CA2420179A1 - Composition and method for inhibiting platelet aggregation - Google Patents

Composition and method for inhibiting platelet aggregation Download PDF

Info

Publication number
CA2420179A1
CA2420179A1 CA002420179A CA2420179A CA2420179A1 CA 2420179 A1 CA2420179 A1 CA 2420179A1 CA 002420179 A CA002420179 A CA 002420179A CA 2420179 A CA2420179 A CA 2420179A CA 2420179 A1 CA2420179 A1 CA 2420179A1
Authority
CA
Canada
Prior art keywords
substituted
formula
alkyl
aralkyl
aryl
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.)
Abandoned
Application number
CA002420179A
Other languages
French (fr)
Inventor
Jose L. Boyer
Gillian M. Olins
Benjamin R. Yerxa
James G. Douglass
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.)
Inspire Pharmaceuticals Inc
University of North Carolina System
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2420179A1 publication Critical patent/CA2420179A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/12Antidiuretics, e.g. drugs for diabetes insipidus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/207Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids the phosphoric or polyphosphoric acids being esterified by a further hydroxylic compound, e.g. flavine adenine dinucleotide or nicotinamide-adenine dinucleotide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids

Abstract

This invention is directed to a method of preventing or treating diseases or conditions associated with platelet aggregation. The method is also directed to a method of treating thrombosis. The method comprises administering to a subject a pharmaceutical composition comprising a therapeutic effective amount of P2Y12 receptor antagonist compound, wherein said amount is effective to bind the P2Y12 receptors on platelets and inhibit ADP-induced platelet aggregation. The P2Y12 receptor antagonist compounds useful for this invention include mononucleoside polyphosphates and dinucleoside polyphosphates of general Formula (I), or salts thereof. The present invention also provides novel compositions comprising mononucleoside polyphosphates and dinucleoside polyphosphates according to Formula (Ia) and (Ib).

Description

COMPOSITION AND METHOD FOR INHIBITING PLATELET AGGREGATION
TECHNICAL FIELD
This invention relates to compounds of mono and dinucleoside polyphosphates and the method of using such compounds in the prevention or treatment of diseases or conditions associated with platelet aggregation, including thrombosis in humans and other mammals.
BACKGROUND OF THE INVENTION
Hemostasis is the spontaneous process of stopping bleeding from damaged blood A 10 vessels. Precapillary vessels contract immediately when cut; within seconds, thrombocytes, or blood platelets, are bound to the exposed matrix of the injured vessel by a process called platelet adhesion. Platelets also stick to each other in a phenomenon known as platelet aggregation to form a platelet plug to stop bleeding quickly.
An intravascular thrombus results from a pathological disturbance of hemostasis.
Platelet adhesion and aggregation are critical events in intravascular thrombosis. Activated under conditions of turbulent blood flow in diseased vessels or by the release of mediators from other circulating cells and damaged endothelial cells lining the vessel, platelets accumulate at a site of vessel injury and recruit further platelets into the developing thrombus.
The thrombus can grow to sufficient size to block off arterial blood vessels.
Thrombi can also form in areas of stasis or slow blood flow in veins. Venous thrombi can easily detach portions of themselves called emboli that travel through the circulatory system and can result in blockade of other vessels, such as pulmonary arteries. Thus, arterial thrombi cause serious disease by local bloclcade, whereas venous thrombi do so primarily by distant blockade, or embolization. These conditions include venous thrombosis, thrombophlebitis, arterial embolism, coronary and cerebral arterial thrombosis, unstable angina, myocardial infarction, stroke, cerebral embolism, kidney embolisms and pulmonary embolisms.
A number of converging pathways lead to platelet aggregation. Whatever the initial stimulus, the final common event is crosslinlcing of platelets by binding fibrinogen to a membrane binding site, glycoprotein IIb/IIIa (GPIIbIDIa). Compounds that are antagonists for GPIIb/Illa receptor complex have been shown to inhibit platelet aggregation (U.S. Patent Nos.
6,037,343 and 6,040,317). Antibodies against GPIlblIIIa have also been shown to have high antiplatelet efficacy (The EPIC investigators, New Engl. J. Med. (1994) 330:956-961).
However, this class of antiplatelet agents sometimes causes bleeding problems.

Thrombin can produce platelet aggregation largely independently of other pathways but substantial quantities of thrombin are unlikely to be present without prior activation of platelets by other mechanisms. Thrombin inhibitors such as hirudin are highly effective antithrombotic agents. However, functioning as both antiplatelet and anti-coagulant agents, thrombin inhibitors again may produce excessive bleeding. (The TIMI 9a investigators, The GUSTO Iia investigators, Circulation, 90: 1624-1630 (1994); Circulation, 90:

(1994); Neuhaus K. L. et al., Circulation, 90: 1638-1642 (1994)) Various antiplatelet agents have been studied for many years as potential targets for inhibiting thrombus formation. Some agents such as aspirin and dipyridamole have come into use as prophylactic antithrombotic agents, and others have been the subjects of clinical investigations. To date, the powerful agents such as disintegrins, and the thienopyridines ticlopidine and clopidogrel have been shown to have substantial side effects, while agents such as aspirin have useful but limited effectiveness (Hass, et al., N. Eragl.
J. Med., 321:501-507 (1989); Weber, et al., Am. J. Car~diol. 66:1461-1468 (1990); Lekstrom and Bell, Medicine 70:161-177 (1991)). In particular, use of the thienopyridines in antiplatelet therapy has been shown to increase the incidence of potentially life threatening thrombotic thrombocytopenic purpura (Bennett, C.L. et al. N. Engl. J. Med, (2000) 342:
1771-1777).
Aspirin, which has a beneficial effect on platelet aggregation (Br. Med. J.
(1994) 308: 81-106;
159-168), acts by inducing blockade of prostaglandin synthesis. Aspirin has no effect on ADP-induced platelet aggregation, and thus has limited effectiveness on platelet aggregation.
Furthermore, its well documented high incidence of gastric side effects limits its usefulness in many patients. Clinical efficacy of some newer drugs, such as ReoPro (7E3), is impressive, but recent trials have found that these approaches are associated with an increased risk of major bleeding, sometimes necessitating blood transfusion (New Eragl. .I. Med.
(1994) 330:956-961). Thus it appears that the ideal "benefit/risk" ratio has not been achieved.
Recent studies have suggested that adenosine 5'-diphosphate (ADP), a common agonist, plays a key role in the initiation and progression of arterial thrombus formation (Bernat, et al., Thf~omb. Haenaostas. (1993) 70:812-826); Maffrand, et al., ThYOrnb.
' Haefraostas. (1988) 59:225-230; Herbert, et al., At°terioscl.
Thr~omb. (1993) 13:1171-1179).
ADP induces platelet aggregation, shape change, secretion, influx and intracellular mobilization of Ca+2, and inhibition of adenylyl cyclase. Binding of ADP to platelet receptors is required for elicitation of the ADP-induced platelet responses. There are at least three P2 receptors expressed in human platelets: a cation channel receptor P2X1, a G
protein-coupled receptor P2Yi, and a G protein-coupled receptor P2Yla (also referred to as P2Ya~ and P2T ).
The P2X1 receptor is responsible for rapid calcium influx and is activated by ATP and by ADP. However, its direct role in the process of platelet aggregation is unclear. The P2Y1 receptor is responsible for calcium mobilization, shape change and the initiation of aggregation. P2Yla receptor is responsible for inhibition of adenylyl cyclase and is required for full aggregation. (Hourani, et al., The Platelet ADP Receptors Meeting, La Thuile, Italy, March 29-31, 2000) Ingall et al. (J. pled. Chem. 42: 213-220, (1999)) describe a dose-related inhibition of ADP-induced platelet aggregation by analogues of adenosine triphosphate (ATP), which is a weak, nonselective but competitive P2Y12 receptor antagonist. Zamecnik (USPN
5,049,550) discloses a method for inhibiting platelet aggregation in a mammal by administering to said mammal a diadenosine tetraphosphate compound of App(CHa)ppA or its analogs.
Kim et al.
(LTSPN 5,681,823) disclose Pi, P4-dithio-P2, P3-monochloromethylene 5', 5"' diadenosine P1, P4-tetraphosphate as an antithrombotic agent. The thienopyridines ticlopidine and clopidogrel, which are metabolized to antagonists of the platelet P2Y12 receptor, are shown to inhibit platelet function in vivo (Quinn and Fitzgerald, Cir~culatiofz 100:1667-1672 (1999);
Geiger, et al., ArteYioscley~. Th~omb. vast. Biol. 19:2007-2011 (1999)).
There is a need in the area of cardiovascular and cerebrovascular therapeutics fox an agent that can be used in the prevention and treatment of thrombi, with minimal side effects, such as unwanted prolongation of bleeding, while preventing or treating target thrombi.
SUMMARY OF THE INVENTION
This invention is directed to a method of preventing or treating diseases or conditions associated with platelet aggregation. The method is also directed to a method of treating thrombosis. The method comprises administering to a subject a pharmaceutical composition comprising a therapeutic effective amount of P2Y12 receptor antagonist compound, wherein said amount is effective to bind the P2Y12 receptors on platelets and inhibit ADP-induced platelet aggregation.
The P2Y12 receptor antagonist compounds useful for this invention include compounds of general Formula I, or salts thereof:

Formula I

A-O P X3 X1-P O D1 B' OM O
.s' S
Z' Y' wherein:
Xl, X2, and X3 are independently oxygen, methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido; .
T1,T2,W, and V are independently oxygen or sulfur;
m= 0,1 or 2;
n=Oorl;
p= 0,1, or 2 ;
where the sum of m+n+p is from 1 to 5;
M = H or a pharmaceutically-acceptable inorganic or organic counterion;
D 1=O or CH2;
B' is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;
Y' = H, OH, or ORI;
Z' = H, OH, or OR2;
with the proviso that Y' and Z' are both not H, both not OH when A=M, and that at least one residue Rl and R2 is present when A--M;
A=M, or A is a nucleoside residue which is defined as:
D~
Y Z

and which is linked to the phosphate chain via the 5' position of the furanose or carbocycle;
wherein:
DZ =O or CH2;
Z = H, OH, or OR3;
Y = H, OH, or OR4;
with the proviso that Z and Y are both not H, and, when Y' and Z' are H or OH, at least one residue R3 or R4 is present;
B is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;
Rl, RZ, R3, and/or Rq are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to Formula III, such that from one to four independent residues of Rl, R2, R3 and R4 falling within the definition of Formula II are present or from one to two independent residues made up of Rl+RZ andlor R3+R4 are present;
The invention also provides novel pharmaceutical compositions comprising compounds of Formula Ia or Ib, which are highly selective antagonists of P2Ylz receptor on platelets. The invention further provides a method of preventing or treating diseases or conditions associated with platelet aggregation; such diseases include venous thrombosis, thrombophlebitis, arterial embolism, coronary and cerebral arterial thrombosis, unstable angina, myocardial infarction, stroke, cerebral embolism, kidney embolisms and pulmonary embolisms.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 shows the effect of inhibition of ADP-induced aggregation by different compounds.
DETAILED DESCRIPTION OF THE INVENTION
This invention is provides a method of preventing or treating diseases or conditions associated with platelet aggregation. The method also provides a method of treating thrombosis. The method comprises administering to a subject a pharmaceutical composition comprising a therapeutic effective amount of P2Y12 receptor antagonist compound, wherein said amount is effective to bind the P2Y12 receptors on platelets and inhibit ADP-induced platelet aggregation.The P2Y12 receptor antagonist compounds useful for this invention include compound of general Formula I and salts thereof Formula I
T2 W V T~
A-O P X3 P X2 P X~-P O p~ B' OM OM OM O
Z' Y' wherein:
Xl, X2, and X3 are independently oxygen, methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;
Tl, T2,W, and V are independently oxygen or sulfur;
m= 0, 1 or 2;
n=Oorl;
p= 0, 1, or 2 ;
where the sum of m+n+p is from 1 to 5;
M =H, or a pharmaceutically-acceptable inorganic or organic counterion;
D1 =O or CH2 B' is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;
Y' =H, OH, or ORI;
Z' =H, OH, or ORZ; with the proviso that at least one of Y' and Z' is ORl or OR2;
Rl and RZ are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to Formula III, Formula lI
/Rs O ~ Rs R~
wherein:
O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;
R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ether; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II is an acyclic acetal or ketal; or RS and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ester or thioester; or RS and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to Formula II is a carbamate or thiocarbamate; or RS and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to Formula II is a carbonate or thiocarbonate; or R7 is not present and RS and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;

Formula III
-O~C~Rs S ~'--O~ ERs wherein:
O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by a common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester;
for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, preferably 3 to 6 atoms;
for cyclical orthoesters, R~ is hydrogen, alkyl, cycloalleyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R 9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy.
When present, the alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl and substituted aryl components of RS to R9may be generally defined as , but are not limited to, the following:
alkyl groups from 1 to 12 carbons, either straight chained or branched, with or without unsaturation and with or without heteroatoms, more preferably from 2 to 8 carbons, and most preferably 2 to 6 carbons;
cycloalkyl groups from 3 to 12 carbons, more preferably from 3 to 10 carbons, and most preferably 3 to 8 carbons, with or without unsaturation, and with or without heteroatoms;
aralkyl groups from 1 to 8 carbons in the alkyl portion, more preferably from 1 to 6 carbons and most preferably 1 to 4 carbons, and are monocyclic or polycyclic moieties from 4 to 8 carbons per ring in the aryl portion , more preferably from 4 to 7 carbons, and most preferably 5 to 6 carbons per ring, with or without heteroatoms;

aryl groups, either monocyclic or polycyclic, from 4 to ~ caxbons per ring, more preferably from 4 to 7 carbons, and most preferably 5 to 6 carbons per ring, with or without heteroatoms; and these groups may or may not bear substituents.
Preferred substituents on the foregoing groups may be, but are not limited to, hydroxy, nitro, methoxy, fluoro, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, thioalkyl, alkoxy, carboxyl, cyano, amino, substituted amino, trifluoromethyl, phenyl, cyclopropyl, cyclopentyl, and cyclohexyl; and preferred heteroatoms are oxygen, nitrogen, acid sulfur.
One embodiment of the invention is that A= M, wherein M =H or a pharmaceutically acceptable inorganic or organic counterion. In such embodiment, the compound is nucleoside diphosphate, nucleoside triphosphate, nucleoside tetraphosphate, nucleoside pentaphosphate, and nucleoside hexaphosphate with the 2'- and/or 3' position of the furanose or carbocycle modified. Most preferred are nucleotide diphosphates, nucleotide triphosphates, and nucleotide tetraphosphates. When T2, W, V, or Tl axe sulfur, the preferred position for this atom is on the terminal phosphorous of the polyphosphate chain (i.e. the phosphorous furthest removed from the nucleoside residue).
Another embodiment of the invention is that A is a nucleoside residue defined as:
B
Y Z
and linked to the phosphate chain via the 5' position of the furanose or carbocycle (dinucleoside polyphosphate with at least one of the 2, 3, 2' and 3' position of the furanose or carbocycle modified);
wherein:
DZ =O or CHI;
B is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;
Z =H, OH, or OR3;
Y =H, OH, or OR4;
R3, and/or R4 axe residues which axe linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to Formula IfI.
When T2, W, V, and/or Tl are sulfur, the preferred positions for this atom are Tl and T2.
S Further provisions are that when D1 and D2 are oxygen, the furanose is preferably in the (3-configuration;and that the furanose is most preferably in the (3-D-configuration.
Preferred compounds of general Formula I are molecules whose structures fall within the definitions of Formula Ia and Formula Ib:
Formula Ia B
T2 W V T~
H H p p ~(3 P X2 P X~-P O
I I I I
OM OM O OM

m n p wherein:
D1=O or CHZ;
D2 =O or CHZ;
B and B' are independently purine or pyrimidine residues according to general formula IV orV;
m and p= 0,1 or 2; n= 0 or l; such that the sum of m+n+p is from 1 to 5, preferably 1 to 4, and most preferably 1 to 3;
Xl, Xa, and X3= are independently O, NH, CHZ, CHF, CHCI, CF2, CC12;
T1, T2, V, and W are independently O or S;
M= H+, NH4+, Nab or other pharmaceutically-acceptable inorganic or organic counter ion;
Y'= H, OH, or ORI;
Z'= OH or OR2;
Z= OH or OR3;
Y= H, OH, or ORø, where Rl, R2, R3 and R4 falls under the definition of general formula II or III, provided that at least one of Y', Z', Z and Y is ORI, OR2, OR3, or OR4_ Preferred compounds of Formula Ia include:
D1 =O or CHa;
DZ =O or CH2;
Xl, Xza and X3=O;
Tl, Ta, V, and W= O; or Dl =O or CH2;
DZ =O or CHZ;
Xl and X3=O;
X2= methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;
T, Tl, T2, V, and W= O; or D1=O or CH2;
D2 =O or CHz;
m, n, and p= 1 ;or Xl and X3=O;
X2= methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;
Tl and Ta= S;
V and W=O.
Formula Ib B~
W V T~
AO P X2 P X~-P O
OM O OM
n Di = O or CHI;
n and p = 0, l, or 2 such that the sum of n+p is from 0 to 3;

A = M; wherein M= H+, NH4+, Na:'~ or other pharmaceutically-acceptable inorganic or organic counterion;
B' is purine or pyrimidine residue according to general Formulae IV and V;
Xl and XZ are independently O, NH, CHa, CHF, CHCI, CF2, CCl2;
Tl, V, and W are independently O or S;
Y'= H, OH, or ORI, Z'= H, OH or ORZ, where Rl and R2 fall under the definitions of general Formulae II
or III; with the proviso that at least ane of Y' and Z' is ORl or OR2, respectively.
Preferred compounds of Formula Ib include:
D1 = O or CHZ;
n and p = 0, 1, or 2 such that the sum of n+p is from 0 to 3, preferably 1 to 2;
Xl and Xa= O;
Tl, V, and W= O; or D1 = O or CH2;
Xl and X2= O;
Tl and V= O;
W=S; or D 1= O or CHI;
p = 0, 1, or 2 such that the sum of n+p is from 1 to 3; n=1;
Xl =O;
X2= methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;
Tl, V, and W= O;
Y'= H, OH, or ORI;
Z'= H, OH or ORZ, where Rl and R2 falls under the definition of general Formula II or III; with the proviso that at least one of Y' and Z' is ORl or ORZ, respectively.
B and B' are independently a purine residue, as in Formula IV, linked through the 9-position, or a pyrimidine residue, as in Formula V, linked through the 1-position. The ribosyl moieties are in the D- configuration, as shown, but may be L-, or D-and L-. The D-configuration is preferred.
Formula IV
,Rlo ., /R11 R~s 5 Formula V

R1g 4,'\ N /R15 N O

wherein:
10 Rlo and R14 are hydroxy, oxo, amino, mercapto, allcylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or Rln and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or 15 when Rlo in a purine or R14 in a pyrimidine has as its first atom nitrogen, Rlo and Rl l or R14 and Rls are taken together to form a 5-membered fused imidazole ring (etheno compounds), optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for RS-R~ above;

J is carbon or nitrogen, with the provision that when nitrogen, Rla is not present;
Rll is hydrogen, O (adenine 1-oxide derivatives) or is absent (adenine derivatives);
Rls is hydrogen, or acyl (e.g. acetyl, benzoyl, phenylacyl, with or without substituents);
R12 is hydrogen, alkyl, bromo, azido, allcylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or w-A(C1_6alkyl)B-, wherein A and B
are independently amino, mercapto, hydroxy or carboxyl;
R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation;
R1G is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl.
Compounds according to Formulae IV and V where Rlo or R14 is acylamino for the most part fall within the scope of Formula VI:
Formula VI
W
-N C/
H
wherein:
NH is the amino residue at the C-6 position in a purine or the amino residue at the C-4 position in a pyrimidine;
C is a carbon atom;
W is oxygen or sulfur;
R17 is amino or mono- or disubstituted amino such that the moiety according to Formula VI is a urea or thiourea; orRl7 is alkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to Formula VI is a carbamate or thiocarbamate; or R17 is alkyl, cycloalkyl, aralkyl, or aryl, with or without substituents or heteroatoms, such that the moiety according to Formula VI is an amide; with definitions of alkyl, cycloalkyl, aralkyl, or aryl groups as previously defined for comparable groups in RS to R9.
The compounds of the present invention may be conveniently synthesized by those skilled in the art using well-known chemical procedures. Mononucloside mono-, di- and triphosphates may be obtained from commercial sources or may be synthesized from the nucleoside using a variety of phosphorylation reactions which may be found in the chemical literature. Symmetrical and unsymmetrical dinucleotide polyphosphates may be prepared by activation of a nucleoside mono-, di- or triphosphate with a coupling agent such as, but not limited to, dicyclohexylcarbodiimide or l, 1'-carbonyldiimidazole, followed by condensation with another nucleoside mono-, di-, or triphosphate, which may be the same or different as the activated moiety. Activation of nucleoside triphosphates with dicyclohexylcarbodiimide gives a cyclical trimetaphosphate as the activated species, which may be advantageously reacted with a variety of nucleophiles to install unique substituents on the terminal phosphate of a triphosphate.
The compounds of the present invention may be prepared by derivatization or substitution at the level of the nucleoside, followed by phosphorylation and condensation as previously described, or the reactions may be carried out directly on the preformed mono- or dinucleotides. In the general Formulae Ia and Ib, the substituents at Y', Z', Y, and Z may be esters, carbamates, or carbonates, which are generally described by Formula II. Esters may be readily prepared by reacting a hydroxyl group of the furanose in a nucleoside or nucleotide with an activated form of an appropriate organic acid, such as an acid halide or acid anyhydride in the presence of an organic or inorganic base. Alternately, use of a suitable coupling reagent such as dicyclohexylcarbodiimide, 1,l'- carbonyldiimidazole and the like to activate the organic acid may be used to achieve the same result.
Carbamates or thiocarbamates may be most conveniently prepared by reaction of a hydroxyl group of the furanose in a nucleoside or nucleotide with any of a number of commercially available isocyanates or isothiocyanates, respectively, in an inert solvent.
Alternately, when a desired isocyanate or isothiocyanate cannot be obtained from commercial sources, they may be prepared from the corresponding amine by the use of phosgene or thiophosgene, respectively, or their chemical equivalents. Carbonates or thiocarbonates may be synthesized by reacting the hydroxyl groups of a furanose in a nucleoside or nucleotide with an appropriate haloformate in the presence of an organic or inorganic base.
In the general Formulae Ia and Ib, the substituents at Y'and Z', and Y and Z, when taken together, may be taken to mean acetals, lcetals or orthoesters, as described in Formula III. Acetals and ketals may be readily prepared by reaction of the neighboring 2' and 3' hydroxyl groups of the furanose in an appropriate nucleoside or nucleotide with an aldehyde or ketone, respectively, or their chemical equivalents, in the presence of an acid catalyst.
Particularly advantageous is the use of an organic acid , which can effect the transformation without affecting the integrity of the rest of the molecule. Alternately, strong acids such as trichloroacetic, p-toluenesulfonic, methanesulfonic and the like may be employed in catalytic amounts, in conjunction with inert solvents. Most preferred is formic acid, which is ideally suited to serve as both solvent and catalyst for these reactions. The discovery of the utility of formic acid for this purpose is one particular aspect of this invention.
Cyclical orthoesters may be prepared by reaction of the neighboring 2' and 3' hydroxyl groups of a furanose with an acylic orthoester, in the presence of an acid. When the nucleoside or nucleotide to be derivatized is a purine that contains a 6-amino functionality or is a pyrimidine that contains a 4-amino functionality, it may be converted to the respective urea or thiourea by treatment with isocyanates or isothiocyanates, respectively, as was previously described for carbamates or thiocarbamates of the 2' or 3' hydroxyls of the furanose. It was found that reactions of the amino group with isocyanates or isothiocyanates could be carried out in the presence of the hydroxyl groups of the furanose, by appropriate manipulation of the stoichiometry of the reaction.
All of the derivitization reactions described may be carried out on preformed dinucleotide polyphosphates, which results in multiple products dependent of reaction stoichiometry and on whether multiple reactive groups are present. When multiple products are obtained, these may be conveniently separated by the use of preparative reverse phase high performance liquid chromatography (HPLC). Particularly advantageous is the use of C18 or phenyl reverse phase columns, in conjunction with gradients that start with ammonium acetate buffer and end with methanol. The use of a buffer provides for nucleotide stability and improved peak shape of the eluting products and the use of methanol allows for . effective desorption of these lipophilic compounds from the column.
Particularly advantageous is the use of ammonium acetate buffer solutions in conjunction with methanol, as these solvents are miscible in all proportions and may be readily removed from the chromatographed products by evaporation, followed by lyophilization.
. While separation of multiple products may be done by HPLC, another strategy is to use nucleosides or nucleotides which contain only a single reactive functionality, whether because only one is present, or by the use of protecting groups to block side reactions at other positions in the molecule. This may be done at the level of preformed dinucleotide polyphosphates, or alternately, may be carried out on nucleoside mono-, di-, or triphosphates, leading to novel products in their own right, or may be coupled to other nucleoside mono-, di, or triphosphates by the methods which have already been described.
The inventors of the present invention have discovered compounds that are antagonists of the effect of ADP on its platelet membrane receptor, the P2Y12 receptor. The compounds provide efficacy as antithrombotic agents by their ability to block ADP from acting at its platelet receptor site and thus prevent platelet aggregation.
Thus, these compounds caal provide a more efficacious antithrombotic effect than aspirin, but with less profound effects on bleeding than antagonists of the fibrinogen receptor.
Since ADP-induced platelet aggregation is mediated by the simultaneous activation of both P2Y12 and P2Y1 receptors, the combined administration of the compounds described here with antagonists of platelet P2Y1 receptors could potentially provide a more efficacious antithrombotic effect at concentrations of each antagonist that are below the effective concentrations to block each receptor subtype in other systems, resulting in a decrease of the potential manifestation of adverse effects. In addition, these compounds can be used in conjunction with lower doses of these other agents which inhibit platelet aggregation by different mechanisms, to reduce the toxicity of these agents. Finally, if the compounds of the present invention have sufficient binding affinity and bear a fluorescent moiety, they can find uses as biochemical probes for the P2Y12 receptor.
The compounds of general Formula I are useful in therapy, in particular in the prevention of platelet aggregation. The compounds of the present invention are thus useful as anti-thrombotic agents, and are thus useful in the treatment or prevention of unstable angina, coronary angioplasty (PTCA) and myocardial infarction.
The compounds of the present invention are also useful in the treatment or prevention of primary arterial thrombotic complications of atherosclerosis such as thrombotic stroke, peripheral vascular disease, myocardial infarction without thrombolysis.

Still further indications where the compounds of the invention are useful are for the treatment or prevention of arterial thrombotic complications due to interventions in atherosclerotic disease such as angioplasty, endarterectomy, stent placement, coronary and other vascular graft surgery.
Still further indications where the compounds of the invention are useful are for the treatment or prevention of thrombotic complications of surgical or mechanical damage such as tissue salvage following surgical or accidental trauma, reconstructive surgery including skin flaps, and "reductive" surgery such as breast reduction.
The compounds of the present invention are also useful for the prevention of mechanically-induced platelet activation in vivo such as cardiopulmonary bypass (prevention of microthromboembolism), prevention of mechanically-induced platelet activation in vitro such as the use of the compounds in the preservation of blood products, e.g.
platelet concentrates, prevention of shunt occlusion such as renal dialysis and plasmapheresis, thrombosis secondary to vascular damage/inflammation such as vasculitis, arteritis, glomerulonephritis and organ graft rejection.
Still further indications where the compounds of the present invention are useful are indications with a diffuse thrombotic/platelet consumption component such as disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, heparin-induced thrombocytopenia and pre-eclampsia/eclampsia.
Still further indications where the compounds of the invention are useful are for the treatment or prevention of venous thrombosis such as deep vein thrombosis, veno-occlusive disease, hematological conditions such as thrombocythemia and polycythemia, and migraine.
In a particularly preferred embodiment of the present invention, the compounds are used in the treatment of unstable angina, coronary angioplasty and myocardial infarction.
In another particularly preferred embodiment of the present invention, the compounds are useful as adjunctive therapy in the prevention of coronary arterial thrombosis during the management of unstable angina, coronary angioplasty and acute myocardial infarction, i.e.
perithrombolysis. Agents commonly used for adjunctive therapy in the treatment of thrombotic disorders may be used, for example heparin and/or aspirin, just to mention a few.
A method of treating a mammal to alleviate the pathological effects of atherosclerosis and arteriosclerosis, acute MI, chronic stable angina, unstable angina, transient ischemic attacks and strokes, peripheral vascular disease, arterial thrombosis, preeclampsia, embolism, restenosis or abrupt closure following angioplasty, carotid endarterectomy, and anastomosis of vascular grafts.
The compounds of this invention may be used in vitro to inhibit the aggregation of platelets in blood and blood products, e.g. for storage, or for ex vivo manipulations such as in diagnostic or research use. This invention also provides a method of inhibiting platelet aggregation and clot formation in a mammal, especially a human, which comprises the internal administration of a compound of Formula (I) and a pharmaceutically acceptable carrier.
Chronic or acute states of hyper-aggregability, such as disseminated intravascular coagulation (DIC), septicemia, surgical or infectious shock, post-operative and post-partum trauma, cardiopulmonary bypass surgery, incompatible blood transfusion, abruptio placenta, thrombotic thrombocytopenic purpura (TTP), snake venom and immune diseases, are likely to be responsive to such treatment.
This invention further provides a method for inhibiting the reocclusion of an artery or vein following fibrinolytic therapy, which comprises internal administration of a compound of Formula (I) and a fibrinolytic agent. When used in the context of this invention, the teen fibrinolytic agent is intended to mean any compound, whether a natural or synthetic product, which directly or indirectly causes the lysis of a fibrin clot. Plasminogen activators are a well known group of fibrinolytic agents. Useful plasminogen activators include, for example, anistreplase, urokinase (UK), pro-urokinase (pUK), streptokinase (SIB), tissue plasminogen activator (tPA) and mutants, or variants thereof, which retain plasminogen activator activity, such as variants which have been chemically modified or in which one or more amino acids have been added, deleted or substituted or in which one or more functional domains have been added, deleted or altered such as by combining the active site of one plasminogen activator or fibrin binding domain of another plasminogen activator or fibrin binding molecule.
Extracorporeal circulation is routinely used for cardiovascular surgery in order to oxygenate blood. Platelets adhere to surfaces of the extracorporeal circuit.
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 of coronary and other arteries and after coronary artery bypass procedures.
The compounds of the present invention also encompass their non-toxic pharmaceutically acceptable salts, such as, but not limited to, an alkali metal salt such as sodium or potassium; an alkaline earth metal salt such as manganese, magnesium or calcium;
or an ammonium or tetraalkyl ammonium salt, i.e., NX~+ (wherein X is C1_4).
Pharmaceutically acceptable salts are salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects.
Those skilled in the art will recognize various synthetic methodologies which may be employed to prepare non-toxic pharmaceutically acceptable salts and acylated prodrugs of the compounds.
The active compounds may be administered systemically to target sites in a subject in need such that the extracellular concentration of a P2Y12 agonist is elevated to block the binding of ADP to P2Y12 receptor, thus inhibit the platelet aggregation. The term systemic as used herein includes subcutaneous inj ection, intravenous, intramuscular, intrasternah injection, intravitreal injection, infusion, inhalation, transdermal administration, oral administration, rectal administration and intra-operative instillation.
For systemic administration such as injection and infusion, the pharmaceutical formulation is prepared in a sterile medium. The active ingredient, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
Adjuvants such as local anesthetics, preservatives and buffering agents can also be dissolved in the vehicle.
The sterile indictable preparation may be a sterile indictable solution or suspension in a non-toxic acceptable diligent or solvent. Among the acceptable vehicles and solvents that may be employed are sterile water, saline solution, or Ringer's solution.
Another method of systemic administration of the active compound involves oral administration, in which pharmaceutical compositions containing active compounds are in the form of tablets, lozenges, aqueous or oily suspensions, viscous gels, chewable gums, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
For oral use, an aqueous suspension is prepared by addition of water to dispersible powders and granules with a dispersing or wetting agent, suspending agent one or more preservatives, and other excipients. Suspending agents include, for example, sodium carboxymethylcellulose, methylcellulose and sodium alginate. Dispersing or wetting agents include naturally-occurring phosphatides, condensation products of an allylene oxide with fatty acids, condensation products of ethylene oxide with long chain aliphatic alcohols, condensation products of ethylene oxide with partial esters from fatty acids and a hexitol, and condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anydrides. Preservatives include, for example, ethyl, and n-propyl p-hydroxybenzoate. Other excipients include sweetening agents (e.g., sucrose, saccharin), flavoring agents and coloring agents. Those skilled in the art will recognize the many specific excipients and wetting agents encompassed by the general description above.
For oral application, tablets are prepared by mixing the active compound with nontoxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
These excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil. Formulation for oral use may also be presented as chewable gums by embedding the active ingredient in gums so that the active ingredient is slowly released upon chewing.
Additional means of systemic achninistration of the active compound to the target platelets of the subject would involve a suppository form of the active compound, such that a therapeutically effective amount of the compound reaches the target sites via systemic absorption and circulation.
For rectal administration, the compositions in the form of suppositories can be prepared by mixing the active ingredient with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the compound. Such excipients include cocoa butter and polyethylene glycols.

The active compounds may also be systemically administered to the platelet aggregation sites through absorption by the skin using transdermal patches or pads. The active compounds are absorbed into the bloodstream through the skin. Plasma concentration of the active compounds can be controlled by using patches containing different concentrations of active compounds.
One systemic method involves an aerosol suspension of respirable particles comprising the active compound, which the subject inhales. The active compound would be absorbed into the bloodstream via the lungs, and subsequently contact the target platelets in a pharmaceutically effective amount. The respirable particles may be liquid or solid, with a particle size sufficiently small to pass through the mouth and larynx upon inhalation; in general, particles ranging from about 1 to 10 microns, but more preferably 1-5 microns, in size are considered respirable.
Another method of systemically administering the active compounds to the platelet aggregation sites of the subject involves administering a liquid/liquid suspension in the form of eye drops or eye wash or nasal drops of a liquid formulation, or a nasal spray of respirable particles that the subject inhales. Liquid pharmaceutical compositions of the active compound for producing a nasal spray or nasal or eye drops may be prepared by combining the active compound with a suitable vehicle, such as sterile pyrogen free water or sterile saline by techniques known to those skilled in the art.
Intravitreal delivery may include single or multiple intravitreal inj ections, or via an implantable intravitreal device that releases P2Yla antagonists in a sustained capacity.
Intravitreal delivery may also include delivery during surgical manipulations as either an adjunct to the intraocular irrigation solution or applied directly to the vitreous during the surgical procedure.
For systemic administration, plasma concentrations of active compounds delivered may vary according to compounds; but are generally 1x10-1°-1x10-5 moles/liter, and preferably 1x10-8-1x10-6moles/liter.
The pharmaceutical utility of P2Yla antagonist compounds of this invention is indicated by their inhibition of ADP-induced platelet aggregation. This widely used assay, as described in S.M.O. Hourani et al. BY. J. Pharmacol. 105, 453-457 (1992) relies on the measurement of the aggregation of a platelet suspension upon the addition of an aggregating agent such as ADP.

The present invention also provides novel compositions of matter. The compositions are pharmaceutically acceptable formulation comprising compounds of Formula I
of high purity, and/or in a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier can be selected by those skilled in the art using conventional criteria. The pharmaceutically acceptable carrier include, but are not limited to, saline and aqueous electrolyte solutions, water polyethers such as polyethylene glycol, polyvinyls such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate and salts such as sodium chloride and potassium chloride.
Preferred compositions of the present invention comprises compounds of Formula Ib (mononucleotide), provided that both Xl and X2 are not O, when n=1, and Xl is not O when n=0; and provided that X2 is independently O, CHz, CHF, CHCI, CFZ, CCIa when Y' = H;
also provided that when Rlo = NHa or O, and when RS and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to ortho-methylamino phenyl; further provided that when n~=1, X2=CHZ and B'=adenosine, then Rl and R2 are not equal to napththylenylinethyl, napthylenylmethylene, or phenylmethylene.
Preferred compositions of the present invention also comprises compounds of Formula Ia, wherein B and B' are independently pyrimidine (pyrimidine/pyrimidine dinucleotide), provided that when m+n+p = l, R16 = CH3, and RS and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to CH3 (Z' does not equal to acetate); also provided that when m+n+p = 3, B and B' = uridine, and RS and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to phenyl for Y'=ORl and 'or Y= OR4 (Y
and Y' does not equal to benzoyl); further provided that when m+n+p = 1, then both R8 and R9 are not CH3 (Z' and Y' taken together do not equal isopropylidine).
Preferred compositions of the present invention also comprises compounds of Formula Ia, wherein B is a purine or residue according to general formula IV, and B' is a pyrimidine residue according to general formula V, ( purinelpyrimidine dinucleotide);
provided that Y' is not equal to OCH3 when Z', Y, or Y' = H or OH; further provided that R8 is not equal to OCHZCH3 when R9 = H (Z' and Y' or Z and Y taken together do not equal to an orthoethylester).
Preferred compositions of the present invention also comprises compounds of Formula Ia, wherein B and B' are independently a purine residue according to general formula IV, (purine/purine dinonucleotide); provided that (a)Y or Y' is not equal to OCH3 when Rlo = NHz or O; (b) R8 is not equal to OCH3 or OCHZCH3 when R9 = H; (c) both R8 and R9 are not equal to CH3; (d) when m+n+p = l, then R$ and R9 does not equal OCH2CH3;
(e) when Rlo = NHaa and when RS and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to ortho-methylaminophenyl; (f) when m+n+p = 1, arid when RS and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to CH(CH2CHZSCH3)NHS(o-N02-Ph) or CH(CHZPh)NHS(o-NOa-Ph).
More preferred compositions of the present invention include the following compounds: 2' or 3' phenylcarbamate UTP, 2',3' di-phenylcarbamate UTP, 2'3' phenylacetaldehyde acetal ADP, di[3'(phenylcarbamate)dUp2dU], 2'3' phenylacetaldehyde acetal Up3U, di 2'3' phenylacetaldehyde acetal Up3U, 2'3' phenylacetaldehyde acetal Up4A, 2'3' phenylacetaldehyde acetal Ap4U, di 2'3' phenylacetaldehyde acetal Ap4U, 2'3' phenylacetaldehyde acetal Ip4U, 2'3' phenylacetaldehyde acetal Up4U, 2'3' phenylacetaldehyde acetal Ip4U, 2'3' phenylacetaldehyde acetal Up4dC, tetraphenylcarbamate Up4U, di 2'3' benzaldehyde acetal Ip4U, di 2',3' benzaldehyde acetal Up4U, 2',3' benzaldehyde acetal Up4U, di 2',3' phenylacetaldehyde acetal Cp4U, 2',3' phenylacetaldehyde acetal Cp4U, 2',3' phenylacetaldehyde acetal Up4C, 2',3' phenylacetaldehyde acetal Up4T, di 2'3' benzaldehyde acetal Cp4U, 2',3' benzaldehyde acetal Ip4U, 2',3' benzaldehyde acetal Up4U, 2',3' benzaldehyde acetal Up4dC, 2'3' benzaldehyde acetal Cp4U, 2'3' benzaldehyde acetal Up4C, 2',3' phenylpropionaldehyde acetal Up4U, di 2',3' phenylpropionaldehyde acetal Up4U,2',3' benzaldehyde acetal Cp4C, Bis MANT Up4U, Mant Up4U, Di 2'/3' benzylacetal Up4U, Mono 2'/3' benzylacetal Up4U, Triphenyl carbamate Up4U, 2' 3' phenylcarbamate Up4U, and monophenylcarbamate Up4U.
Preferred composition also comprises the following Compounds 1-21. In the following structures hydrogens which are understood to be present have been omitted for the sake of simplicity.

O
O ~P~ODP~O N
O --~
O N / N~S' N \ /N
S--~CF3 \ ~
Compound 1 O
O ll~l ~(C~ /O
O ~ PLO ~N
N / N~S' O ,p N~N
S-~CF3 Compound 2 O
O iP'(CF2~P
O O~ ~O ~N
N / N~S' _ N N
O O
S-~CF3 ~s Compound 3 2-(3-trifiluoromethylpropyl)thio-6-(2-methylthio) ethylamino-2',3'-(benzyl)methylenedioxy purine riboside 5'-a,~-difiluoromethylene diphosphate /O O
O-P~O\ //
O Pw0 f--N
O
O N / N~S' _ N N
O O
S--~CF3 Compound 4 O
//
O-P-(CH2)O
O \P
~ ~O ~N
O
O N / N~S' _ N N
O O
S--~CF3 Compound 5 O
//
O_P-( ~%O
O
OP~O /--N
O N / N~S' ~O N~N
S~CF3 Compound 6 O
// O
O-P~O\ //
O PLO ~N
O
O N / N~S' N N
O O
S--~CF3 N
Compound 7 O
y O_PyCH~) O
O
~P~O ~N
O N / N~S' N N
O O
S--~CF3 N
Is Compound 8 p /l o_o (C\~p OP~O ~-N
p N / N~S' ' N \ 'N
O ~O
S-~CF3 N
Compound 9 O
p-P p p O
OP~O ~N
p N / N~S' _ N N
O O
O~ ~p S--~CF3 N N
\ I I /
Compound 10 O
//
~ ~2 ~O
O
~ PLO ~N
N / N~S' N N
O O
O~ ~O S-~/CF3 N N
I
Compound 11 O
//
O_P-~ ~%O
O
~ PLO I-N
O N / N~S' _ N N
O O
O~ ~O S~CF3 N N
I, Compound 12 O ~ O
O .P-O P O-P~O /~
,, O O iP~O -N
O O
0,,,, O N / N~S~
O N
I O 'O N
N S---~CF3 O
Compound 13 O O ~O
O O.P, O O~P~O N
O ~~'~ N i O / I N~S
O~N : ~ N w N
I O' 'O
N S--~CF3 w O
Compound 14 P~-[2-(3-trifluoromethylpropyl)thio-6-(2-methylthio)ethylamino 2',3'-(benzyl)methylene dioxy purine riboside]-P4-(2',3'-(benzyl)methylene dioxy uridine) tetraphosphate O ,P ~O O
/P:0 00_ O O~P
O ~ O ~N
O,, O N / I N~S' O
' N\/
O
O ~O
O N S-~CF3 N
O
Compound 15 O _P ~O O
/P:0 00_ O O~P~
O ~ O ~N
O N / I N~S' O
N \ /N
O O ~O
O_ ,N S~CF3 N ~I
O
Compound 16 O O i0 O O O.P;O -POO-~ O\P
O p ~O N
N O"' 0 ~ N
S
O~N , ~ N \ /N
O I ~O
N S~,/CF3 O
Compound 17 ,p ~O O
N O /P:0 00_ O O~Pw O ~ O N
O,, O N / I N~S' O \ N
I ~ ,,~ O , : N ~
N O N O O
CF
O~ ~
N
O
Compound 18 ,p ~O O
N p~0 s0-P~O //
O p 'O O O PLO N
O
O,, O O O N / I N~S' I ,,w ; , N~N
N O N O O
O_ , ~O S--~CF3 N
N
O
Compound 19 O ,P /O O
P.p ~ O-P~p\ //
p O p O PLO N
O
p O % N~S~
O N ~ IN
N O O O
N O p ~ CF
o \
N
N
O
I /
Compound 20 P~p_p O_P%O
N~O p ,O O p p PLO -N
O
O~'~ p N / I N~S' p ,,: p N w N
N~p p N O~ ~p S~CF3 \ N N
N
p Compound 21 The invention is illustrated further by the following examples that are not to be construed as limiting the invention in scope to the specific procedures described in them.
EXAMPLES
Example 1 2'(3')-O-((phenylaminocarbonyl) -uridine 5'-)triphosphate Uridine 5'- triphosphate, ditributylammonium salt (100 mg, 0.176 mmol;
prepared from the trisodium salt by treatment with Dowex SOWx4 H+ in water, followed by mixing the protonated species with an excess of tributylamine, stripping and lyophilization) was dissolved in dry DMF (1 mL) and phenylisocyanate (19 ~,L, 0.176 mmol) added.
The reaction mixture was heated at 45°C for 15 minutes, at which point a further portion of phenylisocyanate (19 ~,L, 0.176 mmol) was added. The solution was heated at 45°C
overnight and the DMF was removed on a rotary evaporator. The residual oil was partitioned between water (2 mL) and ethyl acetate (2 mL) and the layers were separated.
The aqueous layer was extracted twice more with ethyl acetate (2 mL each) and the water was removed on a rotary evaporator. The residue was dissolved in water (1.5 mL) and the product isolated by repeated injections onto a preparative HPLC column (Alltech Nucleotide/Nucleoside C18, 7um, 10 X 250 mrn, gradient from 0.1 M ammonium acetate to methanol over 30 minutes, 5 mL/min, monitor at 260 run). The yield of the carbamate was 26mg ( 22%, calculated for the tetraammonium salt). 1H NMR showed the product to be a mixture of 2' and 3' carbamates.
The product so obtained can be used for the purposes of this invention per se or can be activated with a suitable coupling agent (e.g. a carbodiimide) and reacted with a variety of nucleotides to generate novel dinucleoside polyphosphates.
1H NMR (D20, 300 MHz): ~ 4.10-4.47 (m, 4H), 5.17 (m, 1H), 5.83 (dd, 1H), 5.96 (m, 1H), 7.04 (t, 1H), 7.25 (m, 4H), 7.79 (m, 1H). 31P NMR (D20,121.47 MHz): ~ -9.54 (m, 1P), -10.20 (m, 1P), -21.87 (m, 1P).
Example 2 2'.(3')-O-(phenylaminocarbonyl) -P1,P4-di(uridine 5'-)tetraphosphate . ["monophenylcarbamate Up4U"], Di- 2'(3')-O-(phenylaminocarbonyl) -P1,P4-di(uridine 5'-)tetraphosphate ["diphenylcarbamate Up4U"] and Tri-2'(3')-O-(phenylaminocarbonyl) -P1,P4-di(uridine 5'-)tetraphosphate ["triphenylcarbamate Up4U"]
P1,P4-Di(uridine 5'-) tetraphosphate, ditributylammonium salt (211 mg, 0.182 mmol;
prepared from the tetrasodium salt by treatment with Dowex SOWx4 H+in water, followed by mixing the protonated species with an excess of tributylamine, stripping and lyophilization) was dissolved in dry DMF (2 mL) and phenylisocyanate (40 ~L, 3.64 mmol) added in a single portion. The homogeneous reaction mixture was heated overnight at 45°C, whereupon TLC (silica gel, 50% isopropanol / 50% ammonium hydroxide) indicated a substantial conversion to two products. The solvent was removed on a rotary evaporator and the residue was partitioned between water (7 mL) and ethyl acetate (10 mL).
The layers were separated, and the aqueous was extracted twice more with ethyl acetate. (10 mL
each). The water was removed from the aqueous extract and the residual oil lyophilized overnight. The solid obtained was reconstituted in water (3 mL) and the two products separated by repeated inj ections onto a semipreparative HPLC column (Alltech Nucleotide/Nucleoside C 18, 7um, ~ 250 mm, gradient from 0.1 M ammonium acetate to methanol over 30 minutes, 5 mL/min, monitor at 260 nm). Stripping and lyophilization gave the mono-phenylcarbamate (48 mg, 27 % yield), di-phenylcarbamate (16 mg, 8%yield) and a trace amount of the triphenylcarbamate, as the tetraammonium salts. All three products were mixtures of the 10 corresponding 2'/3' regiosiomers.
Monophenylcarbamate: 1H NMR (D20, 300 MHz): ~ 4.08-4.65 (m, 9H), 5.14 (d, 1H), 5.75-5.94 (m, 4H), 7.01 (t, 1H), 7.22 (m, 4H), 7.76 (m, 2H). 31P NMR (D20, 121.47 MHz): ~ -10.17 (m, 2P), -21.81 (m, 2P).
Diphenylcarbamate: 1H NMR (D20, 300 MHz): 8 4.13-4.43 (m, 8H), 5.12 (m, 2H), 5.84 (m, 4H), 7.01 (m, 2H), 7.21 (m, 8H), 7.75 (dd, 2H). 31P NMR (D20, 121.47 MHz): 8 -10.19 (m, 2P), -21.65 (m, 2P).
Triphenylcarbamate: 1H NMR (DZO, 300 MHz): b 4.29 (m, 7H), 4.5.10 (m, 1H), 5.27 (m, 2H), 5.87 (m, 4H), 7.09 (m, 15H), 7.76 (d, 2H). 31P NMR (D20, 121.47 MHz): & -10.30 (m, 2P), -21.73 (m, 2P).
Example 3 Pl,P4-Tetra-(2'(3')-O-(phenylaminocarbonyl) di(uridine 5'-)tetraphosphate [tetraphenylcarbamate Up4U"]
This derivative was prepared according to the method of example 2. P1,P~-Di(uridine 5'-) tetraphosphate, ditributylammonium salt (200 mg, 0.172 rrunol) was treated with 16 eq of phenylisocyanate (300 uL, 2.76 mmol) in DMF and stirred overnight at 35°C. The solvent was evaporated and the excess reagents removed by extraction of an aqueous solution of the product with ethyl acetate. Following preparative HPLC as previously described, 93 mg (30% yield) of the tetraphenylcarbamate was obtained.
TetraphenylcarbamatelH NMR (D20, 300 MHz): S 7.75 (d, 2H), 7.11 (m, 16H), 6.94 (m, 4H), 5.95 (d, 2H), 5.80 (d, 2H), 5.32 (m, 2H), 5.23 (m, 2H), 4.42 (m, 2H), 4.25 (m, 2H), 4.16 (m, 2H). 31P NMR (D20, 121.47 MHz): ): 8 -10.30 (m, 2P), -22.32 (m, 2P).

Example 4 2',3'-(benzyl)methylenedioxy-P1,P4-di(uridine 5'-)tetraphosphate ["Mono 2'/3' benzylacetal Up4U"] and Pl,P4-Di-(2',3'-((benzyl)methylenedioxy) di(uridine 5' )tetraphosphate ["Di 2'/3' benzylacetal Up4U"]
P1,P4-Di(uridine 5'-) tetraphosphate, tetrasodium salt (290 mg, 0.332 mmol) was dissolved in 98% formic acid and phenylacetaldehyde, dimethyl acetal (110 uL, 0.662 mmol) added. The reaction was stirred at ambient temperature for 3 days, at which point TLC
(silica gel, 50%
isopropanol / 50% ammonium hydroxide) and HPLC (C18) showed good conversion to two less polar products. The formic acid was removed on a rotary evaporator, and the residue partitioned between 0.7 M ammonium bicarbonate (15 mL) and butyl acetate (15 mL). The layers were separated and the aqueous was washed with a further portion of butyl acetate (10 mL). The aqueous layer was stripped and the residue lyophilized overnight. The crude product was dissolved in water (5 mL) and the components separated by preparative HPLC
(Waters Novapak C18, Gum, 25 X 100 mm, gradient from 0.1 M ammonium acetate to methanol over 30 minutes, 30 mL/min, monitor at 260 nm). The yield of the monoacetal was 88 mg (28%) and of the diacetal 60mg (17%), both as the tetraamrnonium salts.
Monoacetal: 1H NMR (D20, 300 MHz): 8 2.99 (d, 2H), 4.01-4.32 (m, 8H), 4.77 (m, 2H), 5.33 (m, 2H), 5.74 (d, 1H), 5.81 (m, 2H), 7.21 (m, 5H), 7.64 (d, 1H), 7.79 (d, 1H). 31P NMR
(D20, 121.47 MHz): 8 -10.18 (m, 1P), -10.78 (m, 1P), -22.00 (m, 2P).
Diacetal: 1H NMR (DaO, 300 MHz): ~ 2.98 (d, 4H), 3.99 (m, 4H), 4.27 (m, 2H), 5.27 (m, 2H), 5.36 (m, 2H), 5.73 (d, J= 8.1 Hz, 2H), 7.21 (m, 10H), 7.61 (d, J= 8.1 Hz, 2H). 31P NMR
(DaO, 121.47 MHz): 8 -10.57 (m, 2P)., -21.81 (m, 2P).
Example 5 2',3'-((benzyl)methylenedioxy) P1,P3- uridine 5'-)triphosphate ["2'3' phenylacetaldehyde acetal Up3U"] and Pl,P3-Di-(2',3'-((benzyl)methylenedioxy) uridine 5'-)triphosphate ["di 2'3' phenylacetaldehyde acetal Up3U"]
P1,P3-Di(uridine 5'-) triphosphate, trisodium salt (100 mg, 0.129 mmol) was dissolved in 98% formic acid and phenylacetaldehyde, dimethyl acetal (64 uL, 0.386 mmol) added. After overnight stirring at room temperature, the formic acid was removed, and the residue partitioned between 1 M sodium bicarbonate and ethyl acetate. Following removal of the organic layer, the product was purified on preparative HPLC, as previously described.
Following lyophilization, 40 mg (36%) of the monoacetal and 24 mg (19%) of the diacetal were obtained.
Monoacetal: 1H NMR (DaO, 300 MHz): 8 7.7s (d, 2H), 7.54 (d, 2H), 7.16 (s, SH), 5.70 (m, 3H), 5.31 (s, 1H), 5.23 (s, 1H), 4.66 (m, 2H), 4.10 (m, 8H), 2.93 (d, 2H). 31P
NMR (DaO, 121.47 MHz): b -10.30 (m, 1P), 10.81 (m, 1P), -21.99 (m, 1P).
Diacetal: 1H NMR (D20, 300 MHz): 8 7.51 (d, 2H), 7.15 (m, l OH), 5.65 (d, 2H), 5.31 (d, 2H), 5.20 (t, 2H), 4.63 (m, 2H), 4.13 (m, 2H), 3.88 (m, 4H), 2.90 (d, 4H). 31P
NMR (D20, 121.47 MHz): ~ -10.75 (m, 2P), -21.97 (m, 1P).
Example 6 Pl-2',3'-((benzyl)methylenedioxy) (uridine 5'-) P4-( deoxycytidine 5'-) tetraphosphate ["2'3' phenylacetadehyde acetal Up4dC"]
P1- (uridine 5'-) P4-( deoxycytidine 5'-) tetraphosphate, tetrasodium salt (100 mg, 0.16 mmol) was dissolved in 98% formic acid (1 mL), and phenylacetaldehyde, dimethyl acetal (57 uL, 0.384 mmol) added. After overnight stirring, the formic acid was removed and the residue partitioned between 1 M sodium bicarbonate and ethyl acetate. After separation of the layers, the product was purified on preparative HPLC, as previously described. Yield 40 mg (36 %). This product was amenable to subsequent modification of the deoxy cytidine base by the procedures described in examples 9-13, giving rise to lipophilic bifunctional molecules falling within the scope of this invention.
Monoacetal: 1H NMR (D20, 300 MHz): 8 7.98 (d, 1H), 7.62 (d, 1H), 7.21 (m, SH), 6.11 (m, 2H), 5.74 (d, 1 H), 5.39 (d, -1 H), 5.31 (t, 1 H), 4.77 (m, 2H), 4.45 (m, 1 H), 4.32 (m, 1 H), 4.03 (m, SH), 2.99 (d, 2H), 2.29 and 2.21 (M, 2H). 31P NMR (D20, 121.47 MHz): b -10.15 (m, 1P), -10.68 (m, 1P), -21.98 (m, 2P).
Example 7 3'-O-(phenylaminocarbonyl) -2'-deoxy (uridine 5')- monophosphate Deoxyuridine 5'- monophosphate, tetrabutylammonium salt (135 mg , 0.274 mmol;
prepared from the disodium salt by treatment with Dowex SOWx4 H+, followed by stirring the resultant neutral species with excess tributylamine , stripping and lyophilization) was dissolved in dry DMF (1 mL). Phenylisocyanate (60 uL, 0.547 mmol) was added and the mixture heated overnight at 45°C, at which time TLC (silica gel, 50%
isopropanol / 50%
ammonium hydroxide) and HPLC (C18) indicated a substantial conversion to a less polar product. The DMF was stripped on a rotary evaporator and the oily residue partitioned between water (10 mL) and ethyl acetate (10 mL). The layers were separated and the aqueous layer was rewashed with ethyl acetate (2 X 10 mL). The water was removed and the residue was dissolved in water (2 mL). The product was isolated by repeated injections onto semipreparative HPLC (Alltech Nucleotide/Nucleoside C18, 7um, 10 X 250 mm, gradient from 0.1 M ammonium acetate to methanol over 30 minutes, 5 mL/min, monitor at 260 nm).
The yield was 67 mg as the diammonium salt (53 %).
1H NMR (D20, 300 MHz): 8 2.21 (m, 2H), 3.84 (s, 2H), 4.13 (s, 1H), 5.08 (d, 1H), 5.63 (d, 1H), 6.06 (t, 1H), 6.89 (br. t, 1H), 7.10 (m, 4H), 7.72 (d, 1H).
3iP NMR (D20, 121.47 MHz): ~ -2.31 (s).
Pl-(3'-O-(phenylaminocarbonyl)-2'-deoxyuridine 5'-)P4-(uridine 5'-)tetraphosphate Uridine 5'-triphosphate, ditributylammonium salt (prepared from the trisodium salt by treatment with Dowex 50Wx4 H+, followed by stirring the resultant neutral species with excess tributylamine , stripping and lyophilization) is treated with 1.5 equivalents of dicyclohexylcarbodiimide in DMF for 2 hours at room temperature. The dicyclohexylurea is filtered off, and the resultant uridine 5'- cyclical triphosphate is treated with 3'-O-(phenylaminocarbonyl) -2'-deoxy (uridine 5')- monophosphate, which is in the monotributylammonium salt form. The reaction mixture is stirred for several days at 45°C, and the solvent is removed. The products are separated by preparative HPLC, as has been previously described.
Example 8 2'(3')-(2-methylamino)benzoyl-P1,P4-di(uridine 5'-)tetraphosphate ("MANT
Up4U") and Pl,P4-Di-(2'(3')-(2-methylamino)benzoyl uridine 5'-)tetraphosphate ("Bis MANT
Up4U") P1,P4-Di(uridine 5'-) tetraphosphate, tetrasodium salt (800 mg, 0.93 mmol) was dissolved in water (5 mL) and the pH adjusted to 7.6 by the addition of solid sodium bicarbonate. N,N-dimethylformamide (DMF, 5 mL) was added, followed by N-methylisatoic anhydride (231 mg, 1.3 mmol) and the suspension was heated at 50°C for 2.5 hrs. TLC (silica gel, 50% isopropanol, 50% ammonium hydroxide) indicated that the reaction was not done by this time, so a further portion of N-methylisatoic anhydride (100 mg, 0.56 mmol) was added and the reaction heated for another hour. The DMF was removed on a rotary evaporator and the residue was dissolved in a minimum of water and applied to a DEAE Sephadex column (3 X 60 cm). The column was eluted with a stepwise gradient from water to 1 M
ammonium bicarbonate and the eluent monitored with a UV detector set at 254 nm. The two products that eluted were collected separately and the solvent was removed from each and the residue lyophilized overnight. 1H NMR indicated that the first product to elute was the monoacylated compound, while the latter was the diacylated derivative, and that both were mixtures with the acylation at either the 2' or 3' hydroxyls, but without two caxbamates on the same sugar. The yield of the monoaminobenzoylated product was 150 mg ( 16%); the yield of the diaminobenzoylated compound was 91 mg (8.7%).
Monoaminobenzoylated derivative: 1H NMR (D20, 300MHz): S 2.70 (s, 3H), 4.09-4.55(m, 9H), 5.34 (m, 1H), 5.71 (m, 2H), 5.83 (dd, 1H), 6.01 (m, 1H), 6.57 (m, 1H), 6.65 (m, 1H), 7.25 (t, 1H), 7.72 (d, 2H), 7.81 (m, 2H). 31P NMR (D20, 121.47 MHz): S -10.20 (m, 2P), -21.83 (m,2P).
Diaminobenzoylated derivative: 1H NMR (D20, 300 MHz): b 2.69 (s, 6H), 4.15-4.51 (m, 8H), 5.27 (m, 2H), 5.86 (m, 4H), 6.60 (m, 4H), 7.30 (m, 2H), 7.79 (m, 4H). 31P
NMR (D20, 121.47 MHz): 8 -10.16 (m, 2P), -21.76 (m, 2P).
Example 9 Pl-(4-N-(4-methoxyphenyl)aminocarbonylcytidine 5'-) -P4-(uridine 5'-) tetraphosphate Pi-(cytidine 5'-) -P4-(uridine 5'-) tetraphosphate, ditributylammonium salt (50 mg, 0.043mmo1; prepared from the tetraammonium salt by treatment with Dowex SOWx4 H+in water, followed by mixing the protonated species with an excess of tributylamine in methanol, stripping and lyophilization) was dissolved in dry DMF (1mL) and tributylamine (10 uL, 0.43 mmol), and p-methoxyphenylisocyanate (8.4 uL, 0.648 mmol) were added in a single portion. The homogeneous reaction mixture was heated overnight at 35°C, whereupon TLC (silica gel, 50% isopropanol / 50% ammonium hydroxide) and HPLC (C18) indicated a substantial conversion to a single product. The solvent was removed on a rotary evaporator and the residue dissolved in water (1mL). The product was isolated by repeated injections onto a semi-preparative HPLC column (Alltech Nucleotide/Nucleoside C18, 7um, mm, gradient from 0.1 M ammonium acetate to methanol over 30 minutes, 5 mL/min, monitor at 260 nm). Stripping and lyophilization gave the p-methoxyphenyluiea (24 mg, 55 yield), as the tetraammonium salt.
The product so obtained can be derivatized on the 2' and/or 3' hydroxyl groups according to the foregoing methods (e.g. Examples 2-6).
1H NMR (D20, 300 MHz): 8 3.59 (s, 3H), 4.01-4.20 (m, 10H), 5.68 (m, 3H), 6.19 (d, 1H), 6.71 (d, 2H), 7.18 (d, 2H), 7.67 (d, 1H), 8.06 (d, 1H). 31P NMR (D20, 121.47 MHz): 8 -10.13 (m, 2P), -21.76 (m, 2P).
Example 10 Pl-(( 4-bromophenyl)ethenocytidine 5'-) -P4-(uridine 5'-) tetraphosphate Pl-(cytidine 5'-) -P4-(uridine 5'-) tetraphosphate, tetrasodium salt (500 mg, 0.57 mrnol) was dissolved in water (5 mL) and a solution of 2,4'-dibromoacetophenone (792 mg, 2.85 mmol) in DMF (15 mL) added. The mixture was heated overnight at 40°C, and a further portion of the dibromoketone (400 mg, 1.44 mmol) in DMF (5 mL) added.
The rection was heated a further 5 hrs, and the solvents removed by evaporation.
The residue was partitioned between water (20 mL) and ethyl acetate (25 mL) and the layers separated. The aqueous layer was washed with further ethyl acetate (2x15 mL) and the aqueous evaporated to dryness. The residue was dissolved in water (5 mL) and the product was isolated by repeated injections onto a semi-preparative HPLC column (see example 6 for conditions).
The yield of the pure etheno compound was 80 mg (13.5%) 1H NMR (D20, 300 MHz): ~ 4.06 (m, 8H), 4.36 (m, 2H), 5.64 (dd, 2H), 6.07 (d, 1H), 6.74 (d, 1H), 7.45 (d, 2H), 7.54 (d, 2H), 7.59 (d, 1H), 7.63 (d, 1H), 7.93 (s, 1H).
31P NMR (D20, 121.47 MHz): b -10.09 (m, 2P), -21.59 (m, 2P) Example 11 Pl-(( 4-bromophenyl)etheno-2'-deoxycytidine 5'-) -P4-(uridine 5'-) tetraphosphate Example 8 product was prepared from 100 mg P1-(2'-deoxycytidine 5'-) -P4-(uridine 5'-) tetraphosphate, tetrasodium salt and 2,4'-dibromoacetophenone, according to the general method of example 7. Yield= 35 mg (30%).
1H NMR (D20, 300 MHz): b 2.31 (m, 2H), 4.03 (m, 8H), 5.60 (dd, 2H), 6.41 (t, 1H), 6.73 (d, 1H), 7.53 (m, 5H), 7.65 (d, 1H), 7.93 (s, 1H). 31P NMR (D20, 121.47 MHz):
& -10.11 (m, 2P), -21.58 (m, 2P) Example 12 Pl, P4-Di(( 4-bromophenyl)ethenocytidine 5'-) - tetraphosphate Example 9 product was prepared from 50 mg P1,P4-Di(cytidine 5'-) tetraphosphate, tetrasodium salt and 2,4'-dibromoacetophenone, according to the general method of example 7. Yield= 20 mg (29%).
1H NMR (D20, 300 MHz): 8 4.24 (m, l OH), 5.98 (d, 2H), 6.39 (d, 2H), 7.14 (m, 8H), 7.45 (m, 4H). ). 31P NMR (D20, 121.47 MHz): b -10.13 (m, 2P), -21.68 (m, 2P).
Example 13 Pl-(( 4-phenylphenyl)ethenocytidine 5'-) -P4-(cytidine 5'-) tetraphosphate Example 10 was product prepared from 50 mg Pl,P4-Di(cytidine 5'-) tetraphosphate, tetrasodium salt and 2-bromo-4'-phenylacetophenone, according to the general method of example 7. Yield= 15 mg (13%).
1H NMR (DaO, 300 MHz): 8 4.10 (m, l OH), 5.48 (d, 1H), 5.87 (m, 2H), 6.68 (d, 1H), 7.20 (m, 3H), 7.36 (m, 6H), 7.68 (m, 3H). 31P NMR (D20, 121.47 MHz): 8 -10.08 (m, 2P), -21.78 (m, 2P).
The products of examples 7-10 can be further derivatized according to the methods of Examples 2-6, to give bifunctional molecules that fall within the scope of the invention.
Example 14 Inhibition of ADP-Induced Platelet Aggregation Isolation of Platelets: Human blood was obtained from informed healthy volunteers.
Blood was collected into one-sixth volume of ACD (2.5 g of sodium citrate, 1.5 g citric acid, and 2.5 g glucose in 100 ml dH20). Blood was centrifuged at 800 x g for 15 min at room temperature and the platelet-rich plasma removed and incubated for 60 min at 37 °C in the presence of 1 mM acetylsalicylic acid followed by centrifugation at 1000 x g for 10 min at room temperature. The platelet pellet was resuspended at a density of 2 x 10$
cells/ml with HEPES-buffered Tyrode's solution (137 mM NaCI, 2.7 mM ICI, 1 mM MgCl2, 3 mM
NaH2P44, 5 mM glucose, 10 mM HEPES pH 7.4, 0.2% bovine serum albumin, and 0.05 U/ml apyrase).

Aggf°egatiofa Studies: ADP-induced platelet aggregation was determined by measuring the transmission of light through a O.S ml suspension of stirred (900 rpm) aspirin-treated washed platelets in a lumi-aggregometer at 37 °C (Chrono-Log Corp. Havertown, PA). The baseline of the instrument was set using O.S ml of Hepes-buffered Tyrode's solution.
Prior to S aggregation measurements, the platelet suspension was supplemented with 2 mM
CaClz and 1 mg/ml fibrinogen. Platelet aggregation was initiated by the addition of indicated concentrations of ADP or other agonists, and the light transmission continuously recorded for at least 8 min. When inhibitors of platelet aggregation were tested, platelets were incubated for 3-6 min in the presence of indicated concentrations of inhibitor before addition of ADP or other agonists, and the response recorded for at least 8 min. The potency of agonists and inhibitors of platelet aggregation was calculated from both, the rate of aggregation and the maximal extent of aggregation obtained for each determination by fitting the data to a four-parameter logistic equation using the GraphPad software package (GraphPad Corp. San Diego, CA).
1 S The ability of P2Y12 antagonists to inhibit platelet aggregation is presented in this application as the percent inhibition of the aggregation induced by a maximally effective concentration of ADP. When a broad range of concentrations of P2Y12 antagonist was tested (usually from 1nM to 100 ~,M), an ICSO value was also obtained. ICSO values represent the concentration of antagonist needed to inhibit by SO% the aggregation elicited by a given concentration of ADP.
Example 1 S
Effects on Platelet Aggregation Isz T~ivo To evaluate the ability of these compounds to inhibit platelet aggregation in vivo, an 2S experimental protocol similar to the method of R. G. Humphries et al. (Br.
J. Pharmacol.
115:1110-1116, 1995) will be performed.
Surgical P~epaf~ation ahd IustYUfraehtation: Male Sprague-Dawley rats are anesthetized. Body temperature is maintained at 37 ~ 0.S°C with a heating Lamp. Animals breathe spontaneously and a tracheotomy is performed to ensure a patent airway. A cannula containing heparinized saline is introduced into the left femoral artery and connected to a transducer to record blood pressure and heart rate. Cannulae containing non-heparinized saline are introduced into the left common carotid artery and left jugular vein for withdrawal of arterial blood samples and i.v. administration of compounds, respectively.
Experimental Protocol: Either compound or vehicle is administered to each animal as an . infusion. Blood samples axe taken immediately prior to the first infusion, at the end of each infusion and 20 min after cessation of the final infusion for measurement of platelet aggregation ex vivo. hnmediately after sampling, ADP-induced platelet aggregation is measured in duplicate in 0.5 ml blood samples diluted 1:1 with saline and incubated at 37°C
for 4 min. For the final minute of this period, cuvettes are transferred to a lumi-aggregometer and the sample stirred at 900 rpm. ADP (3 ~.M) is added in a volume of 20 ~.1 and the aggregation response is recorded.
Example 16 Inhibition of Thrombus Formation in Anesthetized Rats To evaluate the effect of these compounds on thrombus formation in vivo, the following experimental protocol will be performed.
Rats (CD-1; male; approximately 350 grams; Charles River, Raleigh, NC), are anesthetized with sodium pentobarbital (70 mg/kg i.p.). The abdomens axe shaved and a 22 gauge intravenous catheter is inserted into a lateral tail vein. A midline incision is made and the intestines are wrapped in saline-soaked gauze and positioned so the abdominal aorta is accessible. The inferior vena cave and abdominal aorta are carefully isolated and a section (approx. 1 cm) of the abdominal aorta (distal to the renal arteries proximal to the bifurcation) is dissected. All branches from the aorta in this section are ligated with 4-0 silk suture. A 2.5 mm diameter flow probe connected to a Transonic flow meter is placed on the artery and a baseline (pre-stenosis) flow is recorded. Two clips are placed around the artery decreasing the vessel diameter by approximately 80%. A second baseline flow measurement is taken (post-stenosis) and the hyperemic response is tested. Animals are then treated with either compound or saline i.v., via tail vein catheter. Thrombosis is induced five minutes after treatment by repeated external compressions of the vessel with hemostatic forceps. Two minutes post-injury, the vessel compressions are repeated and a 10 minute period of flow monitoring is started. Animals are monitored continuously for a minimum of the first ten minutes post-injury. After twenty minutes (post-injury), a flow measurement is repeated and the animals are euthanized. The section of the aorta that includes the injured section is harvested and placed in 10% formalin for possible histologic evaluation.
Example 17 Inhibition of Thrombus Formation in Anesthetized Dogs To evaluate the effect of these compounds on dynamic thrombus formation ira vivo, the following experimental protocol similar to the method of J. L. Romson et al. (Tlaronab.
Res. 17:841-853, 1980) will be performed.
Surgical Prepa~atioya and Irzst~~umeratatiou: .Briefly, purpose-bred dogs are anesthetized, intubated and ventilated with room air. The heart is exposed by a left thoracotomy in the fifth intercostal space and suspended in a pericardial cradle. A 2-3 cm segment of the left circumflex coronary artery (LCCA) is isolated by blunt dissection. The artery is instruxnented from proximal to distal with a flow probe, a stimulation electrode, and a Goldblatt clamp. The flow probe monitors the mean and phasic LCCA blood flow velocities. The stimulation electrode and its placement in the LCCA and the methodology to induce an occlusive coronary thrombus have been described previously (J. K.
Mickelson et al., Circulation 81:617-627, 1990; R. J. Shebuski et al., Circulation 82:169-177, 1990; J. F.
Tschopp et al., Coron. Artery Dis. 4:809-817, 1993).
Experimental Protocol. Dogs axe randomized to one of four treatment protocols (n=6 per treatment group) in which the control group receives saline i.v. and the three drug-treated groups are administered compound i.v. Upon stabilization from the surgical interventions, dogs receive either saline or compound. After approximately 30 minutes, an anodal current is applied to the LCCA for 180 min. The number and frequency of cyclic flow variations (CFV) that precede formation of an occlusive thrombus are recorded. These cyclic phenomena are caused by platelet thrombi that form in the narrowed lumen as a result of platelet aggregation (J. D. Folts et al., Circulation 54:365-370, 1976; Bush et al., Circulation 69:1161-1170, 1984). Zero flow in the LCCA for a minimum of 30 minutes indicates a lack of antithrombotic efficacy (L.G. Frederick et al., Circulation 93:129-134, 1996).
Example 18 ADP-Induced Aggregation of Different Compounds Different compounds were tested for their inhibition of ADP-induced aggregation and their ICSo according to the protocols in Example 14; the results are shown in Figure 1. The bar graphs in the figure illustrate the effect of 100 ~,M concentration of the compound on ADP-induced platelet aggregation, and the data are expressed as % inhibition of the ADP
response.
Figure 1 shows the structure and abbreviated name of each compound and its activity.
Where hydrogens are understood to be present, they have been omitted for the sake of simplicity. For example, for the first structure of the figure, it is implied that there are hydrogens at the 3- position of the pyrimidine ring, at the 3' position of the ribose on the oxygen, and on the nitrogen of the carbamate at the 2' position of the ribose.
In addition, as disclosed within the scope of the present invention, it is implied that the oxygens that are not doubly bonded to the phosphorous atoms axe either present in the ionized form as salts with a counterion, or axe bonded to a hydrogen atom. For simplicity, some of the structures in the figure are portrayed in the salt form, but this should not be interpreted as excluding the possibility that hydrogens could be present instead.
Several parent compounds, Up4U, Ip4U, Up3U, and Cp4U, without modifications on the furanose hydroxyl groups, have been included at the end of the figure to illustrate the utility of the present invention. However, these unmodified parent compounds do not inhibit the ADP-induced aggregation and are not within the scope of the present invention.

Claims (23)

1. A method of preventing or treating diseases or conditions associated with platelet aggregation comprising:

administering to a subject a pharmaceutical composition comprising a therapeutic effective amount of P2Y12 receptor antagonist compound, wherein said amount is effective to bind P2Y12 receptors on platelets and inhibit ADP-induced platelet aggregation.
2. The method according to Claim 1, wherein said P2Y12 receptor antagonist compound is a mononucleotide compound of Formula I:

wherein:
X1, X2, and X3 are independently oxygen, methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;
T1, T2, W, and V are independently oxygen or sulfur;

m= 0, 1 or 2;
n= 0 or 1;
p= 0, 1, or 2;
where the sum of m+n+p is from 0 to 5;
M =H, or a pharmaceutically-acceptable inorganic or organic counterion;
A = M;
D1 =O or C;
Y' =H, OH, or OR1;
Z' =H, OH, or OR2; with the proviso that at least one of Y' and Z' is OR1 or OR2;
R1 and R2 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to Formula III, Formula II

wherein:

O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;
R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ether; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II is an acyclic acetal or ketal; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ester or thioester; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to Formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to Formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;

Formula III

wherein:
O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by the common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester; and for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, or for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy;

B' is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;

Formula IV

Formula V

wherein:

R10 and R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or when R10 in a purine or R14 in a pyrimidine has as its first atom nitrogen, R10 and R11 or R14 and R15 are taken together to form a 5-membered fused imidazole ring (etheno compounds), optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;

J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;
R11 is hydrogen, O, or is absent;
R15 is hydrogen, or acyl;
R12 is hydrogen, alkyl, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or .omega.-A(C1-6alkyl)B-, wherein A and B are independently amino, mercapto, hydroxy or carboxyl;

R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation; and R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl.
3. The method according to Claim 1, wherein said P2Y12 receptor antagonist compound is a dinucleotide compound of Formula I:

wherein:
X1, X2, and X3 are independently oxygen, methylene, monochloromethylene, dichloromethylene, monofluoromethylene, difluoromethylene, or imido;

T1, T2,W, and V are independently oxygen or sulfur;

m= 0, 1 or 2;

n= 0 or 1;

p= 0, 1, or 2;

where the sum of m+n+p is from 1 to 5;
M=H or a pharmaceutically-acceptable inorganic or organic counterion;

D1 =O or C;

Y' =H, OH, or OR1;

Z' =H, OH, or OR2; with the proviso that at least one of Y' and Z' is OR1 or OR2;

R1 and R2 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to Formula III, Formula II

wherein:
O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;

R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ether; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II is an acyclic acetal or ketal; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to Formula II is an ester or thioester; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to Formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to Formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;

Formula III

wherein:
O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by the common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester; and for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, or for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy;

A is a nucleoside residue defined as:

and linked to the phosphate chain via the 5' position of the furanose or carbocycle;
wherein:

D2 =O or C;

Z =H, OH, Or OR3;

Y =H, OH, or OR4;

R and R4 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to Formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via the common carbon atom according to Formula III;

B' is a purine or a pyrimidine residue according to general Formulae IV and V
which is linked to the 1' position of the furanose or carbocycle via the 9- or 1-position of the base, respectively;

Formula IV

Formula V

wherein:

R10 and R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or When R10 in a purine or R14 in a pyrimidine has as its first atom nitrogen, R10 and R11 or R14 and R15 are taken together to form a 5-membered fused imidazole ring (etheno compounds), optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;

J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;

R11 is hydrogen, O, or is absent;

R15 is hydrogen, or acyl;

R12 is hydrogen, alkyl, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or .omega.-A(C1-6alkyl)B-, wherein A and B are independently amino, mercapto, hydroxy or carboxyl; and R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation; and R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl.
4. The method according to Claim 1, wherein said pharmaceutical composition reduces the incidence of dose-related adverse side effects of other therapeutic agents that are used to prevent, manage or treat platelet aggregation disorders.
5. The method according to Claim 1, wherein said diseases or conditions associated with platelet aggregation are disorders or procedures characterized by thrombosis, primary arterial thrombotic complications of atherosclerotic disease, thrombotic complications of interventions of atherosclerotic disease, thrombotic complications of surgical or mechanical damage, mechanically-induced platelet activation, shunt occlusion, thrombosis secondary to vascular damage and inflammation, indications with a diffuse thrombotic/platelet consumption component, venous thrombosis, coronary arterial thrombosis, pathological effects of atherosclerosis and arteriosclerosis, platelet aggregation and clot formation in blood and blood products during storage, chronic or acute states of hyper-aggregability, reocclusion of an artery or vein following fibrinolytic therapy, platelet adhesion associated with extracorporeal circulation , thrombotic complications associated with thrombolytic therapy, thrombotic complications associated with coronary and other angioplasty, or thrombotic complications associated with coronary artery bypass procedures.
6. The method according to Claim 5, wherein said disorders or procedures associated with thrombosis are unstable angina, coronary angioplasty, and myocardial infarction; said primary arterial thrombotic complications of atherosclerosis are thrombotic stroke, peripheral vascular disease, and myocardial infarction without thrombolysis; said thrombotic complications of interventions of atherosclerotic disease are angioplasty, endarterectomy, stent placement, coronary and other vascular graft surgery; said thrombotic complications of surgical or mechanical damage are tissue salvage following surgical or accidental trauma, reconstructive surgery including skin flaps, and "reductive" surgery such as breast reduction;
said mechanically-induced platelet activation is caused by cardiopulmonary bypass resulting in microthromboembolism and storage of blood products; said shunt occlusion is renal dialysis and plasmapheresis; said thromboses secondary to vascular damage and inflammation are vasculitis, arteritis, glomerulonephritis and organ graft rejection; said indications with a diffuse thrombotic/platelet consumption component are disseminated intravascular coagulation, thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, heparin-induced thrombocytopenia, and pre-eclampsia/eclampsia; said venous thrombosis are deep vein thrombosis, veno-occlusive disease, hematological conditions, and migraine; and said coronary arterial thrombosis is associated with unstable angina, coronary angioplasty and acute myocardial infarction.
7. The method according to Claim 6, wherein said hematological conditions are thrombocythemia and polycythemia.
8. The method according to Claim 7, wherein said pathological effects of atherosclerosis and arteriosclerosis are arteriosclerosis, acute myocardial infarction, chronic stable angina, unstable angina, transient ischemic attacks, and strokes, peripheral vascular disease, arterial thrombosis, preeclampsia, embolism, restenosis or abrupt closure following angioplasty, carotid endarterectomy, and anastomosis of vascular grafts; said chronic or acute states of hyper-aggregability is caused by DIC, septicemia, surgical or infectious shock, post-operative and post-partum trauma, cardiopulmonary bypass surgery, incompatible blood transfusion, abruptio placenta, thrombotic thrombocytopenic purpura, snake venom and immune diseases;
and said reocclusion of an artery or vein following fibrinolytic therapy is inhibited by internal administration of said compound with a fibrinolytic agent.
9. The method according to Claim 8, wherein said fibrinolytic agent is a natural or synthetic product which directly or indirectly causes lysis of a fibrin clot.
10. The method according to Claim 8, wherein said fibrinolytic agent is a plasminogen activator selected from the group consisting of anistreplase, urokinase, pro-urokinase, streptokinase, tissue plasminogen activator and mutants, or variants thereof, which retain plasminogen activator activity.
11. The method according to Claim 10, wherein said variants are selected from the group consisting of variants which have been chemically modified, variants which one or more amino acids have been added, deleted or substituted, and variants with one or more modified functional domains.
12. The method according to Claim 11, wherein said modified functional domains are added, deleted or altered by combining the active site of one plasminogen activator or fibrin binding domain of another plasminogen activator or fibrin binding molecule.
13. The method according to Claim 1, wherein said administering is systemic administration of said compound to a subject.
14. The method according to Claim 13, wherein said systemic administration is an administration selected from the group consisting of injecting an injectable form of said compound; administering by mouth an oral form of said compound; applying to the skin a transdermal patch or a transdermal pad containing said compound; administering a liquid/liquid suspension of said compound via nose drops or nasal spray;
administering a nebulized liquid of said compound to oral or nasopharyngeal airways;
administering rectally a suppository form of said compound; administering vaginally said compound in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles; administering said compound intravitreally; and administering via intra-operative instillation a gel, cream, powder, foam, crystals, liposomes, spray or liquid suspension form of said compound; such that a therapeutically effective amount of said compound contacts the target platelets of said patient via systemic absorption and circulation.
15. The method according to Claim 13, wherein said systemic administration comprises infusion of said compound to target platelets via a device selected from the group consisting of a pump catheter system and a continuous or selective release device.
16. A composition comprises compound of Formula Ib:

wherein:

D1 = O or CH2;

n and p = 0, 1, or 2 such that the sum of n+p is from 0 to 3;

A = M; wherein M= H+, NH4+, Na+ or other pharmaceutically-acceptable inorganic or organic counterion;

X1 and X2 are independently O, NH, CH2, CHF, CHCl, CF2, CCl2, provided that both X1 and X2 are not O when n=1, and X1 is not O when n=0; and provided that X2 is independently O, CH2, CHF, CHCl, CF2, CCl2 when Y'=H;

T1, V, and W are independently O or S;

Y'= H, OH, or OR1, Z'= H, OH or OR2, where R1 and R2 fall under the definitions of general formulas II
and III;

with the proviso that at least one of Y' and Z' is OR1 or OR2, respectively;

R1 and R2 are residues which are linked directly to the 2' and/or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to formula III;

wherein:

O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;

C is the carbon atom;

R5, R6, and R7 are independently H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ether, provided when R5=R6=H, R7 is not C2-C6 alkyl, or mononitro-substitued or monomethoxy-substittued aryl; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II
is an acyclic acetal or ketal; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is C2-C8 alkyl, cycloalkyl, aralkyl, aryl, hydroxy, methoxy, fluoro9, chloro, bromo, iodo, methyl, ethyl, propyl, butyl, thiioalkyl, alkoxy, carboxyl, cyano, trifluoromethyl, phenyl, cyclopropyl, cyclopenty, or cyclohexyl-substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ester or thioester;
or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;

Formula III

wherein:

O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by a common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester;

for cyclical acetals and ketals, R8 and R9 are independently hydrogen; alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, provided that when R8 is hydrogen or alkyl, R9 is not C1 alkyl;

for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, provided when R8 is hydrogen, R9 is not alkyloxy;

B' is either a purine residue, as in formula IV, linked through the 9-position, or a pyrimidine residue, as in formula V, linked through the 1- position:
wherein:
R10 and R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or When R10 in a purine or R14 in a pyrimidine has as its first atom nitrogen, R10 and R11 or R14 acid R15 are taken together to form a 5-membered fused imidazole ring, optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;
J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;
R11 is hydrogen, O or is absent;
R15 is hydrogen, or acyl; or R12 is hydrogen, alkyl, bromo, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or co-A(C1-6alkyl)B-, wherein A and B
are independently amino, mercapto, hydroxy or carboxyl;
R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation;
R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl;
provided that when R10 = NH2 or O, and when R5 and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to ortho-methylamino phenyl;
further provided that when n=p=1, X2=CH2 and B'=adenosine, then R1 and R2 are not equal to napththylenylmethyl, napthylenylinethylene, or phenylmethylene.
17. A composition comprises compound of Formula Ia:
D1 and D2 are independently O or CH2;
m and p= 0,1 or 2; n= 0 or 1; such that the sum of m+n+p is from 1 to 5;

X1, X2, and X3= are independently O, NH, CH2, CHF, CHCl, CF2, CCl2;
T1, T2, V, and W are independently O or S;
M= H+, NH4+, Na+ or other pharmaceutically-acceptable inorganic or organic counter ion;
Y'= H, OH, or OR1;
Z'= OH or OR2;
Z= OH or OR3;
Y= H, OH, or OR4, where R1, R2, R3 and R4 falls under the definition of general formula II or III, provided that at least one of Y', Z', Z, and Y is OR1, OR2, OR3, or OR4;
R1 and R2 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to formula III, wherein:
O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;
R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ether; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II is an acyclic acetal or ketal; or.
R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ester or thioester; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;
provided that when m+n+p = 1, R16 = CH3, and R5 and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to CH3;
provided that when m+n+p = 3, B and B' = uridine and R5 and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to phenyl for Y' = OR1 and/or Y = OR4;
Formula III
wherein:
O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by a common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester;
for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to fonn a homocyclic or heterocyclic ring composed of 3 to 8 atoms, for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy;

B and B' are independently pyrimidine residues, as in formula V, linked through the 1- position:

wherein:
R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R14 are acylamino, provided that they incorporate an amino residue from the C-position of the purine or the C-4 position of the pyrimidine; or when R14 in a pyrimidine has as its first atom nitrogen, R14 and R15 are taken together to form a 5-membered fused imidazole ring , optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;
R15 is hydrogen, or acyl; or R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl;
provided that when m+n+p =1, then both R8 and R9 are not CH3.
18. A composition comprises compound of Formula Ia:

wherein:
D1 and D2 are independently either O or CH2;
m and p= 0,1 or 2; n= 0 or 1; such that the sum of m+n+p is from 1 to 5;
X1, X2, and X3 are independently O, NH, CH2, CHF, CHCl, CF2, CCl2;
T1, T2, V, and W are independently O or S;
M= H, NH4+, Na+ or other pharmaceutically-acceptable inorganic or organic counterion;
Y'= H, OH, or OR1;
Z'= OH or OR2;
Z= OH or OR3;
Y= H, OH, or OR4, where R1, R2, R3 and R4 falls under the definition of general formula II or III, provided that at least one of Y, Z', Z and Y' is OR1, OR2, OR3, or OR4;
R1 and R2 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to formula III, wherein:

O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;
R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ether; or R5 and R6 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy such that the moiety defined according to formula II is an acyclic acetal or ketal; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ester or thioester; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;

wherein:

O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furanose or carbocycle are linked by a common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester;
for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy;
B is a purine residue, as in Formula IV, linked through the 9- position;
B1 is a pyrimidine residue, as in Formula V, linked through the 1-position:

wherein:

R10 and R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or When R10 in a purine or R14 in a pyrimidine has as its first atom nitrogen, R10 and R11 or R14 and R15 are taken together to form a 5-membered fused imidazole ring, optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;
J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;
R11 is hydrogen, O or is absent;
R15 is hydrogen, or acyl; or R12 is hydrogen, alkyl, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or cu-A(C1-6alkyl)B-, wherein A and B are independently amino, mercapto, hydroxy or carboxyl;
R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation;
R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl;
provided that Y' is not equal to OCH3 when Z', Y, or Z = H or OH;
further provided that R8 is not equal to OCH2CH3 when R9 = H.
19. A composition comprises compound of Formula Ia:

wherein:
D1 and D2 are independently O or CH2;
m and p= 0,1 or 2; n= 0 or 1; such that the sum of m+n+p is from 1 to 5;
X1, X2, and X3= are independently O, NH, CH2, CHF, CHCl, CF2, CCl2;
T1, T2, V, and W are independently O or S;

M= H+, NH4+, Na+ or other pharmaceutically-acceptable inorganic or organic counterion;
Y'= H, OH, or OR1;
Z'= OH or OR2;
Z= OH or OR3;
Y= H, OH, or OR4, where R1, R2, R3 and R4 falls under the definition of general formula II or III, provided that at least one of Y, Z, Y' and Z' is OR1, OR2, OR3, or OR4;
provided that Y or Y' is not equal to OCH3 when R10 = NH2 or O;
R1 and R2 are residues which are linked directly to the 2' and /or 3' hydroxyls of the furanose or carbocycle via a carbon atom according to formula II, or linked directly to two of the 2' and 3' hydroxyls of the furanose or carbocycle via a common carbon atom according to formula III, wherein:
O is the corresponding 2' and/or 3' oxygen of the furanose or carbocycle;
C is the carbon atom;
R5, R6, and R7 are H, an alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ether; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety defined according to formula II is an ester or thioester; or R5 and R6 are taken together as oxygen or sulfur doubly bonded to C, and R7 is amino or mono- or disubstituted amino, where the substituents are alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, such that the moiety according to formula II is a carbamate or thiocarbamate; or R5 and R6 are taken together to mean oxygen or sulfur doubly bonded to C, and R7 is alkoxy, cycloalkoxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy, such that the moiety according to formula II is a carbonate or thiocarbonate; or R7 is not present and R5 and R6 are taken together as oxygen or sulfur doubly bonded to C and both the 2' and 3' oxygens of the furanose are directly bound to C to form a cyclical carbonate or thiocarbonate;
provided that when m+n+p = 1, and when R5 and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to CH(CH2CH2SCH3)NHS(o-NO2-Ph) or CH(CH2Ph)NHS (o-NO2-Ph);

wherein:
O is the 2' and 3' oxygens of the furanose or carbocycle; and the 2' and 3' oxygens of the furasnose or carbocycle are linked by a common carbon atom (C) to form a cyclical acetal, cyclical ketal, or cyclical orthoester;
for cyclical acetals and ketals, R8 and R9 are independently hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, substituted aryl, or may be joined together to form a homocyclic or heterocyclic ring composed of 3 to 8 atoms, for cyclical orthoesters, R8 is hydrogen, alkyl, cycloalkyl, aralkyl, aryl, substituted aralkyl, or substituted aryl, R9 is alkyloxy, cycloalkyloxy, aralkyloxy, aryloxy, substituted aralkyloxy, or substituted aryloxy;
provided that R8 is not equal to OCH3 or OCH2CH3 when R9 = H; and provided that both R8 and R9 are not equal to CH3;
further provided that when m+n+p = 1, then R8 and R9 does not equal OCH2CH3;
B and B' are independently a purine residue, as in formula IV, linked through the 9-position:

wherein:

R10 is hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 is acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or when R10 in a purine has as its first atom nitrogen, R10 and R11 are taken together to form a 5-membered fused imidazole ring (etheno compounds), optionally substituted on the etheno ring with alkyl, cycloalkyl, aralkyl, or aryl moieties, as described for R5-R9 above;
J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;
R11 is hydrogen, O or is absent;
R12 is hydrogen, alkyl, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or .omega.-A(C1-6alkyl)B-, wherein A and B are independently amino, mercapto, hydroxy or carboxyl;
provided that when R10 = NH2, and when R5 and R6 are taken together as oxygen doubly bonded to C, then R7 is not equal to ortho-methylaminophenyl.
20. A compound of 2' or 3' phenylcarbamate UTP, 2',3' di-phenylcarbamate UTP, 2'3' phenylacetaldehyde acetal ADP, di[3'(phenylcarbamate)dUp2dU], 2'3' phenylacetaldehyde acetal Up3U, di 2'3' phenylacetaldehyde acetal Up3U, 2'3' phenylacetaldehyde acetal Up4A, 2'3' phenylacetaldehyde acetal Ap4U, di 2'3' phenylacetaldehyde acetal Ap4U, 2'3' phenylacetaldehyde acetal Ip4U, 2'3' phenylacetaldehyde acetal Up4U, 2'3' phenylacetaldehyde acetal Ip4U, 2'3' phenylacetaldehyde acetal Up4dC, tetraphenylcarbamate Up4U, di 2'3' benzaldehyde acetal Ip4U, di 2',3' benzaldehyde acetal Up4U, 2',3' benzaldehyde acetal Up4U, di 2',3' phenylacetaldehyde acetal Cp4U, 2',3' phenylacetaldehyde acetal Cp4U, 2',3' phenylacetaldehyde acetal Up4C, 2',3' phenylacetaldehyde acetal Up4T, di 2'3' benzaldehyde acetal Cp4U, 2',3' benzaldehyde acetal Ip4U, 2',3' benzaldehyde acetal Up4U, 2',3' benzaldehyde acetal Up4dC, 2'3' benzaldehyde acetal Cp4U, 2'3' benzaldehyde acetal Up4C, 2',3' phenylpropionaldehyde acetal Up4U, di 2',3' phenylpropionaldehyde acetal Up4U, 2',3' benzaldehyde acetal Cp4C, Bis MANT Up4U, Mant Up4U, Di 2'/3' benzylacetal Up4U, Mono 2'/3' benzylacetal Up4U, Triphenyl carbamate Up4U, 2'3' phenylcarbamate Up4U, or monophenylcarbanlate Up4U.
21. A compound of P1-[2-(3-trifluoromethylpropyl)thio-6-(2-methylthio)ethylamino 2', 3'-(benzyl)methylene dioxy purine riboside]-P4-(2',3'-(benzyl)methylene dioxy uridine) tetraphosphate, or 2-(3-trifluoromethylpropyl)thio-6-(2-methylthio) ethylamino-2',3'-(benzyl)methylenedioxy purine riboside 5'-.alpha., .beta.-difluoromethylene dipohsphate.
22. A compound of Formula Ia':

wherein:
D1 =O or CH2;
D2 =O or CH2;
B and B' are independently purine or pyrimidine residues according to general formula IV or V;
m and p= 0, 1 or 2; n= 0 or 1; such that the sum of m+n+p is from 1 to 5;
X1, X2, and X3 are independently O, NH, CH2, CHF, CHCl, CF2, or CCl2;

T1, T2, V, and W are independently O or S;

M= H+ or NH4+, Na or other pharmaceutically-acceptable inorganic or organic counter ion;

Y'= H or OH;

Z'= OH;

Z= OH;

Y= H or OH;

Formula IV

Formula V

wherein:
R10 and R14 are hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle; or R10 and R14 are acylamino, provided that they incorporate an amino residue from the C-6 position of the purine or the C-4 position of the pyrimidine; or when R10 in a purine or R14 in a pyrimidine has as its first atom nitrogen, R10 and R11 or R14 and R15 are taken together to form a 5-membered fused imidazole ring;

R11 is hydrogen, O, or is absent;

R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation;

R15 is hydrogen, or acyl; and R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl.
23. A compound of Formula Ia":

wherein:

D1 =O or CH2;

D2 =O or CH2;

m and p= 0, 1 or 2; n= 0 or 1; such that the sum of m+n+p is from 1 to 5;

X1, X2, and X3 are independently O, NH, CH2, CHF, CHCl, CF2, or CCl2;

T1, T2, V, and W are independently O or S;

M= H+, NH4+, Na+ or other pharmaceutically-acceptable inorganic or organic counter ion;

Y'= H or OH;

Z'= OH;

Z= OH;

Y= H or OH;

B and B' are independently a purine residue, as in Formula IV, linked through the 9-position, or a pyrimidine residue, as in Formula V, linked through the 1-position;

Formula IV

Formula V

wherein:

R10 is hydroxy, oxo, mercapto, alkylthio, alkyloxy, aryloxy, diaralkylamino, diarylamino, or dialkylamino, where the alkyl groups are optionally linked to form a heterocycle;

R14 is hydroxy, oxo, amino, mercapto, alkylthio, alkyloxy, aryloxy, alkylamino, cycloalkylamino, aralkylamino, arylamino, diaralkylamino, diarylamino, dialkylamino, or acylamino, where the alkyl groups are optionally linked to form a heterocycle;
or when R14 has as its first atom nitrogen, R14 and R15 are taken together to form a 5-membered fused imidazole ring;

J is carbon or nitrogen, with the provision that when nitrogen, R12 is not present;

R11 is hydrogen, O, or is absent;

R12 is hydrogen, alkyl, bromo, azido, alkylamino, arylamino or aralkylamino, alkoxy, aryloxy or aralkyloxy, alkylthio, arythio or aralkylthio, or .omega.-A(C1-6alkyl)B-, wherein A and B
are independently amino, mercapto, hydroxy or carboxyl;

R13 is hydrogen, chlorine, amino, monosubstituted amino, disubstituted amino, alkylthio, arylthio, or aralkylthio, where the substituent on sulfur contains up to a maximum of 20 carbon atoms, with or without unsaturation;

R15 is hydrogen, or acyl; and R16 is hydrogen, methyl, alkyl, halo, alkyl, alkenyl, substituted alkenyl, alkynyl, or substituted alkynyl.
CA002420179A 2000-08-21 2001-08-20 Composition and method for inhibiting platelet aggregation Abandoned CA2420179A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/643,138 2000-08-21
US09/643,138 US7018985B1 (en) 2000-08-21 2000-08-21 Composition and method for inhibiting platelet aggregation
PCT/US2001/041818 WO2002016381A2 (en) 2000-08-21 2001-08-20 Composition and method for inhibiting platelet aggregation

Publications (1)

Publication Number Publication Date
CA2420179A1 true CA2420179A1 (en) 2002-02-28

Family

ID=24579508

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002420179A Abandoned CA2420179A1 (en) 2000-08-21 2001-08-20 Composition and method for inhibiting platelet aggregation

Country Status (12)

Country Link
US (2) US7018985B1 (en)
EP (1) EP1311525A2 (en)
JP (1) JP2004521074A (en)
KR (1) KR20040014934A (en)
CN (1) CN100465186C (en)
AR (1) AR032631A1 (en)
AU (2) AU8547001A (en)
BR (1) BR0113402A (en)
CA (1) CA2420179A1 (en)
MX (1) MXPA03001518A (en)
TW (1) TWI265931B (en)
WO (1) WO2002016381A2 (en)

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6864243B1 (en) * 2000-05-12 2005-03-08 Inspire Pharmaceuticals, Inc. Method for treating retinal degeneration with purinergic receptor agonists
US7452870B2 (en) 2000-08-21 2008-11-18 Inspire Pharmaceuticals, Inc. Drug-eluting stents coated with P2Y12 receptor antagonist compound
US7018985B1 (en) 2000-08-21 2006-03-28 Inspire Pharmaceuticals, Inc. Composition and method for inhibiting platelet aggregation
US7132408B2 (en) 2000-08-21 2006-11-07 Inspire Pharmaceuticals, Inc. Composition and method for inhibiting platelet aggregation
US7084128B2 (en) 2002-01-18 2006-08-01 Inspire Pharmaceuticals, Inc. Method for reducing intraocular pressure
EP1348466A3 (en) * 2002-02-01 2003-10-29 Inspire Pharmaceuticals, Inc. Method for treating pain with adenosine-tetraphosphates
US7435724B2 (en) * 2002-02-27 2008-10-14 Inspire Pharmaceutical, Inc. Degradation-resistant mononucleoside phosphate compounds
JP4699028B2 (en) * 2002-11-08 2011-06-08 ザ ブライアム アンド ウィミンズ ホスピタル インコーポレーテッド Compositions and methods for prolonging platelet survival
CA2734052A1 (en) * 2003-05-30 2005-01-13 Pharmasset, Inc. Modified fluorinated nucleoside analogues
US7749981B2 (en) 2003-10-21 2010-07-06 Inspire Pharmaceuticals, Inc. Drug-eluting stents coated with non-nucleotide P2Y12 receptor antagonist compound
EP1685135B1 (en) 2003-10-21 2010-05-26 Inspire Pharmaceuticals, Inc. TETRAHYDRO-FURO[3,4-d]DIOXOLE COMPOUNDS AND COMPOSITIONS AND METHOD FOR INHIBITING PLATELET AGGREGATION
US7504497B2 (en) 2003-10-21 2009-03-17 Inspire Pharmaceuticals, Inc. Orally bioavailable compounds and methods for inhibiting platelet aggregation
US7335648B2 (en) 2003-10-21 2008-02-26 Inspire Pharmaceuticals, Inc. Non-nucleotide composition and method for inhibiting platelet aggregation
AU2004285486A1 (en) * 2003-10-22 2005-05-12 University Of Rochester Use of peroxisome proliferator-activated receptor gamma (PPARgamma) and/or retinoic acid receptor (RXR) agonists to inhibit platelet functions
US7326683B2 (en) 2004-05-06 2008-02-05 Molichem Medicines, Inc. Treatment of membrane-associated diseases and disorders using lantibiotic containing compositions
EP1753445A4 (en) 2004-05-06 2009-05-20 Molichem Medicines Inc Treatment of ocular diseases and disorders using lantibiotic compositions
CN101023094B (en) * 2004-07-21 2011-05-18 法莫赛特股份有限公司 Preparation of alkyl-substituted 2-deoxy-2-fluoro-d-ribofuranosyl pyrimidines and purines and their derivatives
WO2006044790A2 (en) * 2004-10-15 2006-04-27 Zymequest, Inc. Compositions and methods for prolonging survival of platelets
ES2376777T3 (en) * 2004-09-07 2012-03-16 Velico Medical, Inc. Apparatus for prolonging platelet survival
PL3109244T3 (en) 2004-09-14 2019-09-30 Gilead Pharmasset Llc Preparation of 2'fluoro-2'-alkyl-substituted or other optionally substituted ribofuranosyl pyrimidines and purines and their derivatives
AU2006204159A1 (en) * 2005-01-06 2006-07-13 Astrazeneca Ab Novel pyridine compounds
JP2008537745A (en) * 2005-03-30 2008-09-25 インスパイアー ファーマシューティカルズ,インコーポレイティド Process for the preparation of 4,6-disubstituted-tetrahydro-furo, thieno, pyrrolo and cyclopenta- [3,4] [1,3] dioxole
US7932376B2 (en) 2005-05-05 2011-04-26 Inspire Pharmaceuticals, Inc. Pyrimidine-based non-nucleotide composition and method for inhibiting platelet aggregation
WO2007008140A1 (en) * 2005-07-13 2007-01-18 Astrazeneca Ab New pyridine analogues
ITRM20050391A1 (en) 2005-07-22 2007-01-23 Giuliani Spa SIMPLE COMPOUNDS OF 6-THIOPHOUANOSINE TRIPOSPHATE, THEIR USE IN THE MEDICAL AREA AND PROCEDURE FOR THEIR PREPARATION.
JPWO2007020935A1 (en) * 2005-08-17 2009-02-26 小野薬品工業株式会社 Pain therapeutic agent comprising P2Y12 receptor and / or P2Y14 receptor blocker
EP1954304A4 (en) * 2005-10-14 2010-03-10 Zymequest Inc Compositions and methods for prolonging survival of platelets
GB0525970D0 (en) 2005-12-21 2006-02-01 Secr Defence Supramolecular polymers
WO2007140333A2 (en) * 2006-05-26 2007-12-06 Inspire Pharmaceuticals, Inc. Mononucleoside phosphonate compounds
TW200815426A (en) * 2006-06-28 2008-04-01 Astrazeneca Ab New pyridine analogues II 333
AU2007270081A1 (en) * 2006-07-04 2008-01-10 Astrazeneca Ab New pyridine analogues
TW200811133A (en) * 2006-07-04 2008-03-01 Astrazeneca Ab New pyridine analogues III 334
US20090286834A1 (en) * 2006-07-04 2009-11-19 Astrazeneca Ab Pyridine Analogues VI
US20080032992A1 (en) * 2006-07-04 2008-02-07 Astrazeneca Ab New Pyridine Analogues V
RU2009123928A (en) * 2007-01-12 2011-02-20 Астразенека Аб (Se) PYRIDINE COMPOUNDS AND THEIR APPLICATION AS P2Y12 ANTAGONISTS
US20080176827A1 (en) * 2007-01-12 2008-07-24 Astrazeneca Ab New Pyridine Analogues VII 543
CL2008000093A1 (en) * 2007-01-12 2008-08-22 Astrazeneca Ab COMPOUNDS DERIVED FROM PIRIDINA, INHIBITORS OF P2Y12; PHARMACEUTICAL COMPOSITION THAT INCLUDES SUCH COMPOUNDS; AND ITS USE FOR THE TREATMENT OF A PLAQUETARY AGREGATION DISORDER.
US7964580B2 (en) 2007-03-30 2011-06-21 Pharmasset, Inc. Nucleoside phosphoramidate prodrugs
UY30865A1 (en) * 2007-07-13 2009-03-02 NEW ANALOGS OF PIRIDINA X 161
US8518912B2 (en) * 2007-11-29 2013-08-27 Actelion Pharmaceuticals Ltd. Phosphonic acid derivates and their use as P2Y12 receptor antagonists
US8173621B2 (en) 2008-06-11 2012-05-08 Gilead Pharmasset Llc Nucleoside cyclicphosphates
US8168597B2 (en) * 2008-10-22 2012-05-01 Inspire Pharmaceuticals, Inc. Method for treating cystic fibrosis
EP2364086B1 (en) * 2008-11-20 2016-09-14 Glsynthesis Inc. Novel antithrombotic diadenosine tetraphosphates and related analogs
CN102753563A (en) 2008-12-23 2012-10-24 吉利德制药有限责任公司 Nucleoside analogs
PT2376088T (en) 2008-12-23 2017-05-02 Gilead Pharmasset Llc 6-o-substituted-2-amino-purine nucleoside phosphoramidates
AR074977A1 (en) 2008-12-23 2011-03-02 Pharmasset Inc SYNTHESIS OF PURINE NUCLEOSIDS
TWI576352B (en) 2009-05-20 2017-04-01 基利法瑪席特有限責任公司 Nucleoside phosphoramidates
US8618076B2 (en) 2009-05-20 2013-12-31 Gilead Pharmasset Llc Nucleoside phosphoramidates
CN102858790A (en) 2010-03-31 2013-01-02 吉利德制药有限责任公司 Nucleoside Phosphoramidates
US8563530B2 (en) 2010-03-31 2013-10-22 Gilead Pharmassel LLC Purine nucleoside phosphoramidate
US9156873B2 (en) 2010-04-28 2015-10-13 Isis Pharmaceuticals, Inc. Modified 5′ diphosphate nucleosides and oligomeric compounds prepared therefrom
TW201242974A (en) 2010-11-30 2012-11-01 Gilead Pharmasset Llc Compounds
WO2013033178A1 (en) 2011-08-30 2013-03-07 University Of Utah Research Foundation Methods and compositions for treating nephrogenic diabetes insipidus
US8889159B2 (en) 2011-11-29 2014-11-18 Gilead Pharmasset Llc Compositions and methods for treating hepatitis C virus
EA201690473A1 (en) 2013-08-27 2017-03-31 ГАЙЛИД ФАРМАССЕТ ЭлЭлСи COMBINED COMPOSITION OF TWO ANTI-VIRUS COMPOUNDS
CN103772461A (en) * 2014-01-06 2014-05-07 南京正科制药有限公司 Cangrelor crystal form I
US11530234B2 (en) 2018-09-11 2022-12-20 Risen (Suzhou) Pharma Tech Co., Ltd. CD73 inhibitors and pharmaceutical uses thereof
EP3849992A4 (en) 2018-09-11 2022-06-29 Risen (Suzhou) Pharma Tech Co., Ltd. Cd73 inhibitors and pharmaceutical uses thereof

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3321462A (en) 1963-07-15 1967-05-23 Syntex Corp Process for the preparation of nucleoside polyphosphates
GB1407903A (en) 1973-03-21 1975-10-01 Ici Ltd Nucleotide derivatives
JPS5973524A (en) 1982-10-21 1984-04-25 Meito Sangyo Kk Remedy for thrombosis
WO1989004321A1 (en) 1987-11-05 1989-05-18 The Worcester Foundation For Experimental Biology Diadenosine 5', 5'''-p1, p4-tetraphosphate and analogs thereof as antithrombotic agents
US5049550A (en) 1987-11-05 1991-09-17 Worcester Foundation For Experimental Biology Diadenosine 5', 5'"-p1, p4,-tetraphosphate analogs as antithrombotic agents
GB9015684D0 (en) 1990-07-17 1990-09-05 Medical Res Council Synthesis and uses of spin labelled ribonucleosides and ribonucleotides
NZ239846A (en) 1990-09-27 1994-11-25 Merck & Co Inc Sulphonamide derivatives and pharmaceutical compositions thereof
WO1992017488A1 (en) 1991-04-06 1992-10-15 Fisons Plc Atp analogues
US5654285A (en) 1991-04-06 1997-08-05 Astra Pharmaceuticals Limited ADP and ATP analogues, process for making and administration to inhibit ADP-induced platelet aggregation
US5292498A (en) 1991-06-19 1994-03-08 The University Of North Carolina At Chapel Hill Method of treating lung disease with uridine triphosphates
AU679721B2 (en) 1993-02-10 1997-07-10 Astrazeneca Ab N-alkyl-2-substituted ATP analogues
WO1995010538A1 (en) 1993-10-15 1995-04-20 The University Of North Carolina At Chapel Hill Dna encoding the human p2u receptor and null cells expressing p2u receptors
US5814609A (en) * 1993-10-22 1998-09-29 University Of Southern California Compositions containing a disintegrin and methods for its use in preventing metastasis and other conditions
US5656256A (en) 1994-12-14 1997-08-12 The University Of North Carolina At Chapel Hill Methods of treating lung disease by an aerosol containing benzamil or phenamil
EP0796098A4 (en) 1994-12-22 1998-04-29 Smithkline Beecham Corp Fibrinogen receptor antagonists
US5635160A (en) 1995-06-07 1997-06-03 The University Of North Carolina At Chapel Hill Dinucleotides useful for the treatment of cystic fibrosis and for hydrating mucus secretions
SK283206B6 (en) 1995-07-11 2003-03-04 Astra Pharmaceuticals Ltd. New inhibitors of platelet aggregation, their preparation method, pharmaceutical composition containing them and use
US5628984A (en) 1995-07-31 1997-05-13 University Of North Carolina At Chapel Hill Method of detecting lung disease
JP3519199B2 (en) 1996-02-06 2004-04-12 株式会社ソニー・コンピュータエンタテインメント Image generation device
US5837861A (en) 1997-02-10 1998-11-17 Inspire Pharmaceuticals, Inc. Dinucleotides and their use as modulators of mucociliary clearance and ciliary beat frequency
DE69736468T2 (en) 1996-03-27 2007-03-29 Inspire Pharmaceuticals, Inc. METHOD FOR TREATING CILIARY DYSKINESIS WITH URIDINTRIPHOSPHATES AND RELATED COMPOUNDS
US5900407A (en) 1997-02-06 1999-05-04 Inspire Pharmaceuticals, Inc. Method of treating dry eye disease with uridine triphosphates and related compounds
US5681823A (en) 1996-05-02 1997-10-28 Prp Inc. P1, P4 -dithio-P2 -P3 -monochloromethylene 5', 5'"-diadenosine P1, P4 -tetraphosphate as antithrombotic agent
US5968913A (en) 1996-07-03 1999-10-19 Inspire Pharmaceuticals, Inc. Pharmaceutical compositions of uridine triphosphate
US5789391A (en) 1996-07-03 1998-08-04 Inspire Pharmaceuticals, Inc. Method of treating sinusitis with uridine triphosphates and related compounds
US5962432A (en) 1996-07-03 1999-10-05 Inspire Pharmaceuticals, Inc. Sterilized, isotonic and pH-adjusted pharmaceutical formulation of uridine triphosphate
US5763447C1 (en) 1996-07-23 2002-05-07 Inspire Pharmaceuticals Method of preventing or treating pneumonia in immobilized patients with uridine triphosphates and related compounds
AU4978897A (en) 1996-10-08 1998-05-05 Inspire Pharmaceuticals, Inc. Method of early lung cancer detection via sputum induction and analysis of sputum to detect cancer associated substances
US6159952A (en) 1996-11-07 2000-12-12 Inspire Pharmaceuticals, Inc. Method of treating bronchitis with uridine triphosphate and related compounds
EP0946561B1 (en) 1996-12-20 2002-02-13 AstraZeneca AB Triazolo(4,5-d)pyrimidinyl derivatives and their use as medicaments
JP4531867B2 (en) 1997-02-06 2010-08-25 インスパイアー ファーマシューティカルズ,インコーポレイティド Certain dinucleotides and their use as modulators of mucociliary clearance and cilia movement frequency
SE9702774D0 (en) 1997-07-22 1997-07-22 Astra Pharma Prod Novel compounds
US6462028B2 (en) 1997-07-25 2002-10-08 Inspire Pharmaceuticals, Inc. Method of promoting cervical and vaginal secretions
JP3723227B2 (en) 1997-07-25 2005-12-07 インスパイアー ファーマシューティカルズ,インコーポレイティド Process for large-scale production of di (uridine 5 ')-tetraphosphate and its salts
BR9811612A (en) 1997-08-29 2000-08-08 Univ North Carolina Use of uridine 5'-diphosphate and the like for the treatment of lung diseases
US6528640B1 (en) 1997-11-05 2003-03-04 Ribozyme Pharmaceuticals, Incorporated Synthetic ribonucleic acids with RNAse activity
JP4633927B2 (en) 1998-05-22 2011-02-16 インスパイアー ファーマシューティカルズ,インコーポレイティド Therapeutic dinucleotides and derivatives
SE9804175D0 (en) 1998-12-02 1998-12-02 Astra Pharma Prod New assay
TWI229674B (en) 1998-12-04 2005-03-21 Astra Pharma Prod Novel triazolo[4,5-d]pyrimidine compounds, pharmaceutical composition containing the same, their process for preparation and uses
EP1140967A1 (en) 1998-12-23 2001-10-10 University Of North Carolina At Chapel Hill Targeted gene transfer using g protein coupled receptors
BR0008498A (en) 1999-02-26 2002-02-05 Inspire Pharmaceuticals Inc Method of promoting hydration of mucous membranes with certain uridine, adenine and cytidine diphosphates and their analogs
SE9903290D0 (en) 1999-09-15 1999-09-15 Astra Pharma Prod Novel compounds
SE9904129D0 (en) 1999-11-15 1999-11-15 Astra Pharma Prod Novel compounds
SE9904377D0 (en) 1999-12-01 1999-12-01 Astra Pharma Prod Pharmaceutical combinations
US7018985B1 (en) 2000-08-21 2006-03-28 Inspire Pharmaceuticals, Inc. Composition and method for inhibiting platelet aggregation

Also Published As

Publication number Publication date
AR032631A1 (en) 2003-11-19
US7018985B1 (en) 2006-03-28
US7101860B2 (en) 2006-09-05
JP2004521074A (en) 2004-07-15
AU2001285470B2 (en) 2007-01-25
CN1501940A (en) 2004-06-02
BR0113402A (en) 2005-05-31
KR20040014934A (en) 2004-02-18
WO2002016381B1 (en) 2002-08-15
US20020052337A1 (en) 2002-05-02
EP1311525A2 (en) 2003-05-21
AU8547001A (en) 2002-03-04
CN100465186C (en) 2009-03-04
MXPA03001518A (en) 2004-07-30
WO2002016381A2 (en) 2002-02-28
WO2002016381A3 (en) 2002-05-10
TWI265931B (en) 2006-11-11

Similar Documents

Publication Publication Date Title
AU2001285470B2 (en) Composition and method for inhibiting platelet aggregation
US7132408B2 (en) Composition and method for inhibiting platelet aggregation
AU2001285470A1 (en) Composition and method for inhibiting platelet aggregation
US7452870B2 (en) Drug-eluting stents coated with P2Y12 receptor antagonist compound
JP3083156B2 (en) N-alkyl-2-substituted ATP analogs
CA1333390C (en) Fluorinated nucleosides and process for treating retrovirus infections therewith
US7504497B2 (en) Orally bioavailable compounds and methods for inhibiting platelet aggregation
WO2005040174A1 (en) Tetrahydro-furo`3,4-d!dioxole compounds and compositions and method for inhibiting platelet aggregation
US7612047B2 (en) Degradation-resistant mononucleoside phosphate compounds
US5153180A (en) Fluorinated nucleosides and process for treating retrovirus infections therewith
WO2007140333A2 (en) Mononucleoside phosphonate compounds
US20060258614A1 (en) Pyrimidine-based non-nucleotide composition and method for inhibiting platelet aggregation

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued