AU719102B2 - Fibrinogen receptor antagonist prodrugs - Google Patents

Fibrinogen receptor antagonist prodrugs Download PDF

Info

Publication number
AU719102B2
AU719102B2 AU35037/97A AU3503797A AU719102B2 AU 719102 B2 AU719102 B2 AU 719102B2 AU 35037/97 A AU35037/97 A AU 35037/97A AU 3503797 A AU3503797 A AU 3503797A AU 719102 B2 AU719102 B2 AU 719102B2
Authority
AU
Australia
Prior art keywords
compound according
inhibiting
mammal
fibrinogen
composition according
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.)
Ceased
Application number
AU35037/97A
Other versions
AU3503797A (en
Inventor
Melissa S. Egbertson
George D. Hartman
Laura A. Libby
Donald E Slaughter
Steven D. Young
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.)
Merck and Co Inc
Original Assignee
Merck and Co 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 GBGB9617899.1A external-priority patent/GB9617899D0/en
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of AU3503797A publication Critical patent/AU3503797A/en
Application granted granted Critical
Publication of AU719102B2 publication Critical patent/AU719102B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • 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
    • C07D213/73Unsubstituted amino or imino radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • C07D209/46Iso-indoles; Hydrogenated iso-indoles with an oxygen atom in position 1
    • 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
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/34Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • C07D211/72Heterocyclic 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 hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D211/74Oxygen atoms
    • C07D211/76Oxygen atoms attached in position 2 or 6
    • 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
    • C07D295/14Heterocyclic 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 carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/155Heterocyclic 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 carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Description

WO 98/00401 PCT/US97/11047 -1- TITLE OF THE INVENTION FIBRINOGEN RECEPTOR ANTAGONIST PRODRUGS BACKGROUND OF THE INVENTION The invention relates generally to modulating cell adhesion and to inhibiting the binding of fibrinogen and other proteins to blood platelets, and inhibiting the aggregation of blood platelets specifically to the gp IIb/IIa fibrinogen receptor site. Fibrinogen is a glycoprotein present in blood plasma that participates in platelet aggregation and in fibrin formation. Platelets are cell-like anucleated fragments, found in the blood of all mammals, that also participate in blood coagulation.
Interaction of fibrinogen with the IIb/IIIa receptor site is known to be essential for normal platelet function.
When a blood vessel is damaged by an injury or other causative factor, platelets adhere to the disrupted subendothethial surface. The adherent platelets subsequently release biologically active constituents and aggregate. Aggregation is initiated by the binding of agonists, such as thrombin, epinephrine, or ADP to specific platelet membrane receptors. Stimulation by agonists results in exposure of latent fibrinogen receptors on the platelet surface, and binding of fibrinogen to the glycoprotein IIb/IIIa receptor complex.
Attempts have been made to use natural products and synthetic peptides to determine the mechanism of adhesion and platelet aggregation. For example, Rouslahti and Pierschbacher in Science, 238, 491-497 (1987), describe adhesive proteins such as fibronectin, vitronectin, osteopontin, collagens, thrombospondin, fibrinogen, and von Willebrand factor that are present in extracellular matrices and in blood. The proteins contain the tripeptide arginine-glycine-aspartic acid (RGD) as their glycoprotein IIb/IIa recognition site. These arginineglycine-aspartic acid containing tripeptides are recognized by at least one member of a family of structurally related receptors, integrins, which are heterodimeric proteins with two membrane-spanning subunits. The authors state that the conformation of the tripeptide WO 98/00401 PCT/US97/11047 -2sequence in the individual proteins may be critical to recognition specificity.
Cheresh.in Proc. Nat'l Acad. Sci. 84, 6471-6475, (1987), describes an Arg-Gly-Asp directed adhesion receptor expressed by human endothelial cells that is structurally similar to the IIb/lIa complex on platelets but is antigenically and functionally distinct. This receptor is directly involved in endothelial cell attachment to fibrinogen, von Willebrand factor, and vitronectin.
Pierschbacher and Rouslahti, in J. of Biol. Chem., 262, 17294-17298 (1987) hypothesized that the Arg-Gly-Asp sequence alone would be a sufficient signal for receptor recognition and binding and that, therefore, the conformation of the tri-peptide sequence would be determinative. Various synthetic peptides were produced and the authors concluded that the stereochemical conformation of Arg-Gly-Asp as influenced by enantiomeric substitutions or additions to this sequence significantly influenced receptor-ligand interaction. The authors further showed that cyclization of a decapeptide by forming a disulfide bridge between non-terminal residues Pen and Cys, rendered the peptide much less effective at inhibiting attachment to fibronectin.
In Proc. Nat'l Acad. Sci. 81, 5985-5988 (1984), the same authors describe tetrapeptide variants of the cell recognition site of fibronectin that retain attachment-promoting activity. Peptides having a tetrapeptide recognition site are described in U.S. Pat. Nos. 4,589,881 and 4,614,517. A number of large polypeptide fragments in the cellbinding domain of fibronectin have cell-attachment activity. For example, see U.S. Pat. Nos. 4,517,686, 4,661,111 and U.S. Pat. No.
4,578,079.
Ruggeri et al., Proc. Nat'l Acad. Sci. 83, 5708- 5712 (1986) explore a series of synthetic peptides designed in lengths to 16 residues, that contain RGD and a valine attached to the aspartic acid residue of RGD that inhibit fibrinogen binding to platelets. See also Koczewiak et al., Biochem. 23, 1767-1774 (1984); Ginsberg et al., WO 98/00401 PCT/US97/11047 -3- J. Biol. Chem. 260(7), 3931-3936 (1985); and Haverstick et al., Blood 66(4), 946-952 (1985). Other inhibitors are disclosed in Eur. Pat. App.
Nos. 275,748 and 298,820.
A number of low molecular weight polypeptide factors have been isolated from snake venom. These factors apparently have high affinity for the gp Ib/IIIa complex. For example, Huang et al., J.
Biol Chem., 262, 16157-16163 (1987); Huang et al., Biochemistry, 28, 661-666 (1989) describe the primary structure of the venom trigramin which is a 72 amino acid polypeptide that contains the RGD subunit.
Echistatin is another compound which has high affinity for the gp IIb/IIIa complex. This polypeptide contains 49 amino acids and has the RGD subunit and various disulfide bridges. Gan et al., J. Biol. Chem., 263, 19827-19832 (1988). See also, Dennis et al., Proc. Nat'l Acad.
Sci. USA, 87, 2471-2475 (1989). However, these snake venom factors also have high affinity for other members of the adhesive protein receptor family including the vitronectin and fibronectin receptors so are not selective for the gp lb/IIIa complex.
While it is known that the tripeptide sequence Arg-Gly-Asp is present in certain polypeptides that can duplicate or inhibit the cell attachment-promoting effects of fibronectin and vitronectin, the tripeptide Arg-Gly-Asp has low activity. At present, there is little understanding of how other amino acids coupled to this sequence influence binding specificity. U.S. Pat. No 5,023,233 discloses small cyclic hexapeptides which contain the sequence Arg-Gly-Asp and are useful platelet aggregation inhibitors. U.S. Pat. No. 5,037,808 discloses the use of indolyl platelet-aggregation inhibitors which are believed to act by antagonizing interactions between fibrinogen and/or extracellular matrix proteins and the platelet gp IIb/IIIa receptor. U.S. Pat. No.
5,037,808 discloses guanidino peptide mimetic compounds that retain an Asp residue which inhibit platelet aggregation. W09014103 describes the use of antibody-poly-peptide conjugates wherein said polypeptides contain the Arg-Gly-Asp (RGD) sequence.
W09111458 discloses the use of large cyclic peptides containing RGD flanked by proline residues which are platelet aggregation inhibitors. WO 9101331 discloses small cyclic platelet aggregation inhibitors which are synthetic cyclic pentapeptides containing the tripeptide sequence Arg-Gly-Asp and a thioether linkage in the cycle. U.S. Patent No. 5,051,405 also discloses the use of peptides pseudopeptides such as Namidino-piperidine-3-carboxylgylcyl-L-aspartyl-L-valine that inhibit platelet aggregation and thrombus formation in mammalian blood. EP 445 796 discloses linear compounds which can include internal piperazinyl or piperidinyl derivatives. EP 437 367 discloses linear polypeptide fibrinogen receptor antagonists. U.S. Patent No. 5,256,812 discloses compounds of the R 1 COOR wherein R 1 is a guandidino or amidino moiety and A and B are chosen from specific monosubstituted aryl or heterocyclic moieties.
While a multitude of compounds or peptide analogs believed to inhibit platelet aggregation by inhibiting binding to a blood platelet by fibrinogen are known, the present invention provides novel fibrinogen receptor antagonist prodrugs of antagonists that have significant binding activity and are, therefore, useful for the reasons stated herein.
Summary of the Invention 5 The invention relates to compounds having the formula 5 0
X-W-Y-(CH
2 )u-C-NH-(A)r-B or pharmaceutically acceptable salt(s) thereof, wherein e::o u is 0or 1; X is "i H 2
N
H N
H
2 W is wherein q is 0 or 2; Y is N N O O X and Y, provided that q=O, together form [R:\LIBAA]07896.dOC tab A is
CH
3 risOor1; s B is -O(CH 2 2
NH
2
-CH
2 C(OPh)HCH 2 NH2, or -CH(CH 3
)(CH
2 )2NH2.
The compounds are useful as prodrugs of fibrinogen receptor antagonists.
The invention also includes the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting the aggregation of blood platelets, preventing platelet thrombosis, preventing thromboembolism or preventing reocclusion, in a 10o mammal.
ft t [R:\LIBAA07896.doc:tab 6 to 24 THIS PAGE HAS BEEN LEFT INTENTIONALLY BLANK.
.00.
0000 to9 0-0 [R:\LlBAA]07896.doctab Detailed Description of the Invention The invention relates to compounds having the formula 0 11 X -W-Y-(CH 2 )u-C NH-(A)r-B or pharmaceutically acceptable salt(s) thereof, wherein u is 0orl1; X is
H
2
N
HN N ,or H N HN
H
2 W is wherein q is 0 or 2; Y is N or
HN
A is HN3 ris 0or 1 i :OC2)N2 NC2(~)C2Hor-HC3(H)N andS Hl te usiun rea rvosydfnd [R:\LIBAA]07896.doc:tab Specific exemplifications of this group are shown below: HN N-
H
3
C
a *4.
a. a.
a a a.
a a a a. a a a i a 9 a. a a a.
9 a.
a 9 a 9 9 [R:\LIBAAj07896.dOC~tab WO 98/00401 PCT/US97/11047 -27 H2N S- 0 N\ -O
NH
2
H
a
C
0 HNO HN N N O NH 2
HN
N
HH
H
3C r^ 0 N N
NH
2 N-N 0 H 0 HN O
H
and pharmaceutically acceptable salts.
The active acids of these compounds have been evaluated in vitro and found to have an IC50 for inhibiting platelet aggregation of between about 8nM and The prodrugs may be administered in low amounts relative to the amounts of antagonist that would ordinarily be administered. The prodrugs may be administered orally. The prodrugs retain structural WO 98/00401 PCT/US97/11047 -28integrity while passing though the gastrointestinal system, and are effectively delivered to cells. They are subjected to metabolic reactions to form the active acid which then interacts with the platelet receptor site.
A number of very serious diseases and disorders involve hyperthrombotic complications which lead to intravascular thrombi and emboli. Myocardial infarction, stroke, phlebitis and a number of other serious conditions create the need for novel and effective fibrinogen receptor antagonists.
One test which is used to evaluate fibrinogen receptor antagonist activity is based on evaluation of inhibition of ADPstimulated platelets. Aggregation requires that fibrinogen bind to and occupy the platelet fibrinogen receptor site. Inhibitors of fibrinogen binding inhibit aggregation. In the ADP-stimulated platelet aggregation assay used to determine inhibition associated with the acids of the compounds claimed in the instant invention, human platelets are isolated from fresh blood, collected into acid citrate/dextrose by differential centrifugation followed by gel filtration on Sepharose 2B in divalent ion-free Tyrode's buffer (pH 7.4) containing 2% bovine serum albumin.
Platelet aggregation is measured at 37°C in a Chronolog aggregometer. The reaction mixture contains gel-filtered human platelets (2 x 108 per ml), fibrinogen (100 micrograms per ml (ug/ml)), Ca 2 (1 mM), and the compound to be tested. The aggregation is initiated by adding 10 mM ADP 1 minute after the other components are added. The reaction is then allowed to proceed for at least 2 minutes. The extent of inhibition of aggregation is expressed as the percentage of the rate of aggregation observed in the absence of inhibitor. The IC50 is the dose of a particular compound inhibiting aggregation by 50% relative to a control lacking the compound.
Additionally, these compounds are useful for treating mammals suffering from a bone condition caused or mediated by increased bone resorption, who are in need of such therapy.
Pharmacologically effective amounts of the compounds, including WO 98/00401 PCT/US97/11047 -29pharamaceutically acceptable salts thereof, are administered to the mammal, to inhibit the activity of mammalian osteoclasts.
Additionally, these compounds are useful for treating angiogenesis (formation of new blood vessels). It has been postulated that the growth of tumors depends on an adequate blood supply, which in turn is dependent on the growth of new vessels into the tumor.
Inhibition of angiogenesis can cause tumor regression in animal models.
(See, Harrison's Principles of Internal Medicine, 12th ed., 1991). These compounds are therefore useful in the treatment of cancer for inhibiting tumor growth. (See Brooks et al., Cell, 79:1157-1164 (1994)).
The term "pharmaceutically acceptable salts" shall mean non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid. Representative salts include the following salts: 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, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamaote, palmitate, panthothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, valerate.
Compounds of the present invention may be chiral; included within the scope of the present invention are racemic mixtures and separated enantiomers of the general formula. Furthermore, all diastereomers, including E, Z isomers, of the general formula are included in the present scope. Furthermore, hydrates as well as anhydrous compositions and polymorphs of the general formula are within the present invention.
The compounds of the invention are prodrugs of active acids which inhibit fibrinogen binding to the gpllb/IIIa platelet receptor WO 98/00401 PCT/US97/11047 site. These acids form in vivo, subsequent to administration to the patient, according to successive metabolic reactions, for example: 0
MAO
R'-CH2NH 2 MAO- R'-CH aldehyde 0 dehydrogenase R'-CH
R'-COOH
NAD+
NADH
Compounds of the invention of the general formula R'-CH2NH2, may form aldehydes that metabolize into the active acid.
The term "pharmaceutically effective amount" shall mean that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system or animal that is being sought by a researcher or clinician. The term "anti-coagulant" shall include heparin, and warfarin. The term "thrombolytic agent" shall include agents such as streptokinase and tissue plasminogen activator.
The term "platelet anti-aggregation agent" shall include agents such as aspirin and dipyridamole.
The term "alkyl" means straight or branched alkane containing 1 to about 10 carbon atoms, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexy, octyl radicals and the like, straight or branched alkene containing 2 to about 10 carbon atoms, propylenyl, buten-1-yl, isobutenyl, pentenylen-1-yl, 2,2-methylbuten-1-yl, 3-methylbuten-l-yl, hexen-1-yl, hepten-l-yl, and octen-l-yl radicals and the like, or straight or branched alkyne containing 2 to about 10 carbon atoms, ethynyl, propynyl, butyn-1-yl, butyn-2-yl, pentyn-1-yl, pentyn-2-yl, 3-methylbutyn-1-yl, hexyn-1-yl, hexyn-2-yl, hexyn-3-yl, 3,3-dimethylbutyn-l-yl radicals and the like.
WO 98/00401 PCT/US97/11047 -31 The term "aryl" means a 5- or 6-membered aromatic ring containing 0, 1, or 2 heteroatoms selected from O, N, and S, e.g.
phenyl, pyridine, pyrimidine, imidazole, thiophene, oxazole, isoxazole, thiazole, and amino- and halogen- substituted derivatives thereof.
The terms "alkyloxy" or "alkoxy" include an alkyl portion where alkyl is as defined above, methyloxy, propyloxy, and butyloxy.
The terms "arylalkyl" and "alkylaryl" include an alkyl portion where alkyl is as defined above and to include an aryl portion where aryl is as defined above. The CO-n or Cl-n designation where n may be an integer from 1-10 or 2-10 respectively refers to the alkyl component of the arylalkyl or alkylaryl unit. Examples of arylalkyl include benzyl, fluorobenzyl, chlorobenzyl, phenylethyl, phenylpropyl, fluorophenylethyl, chlorophenylethyl, thienylmethyl, thienylethyl, and thienylpropyl. Examples of alkylaryl include toluene, ethylbenzene, propylbenzene, methylpyridine, ethylpyridine, propylpyridine, butylpyridine, butenylpyridine, and pentenylpyridine.
The term "halogen" includes fluorine, chlorine, iodine and bromine.
The term "oxy" means an oxygen atom. The term "thio" means a sulfur atom. Under standard nonmenclature 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. For example, a C1-6 alkyl substituted with alkyl-carbonylamino is equivalent to
HO
I II C1-6-alkyl-N-C-C1 In the schemes and examples below, various reagent symbols have the following meanings: WO 98/00401 PCT/US97/11047 -32-
BOC
(or Boc): Pd-C:
DMF:
DMSO:
CBZ:
CH2C12: CHC13: EtOH: MeOH: EtOAc: HOAc:
BOP:
EDC:
Oxone:
LDA:
PYCLU:
t-butyloxycarbonyl PalladiuUn on activated carbon catalyst Dimethylformamide Dimethylsulfoxide Carbobenzyloxy Methylene chloride chloroform ethanol methanol ethyl acetate acetic acid Benzotriazol-1 -yloxytris(dimethylamino)phosphonium, hexafluorophosphate 1-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride potassium peroxymonosulfate Lithium diisopropylamide Chloro-N,N,N',N'-bis(pentamethylene)formamidinium hexafluorophosphate The compounds of the present invention can be administered in such oral forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions.
Likewise, they may be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramusculsar form, all using forms well known to those of ordinary skill in the pharmaceutical arts. An effective but non-toxic amount of the compound desired can be employed as an anti-aggregation agent.
Compounds of the invention, or pharmaceutically acceptable salts thereof, are useful in the manufacture of a medicament for inhibiting binding of fibrinogen to the platelet membrane glycoprotein complex Ib/IIIa receptor, preventing platelet thrombosis, thromboembolism and reocclusion during and after thrombolytic WO 98/00401 PCT/US97/11047 -33 therapy or after angioplasty or coronary artery bypass procedures, and preventing myocardial infarction in a mammal.
Compounds of the invention may be administered to patients where prevention of thrombosis by inhibiting binding of fibrinogen to the platelet membrane glycoprotein complex IIb/IIIa receptor is desired. They are useful in surgery on peripheral arteries (arterial grafts, carotid endarterectomy) and in cardiovascular surgery where manipulation of arteries and organs, and/or the interaction of platelets with artificial surfaces, leads to platelet aggregation and consumption. The aggregated platelets may form thrombi and thromboemboli. Compounds of this invention may be administered to these surgical patients to prevent the formation of thrombi and thromboemboli.
Extracorporeal circulation is routinely used for cardiovascular surgery in order to oxygenate blood. Platelets adhere to surfaces of the extracorporeal circuit. Adhesion is dependent on the interaction between gp IIb/IIIa on the platelet membranes and fibrinogen adsorbed to the surface of the circuit. (Gluszko et al., Amer.
J. Physiol., 252(H), 615-621 (1987)). Platelets released from artificial surfaces show impaired hemostatic function. Compounds of the invention may be administered to prevent adhesion.
Other applications of these compounds include prevention of platelet thrombosis, thromboembolism and reocclusion during and after thrombolytic therapy and prevention of platelet thrombosis, thromboembolism and reocclusion after angioplasty or coronary artery bypass procedures. They may also be used to prevent myocardial infarction.
The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of WO 98/00401 PCT/US97/11047 -34the drug required to prevent, counter, or arrest the progress of the condition.
Oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day and preferably 0.01-100 mg/kg/day and most preferably 0.01-20 mg/kg/day. For example, a typical 90 kg patient would receive oral dosages ranging between about 0.9 mg/day and about 9 g/day, most preferably between about 0.9 mg/day and 1.8 g/day. Suitable pharmaceutical oral compositions such as tablets or capsules may contain, for example, mg, 100 mg, 200 mg and 500 mg. Intravenously, the most preferred doses will range from about 1 to about 10 mg/kg/minute during a constant rate infusion.
Advantageously, compounds of the present invention may be administered in divided doses of two, three, or four times daily.
Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration will, or course, be continuous rather that intermittent throughout the dosage regime.
In the methods of the present invention, the compounds herein described in detail are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "carrier" materials) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with convention pharmaceutical practices.
For instance, for oral administration in the form of a tablet or capsule, the compound can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral WO 98/00401 PCT/US97/11047 administration in liquid form, the oral prodrug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanpl, glycerol, water and the like. Moreover, when desired or necessary, suitable binders, lubricants, distintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or betalactose, corn-sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch methyl cellulose, agar, bentonite, xanthan gum and the like.
Active drug can also be co-administered with the usual doses of suitable anticoagulation agents, such as heparin or warfarin (typically given in tablet doses between 1 and 20 mg daily during administration of the active drug), or thrombolytic agents such as tissue plasminogen activator (typically given in i.v. doses of between 20 and 150 mg over two hour period prior to or during administration of the active drug), to achieve beneficial effects in the treatment of various vascular pathologies. Such co-administration also includes administration if the active drug with doses of anticoagulant agents or thrombolyric agents less than the usual doses of those agents.
Compounds of the invention may prepared according to a number of methods familiar to persons skilled in the art. For example, in one method, a fused, protected tricyclic ring system having a bromine substituent, e.g. 2-(1,1-Dimethylethoxycarbonyl)-7-bromo- 1,2,3,4,-tetrahydro-9H-pyrido[3,4-b]indole, is converted to the corresponding carboxylic acid, e.g. 2-(1,1-Dimethylethoxycarbonyl)- 1,2,3,4,-tetrahydro-9H-pyrido[3,4-blindole-7-yl carboxylic acid, which is then combined with a 4-amino phenoxy compound, e.g. 1-(1,1- Dimethylethoxycarbonylamino)-2-(4-amino-3-methylphenoxy)ethane, to produce amine prodrugs of the invention.
WO 98/00401 PCT/US97/11047 -36 In another general procedure, an isoindol carboxylic acid is reacted with 1,3-Diaminopropane to produce amine prodrugs of the invention.
In another general procedure, a protected 4-amino phenol, e.g. 4-amino-3-methylphenol is converted to a phenoxyacetate with bromoacetate. The corresponding phenoxyacetamide is formed using dimethylamine. The phenoxyacetamide is reacted with a piperazinylbenzoic acid to form amine prodrugs of the invention.
These and other methods, including those exemplified below, may be used to prepare prodrugs of the invention.
WO 98/00401 WO 9800401PCTIUS97/1 1047 37 EXAMPLE 1 Br HN
N
H
B0C 2 0 BOCN I
B
H
2. CH 3 MgCl 2.tefl BuLi, 1-1 J. Am. Chem. Soc.
1987 109, 3378.
THF, -78 0
C
RT
-O\"NHBOC
PYCLU, 0H 2 01 2 i- Pr 2 NEt 1-4
CH
3 0 HN I N Na H
H
WO 98/00401 PCT/US97/11047 -38 Br tBuO N 1-2 YW N O
H
1,1 -Dimethylethoxycarbonyl)-7-bromo-1,2,3,4,-tetrahydro-9Hpyridor3,4-blindole 1-2 A suspension of 1-1, prepared by the method of Rinehart et al. Am. Chem. Soc., 1987 109, p 3378-3387) (0.366 g, 1.46 mmol) in CH2C12 (8 mL) was treated with triethylamine (0.61 mL, 4.4 mmol) followed by di-tert-butyldicarbonate (0.38 g, 1.7 mmol) for 1 hour at room temperature. The solution was concentrated and the residue chromatographed (20% EtOAc/hexanes) to give 1-2 as a white solid.
EtOAc/hexanes)=0.28 1 H NMR (400 MHz, CDC13) d 8.0-7.6 1H), 7.46 IH), 7.33 (d, 1H), 7.2 1H), 4.6 (bs, 2H), 3.78 (bs, 2H), 2.76 (bs, 2H), 1.5 9H).
tBuO N OH 1-
H
O
1,1 -Dimethylethoxycarbonyl)- 1,2,3,4,-tetrahydro-9H-pyrido [3,4blindole-7-yl carboxylic acid 1-3 A solution of 1-2 (0.26 g, 0.734 mmol) in THF (10 mL) was cooled to 0°C and treated with methylmagnesium chloride (3.0M in THF, 0.29 mL, 0.87 mmol) to give a pale yellow solution. After minutes the solution was cooled to -78°C and treated with t-BuLi (1.7M in pentane, 4.35 mL, 7.39 mmol) to give a bright yellow solution.
After 10 minutes C02 gas was bubbled vigorously through the solution for 10 minutes. Saturated NH4C1, water and enough 6N NaOH to reach pH12 were added and the solution extracted with EtOAc. The EtOAc WO 98/00401 WO 9800401PCTIUS97/1 1047 39 layer was back extracted with 0.5 NaOH and the aqueous layers combined, acidified to pH 7 and extracted with EtOAc, the EtOAc layer was dried (Na2SO4)filtered and concentrated to give 1-3 as an offwhite solid.
Rf(75:25:1 CHC13JMeOHIHOAc)=0.48 IH NMR (400 MHz, DMSO-d6) d 12.0 bs, 1H), 11.2 1H), 7.93 (s, 1H), 7.6 IH), 7.45 IHI), 4.6 2H), 3.68 (in, 2H), 2.7 (mn, 2H), 1.4 9H).
0 BOON IN X/\1-4 Na2HI
NHBOC
H CH 3
CH
N-(3 -Methyl-4-(2-(, 1, -dimethylethoxycarbonylamino)ethoxy)phenyl)- 1,1 -dimnethylethoxycarbonyl)- I ,2,3,4,-tetrahydro-9H-pyrido[3, 4-blindol)carboxyamide 1-4 A solution of 1-3 (0.240 g, 0.759 mnmol) and 4-6 (0.202 g, 0.758 inmol) in CH2Cl2 was treated with diisopropylethylamine (0.4 mL, 2.3 mmol) and PYCLU (0.304 g, 0.843 rninol) and stirred at room temperature for three days. The solution was concentrated and the residue was absorbed to silica gel and chromatographed in a gradient of 40 to 60% EtOAc/hexanes to give 1-4 as a white solid.
Rf (60% EtOAcfhexanes)=0.30 I H NMR (400 MHz, CDCl3) d 8.04 I1H), 7.71 I1H), 7.63 I1H), 7.53 2H), 6.79 2H), 5.0 1H), 4.69 2H), 4.01 2H), 3.79 (t, 2H), 3.51 2H), 2.82 2H), 2.32 3H), 1.52 9H), 1.46 9H).
WO 98/00401 WO 9800401PCT/US97/11047 40 0 HN I N H H 3 N-(3-Methyl-4- (2-aminoethoxy)phenyl)-7 ,2 -tetrahydro-9Hpyrido[3,4-blindol)carboxyamide A solution of 1-4 in dioxane was cooled to 0 0 C and treated with HCl over 1.5 min. The mixture was stirred at 0 0 C for Ilh and concentrated in vacuc. The residue was chromatographed (18:1:1 EtOH/H2OINH4OH) to afford a white solid, which was then suspended in EtOAc and treated with HCl to afford 1-5 as a white solid.
Rf (18: 1:1 EtOH/H20/NI-40H)=0.26 I H NMR (400 MHz, D20) d 7.96 1H), 7.6 (in, 2H), 7.17 2H), 6.95 1H), 6.86 IR), 4.46 2H), 4.23 2H), 3.56 2H), 3.37 2H), 3.06 2H), 2.18 3H).
0 BOH 0
,CH
3
N
OH
3 1-OH 3 N-(3-Methyl-4-(2-(dimethylamino)ethoxy)phenyl)-7-(2-( 1,1dimethylethoxycarbonyl)- 1,2,3 ,4,-tetrahydro-9H-pyrido [3,4blindol)carboxyamide 1-6 1-3 (200 mig, 0.63 imol) and 5-4 (0.63 mmol, 122 mg) were dissolved in methylene chloride. PYCLU (0.69 inmol, 250 mg) was added followed by DIPEA (2.52 inmol, 0.44 miL). The reaction was stirred at room temperature overnight. The reaction mixture was diluted with EtOAC and washed with H20, saturated NaHCO3 and brine. T'he organic layer was dried (MgSO4), filtered and concentrated WO 98/00401 PCT/US97/11047 -41 to yield a tan solid. Flash chromatography MeOH/CHCl3 sat. NH3) gave 1-6 as a yellow oil.
Rf (10% MeOH/CHNH3 sat. C13)=0.50 1 H NMR (400 MHz; CDC13) d 8.00 (bs, 1H); 7.52 1H); 7.48 1H); 7.5 1H); 6.51-6.55 3H); 4.61 (bs, 2H); 3.99- 4.00 2H); 3.79- 3.81 (bt, 2H); 2.79-2.81 (bt, 2H); 2.61-2.63 2H); 2.32 3H); 2.34 3H); 2.30 3H); 1.50 9H).
0 HN.. N O, CH 3
N
H C~u CH, H CH 3 1-7 N-(3-Methyl-4-(2-(dimethylamino)ethoxy)phenyl)-7-(1,2,3,4,tetrahvdro-9H-pyrido 3,4-blindol)carboxvamide 1-7 1-6 was dissolved in EtOAc and cooled to HCI (g) was bubbled through until the solution was saturated. The reaction mixture was warmed to 00 and stirred for 15 minutes. The reaction mixture was concentrated to yield a brown solid which was purified by flash chromatography (gradient 10% MeOH/CHCI3 saturated with NH3 to 60% MeOH/CHC13 saturated with NH3)) to yield 1-7 as a white solid.
Rf (10% MeOH/CHCl3 sat. NH3)=0.19 1 H NMR (400 MHz; DMSO- d6) d 9.73 1H); 9.45-9.55 (bs, 1H); 8.05 1H); 7.69 1H); 7.57 1H); 7.24-7.26 IH); 4.40 2H); 4.35 2H); 3.47-3.5 4H); 2.99-3.1 2H); 2.50 6H); 2.22 (s, 3H).
WO 98/00401 WO 9800401PCTIUS97/11047 42 EXAMPLE 2 Br \NHNH 2 0 0% PPA, D C0 2 Et NaH DMF
CICH
2
CN
B r O 2 E t
-~N
ON
LAH Et 2
O
2-4 WO 98/00401 WO 9809401PCTIUS97/1 1047 43 SCHEME 2 (CONTD) 2-4 B0C 2 0 TEA BOCN
B
Pd(OAc) 2 dppp/ DMF 00(g) CH 3 0H WO 98/00401 WO 9800401PCTIUS97/1 1047 44 rN CO 2
CH
3 BOON NaBH 3 CN EtOAc r N 00 2 0H 3 BOOND, 2-7 LiOH MeOH H 2 0fTHF f-N C0 2
H
BOON::,
Lit 1flfl
PYOL
H
2 N O~~1U
U
ICH
2 01 2
H
3
C
i Pr 2 NEt 0' 0
NHBOC
BO N: H
H
3
C
WO 98/00401 WO 9800401PCTJIUS97/11047 .0,,,NHBOC EB0CN HOI/ EtOAc
NH
2 2z-IQ
HN,
WO 98/00401 PCT/US97/11047 -46- H CH3 2-1 Br- N-N Et 0 Ethyl 2-(4-bromo- 1-hvdrazinimine)propanoate 2-1 A mixture of 4-bromophenylhydrazine (Aldrich, 0.5 g, 2.2 mmol) and ethyl acetoacetate (Aldrich, 0.24 mL, 2.2 mmol) in pyridine (0.6 mL) was heated to reflux overnight. The reaction was cooled, diluted with water and the precipitate that resulted was collected and washed with water, dried under vacuum to give 2-1.
MeOH/CHCl3 saturated with NH3)=0.86 1H NMR (400 MHz, CDC13) d 7.64 1H); 7.41 2H); 7.39 2H); 4.30-4.34 2H); 2.10 (s 3H); 1.36 3H).
B
N
CO
2 Et H 2 5-Bromo-2-ethoxvcarbonvl indole 2-2 A mixture of 2-1 (0.54 g, 1.9 mmol) and polyphosphoric acid (1.6 mL) was heated to 115 0 C for 10 minutes, then diluted with cold water and extracted with EtOAc. The layers were separated and the aqueous layer extracted with EtOAc. The organic layers were combined, washed with brine, dried with MgSO4, filtered and concentrated to give 2-2 as a brown solid.
EtOAc/hexanes)=0.45 1H NMR (400 MHz, CDC13) d 8.95( bs, 1H); 7.82 1H); 7.41 1H); 7.30 1H); 7.15 1H); 4.44 2H); 1.40 3H).
WO 98/00401 PCT/US97/11047 -47- Br C02Et N 2-3
CN
1-(Cvanomethvl)-2-ethoxycarbonyl-5-bromo-indole 2-3 A solution of 2-2 (11.2 g, 44.4 mmol) in DMF (400 mL) was treated with NaH (3.2 g of 60% dispersion in oil, 66.6 mmol) for hour and then chloroacetonitrile (Aldrich, 5.6 mL, 88.8 mmol) was added and the reaction was stirred overnight. The solvent was removed in vacuo and the residue was partitioned between water and EtOAc.
The water layer was extracted with EtOAc, the organic layers were combined, washed with water, brine, dried with MgSO4, filtered and evaporated to give 2-3 as a brown solid.
EtOAc/hexanes)=0.46 1H NMR (400 MHz, CDC13) d 7.82-7.83 (bs, 1H); 7.51-7.54 (dd, 1H); 7.30-7.32 (bd, 2H); 5.60 2H); 4.41-4.43 2H); 1.41-1.43 3H).
BrNH 'N
NH
8-Bromo-2.3.4,5-tetrahydropyrazino- 1,2-alindole 2-4 A slurry of 2-3 (12.2 g, 39.7 mmol) in diethyl ether (400 mL) was added via dropping funnel to a solution of LAH in ether (79.4 mL, 1 M in ether, 79.4 mmol) and stirred at room temperature overnight. The slurry was diluted with saturated sodiumpotassium tartrate (Rochelle's salt) and stirred for 15 minutes, then transferred to a separatory funnel containing EtOAc and the layers separated. The aqueous layer was extracted with EtOAc, the organic layers were combined, washed with water, brine, dried with MgSO4, filtered and evaporated to give 2-4 as a brown solid.
WO 98/00401 WO 9800401PCTIUS97/11047 48 MeOH/CHC13 saturated with NH3)=O.42 I H NMR (400 MHz, CDCl3) d 8.38-8.41 (bs, I1H); 7.66 I1H); 7.21 7.22 (dd, 2H); 7.14 2H), 6.14 I1H); 4.22 2H); 3.99 2H); 3.36-3.37 2H).
BOCNK N r 8-Bromo-3-( 1,1 -dinmethylethoxycarbonyl)-2,3 pvrazino-[l,2-alindole A solution of 2-4(10 g, 40 mmol) in CH2Cl2 (200 mL) was cooled to 0 0 C and treated with di-tertbutyldicarbonate (8.7 g, mmol) and triethylamine (5.6 m1L, 40 mimol). The solution was allowed to warm slowly and after 48 hours was concentrated and the residue dissolved in EtOAc, washed with water and brine, dried over Na2SO4, filtered and evaporated. The residue was chrornatographed EtOAc/hexanes) to give 2-5 as a solid.
EtOAc/hexanes)=0.22 IH NMR (400 MHz, CDC13) d 7.67 1H); 7.23 IH); 7.13 1H); 6.21 IH); 4.80 2H); 4.04 2H), 3.93 2H); 1.50 9H).
WO 98/00401 PCT/US97/11047 -49- N'N C0 2
CH
3
BOCN
8-Methoxycarbonyl-3-(1,1 -dimethylethoxycarbonyl)-2,3,4,5tetrahydropvrazino-[ 1,2-alindole 2-6 A solution of 2-5 (3.0 g, 8.5 mmol) in MeOH (60 mL) and DMSO (20 mL) was treated with triethylamine (3.55 mL, 25.5 mmol), 1,3-Bis(diphenylphosphino)propane (1.75 g, 4.25 mmol) and palladium (II) acetate (0.952 g, 4.25 mmol). Carbon monoxide was bubbled through the solution while it was heated to reflux for 2 hours. The reaction was heated at reflux overnight under a balloon atmosphere of carbon monoxide. Additional 1,3-Bis(diphenylphosphino)propane (0.8 g, 2.12 mmol) and palladium acetate (0.476 g, 2.12 mmol) were added and the reaction was heated at reflux for 48 hours under a balloon atmosphere of carbon monoxide. The reaction was cooled to room temperature, the residue was partitioned between water and EtOAc.
The water layer was extracted with EtOAc, the organic layers were combined, washed with water, brine, dried with MgSO4, filtered and evaporated. The residue was chromatographed (25% EtOAc/hexanes) to give 2-6 as a yellow solid.
Rf(30% EtOAc/hexanes)=0.21 1H NMR (400 MHz, CDC13) d 8.32 1H); 7.90 1H); 7.26 1H); 6.38 1H); 4.83 2H); 4.12 2H); 3.96-3.93 5H); 1.50 9H).
WO 98/00401 PCT/US97/11047 C0 2
CH
3 2-7 BOCN,. J 8-Methoxycarbonyl-3-( 1,1 -dimethylethoxycarbonyl)- ,1 a,2, 3,4,5-hexahvdropvrazino-[ 1,2-alindole 2-7 2-6 (0.091 mmol, 30 mg) was dissolved in EtOAc and cooled to 0°C. NaBH3CN (0.45 mmol, 28 mg) was added portion-wise and the reaction was warmed to room temperature for 15 min. The reaction mixture was basified with saturated NaHCO3 and extracted into EtOAc. The organic layer was washed with brine, dried (MgSO4), filtered and concentrated to yield 2-7 as a colorless oil.
Rf (2:1 hexane/ EtOAc)=0.4 1H NMR (400 MHz; CDC13) d 7.83-7.82 1H); 7.73 1H); 6.40- 6.38 1H); 4.15-4.0 (bs, 2H); 3.85 3H); 3.60-3.56 2H); .305- 2.65 4H); 2.60-2.58 (dd, 1H); 1.50 9H).
'NH CO 2
H
BOCN,
3-(1,1-Dimethylethoxycarbonyl)- 1,1a,2,3,4,5-hexahydropyrazino-[ 1,2-alindole-8-carboxylic acid 2-8 2-7 (0.90 mmol, 300 mg) was slurried in (2mL/2/2). LiOH (1.8 mmol, 76 mg) was added and the reaction mixture was heated to 50°C. After 0.5 hours, the reaction mixture became homogeneous, and was then stirred at room temperature for an additional 2 hours. The reaction mixture was diluted with 10% citric acid and EtOAc. The layers were separated, and the organic layer was washed with H20 and brine. Drying (MgSO4), filtering and concentrating gave 2-8 as a yellow solid.
Rf (97/3/1 CHCl3/MeOH/HOAC)=0.70 WO 98/00401 WO 9800401PCTIUS97/11047 51 lH NMR (400 MHz; CDCl3) d 7.90 I1H); 7.87 I1H); 6.41-6.3 9 (d, 1H); 4.25-4.0 (bs, 2H); 3.65-3.57 (in, 2H); 3.10-3.0 (dd, 1H); 3.02-2.98 d, 1H); 2.98-2.91 (bs, 1H); 2.69-2.66 (bs, 1H); 2.62-2.60 (dd, LH); 1.50 9H).
0 BOCN: Z
NHQ\
H
3
C
N-(3 -Methyl.-4-(2-( 1,1-dimethylethoxycarbonylamino )ethoxy)phenyl)-3-( 1,1 -dimethylethoxycarbonyl)- 1.1 a, 2,3.4,5 -hex ahydropvyrazino- r 1 .2-al indole- 8-carbox amide 2-9 2-8 (0.63 mmcl, 200 mg) and 4-6 (0.63 mmcl, 167 mg) were slurried in CH2CI2. PYCLU (0.70 mmol, 252 mg) was added followed by Dilsopropylethylamine. (2.5 mnmol, 0.44 mL). The slurry was stirred at room temperature overnight. The reaction mixture was diluted with EtOAc and washed with water and brine, dried (MgSO4), filtered and concentrated to yield a tan solid. Flash chromatography EtOAc/hexanes) gave 2-9 contaminated with bispiperidine urea by-product which was removed by tniturations with ether.
Rf (70% EtOAc/hexanes)=0.50 IH NMR (400 MHz; CDCl3) 7.65-7.63 (in, 3H); 7.36 1H); 6.77-6.75 (in, 2H), 6.45-6.43 2H); 5.0 (bs, IH); 4.05-4.25 (bs, 2H); 4.02-3.99 2H); 3.60-3.57 (in, 4H); 3.05-2.63 (in, 2.63-2.59 (dd, 1H); 2.28 3H); 1.50 9H).
WO 98/00401 WO 9800401PCTIUS97/11047 52 N 0 N 0
H
2 2-10 HND
H
H
3 0 N-(3-Methyl-4-(2-aminoethoxy)phenyl)- 1,1a,2,3 ,4 hexahydropvyrazino-[ 1 2-alindole-8-carboxamide 2-10 2-9 (180 mg) was dissolved in EtOAc and cooled to -78'C.
HC1(g) was bubbled through until the solution was saturated. The reaction mixture was stirred at 0' for 1 hour and then at room temperature for an additional hour. The reaction mixture was concentrated to yield a yellow solid which was purified by flash chromatography (10/0.5/0.5 EtOH/NH4OH/H20) to yield 2-10 as an off-white solid.
Rf (10/1/1 EtOHINII4OH/H20)=0.33 IH NMR (400 MHz; DMSO- d6) d 9.28 1H); 7.71-7.69 IH); 7.65 1H); 7.14-7.12 6.82-6.81 1H); 6.76-6.73 (dd, IH); 6.50- 6.48 1H); 3.92-3.89 2H); 3.62-3.57 (bd, 2H); 3.52-4.45 (in, IH); 2.97-2.81 (mn, 6H); 2.58-2.50 1H); 2.15 3H).
WO 98/00401 WO 9800401PCT/US97/11047 53 EXAMPLE 3
H
2 Nj NH 2 I
BOC
2 0 H2N,,, NHBOC 0 0
OH
BOCN
N
3-2
BOP
CH
3
CN
NMM
N ~N H BOC
H
HO /EtOAc
H
3-4 WO 98/00401 PCT/US97/11047 -54- H2N
NHBOC
3-1 3-(1,1-Dimethvlethoxycarbonylamino)propylamine 3-1 1,3-Diaminopropane [Aldrich] (40.5 mmol, 3.4 mL) was dissolved in CHC13 and cooled to o0C. Di-tert-butyldicarbonate (13.5 mmol, 2.9 g) was added and the reaction mixture was stirred for two hours at 0°C. The reaction mixture was washed with 10% KHSO4. The aqueous layers were combined, basified to pHO0 with saturated NaHCO3, and extracted with EtOAc and CHC13. The organic layers were combined, dried (Na2SO4), filtered and concentrated to yield pure 3-1 as a tan oil.
Rf (20% MeOH/CHCl3 sat. NH3)=0.3 1H NMR (400 MHz; CDC13) d 5.0 (bs, 1H); 3.18-3.17 (bq, 2H); 2.75- 2.71 2H); 1.68 (bs, 2H); 1.60-1.57 2H); 1.40 9H).
0 N N NNHBOC N H BOCN I H 3-3 -Dimethylethoxycarbonylamino)propyl)-2-(( 1- (1,1 dimethylethyoxycarbonyl)piperidine-4-yl)ethyl)- 1,3dihydroisoindol-l-one-6-carboxamide 3-3 3-2 (Prepared as described in EP 0540334) (0.65 mmol, 250 mg) and 3-1 (0.97 mmol, 167 mg) were slurried in CH3CN. NMM (0.65 mmol, 0.079 mL) was added, followed by BOP reagent (0.84 mmol, 370 mg). The homogenous reaction mixture was stirred for 48 hours at room temperature. The reaction mixture was diluted with EtOAc and washed with water, 10% KHSO4, saturated NaHCO3, and WO 98/00401 PCT/US97/11047 brine. The organic layer was dried (MgSO4), filtered and concentrated to yield an oil. Flash chromatography (gradient 50% EtOAc/hexanes to 100 EtOAc) yielded 3-3 as a yellow solid.
Rf(EtOAc)=0.45 IH NMR (400 MHz, CDC13) d 8.21 1H); 8.13 1H); 7.54 1H); 4.95 (bt, 1H); 4.41 2H); 4.13-4.10 (bd, 2H); 3.70-3.65 2H); 3.54- 3.51 2H); 3.25-3.23 2H); 2.67-2.64 (bt, 2H); 1.76-1.73 3H); 1.65-1.60 6H); 1.5 18H); 1.14-1.16 2H).
0 N NH2 N
H
HN -3-4 N-(3-aminopropyl)-2-(4-piperidinyl)ethyl- 1,3-dihydroisoindol- 1-one- 6-carboxamide 3-4 3-3 was dissolved in EtOAc and cooled to -78 0 C. HCI (g) was bubbled through until the solution was saturated. The reaction was allowed to stir for 1 hour at 00. The reaction mixture was then concentrated to yield pure 3-4 as a white solid.
Rf (10/1/1 EtOH/NH40H/H20)=0.93 1 H NMR (400 MHz; DMSO-d6) 8.6 (bd, 1H); 8.19 1H); 8.10 (d, 1H); 7.92 (bs, 2H); 7.77-7.69 1H); 4.55 2H); 3.58 2H); 3.23 (bd, 2H); 1.90-1.82 4H); 1.60-1.56 3H); 1.27-1.06 2H).
WO 98/00401 WO 9800491PCT/US97/1 1047 56 EXAMPLE 4
OH
CH
3
NH
2 BrCH 2 C0NH 2 30020 CHCI 3
OH
3
CS
2 00 3
DMF
'CH
3
CF
3 C00H 000 RT 4-3 WO 98/00401 WO 9809401PCTIUJS97/11047 57 NaH 2
AI(OCH
2
CH
2
OCH
3 2
-CH
3 THF reflux
OH
3
NH
2
BOC
2 0 CHC1 3
/RT
NH
2 WO 98/00401 WO 9800401PCT/US97/1 1047 58 0 N N-N\COOH cl PF6ill No
CH
2
CI
2 i-P r 2 NEt 4-7
H
3
C
HOI (gas) EtOAc 0 0
C
.0
NH
2 -2H01 WO 98/00401 WO 9800401PCTIUS97/11047 59
OH
~CH
3 0 NH 4-2 1.1-Dimethylethoxycarbonyl)amino-3-methylphenoI (4-2) To a 1 L round bottomed flask with a stirring bar, reflux condenser and an argon inlet was added 4-amino-3-methylphenol (15.00 g, 121.79 mmol), di-tert-butyldicarbonate (27.25 g, 124.84 mmol) and CHC13 (300 mL). This heterogeneous mixture was heated at reflux for 24 h during which time all of the solids dissolved. The mixture was cooled to room temperature and the solid product was collected by filtration. The material was triturated with a mixture of Et2O-hexanes collected on a frit and dried in vacuo to give 21.25 g of 4- (1,1 -dimethylethoxycarbonyl)amino-3 -methylphenol mp: 143- 144 0
C.
I H NMR (CDCI3): d 1.51 9H), 2.14 3H), 6.08 (br s, 1H), 6.48 (in, 2H), 6.60 (br s, lH), 7.20 j=8.5Hz, 1H).
WO 98/00401 PCT/US97/11047 O CONH 2
CH
3 0 NH 4-3 4-(1,1-Dimethylethoxycarbonyl)amino-3-methylphenoxyacetamide (4-3) To a 200 mL round bottomed flask with a stirring bar, and an argon inlet was added 4-(1,1 -dimethylethoxycarbonyl)amino-3methylphenol (5.00 g, 22.39 mmol), Cs2CO3 (14.59 g, 44.78 mmol), DMF (50 mL), and bromoacetamide (3.24 g, 23.51 mmol). This mixture was stirred vigorously at ambient temperature for 24 h. The mixture was filtered through a frit and the DMF was removed under high vacuum. The residue was dissolved in EtOAc (300 mL) and washed with H20 (2x) and brine Drying (MgSO4), filtration, and removal of the solvent in vacuo gave a solid. This material was triturated with EtOAc, the solid was collected by filtration and dried in vacuo to give 4.91 g of 4-(1,1-dimethyl-ethoxycarbonyl)amino- 3-methylphenoxyacetamide as a white, crystalline solid.
1H NMR (CDC13): d 1.50 9H), 2.22 3H), 4.42 2H), 5.81 (br s, 1H), 6.18 1H), 6.51 (br s, 1H), 6.78 2H), 7.58 1H).
WO 98/00401 PCT/US97/11047 -61 O CONH 2
SCH
3
NH
2 4-4 4-Amino-3-methylphenoxyacetamide (4-4) To a 200 mL round bottomed flask with a stirring bar and an argon inlet was added 4-(1,1 -dimethylethoxycarbonyl)amino-3methylphenoxyacetamide (4.91 g, 17.52 mmol) and trifluoroacetic acid mL). This solution was stirred at 0°C for 5h. The trifluoroacetic acid was removed in vacuo and the residue was partitioned between EtOAc and aqueous NaHCO3 solution. The layers were separated and the organic phase was washed with brine, dried (MgSO4), filtered and concentrated in vacuo to give 1.60 g of 4-amino-3-methylphenoxyacetamide as a white solid.
1 H NMR (CDC13): d 2.18 3H), 3.41 (br s, 2H), 4.40 2H), 5.71 (br s, 1H), 6.61 2H), 6.64 1H).
WO 98/00401 PCT/US97/11047 -62- O~ NH2
CH
3
NH
2 2-(4-Amino-3-methylphenoxv)ethvlamine To a 200 mL round bottomed flask with a stirring bar, reflux condenser and an argon inlet was added 4-amino-3-methylphenoxyacetamide (1.60 g, 8.88 mmol) and dry THF (100 mL). To this solution was added a solution of sodium bis(2methoxyethoxy)aluminium hydride (10.0 mL of a 3.4 M solution in toluene, 6.87 mmol). This solution was heated at reflux for 4h. The cooled reaction mixture was treated with saturated aqueous sodium potassium tartrate solution and extracted with EtOAc. The combined EtOAc extracts were dried (MgSO4), filtered and concentrated in vacuo. The crude 2-(4-amino-3-methylphenoxy)ethylamine was used in the next step without further purification.
WO 98/00401 PCT/US97/1 1047 63
H
N 0( 0
OH
3
NH
2 4-6 1 1 -Dimethylethoxycarbonylamino)-2-(4-amino-3methylphenoxv)ethane (4-6) To a 100 mL round bottomed flask with a stirring bar and an argon inlet was added 2-(4-amino-3-methylphenoxy)ethylamine (0.997 g, 6.00 mmol), chlorofom-n and di-tert-butyldicarbonate (1.31 g, 6.00 mmol). This solution was stirred at ambient temperature 2 h. The solvent was removed in vacuo and the residue was chromatographed on 75 g of silica gel using EtoAc-hexane as eluant. There was obtained 1.56 g of 1-(1,1-dimethylethoxycarbonylamino)-2-(4amino-3-methylphenoxy)ethane as a white crystalline solid. 1 H NMR (CDC13): d 1.42 9H), 2.19 3H), 3.35 (in, 2H), 3.48 (in, 2H), 3.92 (in, 2H), 4.98 (br s, 1H), 6.60 2H), 6.63 1H).
WO 98/00401 WO 98/040 1PCT/US97/11047 64 Preparation of 4-9
H
2 N \CO 2
CH
3
HCI*HN
n-butanol
A
HN N \C0 2 0H 3 4-9c IBOCO, NEt,
DMF
BOC-N N N 00 2
CH
3 1N NaO EtOH BOO-N N C0 2
H
4-9d WO 98/00401 PCT/US97/11047 HN C0 2
CH
3 4-9c 4-(N-Piperazine)benzoic acid methyl ester (4-9c) A solution of amine 4-9a (20.0 g, 132 mmol), amine 4-9b (23.6 g, 132 mmol) and n-butanol (500 ml) was refluxed for 168 h.
The solution was allowed to cool to ambient temperature. The crystals were collected, washed with Et20 and dried in vacuo to give ester 4-9c as a white solid.
1 H NMR (CD30D): 6 7.86 J=9Hz, 2H), 7.98 J=9Hz, 2H), 3.78 3H), 3.53 4H), 3.31 4H).
BocN N- C02CH 3 4-9d 4-(N-Boc-Piperazine)benzoic acid methyl ester (4-9d) To a stirred solution of amine 4-9c (15.0 g, 61.1 mmol), NEt3 (7.42 g, 73.4 mmol) and DMF (150 ml) was added Boc20 (14.7 g, 67.2 mmol). After 1.0 h, the solution was diluted with EtOAc and then washed with H20, 10% KHSO4, brine, dried (MgSO4) and concentrated to furnish ester 4-9d as a yellow solid.
TLC Rf 0.63 (silica, 40% EtOAc/hexanes) 1 H NMR (CD30D): 6 7.91 J=9Hz, 2H), 7.01 J=9Hz, 2H), 3.88 3H), 3.59 4H), 3.38 4H).
WO 98/00401 PCT/US97/1 1047 66 BocN \/NOC2 4-(N-Boc-Pip~erazine)benzoic acid (4-9) Asolution of ester1-4 (21.1 g,6 1.1 mmol) 1 N NaOH (100 ml, 100 mmol) and EtOH (200 ml) was heated to 60'C for 2.0 h.
The solution was acidifed with 10% KHS04 and then extracted with EtOAc. The EtOAc phase was washed with brine, dried (MgSO4) and concentrated to furnish acid 4-9 as a white solid.
H NMR (CD3OD): 6 7.81 J=9Hz, 2H), 6.88 J=9Hz, 2H), 3.49 (in, 4H), 3.24 (in, 4H), 1.40 9H).
00 N -0 N-P N0/
H
3
C
4-7 N-(2-Methyl-4-(2-(, 1, -diinethylethoxycarbonylamino)ethoxy)phenyl)- 4-4-(1 11 -dimethylethoxycarbonyl)p2iperazin- 1 -yl)benzamide 4-7) To a 100 mL round bottomed flask with a stirring bar and an argon inlet was added 1 -dimethylethoxycarbonyl)piperazin- 1 -yl)benzoic acid (0.863 g, 2.82 mmol), 1 -(1,1I -diinethylethoxycarbonylamino)-2-(4-ainino-3-methylphenoxy)ethane (0.75 g, 2.82 mmol), chloro-N,N (pentamethylene)formamidinium hexafluorophosphate (1.068 g, 2.96 mmiol) and CH2C12 (30 mL).
When all of the solids had dissolved dilsopropylethylamine (1.57 mL, 9.00 inmol) was added. The resulting mixture was stiffed at ambient temperature for 48 h. The mixture was diluted with CHC13 and washed with 10% aqueous citric acid, NaHCO3 solution and brine. Drying WO 98/00401 PCT/US97/11047 67 (MgSO4), filtration and removal of the solvent in vacuo gave the crude product. This material was chromatographed on 75 g of silica gel using EtOAc/hexane as eluant There was obtained a white solid. This material was recrystallized from hot EtOAc-hexane to give 0.914 g of N-(2-methyl-4-(2-(1,1 -dimethylethoxycarbonylamino)ethoxy)phenyl)-4-(4-( 1,1 -dimethylethoxycarbonyl)piperazin- 1yl)benzamide as white crystals. mp: 145-146 0 C. 1 H NMR (CDCI3): d 1.46 9H), 1.49 9H), 2.28 3H), 3.28 2H), 3.51 1H), 3.58 2H), 4.02 1H), 4.98 (br s, 1H), 6.76 2H), 6.93 j=9 Hz, 2H), 7.43 1H), 7.66 (br s, 1H), 7.78 j=9 Hz, 2H).
HN O NH 2 *2HCI O NH
H
3
C
4-8 N-(2-Methyl-4-(2-aminoethoxy)phenyl)-4-(1 -piperazinyl)benzamide, dihvdrochloride (4-8) To a 200 mL round bottomed flask with a stirring bar and a gas dispersion tube was added N-(2-methyl-4-(2-(1,1-dimethylethoxycarbonylamino)ethoxy)phenyl)-4-(4-( 1,1 -dimethylethoxycarbonyl)piperazin-1-yl)benzamide (0.912 g, 1.64 mmol) and 100 mL of dry EtOAc. This well stirred solution was cooled in an ice bath and saturated with HCI gas over 15 min. The mixture was aged 1 h at OC and the excess HCI was then removed with a stream of argon. The EtOAc was removed in vacuo and the crude product was recrystallized from MeOH-EtOAc to give 0.70 g of N-(2-methyl-4-(2aminoethoxy)phenyl)-4-( 1 -piperazinyl)benzamide, dihydrochloride as a white solid. mp:>250 0 C. 1H NMR (CD30D): d 2.27 3H), 3.29 (m, 1H), 3.30 6H), 3.57 4H), 4.24 j=5 Hz, 3H), 6.87 (dd, j=5,9 WO 98/0040 1 PCTIUS97/11047 68 Hz, IH), 6.94 j=5 Hz, 1H), 7.12 j=9 Hz, 2H), 7.22 j=9 Hz, 1H), 7.91 j=9 Hz, 2H).
WO 98/00401 WO 9800401PCT/US97/11047 69 EXAMPLE
BOC
2 0/ CHC1 3
OH
CH 3 BrCH 2 000Et 0S 2 00 3 DMF CH 3 O NH Me 2 NH neat 21 0
C
O OBut 5-1 C F 3
COOH
000 RT 0H 3 5-2 WO 98/00401 WO 9800401PCTIVS97/11047 70
OCH
3 Na 2
AIH
2
(OCH
2
CH
2 00H 3 2 THF, reflux 0H3
OH
3
~CH
3
NH
2 o0 N N
COOH
Bu t O PYCLU, i-Pr 2 NEt, OH 2 01 2
N-OH
3
OH
3 HOI EtOAc
F-N
N-CH
3 6H,
H
3 0 WO 98/00401 PCT/US97/11047 -71 O COOEt
CH
3
NH
O0 OBu t 5-1 Ethyl 4-(1,1-dimethylethoxycarbonyl)amino-3-methylphenoxyacetate (5-1) To a 200 mL round bottomed flask with a stirring bar, and an argon inlet was added 4-(1,1-dimethylethoxycarbonyl)amino-3methylphenol (5.00 g, 22.39 mmol), Cs2CO3 (14.59 g, 44.78 mmol), DMF (50 mL), and ethyl bromoacetate (2.61 mL, 23.51 mmol). This mixture was stirred vigorously at ambient temperature for 24 h. The mixture was filtered through a frit and the DMF was removed under high vacuum. The residue was dissolved in EtOAc (300 mL) and washed with H20 (2x) and brine Drying (MgSO4), filtration, and removal of the solvent in vacuo gave a solid. This material was triturated with 5% Et20-hexane, the solid was collected by filtration and dried in vacuo to give 5.40 g of ethyl 4-(1,1-dimethylethoxycarbonyl)amino-3-methylphenoxyacetate as a white, crystalline solid.
1H NMR (CDC13): d 1.29 j=7.2Hz, 3H), 1.51 9H), 2.22 3H), 4.26 j=7.2Hz, 2H), 4.57 2H), 6.08 (br s, 1H), 6.72 2H), 7.56 1H).
WO 98/00401 PCT/US97/11047 -72 CH3 CH3
NH
O0 OBu 5-2 N,N-Dimethyl 4-(1,1-dimethylethoxycarbonyl)amino-3methylphenoxvacetamide (5-2) To a 100 mL pressure vessel with a stirring bar was added ethyl 4-(1,1 -dimethylethoxycarbonyl)amino-3-methylphenoxyacetate (1.00 g, 3.24 mmol). The vessel was cooled in a Dry Ice® 2-propanol bath to -78C and dimethylamine was condensed onto the solid to a final volume of -30 mL. The reaction vessel was sealed, allowed to warm to ambient temperature and stirred for 13 days. The excess dimethylamine was vented and the residue was dissolved in CHC13 and concentrated, twice to remove the last traces of dimethylamine. The crude product was chromatographed on 75 g of silica gel using 75/25 EtOAC-hexane as eluant to provide 1 g (100% yield) N,N-dimethyl-4-(1,1dimethylethoxycarbonyl)amino-3-methylphenoxyacetamide as a white, crystalline solid. 1H NMR (CDC13): d 1.50 9H), 2.22 3H), 2.96 3H), 3.07 3H), 4.63 2H), 6.09 (br s, 1H), 6.77, 2H), 7.52 (br s, 1H).
WO 98/00401 WO 9800401PCTIUS97/1 1047 73 9 H3 0 CH
~CH
3
NH
2 5-3 N.N-Dimethvl 4-amino-3 -methvlphenoxvacetamide (5-3) To a 100 mL round bottomed flask with a stirring bar and a drying tube was added N,N-dimethyl 4-(1 ,1 -dimethylethoxycarbonyl)amino-3-methylphenoxyacetamide (1.00 g, 3.24 mmol) and trifluoroacetic acid (20 mL). This solution was stirred at ambient temperature 48 h. The trifluoroacetic. acid was removed in vacuc and the residue was dissolved in 200 ml, of EtOAc. This solution was washed with NaHCO3 solution and brine. Drying (MgSO4), filtration and removal of the solvent in vacuo gave 0.493 g (73% yield) of N,Ndimethyl 4-amino-3-methylphenoxyacetamide as white crystals. IH NMR (CDC13): d 2.15 3H1), 2.97 3H), 3.08 3H), 3.27 (br s, 2H), 4.59 211), 6.71 (in, 3H1).
WO 98/00401 PCT/US97/11047 -74- CH3 O NCH3
CH
3
NH
2 5-4 N,N-Dimethvl-2-(4-amino-3-methylphenoxyvethylamine (5-4) To a 200 mL round bottomed flask with a stirring bar, reflux condenser and an argon inlet was added N,N-dimethyl-4-amino- 3-methylphenoxyacetamide (0.493 g, 2.37 mmol) and dry THF mL). To this solution was added a solution of sodium bis(2-methoxyethoxy)aluminium hydride (2.78 mL of a 3.4 M solution in toluene, 9.47 mmol). This solution was heated at reflux for 3h. The cooled reaction mixture was treated with saturated aqueous sodium potassium tartrate solution and extracted with EtOAc. The combined EtOAc extracts were dried (MgSO4), filtered and concentrated in vacuo. The crude N,N-dimethyl-2-(4-amino-3-methylphenoxy)ethylamine was used in the next step without further purification.
WO 98/00401 WO 9800401PC/US9711047 75 0 0 Buto H \/0f N-OH 3
CH
3
H
3
C
N-(2-Methyl-4-(2-(N,N-dimethylamino)ethoxy)phenyl)-4-(4-(1 ,l dimethvlethoxycarbonvl)piperazin-1I-yl)benzamide To a 100 mL round bottomed flask with a stirring bar and an argon inlet was added 1 -dimethylethoxycarbonyl)piperazin- 1-yl)benzoic acid (0.726 g, 2.37 mmol), N,N-dimethyl-2-(4-amino-3methylphenoxy)ethylamine (0.46 g, 2.37 mmol), chloro-N ,N,N t bis(pentamnethylene)formamidinium hexafluorophosphate (0.937 g, 2.60 mmol) and CH2Cl2 (30 mL). When all of the solids had dissolved dilsopropylethylamine (1.57 mL, 9.00 minol) was added. The resulting mixture was stirred at ambient temperature for 48 h. The mixture was diluted with CHC13 and washed with 10% aqueous citric acid, NaHCO3 solution and brine. Drying (MgSO4), filtration and removal of the solvent in vacuc gave a the crude product. This material was chromatographed on 75 g of silica gel using 2.5% 2-propanol in ammonia saturated CHC13 as eluant. There was obtaine d 0.45 g of a white solid. This material was tritutrated with Et2O and collected on a frit to give 0.300 g of N-(2-methyl-4-(2-(N,N-dimethylamnino)ethoxy)phenyl)-4-(4-(, 1, -dimethylethoxycarbonyl)piperazin- I1yl)benzamide as white crystals. I H NMR (CDCl3): d 1.46 9H), 2.28 3H), 2.69 6H), 2.2.71 j=6 Hz, 2H), 3.28 (in, 4H), 3.59 (in, 4H), 4.07 j=6 Hz, 2H), 6.76 2H), 6.93 j=9 Hz, 2H), 7.43 (s, IH), 7.66 (br s, IR), 7.78 j=9 Hz, 2H).
WO 98/00401 PCT/US97/11047 -76- HN N0 H 0 N-OH 3 N O CH 3
H
3
C
5-6 N-(2-Methyl-4-(2-(N,N-dimethyl)aminoethoxy)phenyl)-4-(1piperazinl)benzamide (5-6) To a 200 mL round bottomed flask with a stirring bar and a gas dispersion tube was added N-(2-methyl-4-(2-(N,N-dimethylamino)ethoxy)phenyl)-4-(4-( 1,1 -dimethylethoxycarbonyl)piperazin- 1yl)benzamide (0.295 g, 0.61 mmol) and 50 mL of dry EtOAc. This well stirred solution was cooled in an ice bath and saturated with HCI gas over 15 min. The mixture was aged 1 h at OoC and the excess HCI was then removed with a stream of argon. The EtOAc was removed in vacuo and the crude product was partitioned between EtOAc and NaHCO3 solution. The layers were separated, the organic phase was washed with brine and dried (MgSO4). Filtration, removal of the solvent in vacuo and recrystallization from EtOAc gave 0.145 g of N- (2-methyl-4-(2-(N,N-dimethyl)aminoethoxy)phenyl)-4-( 1piperazinyl)benzamide, as a white crystals. mp: 139-141C. 1H NMR (CDC13): d 1.66 (br s, 1H), 2.28 3H), 2.33 6H), 2.72 j=6 Hz, 2H), 3.05 4H), 3.27 4H), 4.05 j=6 Hz, 2H), 6.77 2H), 6.93 j=9 Hz, 2H), 7.45 IH), 7.63 j=9 Hz, 1H), 7.77 j=9 Hz, 2H).
WO 98/00401 WO 9800401PCTIUS97/11047 77 EXAMPLE 6
MN
2 neat 2100C
H
o-rN, OH 3 0 CF 3 000H
OH
3 0OC-> RT
NH
o J OBut oJ OBut 5-1
OH
3 0
OCH
3 Na 2
AIH
2
(OCH
2
CH
2 0CH 3 2 THF, ref lux
NH
2 6-2 WO 98100401 WO 98/040 1PCTIUS97/11047 78 Oy OButI
BOC
2 0 CHC1 3
NH
2
NH
2 PYCLU, i-Pr 2 NEt, CH 2 Ct 2
N-CH
3 0:kOBut
H
3
C
HCOI EtOAc .0
N-OH
3
H
6-6 WO 98/00401 PCT/US97/11047 -79-
H
I
O CH 3 O H 3
NH
O0 OBut 6-1 N-Methyl-4-(1,1 -dimethylethoxycarbonyl)amino-3-methylphenoxyacetamide (6-1) To a 50 mL glass pressure vessel with a stirring bar was placed 4-(1,1 -dimethylethoxycarbonyl)amino-3-methylphenoxyethylethoxycarbonyl (3.00 g, 9.7 mmol). The vessel was cooled to -78 0 C in a dry ice/actone bath. 25 mL of methylamine was condensed into the vessel and was sealed with a screw cap. Ice bath was removed and the mixture was stirred at ambient temperature 48 h. The excess methylamine was evaporated and the residue was dissolved in CHC13 then concentrated in vacuo to remove traces of methylamine.
Triturated the solid with Et20. Collected by filtration to give 2.65g of the title compound above as a white crystalline solid.
1 H NMR (CDC13): d 1.5 9H), 2.23 3H), 2.90 3H), 4.45 (s, 2H), 6.09 (br s, 1H), 6.55 (br s, IH), 6.72 IH), 6.74 1H), 7.57 (br s, 1H).
WO 98/00401 PCTIUS97/11047
NH
2 6-2 N-Methyl-2-(4-amino-3-methylphenoxv)acetamide (6-2) To a 100 mL round bottomed flask with a stirring bar and an argon inlet was added N-methyl-4-(1,1 -dimethylethoxycarbonyl)amino-3-methylphenoxyacetamide (2.5 g, 8.49 mmol) and trifluoroacetic acid (25 mL). This solution was stirred at 0°C initially for 30 min then was warmed to room temperature and stirred 18 h.
The trifluoroacetic acid was removed in vacuo and the residue was partitioned between EtOAc and aqueous NaHCO3 solution. The layers were separated and the organic phase was washed with water and brine, dried (MgS04), filtered and concentrated in vacuo to afford 1.43 g of the title compound above as a brown oil.
1H NMR (CDC13): d 2.15 3H), 2.90 3H), 4.41 2H), 6.61 (s, 2H), 6.66 1H).
WO 98/00401 PCT/US97/11047 -81
H
OJ N'CH 3
^CH
3
NH
2 6-3 2-(4-Amino-3-methylphenoxy) N-methylethylamine (6-3) To a 100 mL round bottomed flask with a stirring bar, refluxed condenser and an argon inlet was added N-methyl-2-(4-amino- 3-methylphenoxy)acetamide(1.43 g, 7.36 mmol) and dry THF (50 mL).
To this solution was added a solution of sodium bis(2methoxy)aluminum hydride (7.57 mL of a 3.4 M solution in toluene, 25.76 mmol). This suspension was heated to reflux for 3 h. The cooled solution was quenched with saturated aqueous sodium potassium tartrate solution and extracted with EtOAc Combined EtOAc layers were washed with water and brine. Dried (MgSO4), filtered and in vacuo to give a crude brown oil which was chromatographed on silica gel using 5% IPA/NH3 saturated CHC13 as eluant. Collected 0.61 g of a brown oil as the title compound above.
1 H NMR (CDC13): d 2.15 3H), 2.49 3H), 2.92 j=5 Hz, 2H), 3.35 (br s, 2H), 3.99 j=5 Hz, 2H), 6.62 2H), 6.67 1H).
WO 98/00401 WO 9800401PCT/US97/11047 82 0 yOBut N H
CH
3
NH
2 6-4 N-Methyl-N-(, 1, -dimethylethoxycarbonyl)-2-(4-amino-3 methvlphenoxv)ethylamine (6-4) To a 100 mL round bottomed flask with a stirring bar and argon inlet was added 2-(4-amino-3-methylphenoxy) Nmethylethylamine (.600 g, 3.33 mmol), CHC13 (35 mL) and di.-terbutyldicarbonate (.726 g, 3.33 mmol). The solution was stiffed at ambient for overnight. The solvent was removed in vacuc. The crude N-methyl-N-( 1,1 -dimethylethoxycarbonyl)-2-(4-amino-3methylphenoxy)ethylamine was used without further purification.
WO 98/00401 PCT/US97/11047 -83 0 7 0 tO N N O ButO H 0 N-CH 3 H 0 k Bu'
H
3 0 N-(2-Methyl-4-(2-(1,1 -dimethylethoxycarbonyl(methyl)amino)ethoxy)phenyl)-4-(4-( 1,1 -dimethylethoxycarbonyl)piperazin- 1-vl)benzamide To a 100 mL round bottomed flask with a stirring bar and an argon inlet was added 4-(4-(1,1-dimethylethoxycarbonyl)piperazine- 1-yl)benzoic acid (0.5 g, 1.63 mmol), N-methyl-N-(1,1-dimethylethoxycarbonyl)-2-(4-amino-3-methylphenoxy)ethylamine (0.37 g, 1.32 mmol), chloro-N,N,N',N'-bis(pentamethylene)formamidinium hexafluorophosphate (0.625 g, 1.73 mmol) and CH2Cl2 (30 mL). To the mixture solution was then added diisopropylethylamine (1.0 mL, 5.8 mmol). The resulting solution was stirring at room temperature for 24h. Removed solvent in vacuo. Partitioned the residue between EtOAc and 10% aqueous citric acid, NaHCO3 solution and brine. Dried (MgSO4), filtration and removal of solvent in vacuo gave a crude brown oil. This material was flash chromatographed on silica gel with NH3 saturated CHCl3 as eluant to afford 0.62 g of the title compound above.
1 H NMR (CDCl3): d 1.46 9H), 1.49 9H), 2.28 3H), 2.98 (m, 3H), 3.16 6H), 3.59 4H), 4.06 (br s, 2H), 6.76 2H), 6.93 (d, j=9 Hz, 2H), 7.43 1H), 7.65 j=9 Hz, 2H), 7.80 j=9 Hz, 2H).
WO 98/00401 PCT/US97/11047 -84- HN N N-CH 3 H H
H
3
C
6-6 N-(2-Methyl-4-(2-methylaminoethoxy)phenyl)-4-( 1-piperazinyl)benzamide (6-6) To a 100 mL round bottomed flask with a stirring bar and a gas dispersion tube was added N-(2-methyl-4-(2-(1,1-dimethylethoxycarbonyl(methyl)amino)ethoxy)phenyl)-4-( 4 -dimethylethoxycarbonyl)piperazin-l-yl)benzamide (0.620 g, 1.09 mmol) and mL of dry EtOAc. This well stirred solution was cooled to 0°C in an ice bath and was saturated with HC1 gas for over 15 min. The reaction was aged for an hour and excess HCI was removed with a stream of argon. Removal of EtOAc in vacuo and the crude HC1 salt was converting to freebase by dissolving in saturated NaHCO3 solution.
Extracted with EtOAc and the layers were separated. A white solid precipitated out from the aqueous and was collected via suction filtration. This crude product was recrystallized from hot to give 0.175 g of N-(2-methyl-4-(2-methylaminoethoxy)phenyl)-4-(1piperazinyl)benzamide as a white solid. mp: 136-137 0
C.
1H NMR (DMSO-d6): d 2.28 3H), 2.50 3H), 2.95 2H), 3.03 4H), 3.27 4H), 4.06 2H), 6.78 j=6 Hz, 2H), 6.93 j=9 Hz, 2H), 7.41 1H), 7.65 j=9 Hz, 1H), 7.79 j=9 Hz, 2H).
WO 98/00401 PCT/US97/11047 EXAMPLE 7 Tablet Preparation Tablets containing 25.0, 50.0, and 100.0 mg., respectively, of the active compound N-(2-Methyl-4-(2-(N,Ndimethyl)aminoethoxy)- phenyl)-4-(l-piperazinyl)benzamide are prepared as illustrated below: TABLE FOR DOSES CONTAINING FROM 25-100MG OF THE ACTIVE COMPOUND Amount-mg Active Compound 25.0 50.0 100.0 Microcrystalline cellulose 37.25 100.0 200.0 Modified food corn starch 37.25 4.25 Magnesium stearate 0.50 0.75 All of the active compound, cellulose, and a portion of the corn starch are mixed and granulated to 10% corn starch paste. The resulting granulation is sieved, dried and blended with the remainder of the corn starch and the magnesium stearate. The resulting granulation is then compressed into tablets containing 25.0, 50.0, and 100.0 mg, respectively, of active ingredient per tablet.
WO 98/00401 PCT/US97/11047 -86- EXAMPLE 8 Intravenous formulations An intravenous dosage form of the above-indicated active compound is prepared as follows: Active Compound Sodium Citrate Citric Acid Sodium Chloride Water for Injection (USP) 0.5-10.0mg 5-50mg 1-15mg 1-8mg q.s. to 1 L Utilizing the above quantities, the active compound is dissolved at room temperature in a previously prepared solution of sodium chloride, citric acid, and sodium citrate in Water for Injection (USP, see page 1636 of United States Pharmacopeia/National Formulary for 1995, published by United States Pharmacopeial Convention, Inc., Rockville, Maryland, copyright 1994.

Claims (28)

1. A compound having the formula 0 11 X-W -Y-(CH -NH-(A)r B or pharmaceutically acceptable salt(s) thereof, wherein u is 0orl1; X is H 2 N HN Nf N Nor HN H 2 W is wherein q isO0 or 2; Y is N* N 00* 0 1000 or X and Y, provided that q=O, together form HNH A is CH 3 r is 0ori1; B is -O(CH 22 NH 2 -CH 2 C(OPh)HCH2NH2, or -CH(CH 3 )(CH 2 2 NH2. [R:\LDAA]07896.dc:tab
2. A compound of claim 1 selected from the group consisting of O NH 2 O NH 2 *4 0 9 @0 0 6 *9 9 9 @0 *0 @9 0 9 0 NH 2 H 3 C NH 2 CH 3 N NH 2 H or pharmaceutically acceptable salt(s) thereof.
3. A fibrinogen receptor antagonist prodrug, substantially as hereinbefore described with reference to any one of the examples.
4. A composition comprising a compound of any one of claims 1 to 3 and a pharmaceutically acceptable carrier.
5. A composition of claim 4, further comprising an efficacious amount of one or more egents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent. [R:\LIBAA]07896.doc:tab 89
6. A method for inhibiting the binding of fibrinogen to blood platelets in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3, or of a composition according to claim 4.
7. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in inhibiting the binding of fibrinogen to blood platelets.
8. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for inhibiting the binding of fibrinogen to blood platelets.
9. A method for inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at its receptor site in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3, or of a composition according to claim 4 or claim A compound according to any one of claims 1 to 3 or a composition according to claim 4 or claim 5 when used in inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at its receptor site.
11. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at tis receptor site.
12. A method for inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at its receptor site in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent, or of a composition according to claim
13. A compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent or a 25 composition according to claim 5 when used in inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at its receptor site.
14. The use of a compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent for the manufacture of a medicament for inhibiting the aggregation of blood platelets by blocking fibrinogen from acting at its receptor site. A method for inhibiting the binding of fibrinogen to blood platelets by blocking fibrinogen from acting at its receptor site in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and A51.an antiplatelet agent, or of a composition according to claim [R:\LIBAA]07896.doc:tab
16. A compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent or a composition according to claim 5 when used in inhibiting the binding of fibrinogen to blood platelets by blocking fibrinogen from acting at its receptor site.
17. The use of a compound according to any one of claims 1 to 3 in combination with one or more agents selected from a thrombolytic agent, an anticoagulant agent, and an antiplatelet agent for the manufacture of a medicament for inhibiting the binding of fibrinogen to blood platelets by blocking fibrinogen from acting at its receptor site.
18. A method for inhibiting osteoclast mediated bone resorption in a mammal, which method io includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3, or of a composition according to claim 4.
19. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in inhibiting osteoclast mediated bone resorption. The use of a compound according to any one of claims 1 to 3 for the manufacture of a S 15 medicament for inhibiting osteoclast mediated bone resorption.
21. A method for inhibiting angiogenesis in a mammal, which method includes or consists of SS administering to said mammal an effective amount of at least one compound according to any one of Sclaims 1 to 3, or of a composition according to claim 4.
22. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in inhibiting angiogenesis. S. 23. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for inhibiting angiogenesis •o*S
24. A method for inhibiting tumour growth in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one 25 of claims 1 to 3 or of a composition according to claim 4. A compound according to any one of claims 1 to 3 or a composition according to claim 7 when used in inhibiting tumour growth.
26. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for inhibiting tumour growth.
27. A method for preventing platelet thrombosis in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3 or of a composition according to claim 4.
28. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in preventing platelet thrombosis.
29. The use of a compound according to any one of claims 1 to 3 for the manufacture of a edicament for preventing platelet thrombosis. [R:\LIBAA]07896.doc:tab 91 A method for preventing thromboembolism in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3 or of a composition according to claim 4.
31. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in preventing thromboembolism.
32. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for preventing thromboembolism.
33. A method for preventing reocclusion in a mammal, which method includes or consists of administering to said mammal an effective amount of at least one compound according to any one of claims 1 to 3 or of a composition according to claim 4.
34. A compound according to any one of claims 1 to 3 or a composition according to claim 4 when used in preventing reocclusion. The use of a compound according to any one of claims 1 to 3 for the manufacture of a medicament for preventing reocclusion. 15 Dated 1 March, 2000 Merck Co., Inc. Patent Attorneys for the Applicant/Nominated Person SPRUSON FERGUSON o* S o [R\LIBAA]07896.doc:tab
AU35037/97A 1996-06-28 1997-06-25 Fibrinogen receptor antagonist prodrugs Ceased AU719102B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US2087796P 1996-06-28 1996-06-28
US60/020877 1996-06-28
GBGB9617899.1A GB9617899D0 (en) 1996-08-28 1996-08-28 Fibrinogen receptor antagonist prodrugs
GB9617899 1996-08-28
PCT/US1997/011047 WO1998000401A1 (en) 1996-06-28 1997-06-25 Fibrinogen receptor antagonist prodrugs

Publications (2)

Publication Number Publication Date
AU3503797A AU3503797A (en) 1998-01-21
AU719102B2 true AU719102B2 (en) 2000-05-04

Family

ID=26309930

Family Applications (1)

Application Number Title Priority Date Filing Date
AU35037/97A Ceased AU719102B2 (en) 1996-06-28 1997-06-25 Fibrinogen receptor antagonist prodrugs

Country Status (5)

Country Link
EP (1) EP0912513A1 (en)
JP (1) JP2000513375A (en)
AU (1) AU719102B2 (en)
CA (1) CA2257950A1 (en)
WO (1) WO1998000401A1 (en)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU721130B2 (en) * 1996-06-28 2000-06-22 Merck & Co., Inc. Fibrinogen receptor antagonists
WO2001052549A1 (en) 2000-01-14 2001-07-19 Koninklijke Philips Electronics N.V. Transcoding method and device
HUP0304058A2 (en) 2001-01-30 2004-04-28 Bristol-Myers Squibb Company Sulfonamide lactam inhibitors of fxa and their use and pharmaceutical compositions containing them
GB0112836D0 (en) 2001-05-25 2001-07-18 Smithkline Beecham Plc Medicaments
GB0421908D0 (en) 2004-10-01 2004-11-03 Angeletti P Ist Richerche Bio New uses
CN101296914B (en) 2005-08-26 2012-07-18 盐野义制药株式会社 Derivative having PPAR agonistic activity
JP5501251B2 (en) 2008-01-11 2014-05-21 アルバニー モレキュラー リサーチ, インコーポレイテッド (1-Azinone) substituted pyridoindoles as MCH antagonists
US8273900B2 (en) 2008-08-07 2012-09-25 Novartis Ag Organic compounds
US8629158B2 (en) 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
WO2011003021A1 (en) 2009-07-01 2011-01-06 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine mch-1 antagonists, methods of making, and use thereof
WO2011003012A1 (en) 2009-07-01 2011-01-06 Albany Molecular Research, Inc. Azinone-substituted azapolycycle mch-1 antagonists, methods of making, and use thereof
EP2448585B1 (en) 2009-07-01 2014-01-01 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine mch-1 antagonists, methods of making, and use thereof
WO2012088038A2 (en) 2010-12-21 2012-06-28 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline mch-1 antagonists, methods of making, and uses thereof
US8993765B2 (en) 2010-12-21 2015-03-31 Albany Molecular Research, Inc. Tetrahydro-azacarboline MCH-1 antagonists, methods of making, and uses thereof
GB201106817D0 (en) 2011-04-21 2011-06-01 Astex Therapeutics Ltd New compound
EA026907B1 (en) 2012-03-16 2017-05-31 Вайтаи Фармасьютиклз, Инк. Liver x receptor modulators
RS55532B1 (en) 2012-03-16 2017-05-31 Vitae Pharmaceuticals Inc Liver x receptor modulators
US9980973B2 (en) 2012-10-19 2018-05-29 Astex Therapeutics Limited Bicyclic heterocycle compounds and their uses in therapy
GB201218862D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
GB201218864D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
GB201218850D0 (en) 2012-10-19 2012-12-05 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
MY186311A (en) 2013-12-20 2021-07-08 Astex Therapeutics Ltd Bicyclic heterocycle compounds and their uses in therapy
EP3087069B1 (en) 2013-12-23 2019-01-30 Norgine B.V. Compounds useful as ccr9 modulators
AU2016315720B2 (en) 2015-09-02 2021-02-25 Trevena, Inc. 6-membered AZA-heterocyclic containing delta-opioid receptor modulating compounds, methods of using and making the same
WO2017100593A1 (en) 2015-12-09 2017-06-15 Luc Therapeutics Thienopyrimidinone nmda receptor modulators and uses thereof
LT3386591T (en) 2015-12-09 2020-10-12 Cadent Therapeutics, Inc. Heteroaromatic nmda receptor modulators and uses thereof
CA3047403A1 (en) 2016-12-22 2018-06-28 Cadent Therapeutics, Inc. Nmda receptor modulators and uses thereof
CN110446495B (en) 2017-02-17 2023-09-05 特维娜有限公司 Delta opioid receptor modulating compounds containing 7-membered aza heterocycles and methods of use and preparation thereof
AU2018221148B2 (en) 2017-02-17 2022-05-05 Trevena, Inc. 5-membered aza-heterocyclic containing delta-opioid receptor modulating compounds, methods of using and making the same
HUE062566T2 (en) 2018-08-03 2023-11-28 Novartis Ag Heteroaromatic nmda receptor modulators and uses thereof
US11091447B2 (en) 2020-01-03 2021-08-17 Berg Llc UBE2K modulators and methods for their use

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2276163A (en) * 1993-03-17 1994-09-21 Glaxo Group Ltd Pyridine compounds.
AU3936595A (en) * 1994-11-28 1996-06-19 Taisho Pharmaceutical Co., Ltd. Pyridine derivative

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994012181A1 (en) * 1992-12-01 1994-06-09 Merck & Co., Inc. Fibrinogen receptor antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2276163A (en) * 1993-03-17 1994-09-21 Glaxo Group Ltd Pyridine compounds.
AU3936595A (en) * 1994-11-28 1996-06-19 Taisho Pharmaceutical Co., Ltd. Pyridine derivative

Also Published As

Publication number Publication date
EP0912513A1 (en) 1999-05-06
AU3503797A (en) 1998-01-21
JP2000513375A (en) 2000-10-10
WO1998000401A1 (en) 1998-01-08
CA2257950A1 (en) 1998-01-08

Similar Documents

Publication Publication Date Title
AU719102B2 (en) Fibrinogen receptor antagonist prodrugs
AU721130B2 (en) Fibrinogen receptor antagonists
AU699167B2 (en) Fibrinogen receptor antagonists
AU713676B2 (en) Integrin receptor antagonists
US5932582A (en) Fibrinogen receptor antagonist prodrugs
AU709631B2 (en) Fibrinogen receptor antagonist prodrugs
AU747293B2 (en) Fibrinogen receptor antagonist prodrugs
US5990107A (en) Fibrinogen receptor antagonist prodrugs
AU689961B2 (en) Fibrinogen receptor antagonists
US5952306A (en) Integrin receptor antagonists
AU742197B2 (en) Fibrinogen receptor antagonists
US5981584A (en) Fibrinogen receptor antagonist prodrugs
US5789421A (en) Fibrinogen receptor antagonist
US6117842A (en) Fibrinogen receptor antagonists
AU719495B2 (en) Fibrinogen receptor antagonists
US5945545A (en) Fibrinogen receptor antagonists
AU702025B2 (en) Fibrinogen receptor antagonist

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired