, optionally containing one to three additional heteroatoms independently selected from the group consisting of O, N and S; or a nine to ten membered saturated, partially unsaturated or partially aromatic bicyclic ring system containing at least one heteroatom selected from the group consisting of O, N and S, optionally containing one to four additional heteroatoms independently selected from the group consisting of O, N and S. The heterocycloalkyl group may be attached at any heteroatom or carbon atom of the ring such that the result is a stable structure.
Examples of suitable heterocycloalkyl groups include, but are not limited to, pyrrolinyl, pyrrolidinyl, dioxalanyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, dioxanyl, morpholinyl, dithianyl, thiornorpholinyl, piperazinyl, trithianyl, indolinyl, chromenyl, 3,4-methyIenedioxyphenyl and 2,3- dihydrobenzofuryl and the like.
When a particular group is "substituted" (e.g., cycloalkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl), that group may have one or more substituents, preferably from one to five substituents, more preferably from one to three substituents, most preferably from one to two substituents, independently selected from the list of substituents.
With reference to substituents, the term "independently" means that when more than one of such substituents is possible, such substituents may be the same or different from each other.
Under standard nomenclature used throughout this disclosure, the terminal portion of the designated side chain is described first, followed by the adjacent functionality toward the point of attachment. Thus, for example, a "phenylalkylaminocarbonylalkyl" substituent refers to a group of the formula
Names for chemical entities of the present invention may be generated using nomenclature rules known in the art or may alternatively be generated using commercial chemical naming software, for example ACD/lndex Name
(Advanced Chemistry Development, Inc., Toronto, Ontario)
For use in medicine, the salts of the compounds of this invention refer to non-toxic "pharmaceutically acceptable salts." Other salts may, however, be useful in the preparation of compounds according to this invention or of their pharmaceutically acceptable salts. Suitable pharmaceutically acceptable salts of the compounds include acid addition salts which may, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, e.g., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts. Thus, representative pharmaceutically acceptable salts include the following: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, Iactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, oleate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide and valerate.
The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention. Furthermore, some of the crystalline forms for the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e., hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention.
The term "subject" as used herein, refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
The term "therapeutically effective amount" as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.
Abbreviations used in the specification, particularly the Schemes and
Examples, are as follows: MEM = Minimum Essential Medium
DCM = Dichloromethane
DIPEA = Diisopropylethylamine
DMF = N,N-Dimethylformamide
DME = Dimethoxyethane
DMSO = Dimethylsulfoxide
Et3N = Triethylamine
EtOAc = Ethyl Acetate
LHMDS = Lithium hexamethyldisilaz.de
MeOH = Methanol
NBS = 1 -bromo-2,5-pyrrolidinedione
Ph = Phenyl
RT or rt = Room temperature
TEA = Triethylamine
THF = Tetrahydrofuran
Compounds of the general formula (I) wherein X is O, may be prepared according to the process outlined in Scheme 1.
Scheme 1
More particularly, a compound of formula (III), a known compound or compound prepared by known methods, wherein Q is bromo or iodo, is reacted with a suitably substituted compound of formula (IV), in the presence of a base such as potassium carbonate, TEA, NaOH, NaH, DIPEA, and the like, in an organic solvent such as THF, DMF, DCM, and the like, to yield the corresponding compound of formula (V).
The compound of formula (V) is reacted with a brominating agent such as 70% NBS, and the like, in an organic solvent such as THF, DMF, DCM, and the like, to yield the corresponding compound of formula (VI).
The compound of formula (VI) is reacted with a suitably substituted amine of formula (VII), in the presence of a base such as TEA, DIPEA, and the like, in an organic solvent such as THF, DMF, and the like, to yield the corresponding compound of formula (VIII).
The compound of formula (VIII) is reacted with a suitably substituted boronic acid of formula (IX), in the presence of a catalyst such as palladium tetrakis(triphenylphosphine) (Pd(PPh3)4), and the like, in the presence of a base such as NaC03, NaOH, and the like, in an organic solvent such as THF, DMF, dioxane, and the like, optionally in a mixture with water, to yield the corresponding compound of formula (la).
Compounds of formula (I) wherein X is S may be prepared according to the process outlined in Scheme 2.
Scheme 2
Specifically, a suitably substituted compound of formula (la), prepared as in Scheme 1 , is reacted with a sulfonating agent such as P4Sιo, Lawesson's reagent, and the like, in an organic solvent such as pyridine, toluene, xylene, and the like, at an elevated temperature in the range of about 60-140°C, to yield the corresponding compound of formula (lb).
Compounds of formula (I) wherein X is N and is taken together with R4 to
form , wherein RD is selected from the group consisting of alkyl, aryl and aralkyl, may be prepared according to the process outlined in Scheme 3.
Scheme 3
More particularly, a suitably substituted compound of formula (lb), prepared as in Scheme 2, is reacted with a compound of formula (X), in an organic solvent such as DMF, DMSO, and the like, at an elevated temperature in the range of about 80-110°C, to yield the corresponding compound of formula (Ic).
The compound of formula (Ic) is optionally further reacted with a compound of formula (XI), wherein RD is selected from the group consisting of alkyl, aryl and aralkyl, in the presence of a strong base such as LHMDS, NaH, potassium t-butoxide, and the like, to yield the corresponding compound of formula (Id).
Alternatively, the compound of formula (Ic) may be further reacted according to known methods to introduce one or more substituents on the pyrazolyl group. Compounds of formula (I) wherein X is NRA may be prepared according to the process outlined in Scheme 4.
Scheme 4
More particularly, a suitably substituted compound of formula (lb), prepared as in Scheme 2, is reacted with a suitably substituted amine of formula (XII), in an organic solvent such as DMF, DMSO, and the like, preferably at an elevated temperature in the range of about 60-120°C, to yield the corresponding compound of formula (le).
Compounds of formula (I) wherein X is N and is taken together with R4 to form a ring structure selected from the group consisting of dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl may be prepared by methods known to those skilled in the art.
For example, compounds of formula (I) wherein X is N and is taken
together with R4 to form dihydropyrazolyl may be prepared by reacting a suitably substituted compound of formula (la), wherein X is O and R4 is an ester of the formula -C02RB, prepared as in Scheme 1 , with a suitable reducing agent, to yield the corresponding compound wherein R4 is an aldehyde of the formula -C(0)H, subsequently reacting the R4 aldehyde with benzylcarbazate to yield the corresponding compound wherein R4 is hydrazone (-CHNNH-C(O)O-benzyl), reducing the R4 hydrazone with hydrogen gas, in the presence of a catalyst such as palladium hydroxide, to yield the corresponding compound wherein R4 is hydrazine (-CH2NHNH2), and then treating the R4 hydrazine with a dehydration reagent such as P2O5, at an elevated temperature to yield the corresponding dihydropyrazole substituted compound of formula (I).
Compounds of formula (I) wherein X is N and is taken together with R4 to
form isoxazolinyl ( ) may be prepared by subjecting a suitably substituted compound of formula (la), wherein X is O and R4 is an ester of the formula -C02RB, prepared as in Scheme 1 , to saponification to convert the R4 ester group to the corresponding carboxy group (-CO2H) and then treating the R4 carboxy group with a suitable reducing agent, to yield the corresponding compound wherein R4 is an alcohol of the formula -CH2OH, converting the X is O carbonyl group to the corresponding X is S thiocarbonyl group, converting the thiocarbonyl group to the corresponding hydroxyamine where X is N-OH, and then affecting ring closure of the X hydroxyamine and R4 alcohol with a dehydrating agent such as P2θ5, at an elevated temperature to yield the corresponding isoxazolinyl substituted compound of formula (I).
Compounds of formula (I) wherein X is N and is taken together with R4 to
form dihydropyrimidinyl ( *• ) or oxo-substituted dihydropyrimidinyl
may be prepared.by reacting a suitably substituted compound of formula (la), wherein R4 is an ester of the formula -C02RB, prepared as in Scheme 1 , with urea, thiourea, guanidine or a suitably subtituted alkyl or aryl amidine, in an organic solvent, at an elevated temperature to yield the corresponding oxo-substituted dihydropyrimidinyl
dihydropyrimidinyl imino-substituted dihydropyrimidinyl
) or alkyl or aryl substituted dihydropyrimidinyl substituted compound of formula (I). The oxo-substituted dihydropyrimidinyl substituted compound of formula (I) may be further optionally, alkylated according to the process described in Scheme 3, to yield the corresponding dihydropyrimidinyl substituted compound of formula (I). Similarly, the thio- or imino-substituted dihydropyrimidinyl substituted compound of formula (I) may be further, optionally reacted according to known methods to displace the thio group (=S) or modify the imino group (=NH), respectively.
Compounds of formula (II), wherein R4 is -C(0)0-R6, X is S or X is N
H
and is taken together with R4 to form " 7 , may be prepared according to the process outlined in Scheme 5.
(lib)
Scheme 5
More particularly, a compound of formula (X), a known compound or compound prepared by known methods, is reacted with a sulfonating agent such as P4Sιo, Lawesson's Reagent, and the like, in the presence of an organic base such as pyridine, TEA, Hϋnig's base (DIEA), and the like, at an elevated temperature in the range of about 60-120°C, preferably at about reflux temperature, to yield the corresponding compound of formula (I la).
The compound of formula (lla) may be optionally further reacted with H2N-NH2, in an organic solvent such as DMF, DMSO, and the like, at an elevated temperature in the range of about 80-110°C, to yield the corresponding compound of formula (lib).
The compound of formula (lib) may be further, optionally reacted according to known methods to introduce one or more substituents on the pyrazolyl group. Compounds of formula (II) wherein X is N and taken together with R4 to form a ring selected from the group consisting of dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl, may be similarly prepared according to the processes described above, with appropriate substitution of a compound of formula (X) for the compound of formula (la).
Compounds of formula (II) wherein X is selected from the group consisting of O, S and NRA, may be prepared from the corresponding compound of formula (X), by methods known to those skilled in the art.
For example, compounds of formula (II) wherein X is O and R4 is other than -C(0)OR6 may be prepared by converting the R4 ester group on a suitably substituted compound of formula (X) to the corresponding R4 carboxy group and then using known quinolone chemistry to convert the R4 carboxy group to the desired R4 functionality.
Compounds of formula (II) wherein X is S and R4 is other than -C(0)OR6, may similarly be prepared by converting the -C(0)OR6 ester on the compound of formula (X) to the desired R4 group as described above and then converting the X is O, (carbonyl group) to the corresponding X is S
(thiocarbonyl group) by reacting with a sulfonating agent, as described in Scheme 5.
Compounds of formula (II) wherein X is NRA and R4 is other than -C(0)OR6, may similarly be prepared by converting the -C(0)OR6 ester on the compound of formula (X) to the desired R4 group as described above and then converting the X is O (carbonyl group) to the corresponding X is NRA (amine group) by reacting with a suitably substituted amine, as described in Scheme 4.
The following Examples are set forth to aid in the understanding of the invention, and are not intended and should not be construed to limit in any way the invention set forth in the claims which follow thereafter. EXAMPLE 1 7-Bromo-6-bromomethyl-1-(2,6-difluorobenzyl)-1 ,4-dihydro-4-oxoquinoline-3- carboxylic acid ethylester
A mixture of 7-bromo-1-(2,6-difluorobenzyI)-1 ,4-dihydro-6-methyl-4- oxoquinoline-3-carboxylic acid ethyl ester (3.5 g, 8 mmol), prepared according to the procedure outlined in PCT application W097/14682, Reference Example 3, NBS (1.5 g, 8.4 mmol) and 2,2'-azobisisobutyronitrile (AIBN, 100 mg) in DCM (200 mL) was stirred at reflux for 4 h. Additional NBS (750 mg) was added and the mixture was refluxed for 4 h. Column chromatography (hexanes:ethyl acetate = 3:7) yielded the product as a white solid. Yield: 2.95 g (72%) m.p. 184-187°C; 1H NMR (CDCI3), δ 1.41 (t, J = 8 Hz, 3 H), 4.40 (q, J = 8 Hz, 2 H), 4.66
(s, 2 H), 5.36 (s, 2H), 7.03 (m, 2 H), 7.39 (m, 1 H), 7.92 (s, 1 H), 8.54 (s, 1 H), 8.68 (ds, 1 H);
MS (m/z): 514 (MH+).
EXAMPLE 2 6-(N-Benzyl-N-methylaminomethyl)-7-bromo-1-(2,6-difluorobenzyl)-1 ,4-dihydro- 4-oxoquinoline-3-carboxylic acid ethylester A mixture of 7-bromo-6-bromomethyl-1-(2,6-difluorobenzyl)-1 ,4-dihydro- 4-oxoquinoline-3-carboxylic acid ethyl ester (110 mg, 0.21 mmol), methylbenzylamine (31 mg, 0.26 mmol) in DIPEA (0.045 mL) and DMF (15 mL) was stirred at room temperature for 16 h. Ethyl acetate and water were added. The organic phase was separated and washed with water, dried with MgS04. The solvent was evaporated and the residue dried under vacuum to yield the product as a white solid.
Yield: 120 mg (100%) 1H NMR (CDCI3), δ 1.41 (t, J = 8 Hz, 3 H), 2.17 (s, 3 H), 3.62 (s, 2 H), 3.67 (s, 2 H), 4.40 (q, J = 8 Hz, 2 H), 5.36 (s, 2H), 7.03 (m, 2 H), 7.25-7.39 (m, 6 H), 7.88 (s, 1 H), 8.59 (s, 1 H), 8.67 (ds, 1 H);
MS (m/z): 555 (MH+).
EXAMPLE 3 6-(N-Benzyl-N-methylaminomethyl)-7-(benzofuran-2-yl)-1-(2,6-difluorobenzyl)- 1 ,4-dihydro-4-oxoquinoline-3-carboxylic acid ethylester Compound #2
A mixture of 7-bromo-6-bromomethyl-1-(2,6-difluorobenzyl)-1 ,4-dihydro- 4-oxoquinoline-3-carboxylic acid ethyl ester (278 mg, 0.5 mmol), benzofuran-2- boronic acid (97 mg, 0.6 mmol), tetrakis(triphenylphosphine)palladium(0) (69 mg, 0.06 mmol) and 2M sodium carbonate (414 mg, 3 mmol) in DME (20 mL) was heated at reflux for 16 h. Ethyl acetate and water were added. The organic phase was separated and washed with water and dried with MgS0 . Column chromatography (ethyl acetate) yielded the product as a yellow solid. Yield: 55 mg (19%)
1H NMR (CDCI3), δ 1.44 (t, J = 8 Hz, 3 H), 2.17 (s, 3 H), 3.63 (s, 2 H), 3.90 (s, 2 H), 4.42 (q, J = 8 Hz, 2 H), 5.49 (s, 2H), 7.00 (m, 2 H), 7.22-7.81 (m,
11 H), 8.23 (s, 1 H), 8.62 (s, 1 H), 8.76 (ds, 1 H); MS (m/z): 593 (MH+).
EXAMPLE 4
2,5-dihydro-7-(4-methoxyphenyl)-5-[(2-methoxyphenyI)methyl]-8- [[methyl(phenylmethyl)amino]methyl]- 3H-pyrazolo[3,4-c lthieno[2,3-ib]pyridin-3- one Compound #8 To a solution of 4,7-dihydro-2-(4-methoxy-phenyI)-7-[(2- methoxyphenyl)methyl]-3-[[methyl(phenylmethyl)amino]methyl]-4-oxo- thieno[2,3-b]pyridine-5-carboxylic acid, ethyl ester (prepared according to the procedure described by Furuya, S, et al., in PCT Application WO95/28405), (387 mg, 0.66 mmol) in pyridine (3 mL) was added P Sιo. The reaction flask was flushed with argon and stirred at reflux for 5 hours. The solvent was removed and the product purified by flash chromatography to yield the corresponding thiocarbonyl.
The thiocarbonyl (118 mg, 0.19 mmol) was dissolved in dry DMF (0.25 mL) and treated with hydrazine monohydrate (0.02 mL, 0.42 mmol). The resulting solution was heated to 80°C for 2 hours, the mixture was cooled and purified by flash chromatography (0-10% MeOH/CHCI3). The product was converted to its hydrochloride salt by treating with HCI to yield the product as a yellow powder.
Yield: 7.5 mg
MS (m/z) 551 (MH+).
EXAMPLE 5
2,5-Dihydro-7-(benzofuran-2-yl)-8-[[methyl(phenylmethyl)amino]methyl]-5-[(2,6- difluorophenyl)methyl]-3H-pyrazolo[4,3-c]quinolin-3-one
Compound #4 To a solution of 7-(benzofuran-2-yl)-8-[[methyl(phenylmethyl)-amino]- methyl]-5-[(2,6-difluorophenyl)methyl]-3-quinolinecarboxylic acid ethyl ester, (500 mg, 0.84 mmol) in pyridine (5 mL) was added P4S-|0 (240 mg, 0.65 eq). The reaction flask was flushed with argon and stirred at reflux for 2 hours, cooled to 100°C and poured into water (100 mL). The product was extracted into chloroform, dried (MgS0 ) and concentrated to yield a red-brown solid.
The solid (337 mg, 0.55 mmol) was dissolved in dry DMF (5 mL) and treated with hydrazine monohydrate (60 mg, 1.1 mmol). The resulting mixture was warmed to 100°C for 3 hours, the mixture was cooled and poured onto water. The resulting yellow precipitate was collected by filtration and dried to yield the product.
Yield: 149 mg 1H NMR (CDCI3) δ 2.09 (s, 3H), 3.57 (s, 2H), 3.86 (s, 2H), 5.84 (s, 2H), 7.18-7.49 (m, 12H), 7.66-7.77 (m, 2H), 8.16 (s, 1 H), 8.33 (s, 1 H), 8.95 (s, 1 H).
EXAMPLE 6
7-(Benzofuran-2-yl)-3-[(phenylmethyl)oxy]-5-[(2,6-difluorophenyl) methyl]-8- [[methyl(phenylmethyl)amino]methyl]-5H-pyrazolo[4,3-c]quinoIine
Compound #5 A solution of 2,5-dihydro-7-(benzofuran-2-yl)-8- [[methyl(phenylmethyl)amino]methyl]-5-[(2,6-difluorophenyl)methyl]-3H- pyrazolo[4,3-c]quinolin-3-one (120 mg, 0.21 mmol) in dry DMF (5 mL) was treated with a solution of lithium hexamethyldisilazide (0.25 mL, 0.25 mol, 1.0 M) in tetrahydrofuran (THF). Benzyl bromide (40 mg, 0.22 mmol) was introduced via syringe and the mixture was stirred overnight. One equivalent hydrochloric acid in ether was added and the solvent evaporated to yield the corresponding hydrochloride salt product as a yellow solid. Yield: 47 mg
1H NMR (CDCI3) δ 2.08 (s, 3H), 3.55 (s, 2H), 3.87 (s, 2H), 5.13 (s, 2H), 5.88 (s, 2H), 7.12-7.51 (m, 16 H), 7.66-7.70 (m, 2H), 8.16 (s, 1 H), 8.37 (s, 1 H), 9.10 (s, 1 H).
EXAMPLE 7 6-(N-Benzyl-N-methylaminomethyl)-7-(thien-3-yl)-1-(2,6-difluorobenzyl)-1 ,4- dihydro-4-oxoquinoline-3-carboxylic acid ethyl ester
Compound #1 Following the procedure described in Example 3, the compound prepared in Example 2 (6-(N-Benzyl-N-methylaminomethyl)-7-bromo-1-(2,6- difluorobenzyl)-1 ,4-dihydro-4-oxoquinoline-3-carboxylic acid ethylester) (150 mg, 0.27 mmol) was reacted with thiophene-3-boronic acid (38.4 mg, 0.30 mmol), to yield the product as a yellow solid. Yield: 48 mg MS(m/z) 559 (MH+).
EXAMPLE 8 2,5-Dihydro-7-bromo-8-[[methyl(phenylmethyl)amino]methyl]-5-[(2,6- difluorophenyl)methyl]-3H-pyrazolo[4,3-c]quinolin-3-one
Compound #3 Following the procedure described in Example 4, 6-(N-benzyl-N- methylaminomethyl)-7-bromo-1-(2,6-difluorobenzyI)-1 ,4-dihydro-4- oxoquinoline-3-carboxylic acid ethyl ester (0.6g, 1.1 mmol) was converted to the title compound and isolated as the corresponding hydrochloride salt, as a yellow powder.
Yield: 0.15 g MS (m/z) 524 (MH+)
EXAMPLE 9 7-(Benzofuran-2-yl)-3-ethoxy-5-[(2,6-difluorophenyl)methyl]-8 [[methyI(phenylmethyl)amino]methyl]-5H-pyrazolo[4,3-c]quinoline HCI Compound #6
Following the procedure described in Example 6. 2,5-dihydro-7- (benzofuran-2-yl)-8-[[methyl(phenylmethyl)amino]methyl]-5-[(2,6- difluorophenyl)methyl]-3H-pyrazolo[4,3-c]quinolin-3-one (0.05 g, 0.09 mmol) was reacted with ethyl iodide (0.018 g, 0.116 mmol) to yield the title compound, which was isolated as its corresponding hydrochloride salt, as a yellow powder.
Yield: 0.05 g MS (m/z) 589 (MH+). EXAMPLE 10
4,7-Dihydro-2-(4-methoxyphenyl)-7-[(2-methoxyphenyl)methyl]-3-
[[methyl(phenylmethyl)amino]methyl]-4-oxo-thieno[2,3-b]pyridine-5-carboxylic acid, hydrazide bis-hydrochloride Compound #7
To a solution of 4,7-dihydro-2-(4-methoxyphenyl)-7-[(2- methoxyphenyl)methyl]-3-[[methyl(phenylmethyl)amino]methyl]-4-oxo- thieno[2,3-b]pyridine-5-carboxylic acid, ethyl ester (57 mg, 0.097 mmol) in ethyl alcohol (2 mL) was added hydrazine (0.030 mL, 0.096 mmol). The mixture was heated to reflux for 6 hours and concentrated in vacuo. Trituration of the residue with diethyl ether and collection of the solid by precipitation yielded the product as a pale yellow solid. Yield: 38 mg MS (m/z) 569 (MH+).
EXAMPLE 11 GnRH Receptor Binding Assay A homogenate prepared from an equal mixture of female and male rat pituitaries was used as the source of the membrane-bound GnRH receptor.
The receptor was allowed to interact in solution with [125l]-histrelin alone or in combination with a competitive ligand (the compound being tested). The bound radiolabeled ligand was separated from the free (unbound) radiolabeled ligand by filtration through glass filter mats using a 96-well plate harvesting system (Tomtec Mach II 96). In the absence of a competitive ligand, a maximum amount of radiolabeled ligand is bound to the receptor and trapped by the glass filter mats. When an unlabelled ligand that can compete for the receptor site is present, the amount of radiolabeled ligand bound to the receptor and trapped on the filter mat is proportionally reduced depending on the concentration of the competitor and on the strength of the competitor's affinity for the receptor. The amount of receptor-bound [125l]-histrelin on the filter mat was determined using a Wallac Betaplate™ Liquid Scintillation Counter. Binding was determined as follows:
NSB Non-specific binding
Bo maximum concentration of compound
Average NSB: (NSB1 + NSB2)/2
Average B0: (B01 + B02)/2
Corrected B0: Average B0 - Average NSB
% inhibition of Corrected B0 (or maximum response) was calculated as follows:
% Inhibition = 100-[((Actual counts per minute - Average NSB)
Corrected B0)*100]
EXAMPLE 12
LUCIFERASE Assay FOR GnRH Hek 293 cells with the GnRHR gene were transfected with the hCG promoter and the luciferase reporter system. On day 1 , the cells were plated at a density of 80,000 cells per well on a Poly-D-lysine pre-coated 96 well plate. The plates were incubated at 37°C for 24 hours. On day 2, the spent media was decanted and replaced with fresh media. Test compounds, standard and controls were added to individual wells. All the dilutions were done in 7.5% DMSO/αMEM media. The assay was run in both agonist and antagonist format. For the antagonist format, the assay measurements were run against a standard of 0.6nM Histrelin. On day 3, the levels of luciferase production were measured in a chemiluminescence assay using Enhanced Luciferase Assay Kit. The results were expressed as % Inhibition using the following formula:
RLU Relative Light Units, a measure of chemiluminescence Agonist [RLU value(test compound) -(Background/0.6nM
Histrelin) - Background]*! 00 Antagonist (1-[(RLU value - (Background/0.6nM Histrelin) - Background)^ 00])
The calculated percentages were plotted on a graph using Graph Pad Prizm and the IC50/EC50 values determined.
EXAMPLE 13 Primary Pituitary Cell Culture Assay Male rats (between immature and adult) were sacrificed and the anterior pituitaries were collected from them. The pituitaries were dissociated and the cells were plated at a concentration of 0.33 X 106 cells/well on day 1. On day 3 the media on the cells was flushed and replaced with fresh media. The test compound was then added to the plated cells at a concentration ranging from 1 nM to 1000 nM. The plates were incubated at 37°C at 5% C02 for 2 days. On Day 5 the media was flushed again and replaced with fresh media. To the plates were then added test compound and 1 nM GnRH. The cells were incubated for 4 hours, the media was collected by centrifuging the plates at 1200 rpm Son/all RT7 for 10 minutes, 900μL of supernatant was pipetted from each well and dispensed to a 96 well plate. The deep well plates were covered and stored at 20°C for a day. The plates are then evaluated by ELISA (a radioimmunoassay system) to determine the concentration of lutenizing hormone in the media. The assay was repeated at varying concentrations of the test compounds to determine IC50 values. The IC50 value is defined as the concentration of test compound at which 50% inhibition was achieved.
Following the procedures set forth above, selected compounds of the present invention were tested, with results as listed in Table 3.
Table 2 - Biological Activity
1D # Binding, IC50 Luciferase Assay Pituitary Cell (B%@30μM) IC50 (μM) Assay IC50 (μM)
1 32 μM 10 (antagonist)
While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, it will be understood that the practice of the invention encompasses all of the usual variations, adaptations and/or modifications as come within the scope of the following claims and their equivalents.

Claims (31)

We Claim:
1. A compound of the formula (I)
wherein
L1 is selected from the group consisting of CH2, CH(CH3) and C(CH3)2;
R1 and R2are independently selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the aryl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, -C(0)-alkyI, -C(0)-aryl or -C(0)-cycloalkyl;
X is selected from the group consisting of O, S and NRA; where RA is selected from hydrogen, alkyl, aryl or aralkyl;
R4 is selected from the group consisting of -C(0)-RB, -C(0)0-RB, -C(0)NH2, -C(0)-NHRB, -C(0)-N(RB)2, and -C(0)NHNH2; wherein RB is selected from the group consisting of alkyl, aryl, aralkyl and cycloalkyl; alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc; wherein each Rc is independently selected from the group consisting of oxo, alkyl, alkoxy, amino, alkylamino, dialkylamino, aryl, -O-aryl, aralkyl and - O-aralkyl;
L2 is selected from the group consisting of alkyl;
R3 is selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido;
R5 is selected from the group consisting of halogen, cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido; provided that when X is O, then R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
2. A compound as in Claim 1 wherein L1 is CH2;
R1 and R2 are independently selected from the group consisting of lower alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the aryl, aralkyl, heteroaryl or heterocycloalkyl may be optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl) or N(lower alkyl)2;
X is O;
R4 is selected from the group consisting of -C(0)0-alkyl, -C(0)0-aryl and -C(0)NHNH2; alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, pyrazolinyl, dihydropyridyl and dihydropyrimidyl, wherein the ring structure is optionally substituted with one to two substituents independently selected from oxo, lower alkyl, lower alkoxy, aryl, -O-aryl, aralkyl or -O-aralkyl;
L2 is selected from the group consisting of lower alkyl; R3 is selected from the group consisting of aryl; wherein the aryl group is optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl), N(lower alkyl)2, cyano or sulfonamido;
R5 is selected from the group consisting of halogen, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocycloalkyl wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl is optionally substituted with one to two substituents independently selected from halogen, lower alkyl, lower alkoxy, nitro, NH , NH(lower alkyl), N(lower alkyl)2, cyano or sulfonamido; provided that when X is O, then R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocyloalkyl group is optionally substituted with one to two substituents selected from halogen, lower alkyl, lower alkoxy, nitro, NH2, NH(lower alkyl), N(lower alkyl)2, cyano or sulfonamido; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
3. A compound as in Claim 1 wherein X is NRA or X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc; wherein each Rc is independently selected from selected the group consisting of oxo, alkyl, alkoxy, amino, alkylamino, dialkylamino, aryl, -O-aryl, aralkyl and -O-aralkyl; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
4. A compound as in Claim 2 wherein L is CH2; R1 is selected from the group consisting of lower alkyl;
R2 is selected from the group consisting of aralkyl; X is O;
R4 is selected from the group consisting of -C(0)0-(lower alkyl); alternatively, X is taken together with R4 to form a group selected from
O — (alkyl)
L2 is selected from the group consisting of lower alkyl; R3 is selected from the group consisting of phenyl and substituted phenyl, wherein the substituents on the phenyl are one to two independently selected from halogen;
R5 is selected from the group consisting of halogen and heteroaryl; provided that when X is O, R5 is heteroaryl; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
5. A compound as in Claim 4 wherein L1 is CH2; R1 is methyl;
R2 is benzyl; X is O;
R4 is -C(0)0-CH2CH3; alternatively, X is taken together with R4 to form a group selected from
L2 is CH2;
R3 is 2,6-di-fluorophenyl;
R5 is selected from the group consisting of bromo, 3-theinyl and 2- benzofuryl; provided that when X is O, R5 is selected from 3-theinyl and 2- benzofuryl; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
6. A compound as in Claim 5 selected from the group consisting of 1 -[(2,6-difluorophenyl)methyl]-1 ,4-dihydro-6- [[methyl(phenylmethyl)amino]methyl]-4-oxo-7-(3-thienyl)- 3-quinolinecarboxylic acid ethyl ester;
7-(2-benzofuranyl)-1-[(2,6-difluorophenyl)methyl]-1 ,4-dihydro-6- [[methyl(phenylmethyl)amino]methyl]-4-oxo-3-quinolinecarboxyIic acid, ethyl ester; 7-bromo-5-[(2,6-difluorophenyl)methyl]-2,5-dihydro-8- [[methyl(phenylmethyl)amino]methyl]- 3H-pyrazolo[4,3-c]quinolin-3-one;
7-(2-benzofuranyI)-5-[(2,6-difluorophenyl)methyl]-2,5-dihydro-8- [[methyl(phenylmethyl)amino]methyl]- 3 -/-pyrazolo[4,3-c]quinolin-3-one;
7-(benzofuran-2-yl)-5-[(2,6-difluorophenyI)methyl]-3-[(phenylmethyl)oxy]- -8-[[methyl(phenylmethyl)amino]methyl]-5H-pyrazolo[4,3-c]quinoline
7-(benzofuran-2-yl) -5-[(2,6-difluorophenyl)methyl]-3-ethoxy-8- [[methyl(phenylmethyl)amino]methyl]-5H-pyrazolo[4,3-c]quinoline; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
7. A compound of the formula (II)
wherein
L1 is selected from the group consisting of CH2, CH(CHs) and C(CH3)2;
R1 and R2are independently selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the aryl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, -C(0)-alkyl, -C( )-aryI or -C(0)-cycloalkyl;
X is selected from the group consisting of O, S and NRA; where RA is selected from hydrogen, alkyl, aryl or aralkyl;
R4 is selected from the group consisting of -C(0)-RB, -C(0)O-RB, -C(0)NH2, -C(0)-NHRB, -C(0)-N(RB)2, and -C(0)NHNH2; wherein RB is selected from the group consisting of alkyl, aryl, aralkyl and cycloalkyl; alternatively X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc; wherein each Rc is independently selected from the group consisting of oxo, alkyl, alkoxy, amino, alkylamino, dialkylamino, aryl, -O-aryl, aralkyl and - O-aralkyl;
L2 is selected from the group consisting of alkyl;
R3 is selected from the group consisting of alkyl, cycloalkyl, aryl, aralkyl, heteroaryl and heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents independently selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl),
N(alkyl)2, cyano or sulfonamido;
R5 is selected from the group consisting of halogen, cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl; wherein the cycloalkyl, aryl, aralkyl, heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl),
N(alkyl)2, cyano or sulfonamido; provided that when X is O, then R5 is selected from the group consisting of heteroaryl or heterocycloalkyl; wherein the heteroaryl or heterocycloalkyl group is optionally substituted with one or more substituents selected from halogen, alkyl, alkoxy, nitro, NH2, NH(alkyl), N(alkyl)2, cyano or sulfonamido; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
8. A compound as in Claim 7 wherein X is NRA or X is N and is taken together with R4 to form a ring structure selected from the group consisting of pyrazolyl, dihydropyrazolyl, isoxazolinyl and dihydropyrimidinyl; wherein the ring structure is optionally substituted with one or more Rc; wherein each Rc is independently selected from selected the group consisting of oxo, alkyl, alkoxy, amino, alkylamino, dialkylamino, aryl, -O-aryl, aralkyl and -O-aralkyl; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
9. A compound as in Claim 7 identified as 2,5-dihydro-7-(4- methoxyphenyl)-5-[(2-methoxyphenyl)methyI]-8- [[methyl(phenylmethyl)amino]methyl]- 3 -/-pyrazolo[3,4-c]thieno[2,3-j ]pyridin-3- one; and pharmaceutically acceptable salts, esters and pro-drugs thereof.
10. A compound identified as 4,7-dihydro-2-(4-methoxyphenyl)-7-[(2- methoxyphenyl)methyl]-3-[[methyl(phenylmethyl)amino]methyl]-4-oxo- thieno[2,3-jb]pyridine-5-carboxylic acid hydrazide, and pharmaceutically acceptable salts, esters and prodrugs thereof.
11. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Claim 1.
12. A pharmaceutical composition made by mixing a compound of Claim 1 and a pharmaceutically acceptable carrier.
13. A process for making a pharmaceutical composition comprising mixing a compound of Claim 1 and a pharmaceutically acceptable carrier.
14. A method of treating a disorder or disease which respond to antagonism of the GnRH in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the composition of Claim 11.
15. The method of Claim 14, wherein the disorder or disease is selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH).
16. A method of contraception in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 1.
17. A method of treating a condition selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH) in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 1.
18. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Claim 7.
19. A pharmaceutical composition made by mixing a compound of Claim 7 and a pharmaceutically acceptable carrier.
20. A process for making a pharmaceutical composition comprising mixing a compound of Claim 7 and a pharmaceutically acceptable carrier.
21. A method of treating a disorder or disease which respond to antagonism of the GnRH in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the composition of Claim 18.
22. The method of Claim 21 , wherein the disorder or disease is selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH).
23. A method of contraception in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 7.
24. A method of treating a condition selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH) in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 7.
25. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Claim 10.
26. A pharmaceutical composition made by mixing a compound of Claim 10 and a pharmaceutically acceptable carrier.
27. A process for making a pharmaceutical composition comprising mixing a compound of Claim 10 and a pharmaceutically acceptable carrier.
28. A method of treating a disorder or disease which respond to antagonism of the GnRH in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the composition of Claim 25.
29. The method of Claim 28, wherein the disorder or disease is selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH).
30. A method of contraception in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 10.
31. A method of treating a condition selected from the group consisting of the infertility, prostate cancer and benign prostate hyperplasia (BPH) in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of Claim 10.
AU2002229033A 2000-12-12 2001-12-11 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone Expired AU2002229033B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US25482700P 2000-12-12 2000-12-12
US60/254,827 2000-12-12
US09/992,785 US6583153B2 (en) 2000-12-12 2001-11-14 7-heterocyclyl quinoline and thieno[2,3-b]yridine derivatives useful as antagonists of gonadotropin releasing hormone
US09/992,785 2001-11-14
PCT/US2001/048119 WO2002048112A2 (en) 2000-12-12 2001-12-11 7-heterocyclyl quinoline and thieno[2,3-b]pyridine derivatives useful as antagonists of gonadotropin releasing hormone

Publications (2)

Publication Number Publication Date
AU2002229033A1 true AU2002229033A1 (en) 2002-08-29
AU2002229033B2 AU2002229033B2 (en) 2007-09-13

Family

ID=26944257

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2002229033A Expired AU2002229033B2 (en) 2000-12-12 2001-12-11 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone
AU2903302A Pending AU2903302A (en) 2000-12-12 2001-12-11 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2903302A Pending AU2903302A (en) 2000-12-12 2001-12-11 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone

Country Status (14)

Country Link
US (2) US6583153B2 (en)
EP (2) EP1345907B1 (en)
JP (1) JP4427251B2 (en)
CN (1) CN1288137C (en)
AT (2) ATE450510T1 (en)
AU (2) AU2002229033B2 (en)
CA (1) CA2431617C (en)
CY (2) CY1105712T1 (en)
DE (2) DE60140700D1 (en)
DK (2) DK1345907T3 (en)
ES (2) ES2337898T3 (en)
HK (1) HK1055957A1 (en)
PT (2) PT1520855E (en)
WO (1) WO2002048112A2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6455734B1 (en) 2000-08-09 2002-09-24 Magnesium Diagnostics, Inc. Antagonists of the magnesium binding defect as therapeutic agents and methods for treatment of abnormal physiological states
TW200413351A (en) * 2002-08-21 2004-08-01 Astrazeneca Ab Chemical compounds
WO2004035541A1 (en) * 2002-10-15 2004-04-29 Janssen Pharmaceutica, N.V. Benzyl substituted (piperidin-4-yl) aminobenzamido derivatives as delta-opiod receptor modulators
DE602006015851D1 (en) * 2005-12-05 2010-09-09 Merck Sharp & Dohme POSITIVE ALLOSTERIC CHINOLON M1 RECEPTOR MODULATORS
EP2490692B1 (en) * 2009-10-21 2016-11-16 Merck Sharp & Dohme Corp. Quinolinone-pyrazolone m1 receptor positive allosteric modulators
CN116239498B (en) * 2023-05-11 2023-07-21 北京元延医药科技股份有限公司 Method for preparing clenbuterol intermediate

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4659691A (en) 1985-02-08 1987-04-21 Merck & Co., Inc. Novel cyclic Hexapeptide LHRH antagonists
US5140009A (en) 1988-02-10 1992-08-18 Tap Pharmaceuticals, Inc. Octapeptide LHRH antagonists
US5171835A (en) 1988-10-21 1992-12-15 The Administrators Of The Tulane Educational Fund LHRH antagonists
TW449600B (en) 1994-04-19 2001-08-11 Takeda Chemical Industries Ltd Condensed-ring thiophene derivatives, their production and use
US5607939A (en) 1994-04-28 1997-03-04 Takeda Chemical Industries, Ltd. Condensed heterocyclic compounds, their production and use
JPH10500402A (en) 1994-04-29 1998-01-13 武田薬品工業株式会社 Fused heterocyclic compound or salt thereof, production method and use thereof
CN1072219C (en) 1995-06-01 2001-10-03 武田药品工业株式会社 Tricyclic diazepines, its production and use
CA2229205A1 (en) 1995-10-19 1997-04-24 Shuichi Furuya Thienopyridine derivatives, their productions and use
ATE223902T1 (en) * 1995-10-19 2002-09-15 Takeda Chemical Industries Ltd QUINOLINE DERIVATIVES AS GNRH ANTAGONISTS
WO1997021703A1 (en) 1995-12-14 1997-06-19 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
EE9800180A (en) 1995-12-14 1998-12-15 Merck & Co., Inc. Gonadotropin-releasing hormone antagonist, its use, pharmaceutical composition and method of preparation
EP0900222A1 (en) 1996-04-26 1999-03-10 Takeda Chemical Industries, Ltd. Thienopyridine derivatives, their production and use
CA2250908C (en) 1996-04-30 2012-03-13 Takeda Chemical Industries, Ltd. Combined use of gnrh agonist and antagonist
JP2000511533A (en) 1996-05-20 2000-09-05 メルク エンド カンパニー インコーポレーテッド Gonadotropin-releasing hormone antagonist
AU710926B2 (en) 1996-05-20 1999-09-30 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
AU3071397A (en) 1996-05-20 1997-12-09 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
AU712019B2 (en) 1996-05-20 1999-10-28 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
GB9610813D0 (en) 1996-05-23 1996-07-31 Pharmacia Spa Combinatorial solid phase synthesis of a library of benzufuran derivatives

Similar Documents

Publication Publication Date Title
JP4931314B2 (en) Gonadotropin releasing hormone receptor antagonist and related methods
US20040127492A1 (en) Cyclic pyrazoles for the inhibition of mitogen activated protein kinase-activated protein kinase-2
JPH0717637B2 (en) Angiotensin II antagonists incorporating substituted thiophenes or furans
SK285703B6 (en) 2-Aryl-8-oxodihydropurine derivatives, process for producing the same, medicinal compositions containing the same, and intermediates thereof
AU2007264791A1 (en) Derivatives of ureas of piperidine or pyrrolidine, their preparation and their therapeutical use
US5891882A (en) Benzopyranopyrrole and benzopyranopyridine α-1 adenergic compounds
CA2254768A1 (en) Antagonists of gonadotropin releasing hormone
CA2431617C (en) 7-heterocyclyl quinoline and thieno[2,3-b]pyridine derivatives useful as antagonists of gonadotropin releasing hormone
WO2004056780A2 (en) Tetrahydroquinoline derivatives
JP2002544278A (en) Imidazo and pyrrolo [1,2-a] pyrimid-4-ones as gonadotropin releasing hormone receptor antagonists
AU2002229033A1 (en) 7-heterocyclyl quinoline and thieno(2,3-b)pyridine derivatives useful as antagonists of gonadotropin releasing hormone
JP2005500352A (en) 1,2,4-triazine-3,5-diones as gonadotropin releasing hormone receptor (GnRH) antagonists
NZ250679A (en) 2-phenyl-9h-imidazo[1,2-a] benzimidazole-3-acetamide derivatives; their preparation and pharmaceutical compositions
US8431564B2 (en) Ring-annulated dihydropyrrolo[2,1-α]isoquinolines
EP2459560B1 (en) Ring-annulated dihydropyrrolo[2,l-a]isoquinolines
EP1412363A1 (en) Gonadotropin-releasing hormone receptor antagonists
CN115232144B (en) Nitrogen-containing condensed ring derivative, pharmaceutical composition, and preparation method and application thereof
ZA200504775B (en) Tetrahydroquinoline derivatives
CN115260211B (en) Thiophene condensed ring derivative, pharmaceutical composition, preparation method and application thereof
PL174330B1 (en) Novel derivatives of 3-oxadiazolyl-5,6,7,8-tetrahydro-1,6-naphtyridine, novel derivatives of 5,6,7,8-tetrahydro-1,6-naphtyridine and method of obtaining 1,6-naphtyridine derivatives
CA2358895A1 (en) Pyridobenzodiazepine and pyridobenzoxazepine carboxyamide vasopressin agonists
WO2015007606A1 (en) Spiroindoline derivatives and pharmaceutical compositions thereof
JPH0948777A (en) Diazepinone compounds, their production and therapeutic agent
MXPA01007918A (en) Pyridobenzodiazepine and pyridobenzoxazepine carboxyamide vasopressin agonists