WO2016053455A1 - Pyrimidinones as factor xia inhibitors - Google Patents

Pyrimidinones as factor xia inhibitors Download PDF

Info

Publication number
WO2016053455A1
WO2016053455A1 PCT/US2015/042576 US2015042576W WO2016053455A1 WO 2016053455 A1 WO2016053455 A1 WO 2016053455A1 US 2015042576 W US2015042576 W US 2015042576W WO 2016053455 A1 WO2016053455 A1 WO 2016053455A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
independently selected
methyl
chloro
pharmaceutically acceptable
Prior art date
Application number
PCT/US2015/042576
Other languages
English (en)
French (fr)
Inventor
Andrew K. Dilger
James R. Corte
Indawati De Lucca
Tianan Fang
Yang Wu
Yufeng Wang
Kumar Balashanmuga PABBISETTY
William R. Ewing
Yeheng Zhu
Ruth R. Wexler
Donald J.P. Pinto
Michael J. Orwat
Leon M. Smith Ii
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP20197284.1A priority Critical patent/EP3828186B1/en
Priority to EA201790595A priority patent/EA031590B1/ru
Priority to CN201910882473.1A priority patent/CN110734435B/zh
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to JP2017517766A priority patent/JP6462865B2/ja
Priority to TNP/2018/000229A priority patent/TN2018000229A1/en
Priority to PE2020001966A priority patent/PE20210922A1/es
Priority to MYPI2017701150A priority patent/MY183987A/en
Priority to CN202210580097.2A priority patent/CN114957255A/zh
Priority to DK15747721.7T priority patent/DK3089979T3/en
Priority to CN201580053079.7A priority patent/CN106795161B/zh
Priority to EP17189935.4A priority patent/EP3293186B1/en
Priority to LTEP15747721.7T priority patent/LT3089979T/lt
Priority to KR1020177011233A priority patent/KR101921436B1/ko
Priority to SG11201702576QA priority patent/SG11201702576QA/en
Priority to PL15747721T priority patent/PL3089979T3/pl
Priority to ES15747721.7T priority patent/ES2655884T3/es
Priority to TN2017000112A priority patent/TN2017000112A1/en
Priority to MX2017003695A priority patent/MX2017003695A/es
Priority to NZ731416A priority patent/NZ731416A/en
Priority to AU2015324530A priority patent/AU2015324530B2/en
Priority to EP23185426.6A priority patent/EP4286372A3/en
Priority to EP15747721.7A priority patent/EP3089979B1/en
Priority to CA2963395A priority patent/CA2963395C/en
Priority to KR1020187033199A priority patent/KR102269999B1/ko
Priority to RS20180046A priority patent/RS56786B1/sr
Priority to SI201530123T priority patent/SI3089979T1/sl
Priority to BR112017006702-1A priority patent/BR112017006702B1/pt
Priority to MA40123A priority patent/MA40123A1/fr
Publication of WO2016053455A1 publication Critical patent/WO2016053455A1/en
Priority to IL251434A priority patent/IL251434B/en
Priority to PH12017500580A priority patent/PH12017500580B1/en
Priority to ZA2017/02478A priority patent/ZA201702478B/en
Priority to CONC2017/0003833A priority patent/CO2017003833A2/es
Priority to CY20171101314T priority patent/CY1119678T1/el
Priority to HRP20171950TT priority patent/HRP20171950T1/hr
Priority to AU2020200376A priority patent/AU2020200376B2/en
Priority to PH12020500195A priority patent/PH12020500195A1/en
Priority to IL276470A priority patent/IL276470B/en
Priority to CY20201101149T priority patent/CY1123663T1/el
Priority to AU2021245098A priority patent/AU2021245098B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/18Bridged systems
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41621,2-Diazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory 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
    • 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
    • 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
    • A61P7/06Antianaemics
    • 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
    • A61P9/04Inotropic 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
    • A61P9/06Antiarrhythmics
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/61Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings

Definitions

  • the present invention relates generally to novel macrocyclic compounds, and their analogues thereof, which are inhibitors of factor XIa and/or plasma kallikrein, compositions containing them, and methods of using them, for example, for the treatment or prophylaxis of thromboembolic disorders, or for the treatment of retinal vascular permeability associated with diabetic retinopathy and diabetic macular edema.
  • thromboembolic diseases remain the leading cause of death in developed countries despite the availability of anticoagulants such as warfarin (COUMADIN®), heparin, low molecular weight heparins (LMWH), and synthetic pentasaccharides and antiplatelet agents such as aspirin and clopidogrel (PLAVIX®).
  • the oral anticoagulant warfarin inhibits the post-translational maturation of coagulation factors VII, IX, X and prothrombin, and has proven effective in both venous and arterial thrombosis.
  • its usage is limited due to its narrow therapeutic index, slow onset of therapeutic effect, numerous dietary and drug interactions, and a need for monitoring and dose adjustment.
  • safe and efficacious oral anticoagulants for the prevention and treatment of a wide range of thromboembolic disorders has become increasingly important.
  • Factor XIa is a plasma serine protease involved in the regulation of blood coagulation, which is initiated in vivo by the binding of tissue factor (TF) to factor VII (FVII) to generate factor Vila (FVIIa).
  • the resulting TF:FVIIa complex activates factor IX (FIX) and factor X (FX) that leads to the production of factor Xa (FXa).
  • the generated FXa catalyzes the transformation of prothrombin into small amounts of thrombin before this pathway is shut down by tissue factor pathway inhibitor (TFPI).
  • TFPI tissue factor pathway inhibitor
  • the process of coagulation is then further propagated via the feedback activation of Factors V, VIII and XI by catalytic amounts of thrombin.
  • Plasma prekallikrein is a zymogen of a trypsin-like serine protease and is present in plasma at 35 to 50 ⁇ g/mL.
  • the gene structure is similar to that of factor XI.
  • the amino acid sequence of plasma kallikrein has 58% homology to factor XI.
  • Plasma kallikrein is thought to play a role in a number of inflammatory disorders.
  • the major inhibitor of plasma kallikrein is the serpin C 1 esterase inhibitor. Patients who present with a genetic deficiency in CI esterase inhibitor suffer from hereditary angioedema
  • HAE HAE which results in intermittent swelling of face, hands, throat, gastro-intestinal tract and genitals. Blisters formed during acute episodes contain high levels of plasma kallikrein which cleaves high molecular weight kininogen liberating bradykinin leading to increased vascular permeability. Treatment with a large protein plasma kallikrein inhibitor has been shown to effectively treat HAE by preventing the release of bradykinin which causes increased vascular permeability (Lehmann, A., "Ecallantide (DX-88), a plasma kallikrein inhibitor for the treatment of hereditary angioedema and the prevention of blood loss in on-pump cardiothoracic surgery", Expert Opin. Biol. Ther., 8: 187-199 (2008)).
  • the plasma kallikrein-kinin system is abnormally abundant in patients with advanced diabetic macular edema. It has been recently published that plasma kallikrein contributes to retinal vascular dysfunctions in diabetic rats (Clermont, A. et al., "Plasma kallikrein mediates retinal vascular dysfunction and induces retinal thickening in diabetic rats", Diabetes, 60:1590-1598 (2011)). Furthermore, administration of the plasma kallikrein inhibitor ASP-440 ameliorated both retinal vascular permeability and retinal blood flow abnormalities in diabetic rats. Therefore, a plasma kallikrein inhibitor should have utility as a treatment to reduce retinal vascular permeability associated with diabetic retinopathy and diabetic macular edema. Other complications of diabetes such as cerebral hemorrhage, nephropathy, cardiomyopathy and neuropathy, all of which have
  • association with plasma kallikrein may also be considered as targets for a plasma kallikrein inhibitor.
  • no small molecule synthetic plasma kallikrein inhibitor has been approved for medical use.
  • the large protein plasma kallikrein inhibitors present risks of anaphylactic reactions, as has been reported for Ecallantide.
  • the molecules in the known art feature a highly polar and ionizable guanidine or amidine functionality. It is well known that such functionalities may be limiting to gut permeability and therefore to oral availability.
  • the present invention provides novel macrocyclic compounds, their analogues, including stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof, which are useful as selective inhibitors of serine protease enzymes, especially factor XIa and/or plasma kallikrein.
  • the present invention also provides processes and intermediates for making the compounds of the present invention.
  • the present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.
  • the compounds of the invention may be used in the treatment and/or prophylaxis of thromboembolic disorders.
  • the compounds of the invention may be used in the treatment of retinal vascular permeability associated with diabetic retinopathy and diabetic macular edema.
  • the compounds of the present invention may be used in therapy.
  • the compounds of the present invention may be used for the manufacture of a medicament for the treatment and/or prophylaxis of a thromboembolic disorder.
  • the compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more, preferably one to two other agent(s).
  • the present invention provides, inter alia, compounds of Formula (I):
  • ring A is independently selected from ;
  • ring B is independently selected from
  • Pv 1 is independently selected from H and Ci_ 4 alkyl
  • R is independently selected from H, F, CI, CF 3 , and CHF 2 ;
  • R is independently selected from H, CHF 2 , CD 3 , CH 3 , and
  • R 4 is independently selected from H and F
  • R 5 is independently selected from H, F, CI, CH 3 , and OCH 3 .
  • the present invention provides compounds of Formula (I), or stereoisomers, tautomers, pharmaceutically acceptable salts, solvates, or prodrugs thereof, wherein:
  • ring A is independently selected from and
  • ring B is independently selected from and
  • R 1 is independently selected from H and Ci_ 4 alkyl
  • R is independently selected from F, CI, CF 3 , CHF 2 , and COOH;
  • R 3 is independently selected from H, CHF 2 , CD 3 , and CH 3 ;
  • R 4 is independently selected from H and F
  • R 5 is independently selected from H, F, CI, CH 3 , and OCH 3 .
  • the present invention provides compounds of Formula (II):
  • R 1 is Ci_ 4 alkyl
  • R 2 is independently selected from H, F, CI, CF 3 , and CHF 2 ;
  • R 3 is independently selected from CHF 2 , CD 3 , and CH 3 ;
  • R 4 is H
  • R 5 is independently selected from F and CI.
  • the present invention provides compounds of Formula (II), or stereoisomers, tautomers, pharmaceutically acceptable salts, solvates, or prodrugs thereof, wherein: R 1 is Ci_4 alkyl;
  • R 2 is independently selected from F, CI, CF 3 , and CHF 2 ;
  • R 3 is independently selected from CHF 2 , CD 3 , and CH 3 ;
  • R 4 is H
  • R 5 is independently selected from F and CI.
  • the present invention provides compounds of Formula (III):
  • R 1 is Ci_4 alkyl
  • R 2 is independently selected from H, F, CI, CF 3 , and CHF 2 ;
  • R 3 is independently selected from CHF 2 , CD 3 , and CH 3 ;
  • R 4 is H
  • R 5 is independently selected from F and CI.
  • the present invention provides compounds of Formula (III), or stereoisomers, tautomers, pharmaceutically acceptable salts, solvates, or prodrugs thereof, wherein:
  • R 1 is Ci_4 alkyl
  • R 2 is independently selected from F, CI, CF 3 , and CHF 2 ;
  • R 3 is independently selected from CHF 2 , CD 3 , and CH 3 ;
  • R 4 is H
  • R 5 is independently selected from F and CI.
  • the present invention provides compounds of Formula (IV):
  • R 2 is independently selected from F, CI, CF 3 , and CHF 2 ;
  • R 3 is independently selected from CHF 2 , CD 3 , and CH 3 .
  • the present invention provides compounds of Formula (I), or stereoisomers, tautomers, pharmaceutically acceptable salts, solvates, or prodrugs thereof, wherein:
  • ring A is independently selected from
  • R 1 is independently selected from H and Ci_ 4 alkyl
  • R 2 is COOH
  • R 3 is independently selected from H, CHF 2 , CD 3 , and CH 3 ;
  • R 4 is independently selected from H and F
  • R 5 is independently selected from H, F, CI, CH 3 , and OCH 3 .
  • the present invention provides compounds selected from
  • R 1 is independently selected from the group consisting of
  • R 1 is independently selected from the group consisting of H and methyl, ethyl, and isopropyl.
  • the present invention provides a compound selected from any subset list of compounds exemplified in the present application.
  • the compounds of the present invention have Factor XIa or plasma kallikrein Ki values ⁇ 10 ⁇ .
  • the compounds of the present invention have Factor XIa or plasma kallikrein Ki values ⁇ 1 ⁇ .
  • the compounds of the present invention have Factor XIa or plasma kallikrein Ki values ⁇ 0.5 ⁇ .
  • the compounds of the present invention have Factor XIa or plasma kallikrein Ki values ⁇ 0.1 ⁇ .
  • the present invention provides a composition comprising at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate, thereof.
  • the present invention provides a pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
  • the present invention provides a process for making a compound of the present invention.
  • the present invention provides an intermediate for making a compound of the present invention.
  • the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s).
  • the present invention provides pharmaceutical composition, wherein the additional therapeutic agent(s) are an anti-platelet agent or a combination thereof.
  • the anti-platelet agent(s) are clopidogrel and/or aspirin, or a combination thereof.
  • the present invention provides a method for the treatment and/or prophylaxis of a thromboembolic disorder comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
  • the present invention provides a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, for use in therapy.
  • the present invention provides a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, for use in therapy for the treatment and/or prophylaxis of a thromboembolic disorder.
  • the present invention also provides the use of a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, for the manufacture of a medicament for the treatment and/or prophylaxis of a thromboembolic disorder.
  • the present invention provides a method for treatment and/or prophylaxis of a thromboembolic disorder, comprising: administering to a patient in need thereof a therapeutically effective amount of a first and second therapeutic agent, wherein the first therapeutic agent is a compound of the present invention or a
  • the second therapeutic agent is at least one agent selected from a factor Xa inhibitor such as apixaban, rivaroxaban, betrixaban, edoxaban, an anti-coagulant agent, an anti-platelet agent, a thrombin inhibiting agent such as dabigatran, a thrombolytic agent, and a fibrinolytic agent.
  • a factor Xa inhibitor such as apixaban, rivaroxaban, betrixaban, edoxaban
  • an anti-coagulant agent an anti-platelet agent
  • a thrombin inhibiting agent such as dabigatran, a thrombolytic agent, and a fibrinolytic agent.
  • the second therapeutic agent is at least one agent selected from warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatroban, aspirin, ibuprofen, naproxen, sulindac,
  • the second therapeutic agent is at least one anti-platelet agent.
  • the anti-platelet agent(s) are clopidogrel and/or aspirin, or a
  • the thromboembolic disorder includes arterial cardiovascular thromboembolic disorders, venous cardiovascular thromboembolic disorders, arterial cerebrovascular thromboembolic disorders, and venous cerebrovascular thromboembolic disorders.
  • thromboembolic disorder examples include, but are not limited to, unstable angina, an acute coronary syndrome, atrial fibrillation, first myocardial infarction, recurrent myocardial infarction, ischemic sudden death, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary arterial thrombosis, cerebral arterial thrombosis, cerebral embolism, kidney embolism, pulmonary embolism, and thrombosis resulting from medical implants, devices, or procedures in which blood is exposed to an artificial surface that promotes thrombosis.
  • the present invention provides a method for the treatment and/or prophylaxis of an inflammatory disorder comprising: administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
  • the inflammatory disorder include, but are not limited to, sepsis, acute respiratory distress syndrome, and systemic inflammatory response syndrome.
  • the present invention provides a method for the prophylaxis of a disease or condition in which plasma kallikrein activity is implicated comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
  • the disease or condition in which plasma kallikrein activity is implicated includes, but not limited to, impaired visual acuity, diabetic retinopathy, diabetic macular edema, hereditary angioedema, diabetes, pancreatitis, nephropathy, cardio myopathy, neuropathy, inflammatory bowel disease, arthritis, inflammation, septic shock, hypotension, cancer, adult respiratory distress syndrome, disseminated intravascular coagulation, and cardiopulmonary bypass surgery.
  • the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in therapy.
  • the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in treatment and/or prophylaxis of a thromboembolic disorder.
  • Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by
  • the end products of the present invention are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the invention. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers.
  • Compounds of the present invention, free form and salts thereof may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention.
  • stereoisomer refers to isomers of identical constitution that differ in the arrangement of their atoms in space. Enantiomers and diastereomers are examples of stereoisomers.
  • enantiomer refers to one of a pair of molecular species that are mirror images of each other and are not superimposable.
  • diastereomer refers to stereoisomers that are not mirror images.
  • racemate or “racemic mixture” refers to a composition composed of equimolar quantities of two enantiomeric species, wherein the composition is devoid of optical activity.
  • R and S represent the configuration of substituents around a chiral carbon atom(s).
  • the isomeric descriptors “R” and “S” are used as described herein for indicating atom configuration(s) relative to a core molecule and are intended to be used as defined in the literature (IUPAC Recommendations 1996, Pure and Applied Chemistry, 68:2193-2222 (1996)).
  • chiral refers to the structural characteristic of a molecule that makes it impossible to superimpose it on its mirror image.
  • homochiral refers to a state of enantiomeric purity.
  • optical activity refers to the degree to which a homochiral molecule or nonracemic mixture of chiral molecules rotates a plane of polarized light.
  • alkyl or “alkylene” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • “Ci to C 10 alkyl” or “C 1-10 alkyl” (or alkylene) is intended to include C l s C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , and C 10 alkyl groups.
  • Ci to C 6 alkyl or “Ci-C 6 alkyl” denotes alkyl having 1 to 6 carbon atoms.
  • Alkyl group can be unsubstituted or substituted with at least one hydrogen being replaced by another chemical group.
  • Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl ⁇ e.g. , n-propyl and isopropyl), butyl ⁇ e.g. , n-butyl, isobutyl, t- butyl), and pentyl ⁇ e.g. , n-pentyl, isopentyl, neopentyl).
  • methyl (Me) ethyl
  • Et propyl ⁇ e.g. , n-propyl and isopropyl
  • pentyl ⁇ e.g. , n-pentyl, isopentyl, neopentyl.
  • Alkynyl or “alkynylene” is intended to include hydrocarbon chains of either straight or branched configuration having one or more, preferably one to three, carbon- carbon triple bonds that may occur in any stable point along the chain.
  • C 2 to C 6 alkynyl or “C 2 _ 6 alkynyl” (or alkynylene) is intended to include C 2 , C 3 , C 4 , C 5 , and C 6 alkynyl groups; such as ethynyl, propynyl, butynyl, pentynyl, and hexynyl.
  • alkoxy refers to an -O-alkyl group.
  • Ci to C 6 alkoxy or “Ci_6 alkoxy” (or alkyloxy) is intended to include C l s C 2 , C 3 , C 4 , C 5 , and C 6 alkoxy groups.
  • Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy ⁇ e.g. , n-propoxy and isopropoxy), and t-butoxy.
  • alkylthio or “thioalkoxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example, methyl-S- and ethyl-S-.
  • Halo or halogen includes fluoro, chloro, bromo, and iodo.
  • Haloalkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogens.
  • haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl.
  • haloalkyl also include "fluoroalkyl” that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more fluorine atoms.
  • Haloalkoxy or "haloalkyloxy” represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge.
  • “Ci to C 6 haloalkoxy” or “Ci_ 6 haloalkoxy” is intended to include Ci, C 2 , C 3 , C 4 , C 5 , and C 6 haloalkoxy groups.
  • Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy.
  • haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy.
  • haloalkylthio or “thiohaloalkoxy” represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example, trifluoromethyl-S-, and pentaf uoroethyl-S-.
  • amino refers to -NH 2 .
  • substituted amino refers to the defined terms below having the suffix "amino” such as “arylamino”, “alkylamino”, “arylamino”, etc.
  • alkoxycarbonyl refers to an alkoxy group attached to the parent molecular moiety through a carbonyl group.
  • alkoxy carbonylamino refers to an -NHR wherein R is an alkoxycarbonyl group.
  • alkylamino refers to -NHR, wherein R is an alkyl group.
  • alkylcarbonyl refers to an alkyl group attached to the parent molecular moiety through a carbonyl group.
  • alkylcarbonylamino refers to -NHR wherein R is an alkylcarbonyl group.
  • aminosulfonyl refers to -S0 2 NH 2 .
  • arylalkyl refers to an alkyl group substituted with one, two, or three aryl groups.
  • arylamino refers to -NHR wherein R is an aryl group.
  • arylcarbonyl refers to an aryl group attached to the parent molecular moiety through a carbonyl group.
  • arylcarbonylamino refers to -NHR wherein R is an arylcarbonyl group.
  • carbonyl refers to -C(O)-.
  • cyano refers to -CN.
  • cycloalkylamino refers to -NHR wherein R is a cycloalkyl group.
  • cycloalkylcarbonyl refers to a cycloalkyl group attached to the parent molecular moiety through a carbonyl group.
  • cycloalkylcarbonylamino refers to -NHR wherein R is a cycloalkylcarbonyl group.
  • cycloalkyloxy refers to a cycloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • dialkylamino refers to NR 2 , wherein each R is an alkyl group.
  • the two alkyl groups are the same or different.
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl refers to an alkyl group substituted by one, two, three, or four halogen atoms.
  • haloalkylamino refers to -NHR wherein R is a haloalkyl group.
  • haloalkylcarbonyl refers to a haloalkyl group attached to the parent molecular moiety through a carbonyl group.
  • haloalkylcarbonylamino refers to -NHR wherein R is a haloalkylcarbonyl group.
  • alkylcarbonyl refers to an alkyl or substituted alkyl bonded to a carbonyl.
  • alkoxycarbonyl refers to an alkoxy group attached to the parent molecular moiety through a carbonyl group.
  • hydroxy or “hydroxyl” refers to OH.
  • cycloalkyl refers to cyclized alkyl groups, including mono-, bi- or poly-cyclic ring systems.
  • C 3 to C 7 cycloalkyl or “C 3 _ 7 cycloalkyl” is intended to include C 3 , C 4 , C 5 , C 6 , and C 7 cycloalkyl groups.
  • Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbornyl.
  • Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included in the definition of "cycloalkyl”.
  • carrier or “carbocyclic residue” is intended to mean any stable 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 1 1-, 12-, or 13-membered bicyclic or tricyclic hydrocarbon ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic.
  • carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane,
  • bridged rings are also included in the definition of carbocycle (e.g., [2.2.2]bicyclooctane).
  • Preferred carbocycles are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, and indanyl.
  • a bridged ring occurs when one or more carbon atoms link two non- adjacent carbon atoms. Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.
  • bicyclic carbocycle or "bicyclic carbocyclic group” is intended to mean a stable 9- or 10-membered carbocyclic ring system that contains two fused rings and consists of carbon atoms. Of the two fused rings, one ring is a benzo ring fused to a second ring; and the second ring is a 5- or 6-membered carbon ring which is saturated, partially unsaturated, or unsaturated.
  • the bicyclic carbocyclic group may be attached to its pendant group at any carbon atom which results in a stable structure.
  • the bicyclic carbocyclic group described herein may be substituted on any carbon if the resulting compound is stable.
  • bicyclic carbocyclic group examples include, but not limited to, naphthyl, 1 ,2-dihydronaphthyl, 1 ,2,3,4-tetrahydronaphthyl, and indanyl.
  • Aryl groups refer to monocyclic or polycyclic aromatic hydrocarbons, including, for example, phenyl, naphthyl, and phenanthranyl. Aryl moieties are well known and described, for example, in Lewis, R.J., ed., Hawley's Condensed Chemical Dictionary, 13th Edition, John Wiley & Sons, Inc., New York (1997).
  • heterocycle or “heterocyclic ring” is intended to mean a stable 3-, 4-, 5-, 6-, or 7-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered polycyclic heterocyclic ring that is saturated, partially unsaturated, or fully unsaturated, and that contains carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the nitrogen and sulfur heteroatoms may optionally be oxidized ⁇ i.e., N ⁇ 0 and S(0) p , wherein p is 0, 1 or 2).
  • the nitrogen atom may be substituted or unsubstituted ⁇ i.e., N or NR wherein R is H or another substituent, if defined).
  • the heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure.
  • the heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable.
  • a nitrogen in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1 , then these heteroatoms are not adjacent to one another.
  • heterocycle it is intended to include heteroaryl.
  • heterocycles include, but are not limited to, acridinyl, azetidinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl,
  • Examples of 5- to 10-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyl, benzimidazolyl, lH-indazolyl, benzofuranyl, benzothiofuranyl, benztetrazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, oxindolyl, benzoxazolinyl,
  • Examples of 5- to 6-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.
  • bicyclic heterocycle or "bicyclic heterocyclic group” is intended to mean a stable 9- or 10-membered heterocyclic ring system which contains two fused rings and consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S.
  • one ring is a 5- or 6-membered monocyclic aromatic ring comprising a 5-membered heteroaryl ring, a 6- membered heteroaryl ring or a benzo ring, each fused to a second ring.
  • the second ring is a 5- or 6-membered monocyclic ring which is saturated, partially unsaturated, or unsaturated, and comprises a 5-membered heterocycle, a 6-membered heterocycle or a carbocycle (provided the first ring is not benzo when the second ring is a carbocycle).
  • the bicyclic heterocyclic group may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure.
  • the bicyclic heterocyclic group described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1.
  • bicyclic heterocyclic group examples include quinolinyl, isoquinolinyl, phthalazinyl, quinazolinyl, indolyl, isoindolyl, indolinyl, lH-indazolyl, benzimidazolyl, 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 5,6,7,8- tetrahydro-quinolinyl, 2,3-dihydro-benzofuranyl, chromanyl, 1,2,3,4-tetrahydro- quinoxalinyl, and 1,2,3,4-tetrahydro-quinazolinyl.
  • aromatic heterocyclic group or "heteroaryl” is intended to mean stable monocyclic and polycyclic aromatic hydrocarbons that include at least one heteroatom ring member such as sulfur, oxygen, or nitrogen.
  • Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4- thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl,
  • benzodioxolanyl and benzodioxane.
  • Heteroaryl groups are substituted or unsubstituted.
  • the nitrogen atom is substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined).
  • the nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N ⁇ 0 and S(0) p , wherein p is 0, 1 or 2).
  • Bridged rings are also included in the definition of heterocycle.
  • a bridged ring occurs when one or more atoms (i.e., C, O, N, or S) link two non-adjacent carbon or nitrogen atoms.
  • Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.
  • counterion is used to represent a negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate.
  • a dotted ring When a dotted ring is used within a ring structure, this indicates that the ring structure may be saturated, partially saturated or unsaturated.
  • substituted means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound.
  • 2 hydrogens on the atom are replaced.
  • Keto substituents are not present on aromatic moieties.
  • a ring system e.g., carbocyclic or heterocyclic
  • Ring double bonds are double bonds that are formed between two adjacent ring atoms (e.g.,
  • nitrogen atoms e.g. , amines
  • these may be converted to N-oxides by treatment with an oxidizing agent (e.g. , mCPBA and/or hydrogen peroxides) to afford other compounds of this invention.
  • an oxidizing agent e.g. , mCPBA and/or hydrogen peroxides
  • shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N— >0) derivative.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, and/or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic,
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 18th Edition, Mack Publishing Company, Easton, PA (1990), the disclosure of which is hereby incorporated by reference.
  • compounds of formula I may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent ⁇ i.e. , a compound of formula I) is a prodrug within the scope and spirit of the invention.
  • a prodrug within the scope and spirit of the invention.
  • Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:
  • Bundgaard, H. Chapter 5, “Design and Application of Prodrugs", A Textbook of Drug Design and Development, pp. 1 13-191 , Krosgaard-Larsen, P. et al, eds., Harwood Academic Publishers (1991);
  • Compounds containing a carboxy group can form physiologically hydrolyzable esters that serve as prodrugs by being hydrolyzed in the body to yield formula I compounds per se.
  • Such prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes.
  • Parenteral administration may be used where the ester per se is active, or in those instances where hydrolysis occurs in the blood. Examples of physiologically
  • hydrolyzable esters of compounds of formula I include Ci_ 6 alkyl, Ci_ 6 alkylbenzyl, 4- methoxybenzyl, indanyl, phthalyl, methoxymethyl, Ci_ 6 alkanoyloxy-Ci_ 6 alkyl (e.g.
  • Ci_ 6 alkoxycarbonyloxy- Ci_ 6 alkyl e.g., methoxycarbonyl-oxymethyl or ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyl-2-oxo- 1 ,3-dioxolen-4-yl)-methyl
  • esters may be prepared by conventional techniques known in the art.
  • the present invention is intended to include all isotopes of atoms occurring in the present compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include deuterium and tritium.
  • Deuterium has one proton and one neutron in its nucleus and that has twice the mass of ordinary hydrogen.
  • Deuterium can be represented by symbols such as " 2 H” or "D”.
  • Isotopes of carbon include 13 C and 14 C.
  • Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed. Such compounds have a variety of potential uses, e.g., as standards and reagents in determining the ability of a potential
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. It is preferred that compounds of the present invention do not contain a N-halo, S(0) 2 H, or S(0)H group.
  • solvate means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • the solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement.
  • the solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules.
  • “Solvate” encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
  • the compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis, which are described in more detail in Section VI.
  • blood coagulation is essential to the regulation of an organism's hemostasis, it is also involved in many pathological conditions.
  • a blood clot, or thrombus may form and obstruct circulation locally, causing ischemia and organ damage.
  • embolism the clot may dislodge and subsequently become trapped in a distal vessel, where it again causes ischemia and organ damage.
  • thromboembolic disorders Diseases arising from pathological thrombus formation are collectively referred to as thromboembolic disorders and include acute coronary syndrome, unstable angina, myocardial infarction, thrombosis in the cavity of the heart, ischemic stroke, deep vein thrombosis, peripheral occlusive arterial disease, transient ischemic attack, and pulmonary embolism.
  • thrombosis occurs on artificial surfaces in contact with blood, including catheters, stents, artificial heart valves, and hemodialysis membranes.
  • Some conditions contribute to the risk of developing thrombosis. For example, alterations of the vessel wall, changes in the flow of blood, and alterations in the composition of the vascular compartment. These risk factors are collectively known as Virchow's triad. (Colman, R.W. et al., eds., Hemostasis and Thrombosis, Basic Principles and Clinical Practice, Fifth Edition, p. 853, Lippincott Williams &
  • Antithrombotic agents are frequently given to patients at risk of developing thromboembolic disease because of the presence of one or more predisposing risk factors from Virchow's triad to prevent formation of an occlusive thrombus (primary prevention).
  • primary prevention For example, in an orthopedic surgery setting ⁇ e.g. , hip and knee replacement, an antithrombotic agent is frequently administered prior to a surgical procedure.
  • the antithrombotic agent counterbalances the prothrombotic stimulus exerted by vascular flow alterations (stasis), potential surgical vessel wall injury, as well as changes in the composition of the blood due to the acute phase response related to surgery.
  • an antithrombotic agent for primary prevention is dosing with aspirin, a platelet activation inhibitor, in patients at risk for developing thrombotic cardiovascular disease.
  • well recognized risk factors in this setting include age, male gender, hypertension, diabetes mellitus, lipid alterations, and obesity.
  • Antithrombotic agents are also indicated for secondary prevention, following an initial thrombotic episode.
  • patients with mutations in factor V also known as factor V Leiden
  • additional risk factors ⁇ e.g., pregnancy are dosed with anticoagulants to prevent the reoccurrence of venous thrombosis.
  • Another example entails secondary prevention of cardiovascular events in patients with a history of acute myocardial infarction or acute coronary syndrome.
  • a combination of aspirin and clopidogrel or other thienopyridines may be used to prevent a second thrombotic event.
  • Antithrombotic agents are also given to treat the disease state (i.e., by arresting its development) after it has already started.
  • patients presenting with deep vein thrombosis are treated with anticoagulants (i.e., heparin, warfarin, or LMWH) to prevent further growth of the venous occlusion. Over time, these agents also cause a regression of the disease state because the balance between prothrombotic factors and
  • anticoagulant/pro fibrinolytic pathways is changed in favor of the latter.
  • Examples on the arterial vascular bed include the treatment of patients with acute myocardial infarction or acute coronary syndrome with aspirin and clopidogrel to prevent further growth of vascular occlusions and eventually leading to a regression of thrombotic occlusions.
  • antithrombotic agents are used widely for primary and secondary prevention (i.e., prophylaxis or risk reduction) of thromboembolic disorders, as well as treatment of an already existing thrombotic process.
  • Drugs that inhibit blood coagulation, or anticoagulants are "pivotal agents for prevention and treatment of thromboembolic disorders" (Hirsh, J. et al, Blood, 105:453-463 (2005)).
  • prolylcarboxylpeptidase can activate plasma kallikrein complexed with high molecular weight kininogen in a multiprotein complex formed on the surface of cells and matrices (Shariat-Madar et al., Blood, 108:192-199 (2006)).
  • Contact activation is a surface mediated process responsible in part for the regulation of thrombosis and inflammation, and is mediated, at least in part, by fibrinolytic-, complement-, kininogen/kinin-, and other humoral and cellular pathways (for review, Coleman, R., "Contact Activation Pathway", Hemostasis and Thrombosis, pp.
  • factor XII deficient mice More specifically, factor XII deficient mice were protected from thrombotic vascular occlusion in several thrombosis models as well as stroke models and the phenotype of the XII deficient mice was identical to XI deficient mice (Renne et al, J. Exp. Med., 202:271-281 (2005);
  • factor XI is downstream from factor Xlla, combined with the identical phenotype of the XII and XI deficient mice suggest that the contact activation system could play a major role in factor XI activation in vivo.
  • Factor XI is a zymogen of a trypsin-like serine protease and is present in plasma at a relatively low concentration. Proteolytic activation at an internal R369-1370 bond yields a heavy chain (369 amino acids) and a light chain (238 amino acids). The latter contains a typical trypsin-like catalytic triad (H413, D464, and S557). Activation of factor XI by thrombin is believed to occur on negatively charged surfaces, most likely on the surface of activated platelets. Platelets contain high affinity (0.8 nM) specific sites (130-500/platelet) for activated factor XI. After activation, factor XIa remains surface bound and recognizes factor IX as its normal macromolecular substrate. (Galiani, D., Trends Cardiovasc. Med, 10: 198-204 (2000)).
  • thrombin activates thrombin activated fibrinolysis inhibitor (TAFI), a plasma carboxypeptidase that cleaves C-terminal lysine and arginine residues on fibrin, reducing the ability of fibrin to enhance tissue-type plasminogen activator (tPA) dependent plasminogen activation.
  • TAFI thrombin activated fibrinolysis inhibitor
  • tPA tissue-type plasminogen activator
  • baboon cross-reactive, function blocking antibodies to human factor XI protect against baboon arterial-venous shunt thrombosis (Gruber et al., Blood, 102:953- 955 (2003)).
  • Evidence for an antithrombotic effect of small molecule inhibitors of factor XIa is also disclosed in published U.S. Patent Publication No. 2004/0180855 Al . Taken together, these studies suggest that targeting factor XI will reduce the propensity for thrombotic and thromboembolic diseases.
  • factor XI is not required for normal homeostasis, implying a superior safety profile of the factor XI mechanism compared to competing antithrombotic mechanisms.
  • hemophilia A factor VIII deficiency
  • hemophilia B factor IX deficiency
  • mutations of the factor XI gene causing factor XI deficiency result in only a mild to moderate bleeding diathesis characterized primarily by postoperative or posttraumatic, but rarely spontaneous hemorrhage. Postoperative bleeding occurs mostly in tissue with high concentrations of endogenous fibrinolytic activity (e.g., oral cavity, and urogenital system). The majority of the cases are fortuitously identified by preoperative prolongation of aPTT (intrinsic system) without any prior bleeding history.
  • aPTT intrinsic system
  • factor XI activation can be determined by complex formation with either CI inhibitor or alpha 1 antitrypsin.
  • AMI acute myocardial infarction
  • This study can be viewed as evidence that at least in a subpopulation of patients with AMI, factor XI activation contributes to thrombin formation (Minnema, M.C. et al., Arterioscler. Thromb. Vase. Biol., 20:2489-2493
  • aPTT activated partial thromboplastin time
  • PT prothrombin time
  • patient encompasses all mammalian species.
  • treating cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) inhibiting the disease-state, i.e., arresting it development; and/or (b) relieving the disease-state, i.e., causing regression of the disease state.
  • prophylaxis is the protective treatment of a disease state to reduce and/or minimize the risk and/or reduction in the risk of recurrence of a disease state by administering to a patient a therapeutically effective amount of at least one of the compounds of the present invention or a or a stereoisomer, a tautomer, a
  • Patients may be selected for prophylaxis therapy based on factors that are known to increase risk of suffering a clinical disease state compared to the general population.
  • prophylaxis treatment conditions of the clinical disease state may or may not be presented yet.
  • "Prophylaxis" treatment can be divided into (a) primary prophylaxis and (b) secondary prophylaxis.
  • Primary prophylaxis is defined as treatment to reduce or minimize the risk of a disease state in a patient that has not yet presented with a clinical disease state
  • secondary prophylaxis is defined as minimizing or reducing the risk of a recurrence or second occurrence of the same or similar clinical disease state.
  • risk reduction covers therapies that lower the incidence of development of a clinical disease state.
  • primary and secondary prevention therapies are examples of risk reduction.
  • “Therapeutically effective amount” is intended to include an amount of a compound of the present invention that is effective when administered alone or in combination to inhibit factor XIa and/or plasma kallikrein and/or to prevent or treat the disorders listed herein.
  • the term refers to combined amounts of the active ingredients that result in the preventive or therapeutic effect, whether administered in combination, serially, or simultaneously.
  • thrombosis refers to formation or presence of a thrombus (pi. thrombi); clotting within a blood vessel that may cause ischemia or infarction of tissues supplied by the vessel.
  • emblism refers to sudden blocking of an artery by a clot or foreign material that has been brought to its site of lodgment by the blood current.
  • thromboembolism refers to obstruction of a blood vessel with thrombotic material carried by the blood stream from the site of origin to plug another vessel.
  • thromboembolic disorders entails both "thrombotic” and “embolic” disorders (defined above).
  • thromboembolic disorders as used herein includes arterial pressure
  • cardiovascular thromboembolic disorders also includes specific disorders selected from, but not limited to, unstable angina or other acute coronary syndromes, atrial fibrillation, first or recurrent myocardial infarction, ischemic sudden death, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary arterial thrombosis, cerebral arterial thrombosis, cerebral embolism, kidney embolism, pulmonary embolism, and thrombosis resulting from medical implants, devices, or procedures in which blood is exposed to an artificial surface that promotes thrombosis.
  • the medical implants or devices include, but are not limited to: prosthetic valves, artificial valves, indwelling catheters, stents, blood oxygenators, shunts, vascular access ports, ventricular assist devices and artificial hearts or heart chambers, and vessel grafts.
  • the procedures include, but are not limited to:
  • thromboembolic disorders includes acute coronary syndrome, stroke, deep vein thrombosis, and pulmonary embolism.
  • the present invention provides a method for the treatment of a thromboembolic disorder, wherein the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary arterial thrombosis, cerebral arterial thrombosis, cerebral embolism, kidney embolism, pulmonary embolism, and thrombosis resulting from medical implants, devices, or procedures in which blood is exposed to an artificial surface that promotes thrombosis.
  • the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis,
  • the present invention provides a method for the treatment of a thromboembolic disorder, wherein the thromboembolic disorder is selected from acute coronary syndrome, stroke, venous thrombosis, atrial fibrillation, and thrombosis resulting from medical implants and devices.
  • the present invention provides a method for the primary prophylaxis of a thromboembolic disorder, wherein the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, myocardial infarction, ischemic sudden death, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary arterial thrombosis, cerebral arterial thrombosis, cerebral embolism, kidney embolism, pulmonary embolism, and thrombosis resulting from medical implants, devices, or procedures in which blood is exposed to an artificial surface that promotes thrombosis.
  • the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, myocardial infarction, ischemic sudden death, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease,
  • the present invention provides a method for the primary prophylaxis of a thromboembolic disorder, wherein the thromboembolic disorder is selected from acute coronary syndrome, stroke, venous thrombosis, and thrombosis resulting from medical implants and devices.
  • the present invention provides a method for the secondary prophylaxis of a thromboembolic disorder, wherein the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, recurrent myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombosis, deep vein thrombosis, thrombophlebitis, arterial embolism, coronary arterial thrombosis, cerebral arterial thrombosis, cerebral embolism, kidney embolism, pulmonary embolism, and thrombosis resulting from medical implants, devices, or procedures in which blood is exposed to an artificial surface that promotes thrombosis.
  • the thromboembolic disorder is selected from unstable angina, an acute coronary syndrome, atrial fibrillation, recurrent myocardial infarction, transient ischemic attack, stroke, atherosclerosis, peripheral occlusive arterial disease, venous thrombo
  • the present invention provides a method for the secondary prophylaxis of a thromboembolic disorder, wherein the thromboembolic disorder is selected from acute coronary syndrome, stroke, atrial fibrillation and venous thrombosis.
  • stroke refers to embolic stroke or atherothrombotic stroke arising from occlusive thrombosis in the carotid communis, carotid interna, or intracerebral arteries.
  • thrombosis includes vessel occlusion (e.g., after a bypass) and reocclusion (e.g., during or after percutaneous transluminal coronary angioplasty).
  • the thromboembolic disorders may result from conditions including but not limited to atherosclerosis, surgery or surgical complications, prolonged immobilization, arterial fibrillation, congenital thrombophilia, cancer, diabetes, effects of medications or hormones, and complications of pregnancy.
  • Thromboembolic disorders are frequently associated with patients with atherosclerosis.
  • Risk factors for atherosclerosis include but are not limited to male gender, age, hypertension, lipid disorders, and diabetes mellitus. Risk factors for atherosclerosis are at the same time risk factors for complications of atherosclerosis, i.e., thromboembolic disorders.
  • thromboembolic disorders Similarly, arterial fibrillation is frequently associated with thromboembolic disorders. Risk factors for arterial fibrillation and subsequent thromboembolic disorders include cardiovascular disease, rheumatic heart disease, nonrheumatic mitral valve disease, hypertensive cardiovascular disease, chronic lung disease, and a variety of miscellaneous cardiac abnormalities as well as thyrotoxicosis.
  • Risk factors for the more common type 2 include but are not limited to are family history, obesity, physical inactivity, race/ethnicity, previously impaired fasting glucose or glucose tolerance test, history of gestational diabetes mellitus or delivery of a "big baby", hypertension, low HDL cholesterol, and polycystic ovary syndrome.
  • Risk factors for congenital thrombophilia include gain of function mutations in coagulation factors or loss of function mutations in the anticoagulant- or fibrinolytic pathways.
  • Thrombosis has been associated with a variety of tumor types, e.g., pancreatic cancer, breast cancer, brain tumors, lung cancer, ovarian cancer, prostate cancer, gastrointestinal malignancies, and Hodgkins or non-Hodgkins lymphoma. Recent studies suggest that the frequency of cancer in patients with thrombosis reflects the frequency of a particular cancer type in the general population (Levitan, N. et al., Medicine
  • VTE venous thromboembolism
  • Cancer patients at risk for thrombosis may possess any or all of the following risk factors: (i) the stage of the cancer ⁇ i.e., presence of metastases), (ii) the presence of central vein catheters, (iii) surgery and anticancer therapies including chemotherapy, and (iv) hormones and antiangiogenic drugs.
  • risk factors include cancer, tumors, tumors, tumors, and tumors.
  • heparin or low molecular heparin to prevent thromboembolic disorders.
  • a number of low molecular heparin preparations have been approved by the FDA for these indications.
  • the effectiveness of compounds of the present invention as inhibitors of the coagulation Factors XIa, Vila, IXa, Xa, Xlla, plasma kallikrein or thrombin can be determined using a relevant purified serine protease, respectively, and an appropriate synthetic substrate.
  • the rate of hydrolysis of the chromogenic or fluorogenic substrate by the relevant serine protease was measured both in the absence and presence of
  • Factor XIa determinations were made in 50 mM HEPES buffer at pH 7.4 containing 145 mM NaCl, 5 mM KC1, and 0.1% PEG 8000 (polyethylene glycol; JT Baker or Fisher Scientific). Determinations were made using purified human Factor XIa at a final concentration of 25-200 pM (Haemato logic Technologies) and the synthetic substrate S-2366 (pyroGlu-Pro-Arg-pNA; CHROMOGENIX® or AnaSpec) at a concentration of 0.0002-0.001 M.
  • Factor IXa determinations were made in 0.005 M calcium chloride, 0.1 M sodium chloride, 0.0000001 M Refludan (Berlex), 0.05 M TRIS base and 0.5% PEG 8000 at a pH of 7.4. Refludan was added to inhibit small amounts of thrombin in the commercial preparations of human Factor IXa. Determinations were made using purified human Factor IXa (Haemato logic Technologies) at a final assay concentration of 20-100 nM and the synthetic substrate PCIXA2100-B (CenterChem) or Pefafluor IXa 3688 (H-D-Leu- Ph'Gly-Arg-AMC; CenterChem) at a concentration of 0.0004-0.0005 M.
  • Factor Xa determinations were made in 0.1 M sodium phosphate buffer at a pH of 7.5 containing 0.2 M sodium chloride and 0.5% PEG 8000. Determinations were made using purified human Factor Xa (Haematologic Technologies) at a final assay
  • Factor Xlla determinations were made in 0.05 M HEPES buffer at pH 7.4 containing 0.145 M NaCl, 0.05 M KCl, and 0.1% PEG 8000. Determinations were made using purified human Factor Xlla at a final concentration of 4 nM (American
  • Plasma kallikrein determinations were made in 0.1 M sodium phosphate buffer at a pH of 7.5 containing 0.1-0.2 M sodium chloride and 0.5% PEG 8000. Determinations were made using purified human plasma kallikrein (Enzyme Research Laboratories) at a final assay concentration of 200 pM and the synthetic substrate S-2302 (H-(D)-Pro-Phe- Arg-pNA; CHROMOGENIX®) at a concentration of 0.00008-0.0004 M.
  • Thrombin determinations were made in 0.1 M sodium phosphate buffer at a pH of 7.5 containing 0.2 M sodium chloride and 0.5% PEG 8000. Determinations were made using purified human alpha thrombin (Haematologic Technologies or Enzyme Research Laboratories) at a final assay concentration of 200-250 pM and the synthetic substrate S- 2366 (pyroGlu-Pro-Arg-pNA; CHROMOGENIX® or AnaSpec) at a concentration of 0.0002-0.0004 M.
  • K m The Michaelis constant, K m , for substrate hydrolysis by each protease, was determined at 25 °C or 37 °C in the absence of inhibitor. Values of K; were determined by allowing the protease to react with the substrate in the presence of the inhibitor.
  • Vs/vo A + (B-A)/(l + (I/IC 50 ) n );
  • Ki IC 5 o/(l + S/K m ) for a competitive inhibitor
  • v 0 is the velocity of the control in the absence of inhibitor
  • v s is the velocity in the presence of inhibitor
  • V max is the maximum reaction velocity
  • I is the concentration of inhibitor
  • A is the minimum activity remaining (usually locked at zero);
  • B is the maximum activity remaining (usually locked at 1.0);
  • n is the Hill coefficient, a measure of the number and cooperativity of potential inhibitor binding sites
  • IC50 is the concentration of inhibitor that produces 50% inhibition under the assay conditions
  • Ki is the dissociation constant of the enzyme: inhibitor complex
  • S is the concentration of substrate
  • K m is the Michaelis constant for the substrate.
  • the effectiveness of compounds of the present invention as inhibitors of coagulation can be determined using a standard or modified clotting assay.
  • An increase in the plasma clotting time in the presence of inhibitor is indicative of anticoagulation.
  • Relative clotting time is the clotting time in the presence of an inhibitor divided by the clotting time in the absence of an inhibitor.
  • the results of this assay may be expressed as IC1.5x or IC2x, the inhibitor concentration required to increase the clotting time by 50 or 100 percent, respectively.
  • the IC1.5x or IC2x is found by linear interpolation from relative clotting time versus inhibitor concentration plots using inhibitor concentration that spans the IC 1.5x or IC2x.
  • Clotting times are determined using citrated normal human plasma as well as plasma obtained from a number of laboratory animal species (e.g., rat, or rabbit). A compound is diluted into plasma beginning with a 10 mM DMSO stock solution. The final concentration of DMSO is less than 2%. Plasma clotting assays are performed in an automated coagulation analyzer (SYSMEX®, Dade-Behring, Illinois). Similarly, clotting times can be determined from laboratory animal species or humans dosed with compounds of the invention.
  • SYSMEX® automated coagulation analyzer
  • Activated Partial Thromboplastin Time is determined using ACTIN® FSL
  • Plasma 0.05 mL
  • ACTIN® FSL 0.05 mL
  • Calcium chloride 25 mM, 0.05 mL
  • the clotting time is the time in seconds from the moment calcium chloride is added until a clot is detected.
  • Prothrombin Time is determined using thromboplastin (Thromboplastin C Plus or INNOVIN®, Dade-Behring, Illinois) following the directions in the package insert. Plasma (0.05 mL) is warmed to 37 °C for 1 minute. Thromboplastin (0.1 mL) is added to the plasma to initiate coagulation. The clotting time is the time in seconds from the moment thromboplastin is added until a clot is detected.
  • Equilibrium solubilities were determined in various aqueous solvents buffered to a specific pH. Approximately 1 mg of compound was used for equilibration in 100 to 300 of solvent. Samples were stirred at 300 RPM at room temperature (20 ⁇ 2 °C) for 24 hours. If solubilization of the entire solid was observed, additional compound was added to keep the solid in excess for the duration of the study. After 24 hours, microscopy was used to determine if there was a change in morphology to the excess solid. The supernatants were then filtered through a 0.22 ⁇ PVDF filter plate and diluted with acetonitrile for HPLC analysis. Calibration samples were also provided for HPLC analysis.
  • dialyzed serum samples were diluted with 0.133 M sodium phosphate buffer adjusted to pH 7.4 and dialyzed buffer samples were diluted with human serum to result in the same final serum concentration in each sample. Subsequently, these samples were extracted by protein precipitation in acetonitrile containing two analytical internal standards (200 nM alprenolol and 600 nM tolbutamide). Precipitated proteins and supematants were separated by centrifugation at 4000 x g for 10 minutes.
  • Sample supematants were analyzed by LC-MS/MS and the peak area ratios of compound to the internal standard were determined for initial time zero samples (T 0 [ S erum] and T 0[B uffer]) and for post-equilibrium samples (T 5h [Serum] and T 5h [Buffer]). The percent free (free fraction), percent bound, and percent recovery results were calculated as follows:
  • Matrix interference was assessed by measuring the LC-MS/MS area ratio of analyte/internal standard for assay matrix blank (50:50 serum:buffer). The analytical conditions were deemed acceptable for assessment of percent free when the area ratio of analyte/internal standard for assay matrix blank (50:50 serum:buffer) was less than 20% of the area ratio for the T5h[Buffer] sample.
  • the effectiveness of the compounds of the present invention as antithrombotic agents is also assessed in other assays such as aPTT, solubility, and human protein binding affinity described above.
  • aPTT solubility
  • human protein binding affinity described above.
  • the pyrazolyl P2' macrocycles of the present application exhibited surprising pharmacological activities.
  • the compounds of the present invention possess superior anticoagulant activity, solubility and bioavailability compared to the reference compounds.
  • the rabbit ECAT model described by Wong et al. (J. Pharmacol. Exp. Ther., 295:212-218 (2000)), can be used in this study.
  • Male New Zealand White rabbits are anesthetized with ketamine (50 mg/kg + 50 mg/kg/h IM) and xylazine (10 mg/kg + 10 mg/kg/h IM). These anesthetics are supplemented as needed.
  • An electromagnetic flow probe is placed on a segment of an isolated carotid artery to monitor blood flow. Test agents or vehicle will be given (i.v., i.p., s.c, or orally) prior to or after the initiation of thrombosis.
  • Drug treatment prior to initiation of thrombosis is used to model the ability of test agents to prevent and reduce the risk of thrombus formation, whereas dosing after initiation is used to model the ability to treat existing thrombotic disease.
  • Thrombus formation is induced by electrical stimulation of the carotid artery for 3 min at 4 niA using an external stainless-steel bipolar electrode. Carotid blood flow is measured continuously over a 90-min period to monitor thrombus-induced occlusion. Total carotid blood flow over 90 min is calculated by the trapezoidal rule.
  • Average carotid flow over 90 min is then determined by converting total carotid blood flow over 90 min to percent of total control carotid blood flow, which would result if control blood flow had been maintained continuously for 90 min.
  • the ED50 (dose that increased average carotid blood flow over 90 min to 50% of the control) of compounds are estimated by a nonlinear least square regression program using the Hill sigmoid E max equation (DeltaGraph; SPSS Inc., Chicago, IL).
  • the AV shunt also contains an 8-cm-long 2-0 silk thread (Ethicon, Somerville, NJ). Blood flows from the femoral artery via the AV-shunt into the femoral vein. The exposure of flowing blood to a silk thread induces the formation of a significant thrombus. Forty minutes later, the shunt is disconnected and the silk thread covered with thrombus is weighed.
  • Test agents or vehicle will be given (i.v., i.p., s.c, or orally) prior to the opening of the AV shunt.
  • the percentage inhibition of thrombus formation is determined for each treatment group.
  • the ID 50 values (dose that produces 50% inhibition of thrombus formation) are estimated by a nonlinear least square regression program using the Hill sigmoid E max equation (DeltaGraph; SPSS Inc., Chicago, IL).
  • mice are dosed with a compound of the present invention, Evans Blue dye is injected via the tail vein, and extravasation of the blue dye is determined by
  • the ability of the compounds of the current invention to reduce or prevent the systemic inflammatory response syndrome can be tested in in vitro perfusion systems, or by on-pump surgical procedures in larger mammals, including dogs and baboons.
  • Read-outs to assess the benefit of the compounds of the present invention include for example, reduced platelet loss, reduced platelet / white blood cell complexes, reduced neutrophil elastase levels in plasma, reduced activation of complement factors, and reduced activation and/or consumption of contact activation proteins (plasma kallikrein, factor XII, factor XI, high molecular weight kininogen, CI -esterase inhibitors).
  • the compounds of the present invention may also be useful as inhibitors of additional serine proteases, notably human thrombin, human plasma kallikrein and human plasmin. Because of their inhibitory action, these compounds are indicated for use in the prevention or treatment of physiological reactions, including blood coagulation, fibrinolysis, blood pressure regulation and inflammation, and wound healing catalyzed by the aforesaid class of enzymes.
  • the compounds have utility as drugs for the treatment of diseases arising from elevated thrombin activity of the aforementioned serine proteases, such as myocardial infarction, and as reagents used as anticoagulants in the processing of blood to plasma for diagnostic and other commercial purposes.
  • the compounds of this invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release
  • formulations pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • composition means a composition comprising a compound of the invention in combination with at least one additional pharmaceutically acceptable carrier.
  • a “pharmaceutically acceptable carrier” refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals, including, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • Pharmaceutically-acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent-containing composition is to be administered; the intended route of
  • Pharmaceutically-acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Remington's Pharmaceutical Sciences, 18th Edition (1990).
  • the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
  • a physician or veterinarian can determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress of the thromboembolic disorder.
  • the daily oral dosage of each active ingredient when used for the indicated effects, will range between about 0.001 to about 1000 mg/kg of body weight, preferably between about 0.01 to about 100 mg/kg of body weight per day, and most preferably between about 0.1 to about 20 mg/kg/day.
  • the most preferred doses will range from about 0.001 to about 10 mg/kg/minute during a constant rate infusion.
  • Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
  • Compounds of this invention can also be administered by parenteral
  • the pharmaceutical composition is a solid formulation, e.g., a spray-dried composition, which may be used as is, or whereto the physician or the patient adds solvents, and/or diluents prior to use.
  • Compounds of this invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using transdermal skin patches.
  • suitable intranasal vehicles or via transdermal routes, using transdermal skin patches.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the compounds are typically administered in admixture with suitable
  • pharmaceutical carriers suitably selected with respect to the intended form of administration, e.g., oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
  • the active drug component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral administration in liquid form, the oral drug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate,
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • Compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • soluble polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol,
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Solid dispersions are also called solid-state dispersions.
  • any compound described herein is formulated as a spray dried dispersion (SDD).
  • SDD is a single phase amorphous molecular dispersion of a drug in a polymer matrix. It is a solid solution prepared by dissolving the drug and a polymer in a solvent (e.g. , acetone, methanol or the like) and spray drying the solution. The solvent rapidly evaporates from droplets which rapidly solidifies the polymer and drug mixture trapping the drug in amorphous form as an amorphous molecular dispersion.
  • a solvent e.g. , acetone, methanol or the like
  • Dosage forms suitable for administration may contain from about 1 milligram to about 1000 milligrams of active ingredient per dosage unit.
  • the active ingredient will ordinarily be present in an amount of about 0.1-95% by weight based on the total weight of the composition.
  • Gelatin capsules may contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like.
  • powdered carriers such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like.
  • Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • parenteral solutions In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl-or propyl-paraben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field.
  • a daily dosage may be about 0.1 to about 100 milligrams of the compound of the present invention and about 0.1 to about 100 milligrams per kilogram of patient body weight.
  • the compounds of this invention generally may be present in an amount of about 5 to about 300 milligrams per dosage unit, and the second anti-coagulant in an amount of about 1 to about 500 milligrams per dosage unit.
  • a daily dosage may be about 0.01 to about 300 milligrams of the compound of the present invention and about 50 to about 150 milligrams of the anti-platelet agent, preferably about 0.1 to about 4 milligrams of the compound of the present invention and about 1 to about 3 milligrams of antiplatelet agents, per kilogram of patient body weight.
  • a daily dosage may be about 0.1 to about 100 milligrams of the compound of the present invention, per kilogram of patient body weight and, in the case of the thrombolytic agents, the usual dosage of the thrombolytic agent when administered alone may be reduced by about 50-80% when administered with a compound of the present invention.
  • one active ingredient may be enteric coated.
  • enteric coating one of the active ingredients it is possible not only to minimize the contact between the combined active ingredients, but also, it is possible to control the release of one of these components in the gastrointestinal tract such that one of these components is not released in the stomach but rather is released in the intestines.
  • One of the active ingredients may also be coated with a material that affects a sustained-release throughout the gastrointestinal tract and also serves to minimize physical contact between the combined active ingredients.
  • the sustained-released component can be additionally enteric coated such that the release of this component occurs only in the intestine.
  • Still another approach would involve the formulation of a combination product in which the one component is coated with a sustained and/or enteric release polymer, and the other component is also coated with a polymer such as a low viscosity grade of hydroxypropyl methylcellulose (HPMC) or other appropriate materials as known in the art, in order to further separate the active components.
  • HPMC hydroxypropyl methylcellulose
  • the polymer coating serves to form an additional barrier to interaction with the other component.
  • the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s) selected from potassium channel openers, potassium channel blockers, calcium channel blockers, sodium hydrogen exchanger inhibitors, antiarrhythmic agents, antiatherosclerotic agents, anticoagulants, antithrombotic agents, prothrombolytic agents, fibrinogen antagonists, diuretics, antihypertensive agents, ATPase inhibitors, mineralocorticoid receptor antagonists, phospodiesterase inhibitors, antidiabetic agents, anti-inflammatory agents, antioxidants, angiogenesis modulators, antiosteoporosis agents, hormone replacement therapies, hormone receptor modulators, oral contraceptives, antiobesity agents, antidepressants, antianxiety agents, antipsychotic agents, antiproliferative agents, antitumor agents, antiulcer and gastroesophageal reflux disease agents, growth hormone agents and/or growth hormone secretagogues, thyroid mimetics, anti-infective agents, antiviral agents, antibacterial agents, antifungal agents, anti-
  • the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s) selected from an antiarrhythmic agent, an anti-hypertensive agent, an anti-coagulant agent, an anti-platelet agent, a thrombin inhibiting agent, a thrombolytic agent, a fibrinolytic agent, a calcium channel blocker, a potassium channel blocker, a cholesterol/lipid lowering agent, or a combination thereof.
  • additional therapeutic agent(s) selected from an antiarrhythmic agent, an anti-hypertensive agent, an anti-coagulant agent, an anti-platelet agent, a thrombin inhibiting agent, a thrombolytic agent, a fibrinolytic agent, a calcium channel blocker, a potassium channel blocker, a cholesterol/lipid lowering agent, or a combination thereof.
  • the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s) selected from warfarin, unfractionated heparin, low molecular weight heparin, synthetic pentasaccharide, hirudin, argatroban, aspirin, ibuprofen, naproxen, sulindac, indomethacin, mefenamate, dipyridamol, droxicam, diclofenac, sulfinpyrazone, piroxicam, ticlopidine, clopidogrel, tirofiban, eptifibatide, abciximab, melagatran, ximelagatran, disulfatohirudin, tissue plasminogen activator, modified tissue plasminogen activator, anistreplase, urokinase, and streptokinase, or a combination thereof.
  • additional therapeutic agent(s) selected from warfarin, unfractionated heparin, low molecular weight heparin
  • the present invention provides a pharmaceutical composition wherein the additional therapeutic agent is an antihypertensive agent selected from ACE inhibitors, AT-1 receptor antagonists, beta-adrenergic receptor antagonists, ETA receptor antagonists, dual ETA/ AT-1 receptor antagonists, renin inhibitors (aliskiren) and vasopepsidase inhibitors, an antiarrythmic agent selected from I KUT inhibitors, an anticoagulant selected from thrombin inhibitors, antithrombin-III activators, heparin co-factor II activators, other factor XIa inhibitors, other kallikrein inhibitors, plasminogen activator inhibitor (PAI-1) antagonists, thrombin activatable fibrinolysis inhibitor (TAFI) inhibitors, factor Vila inhibitors, factor IXa inhibitors, and factor Xa inhibitors, or an antiplatelet agent selected from GPIIb/IIIa blockers, GP Ib/IX blockers, protease activated receptor 1 (PAR-1) antagonists,
  • the present invention provides pharmaceutical composition, wherein the additional therapeutic agent(s) are an anti-platelet agent or a combination thereof.
  • the present invention provides a pharmaceutical composition, wherein the additional therapeutic agent is the anti-platelet agent clopidogrel.
  • the compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents.
  • administered in combination or “combination therapy” it is meant that the compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated.
  • each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
  • Compounds that can be administered in combination with the compounds of the present invention include, but are not limited to, anticoagulants, anti-thrombin agents, anti-platelet agents, fibrinolytics, hypolipidemic agents, antihypertensive agents, and anti- ischemic agents.
  • anticoagulant agents include warfarin, heparin (either unfractionated heparin or any commercially available low molecular weight heparin, for example, LOVENOX®), synthetic pentasaccharide, direct acting thrombin inhibitors including hirudin and argatroban, as well as other factor Vila inhibitors, factor IXa inhibitors, factor Xa inhibitors (e.g., ARIXTRA®, apixaban, rivaroxaban, LY-517717, DU-176b, DX-9065a, and those disclosed in WO 98/57951 , WO 03/026652, WO 01/047919, and WO 00/076970), factor XIa inhibitors, and inhibitors of activated TAFI and PAI-1 known in the art.
  • warfarin heparin (either unfractionated heparin or any commercially available low molecular weight heparin, for example, LOVENOX®)
  • anti-platelet agents denotes agents that inhibit platelet function, for example, by inhibiting the aggregation, adhesion or granule-content secretion of platelets.
  • agents include, but are not limited to, the various known non-steroidal anti-inflammatory drugs (NSAIDs) such as acetaminophen, aspirin, codeine, diclofenac, droxicam, fentaynl, ibuprofen,
  • NSAIDs non-steroidal anti-inflammatory drugs
  • NSAIDs indomethacin, ketorolac, mefenamate, morphine, naproxen, phenacetin, piroxicam, sufentanyl, sulfinpyrazone, sulindac, and pharmaceutically acceptable salts or prodrugs thereof.
  • aspirin acetylsalicylic acid or ASA
  • piroxicam are preferred.
  • Suitable platelet inhibitory agents include glycoprotein Ilb/IIIa antagonists (e.g., tirofiban, eptifibatide, abciximab, and integrelin), thromboxane-A2- receptor antagonists (e.g., ifetroban), thromboxane- A-synthetase inhibitors,
  • PDE-III phosphodiesterase-III
  • PDE-V inhibitors such as sildenafil
  • PAR-1 antagonists e.g., E-5555, SCH-530348, SCH-203099, SCH-529153 and SCH-205831
  • Suitable anti-platelet agents for use in combination with the compounds of the present invention, with or without aspirin, are ADP (adenosine diphosphate) receptor antagonists, preferably antagonists of the purinergic receptors P2Yi and P2Yi 2 , with P2Yi 2 being even more preferred.
  • ADP adenosine diphosphate
  • P2Yi 2 receptor antagonists include clopidogrel, ticlopidine, prasugrel, ticagrelor, and cangrelor, and
  • Ticlopidine and clopidogrel are also preferred compounds since they are known to be more gentle than aspirin on the gastro-intestinal tract in use. Clopidogrel is an even more preferred agent.
  • a preferred example is a triple combination of a compound of the present invention, aspirin, and another anti-platelet agent.
  • the anti-platelet agent is clopidogrel or prasugrel, more preferably clopidogrel.
  • thrombin inhibitors denotes inhibitors of the serine protease thrombin.
  • various thrombin- mediated processes such as thrombin-mediated platelet activation (that is, for example, the aggregation of platelets, and/or the secretion of platelet granule contents including serotonin) and/or fibrin formation are disrupted.
  • thrombin inhibitors are known to one of skill in the art and these inhibitors are contemplated to be used in combination with the present compounds.
  • Such inhibitors include, but are not limited to, boroarginine derivatives, boropeptides, heparins, hirudin, argatroban, dabigatran, AZD-0837, and those disclosed in WO 98/37075 and WO 02/044145, and
  • Boroarginine derivatives and boropeptides include N-acetyl and peptide derivatives of boronic acid, such as C-terminal a-aminoboronic acid derivatives of lysine, ornithine, arginine, homoarginine and corresponding isothiouronium analogs thereof.
  • hirudin includes suitable derivatives or analogs of hirudin, referred to herein as hirulogs, such as disulfatohirudin.
  • thrombolytic agents denotes agents that lyse blood clots (thrombi).
  • agents include tissue plasminogen activator (TP A, natural or recombinant) and modified forms thereof, anistreplase, urokinase, streptokinase, tenecteplase (TNK), lanoteplase (nPA), factor Vila inhibitors, thrombin inhibitors, inhibitors of factors IXa, Xa, and XIa, PAI-I inhibitors (i.e., inactivators of tissue plasminogen activator inhibitors), inhibitors of activated TAFI, alpha-2-antiplasmin inhibitors, and anisoylated plasminogen streptokinase activator complex, including pharmaceutically acceptable salts or prodrugs thereof.
  • TP A tissue plasminogen activator
  • TNK tenecteplase
  • nPA lanoteplase
  • factor Vila inhibitors thrombin inhibitors
  • anistreplase refers to anisoylated plasminogen streptokinase activator complex, as described, for example, in European Patent Application No. 028489, the disclosure of which is hereby incorporated herein by reference herein.
  • urokinase as used herein, is intended to denote both dual and single chain urokinase, the latter also being referred to herein as prourokinase.
  • Suitable cholesterol/lipid lowering agents and lipid profile therapies for use in combination with the compounds of the present invention include HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, simvastatin, fluvastatin, atorvastatin, rosuvastatin, and other statins), low-density lipoprotein (LDL) receptor activity modulators (e.g., HOE-402, PCSK9 inhibitors), bile acid sequestrants (e.g., HMG-CoA reductase inhibitors (e.g., pravastatin, lovastatin, simvastatin, fluvastatin, atorvastatin, rosuvastatin, and other statins), low-density lipoprotein (LDL) receptor activity modulators (e.g., HOE-402, PCSK9 inhibitors), bile acid sequestrants (e.g.,
  • cholestyramine and colestipol nicotinic acid or derivatives thereof (e.g., NIASPAN®), GPR109B (nicotinic acid receptor) modulators, fenofibric acid derivatives (e.g., gemfibrozil, clofibrate, fenofibrate and benzafibrate) and other peroxisome proliferator- activated receptors (PPAR) alpha modulators, PPARdelta modulators (e.g., GW-501516), PPARgamma modulators (e.g., rosiglitazone), compounds that have multiple peroxisome proliferator- activated receptors (PPAR) alpha modulators, PPARdelta modulators (e.g., GW-501516), PPARgamma modulators (e.g., rosiglitazone), compounds that have multiple peroxisome proliferator- activated receptors (PPAR) alpha modulators, PPARdelta modul
  • PPARalpha, PPARgamma and PPARdelta functionally modulating the activities of various combinations of PPARalpha, PPARgamma and PPARdelta, probucol or derivatives thereof (e.g., AGI-1067), cholesterol absorption inhibitors and/or Niemann-Pick CI -like transporter inhibitors (e.g., ezetimibe), cholesterol ester transfer protein inhibitors (e.g., CP-529414), squalene synthase inhibitors and/or squalene epoxidase inhibitors or mixtures thereof, acyl coenzyme A: cholesteryl acyltransferase (ACAT) 1 inhibitors, ACAT2 inhibitors, dual AC AT 1/2 inhibitors, ileal bile acid transport inhibitors (or apical sodium co-dependent bile acid transport inhibitors), microsomal triglyceride transfer protein inhibitors, liver-X- receptor (LXR) alpha modulators, LXR
  • omega 3 fatty acids e.g., 3-PUFA
  • plant stanols and/or fatty acid esters of plant stanols e.g., sitostanol ester used in BENECOL® margarine
  • endothelial lipase inhibitors e.g., endothelial lipase inhibitors, and HDL functional mimetics which activate reverse cholesterol transport (e.g., apoAI derivatives or apoAI peptide mimetics).
  • the compounds of the present invention are also useful as standard or reference compounds, for example, as a quality standard or control, in tests or assays involving the inhibition of thrombin, Factor Vila, IXa, Xa, XIa, and/or plasma kallikrein.
  • Such compounds may be provided in a commercial kit, for example, for use in pharmaceutical research involving thrombin, Factor Vila, IXa, Xa, XIa, and/or plasma kallikrein.
  • XIa for example, a compound of the present invention could be used as a reference in an assay to compare its known activity to a compound with an unknown activity. This would ensure the experimenter that the assay was being performed properly and provide a basis for comparison, especially if the test compound was a derivative of the reference compound.
  • compounds according to the present invention could be used to test their effectiveness.
  • the compounds of the present invention may also be used in diagnostic assays involving thrombin, Factor Vila, IXa, Xa, XIa, and/or plasma kallikrein.
  • thrombin, Factor Vila, IXa, Xa, XIa, and/or plasma kallikrein in an unknown sample could be determined by addition of the relevant chromogenic substrate, for example, S2366 for Factor XIa, to a series of solutions containing test sample and optionally one of the compounds of the present invention. If production of pNA is observed in the solutions containing test sample, but not in the presence of a compound of the present invention, then one would conclude Factor XIa was present.
  • Extremely potent and selective compounds of the present invention those having K; values less than or equal to 0.001 ⁇ against the target protease and greater than or equal to 0.1 ⁇ against the other proteases, may also be used in diagnostic assays involving the quantitation of thrombin, Factor Vila, IXa, Xa, XIa, and/or plasma kallikrein in serum samples.
  • the amount of Factor XIa in serum samples could be determined by careful titration of protease activity in the presence of the relevant chromogenic substrate, S2366, with a potent Factor XIa inhibitor of the present invention.
  • the present invention also encompasses an article of manufacture.
  • article of manufacture is intended to include, but not be limited to, kits and packages.
  • the article of manufacture of the present invention comprises: (a) a first container; (b) a pharmaceutical composition located within the first container, wherein the composition, comprises: a first therapeutic agent, comprising: a compound of the present invention or a pharmaceutically acceptable salt form thereof; and, (c) a package insert stating that the pharmaceutical composition can be used for the treatment of a thromboembolic and/or inflammatory disorder (as defined previously).
  • the package insert states that the pharmaceutical composition can be used in combination (as defined previously) with a second therapeutic agent to treat a
  • the article of manufacture can further comprise: (d) a second container, wherein components (a) and (b) are located within the second container and component (c) is located within or outside of the second container. Located within the first and second containers means that the respective container holds the item within its boundaries.
  • the first container is a receptacle used to hold a pharmaceutical composition.
  • This container can be for manufacturing, storing, shipping, and/or individual/bulk selling.
  • First container is intended to cover a bottle, jar, vial, flask, syringe, tube (e.g., for a cream preparation), or any other container used to manufacture, hold, store, or distribute a pharmaceutical product.
  • the second container is one used to hold the first container and, optionally, the package insert.
  • the second container include, but are not limited to, boxes (e.g., cardboard or plastic), crates, cartons, bags (e.g., paper or plastic bags), pouches, and sacks.
  • the package insert can be physically attached to the outside of the first container via tape, glue, staple, or another method of attachment, or it can rest inside the second container without any physical means of attachment to the first container.
  • the package insert is located on the outside of the second container. When located on the outside of the second container, it is preferable that the package insert is physically attached via tape, glue, staple, or another method of attachment. Alternatively, it can be adjacent to or touching the outside of the second container without being physically attached.
  • the package insert is a label, tag, marker, etc. that recites information relating to the pharmaceutical composition located within the first container.
  • the information recited will usually be determined by the regulatory agency governing the area in which the article of manufacture is to be sold (e.g., the United States Food and Drug
  • the package insert specifically recites the indications for which the pharmaceutical composition has been approved.
  • the package insert may be made of any material on which a person can read information contained therein or thereon.
  • the package insert is a printable material (e.g., paper, plastic, cardboard, foil, adhesive-backed paper or plastic, etc.) on which the desired information has been formed (e.g., printed or applied).
  • the compounds of the present invention may be synthesized by many methods available to those skilled in the art of organic chemistry (Maffrand, J.P. et al.,
  • Examples of compounds of the present invention prepared by methods described in the general schemes are given in the intermediates and examples section set out hereinafter.
  • Preparation of homochiral examples may be carried out by techniques known to one skilled in the art.
  • homochiral compounds may be prepared by separation of racemic products by chiral phase preparative HPLC.
  • the example compounds may be prepared by methods known to give enantiomerically enriched products. These include, but are not limited to, the incorporation of chiral auxiliary functionalities into racemic intermediates which serve to control the
  • the compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis.
  • the compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below.
  • the reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.
  • suitably substituted pyrimidin-4-ol derivatives lb can be coupled with an appropriately substituted macrocycle amine lc in the presence of HATU and DBU in a solvent such as CH 3 CN to provide pyrimidinone compounds la.
  • ring A is a SEM- protected imidazole ring
  • an additional deprotection step employing 4M HC1 in dioxane or TFA in DCM is required to afford compounds of this invention.
  • Scheme 2 describes the synthesis of suitably substituted pyrimidin-4-ol derivatives lb.
  • a base such as Hunig's base or potassium phosphate tribasic
  • a solvent mixture such as toluene and ethanol, or THF
  • a precatalyst such as Pd(PPh 3 ) 4 or 2 nd generation XPhos
  • an additional deprotection step employing aqueous HBr at elevated temperatures, is required to provide pyrimidin-4-ol derivative
  • Grignard reagents for example, allylmagnesium bromide
  • sulfinimine 3b can be added to sulfinimine 3b to give a sulfinamide 3c, as a mixture of diastereomers which can be separated at various stages of the sequence.
  • the diastereoselectivity for the addition of ally magnesium bromide to sulfinimine 3b can be improved by employing indium(III) chloride according to a modified procedure of Xu (Xu, M.-H., Org. Lett., 10(6): 1259 (2008)).
  • Protecting group interconversion can be accomplished in two steps to give 3d.
  • the critical subunit coupling is accomplished via methodology developed by Sames (J. Am. Chem. Soc, 131 :3042 (2009)).
  • Treatment of chloropyridine 3d with N-protected nitropyrazole 3e in the presence of catalytic Pd(OAc) 2 and P(nBu)Ad 2 forges the desired arylpyrazole bond, forming 3f.
  • Reduction of this nitropyrazole yields 3g.
  • This aminopyrazole can then be coupled with an appropriately substituted carboxylic acid 3h using T3P® and a base, such as pyridine, to give the amide 3i.
  • the diene can be cyclized via ring-closing metathesis using a catalyst, such as Grubbs (II), in a suitable solvent, such as EtOAc at elevated temperature, to give the pyridine-containing macrocycle 3j .
  • the alkene can be reduced with hydrogen over either palladium on carbon or platinum oxide.
  • the second coupling reaction is then carried out as described in Scheme 1 with pyrimidinol 3k to yield pyrimidinone 31.
  • Subsequent deprotection of the pyrazole with TFA in DCM or 4M HC1 in dioxane provides followed by Ullmann coupling with an aryl iodide affords 3m as a major regioisomer. If 3n is formed, it is the minor component of the product mixture.
  • Reverse phase preparative HPLC was carried out using C 18 columns eluting with gradients of Solvent A (90% water, 10% MeOH, 0.1% TFA) and Solvent B (10% water, 90% MeOH, 0.1% TFA, UV 220 nm) or with gradients of Solvent A (90% water, 10% ACN, 0.1% TFA) and Solvent B (10% water, 90% ACN, 0.1% TFA, UV 220 nm) or with gradients of Solvent A (98% water, 2% ACN, 0.05% TFA) and Solvent B (98% ACN, 2% water, 0.05% TFA, UV 220 nm) (or) SunFire Prep C18 OBD 5 ⁇ 30x100mm, 25 min gradient from 0-100% B.
  • A H 2 0/ACN/TFA
  • B ACN/H 2 0/TFA 90: 10:0.1.
  • Method A Waters SunFire column (3.5 ⁇ C 18, 3.0 x 150 mm). Gradient elution (0.5 mL/min) from 10-100% Solvent B for 12 min and then 100% Solvent B for 3 min was used. Solvent A is (95 % water, 5% acetonitrile, 0.05%> TFA) and Solvent B is (5% water, 95% acetonitrile, 0.05% TFA, UV 254 nm).
  • Method B Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5 :95 acetonitrile: water with 10 mM ammonium acetate; Mobile Phase B: 95 :5 acetonitrile: water with 10 mM ammonium acetate; Temperature: 50 °C; Gradient: 0-100%) B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.1 1 mL/min.
  • Method C Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5 :95 acetonitrile: water with 0.1% TFA; Mobile Phase B: 95 :5 acetonitrile: water with 0.1% TFA; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold atl00% B; Flow: 1.1 1 mL/min.
  • reaction mixture was monitored by LCMS and when complete, the reaction mixture was cooled and then 2.0 g of K 2 CO 3 (1 g for 1 mL AcOH) and 2.0 mL water was added. The reaction mixture was then stirred for 5 min. The reaction mixture was then filtered over a pad of CELITE® and concentrated in vacuo to yield the crude product. The crude product was then partitioned between EtOAc (30 mL) and saturated NaHC0 3 (15 mL) solution. The organic layers were separated, dried over MgS0 4 , filtered and concentrated.
  • Example 9C by replacing tert-butyl N-[(lS)-l-[2-(4-amino-l-methyl-lH-pyrazol-5- yl)pyridin-4-yl]but-3 -en- 1-yl] carbamate, prepared as described in Example 9B, with tert- butyl N-[(l S)- 1 - ⁇ 2-[4-amino- 1 -( 2 H 3 )methyl- lH-pyrazol-5-yl]pyridin-4-yl ⁇ but-3-en- 1 - yl]carbamate.
  • MS(ESI) m/z: 429.08 (M+H) + .
  • 6-methoxypyrimidine (1.46 g, 4.10 mmol) in AcOH (10ml) was added 48% HBr in water (5 ml, 44.2 mmol). The mixture was stirred at 85 °C for 1 h. The reaction was
  • reaction mixture was monitored by LCMS and once reaction is complete, to the cooled reaction mixture was then added 1 g of K 2 CO 3 (1 g for 1 mL AcOH) and 1 mL water. The reaction mixture was then stirred for 5 min. The reaction mixture was then filtered over a pad of CELITE® and concentrated in vacuo to yield the crude product. The crude product was then partitioned between EtOAc (30 mL) and saturated aqueous NaHCC"3 (15 mL) solution. The organic layers were separated, dried over MgS0 4 , filtered and concentrated. The crude product was then purified using normal phase
  • Pd/C (0.016 g, 0.015 mmol) was added to a 100 mL Parr hydrogenation flask containing a solution of tert-hvXyl N-[(9i?,10E,135)-3,9-dimethyl-8-oxo-3,4,7,15- tetraazatricyclo[ 12.3.1.0 2 ' 6 ]octadeca- 1(18),2(6),4, 10,14,16-hexaen- 13 -yl] carbamate (60 mg, 0.151 mmol) in EtOH (6 mL). The flask was purged with N 2 and pressurized to 55 psi of H 2 and allowed to stir for 5 h.
  • Pt0 2 (6.80 mg, 0.030 mmol) was added to a stirring solution of tert-butyl N- [(9R, 10E, 13 S)-3-( 2 H 3 )methyl-9-methyl-8-oxo-3 ,4,7, 15-tetraazatricyclo[ 12.3.1.0 2 ' 6 ] octadeca-l(18),2(6),4,10,14,16-hexaen-13-yl]carbamate (0.120 g, 0.300 mmol) in EtOH (10 ml). The suspension was subjected to a hydrogen atmosphere (55 psi) for 1 h. The catalyst was filtered off through a plug of CELITE® and the filtrate concentrated.
  • Example 20 Preparation of l-(4-chloro-2-(l-((5R,9S)-21,5-dimethyl-4-oxo-21H-3-aza- 1 (2,4)-pyridina-2(5 ,4)-pyrazolacyclononaphane-9-yl)-6-oxo- 1 ,6-dihydropyrimidin-4- yl)phenyl)-lH-l,2,3-triazole- -carboxylic acid trifluoroacetate
PCT/US2015/042576 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors WO2016053455A1 (en)

Priority Applications (39)

Application Number Priority Date Filing Date Title
EP23185426.6A EP4286372A3 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
CN201910882473.1A CN110734435B (zh) 2014-10-01 2015-07-29 作为因子xia抑制剂的嘧啶酮
AU2015324530A AU2015324530B2 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor XIa inhibitors
JP2017517766A JP6462865B2 (ja) 2014-10-01 2015-07-29 第xia因子阻害剤としてのピリミジノン
EP15747721.7A EP3089979B1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
PE2020001966A PE20210922A1 (es) 2014-10-01 2015-07-29 Pirimidinonas como inhibidores del factor xia
EA201790595A EA031590B1 (ru) 2014-10-01 2015-07-29 Пиримидиноны в качестве ингибиторов фактора xia
CN202210580097.2A CN114957255A (zh) 2014-10-01 2015-07-29 作为因子xia抑制剂的嘧啶酮
DK15747721.7T DK3089979T3 (en) 2014-10-01 2015-07-29 PYRIMIDINONES AS A FACTOR XIA INHIBITORS
CN201580053079.7A CN106795161B (zh) 2014-10-01 2015-07-29 作为因子xia抑制剂的嘧啶酮
EP17189935.4A EP3293186B1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
LTEP15747721.7T LT3089979T (lt) 2014-10-01 2015-07-29 Pirimidinonai, kaip faktoriaus xia inhibitoriai
KR1020177011233A KR101921436B1 (ko) 2014-10-01 2015-07-29 인자 XIa 억제제로서의 피리미디논
SG11201702576QA SG11201702576QA (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
PL15747721T PL3089979T3 (pl) 2014-10-01 2015-07-29 Pirymidynony jako inhibitory czynnika xia
ES15747721.7T ES2655884T3 (es) 2014-10-01 2015-07-29 Pirimidinonas como inhibidores del Factor XIa
TN2017000112A TN2017000112A1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
MX2017003695A MX2017003695A (es) 2014-10-01 2015-07-29 Pirimidinonas como inhibidores del factor xia.
NZ731416A NZ731416A (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
EP20197284.1A EP3828186B1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
MYPI2017701150A MY183987A (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
TNP/2018/000229A TN2018000229A1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
CA2963395A CA2963395C (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors
KR1020187033199A KR102269999B1 (ko) 2014-10-01 2015-07-29 인자 XIa 억제제로서의 피리미디논
RS20180046A RS56786B1 (sr) 2014-10-01 2015-07-29 Pirimidinoni kao inhibitori faktora xia
SI201530123T SI3089979T1 (sl) 2014-10-01 2015-07-29 Pirimidinoni kot faktor XIA inhibitorji
BR112017006702-1A BR112017006702B1 (pt) 2014-10-01 2015-07-29 Pirimidinonas inibidoras de fator xia, composição farmacêutica e seu uso
MA40123A MA40123A1 (fr) 2014-10-01 2015-07-29 Pyrimidines utilisées en tant qu'inhibiteurs du facteur xia
IL251434A IL251434B (en) 2014-10-01 2017-03-28 Pyrimidonines as factor xia inhibitors
PH12017500580A PH12017500580B1 (en) 2014-10-01 2017-03-29 Pyrimidinones as factor xia inhibitors
ZA2017/02478A ZA201702478B (en) 2014-10-01 2017-04-07 Pyrimidinones as factor xia inhibitors
CONC2017/0003833A CO2017003833A2 (es) 2014-10-01 2017-04-20 Pirimidinonas como inhibidores del factor xia
CY20171101314T CY1119678T1 (el) 2014-10-01 2017-12-15 Πυριμιδινονες ως αναστολεις τελεστη χια
HRP20171950TT HRP20171950T1 (hr) 2014-10-01 2017-12-18 Pirimidinoni kao inhibitori faktora xia
AU2020200376A AU2020200376B2 (en) 2014-10-01 2020-01-20 Pyrimidinones as factor XIa inhibitors
PH12020500195A PH12020500195A1 (en) 2014-10-01 2020-01-27 Pyrimidinones as factor xia inhibitors
IL276470A IL276470B (en) 2014-10-01 2020-08-03 Pyrimidonines as factor xia inhibitors
CY20201101149T CY1123663T1 (el) 2014-10-01 2020-12-04 Πυριμιδινονες ως αναστολεις τελεστη χια
AU2021245098A AU2021245098B2 (en) 2014-10-01 2021-10-05 Pyrimidinones as factor XIa inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462058316P 2014-10-01 2014-10-01
US62/058,316 2014-10-01

Publications (1)

Publication Number Publication Date
WO2016053455A1 true WO2016053455A1 (en) 2016-04-07

Family

ID=53784023

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/042576 WO2016053455A1 (en) 2014-10-01 2015-07-29 Pyrimidinones as factor xia inhibitors

Country Status (34)

Country Link
EP (4) EP4286372A3 (US07585860-20090908-C00124.png)
JP (4) JP6462865B2 (US07585860-20090908-C00124.png)
KR (2) KR102269999B1 (US07585860-20090908-C00124.png)
CN (3) CN106795161B (US07585860-20090908-C00124.png)
AR (1) AR101367A1 (US07585860-20090908-C00124.png)
AU (3) AU2015324530B2 (US07585860-20090908-C00124.png)
CA (1) CA2963395C (US07585860-20090908-C00124.png)
CL (1) CL2017000712A1 (US07585860-20090908-C00124.png)
CO (1) CO2017003833A2 (US07585860-20090908-C00124.png)
CY (2) CY1119678T1 (US07585860-20090908-C00124.png)
DK (2) DK3293186T3 (US07585860-20090908-C00124.png)
EA (1) EA031590B1 (US07585860-20090908-C00124.png)
ES (3) ES2655884T3 (US07585860-20090908-C00124.png)
HR (2) HRP20171950T1 (US07585860-20090908-C00124.png)
HU (2) HUE052812T2 (US07585860-20090908-C00124.png)
IL (2) IL251434B (US07585860-20090908-C00124.png)
LT (2) LT3089979T (US07585860-20090908-C00124.png)
MA (1) MA40123A1 (US07585860-20090908-C00124.png)
MX (2) MX2017003695A (US07585860-20090908-C00124.png)
MY (1) MY183987A (US07585860-20090908-C00124.png)
NO (1) NO2721243T3 (US07585860-20090908-C00124.png)
NZ (2) NZ731416A (US07585860-20090908-C00124.png)
PE (2) PE20210922A1 (US07585860-20090908-C00124.png)
PH (2) PH12017500580B1 (US07585860-20090908-C00124.png)
PL (1) PL3089979T3 (US07585860-20090908-C00124.png)
PT (2) PT3293186T (US07585860-20090908-C00124.png)
RS (2) RS61183B1 (US07585860-20090908-C00124.png)
SG (2) SG11201702576QA (US07585860-20090908-C00124.png)
SI (2) SI3089979T1 (US07585860-20090908-C00124.png)
TN (2) TN2017000112A1 (US07585860-20090908-C00124.png)
TW (2) TWI692478B (US07585860-20090908-C00124.png)
UY (1) UY36244A (US07585860-20090908-C00124.png)
WO (1) WO2016053455A1 (US07585860-20090908-C00124.png)
ZA (1) ZA201702478B (US07585860-20090908-C00124.png)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9453018B2 (en) 2014-10-01 2016-09-27 Bristol-Myers Squibb Company Pyrimidinones as factor XIa inhibitors
US20170096429A1 (en) * 2014-01-31 2017-04-06 Bristol-Myers Squibb Company Macrocycles with aromatic p2' groups as factor xia inhibitors
WO2018133793A1 (zh) * 2017-01-18 2018-07-26 广东东阳光药业有限公司 凝血因子XIa抑制剂及其用途
US10081623B2 (en) 2014-09-04 2018-09-25 Bristol-Myers Squibb Company Diamide macrocycles that are FXIa inhibitors
US10160750B2 (en) 2015-06-19 2018-12-25 Bristol-Myers Squibb Company Diamide macrocycles as factor XIa inhibitors
US10273236B2 (en) 2014-01-31 2019-04-30 Bristol-Myers Squibb Macrocyclic factor XIa inhibitors bearing heterocyclic groups
US10287288B2 (en) 2015-07-29 2019-05-14 Bristol-Myers Squibb Factor XIa macrocyclic inhibitors bearing alkyl or cycloalkyl P2' moieties
US10550123B2 (en) 2011-09-26 2020-02-04 Katholieke Universiteit Leuven, K.U. Leuven R&D Viral replication inhibitors
US10646469B2 (en) 2016-03-31 2020-05-12 Janssen Pharmaceuticals, Inc. Substituted indole derivatives as dengue viral replication inhibitors
US10669289B2 (en) 2014-10-01 2020-06-02 Merck Patent Gmbh Boronic acid derivatives
US10676477B2 (en) 2015-07-29 2020-06-09 Bristol-Myers Squibb Company Factor XIa macrocycle inhibitors bearing a non-aromatic P2' group
US10689340B2 (en) 2016-04-01 2020-06-23 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US10696632B2 (en) 2015-05-08 2020-06-30 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10730884B2 (en) 2016-04-01 2020-08-04 Janssen Pharmaceuticals, Inc. Substituted indole compound derivatives as dengue viral replication inhibitors
US10752641B2 (en) 2016-03-02 2020-08-25 Bristol-Myers Squibb Company Diamide macrocycles having factor XIa inhibiting activity
US10765662B2 (en) 2015-09-16 2020-09-08 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10786484B2 (en) 2015-09-16 2020-09-29 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
WO2020210629A1 (en) * 2019-04-11 2020-10-15 Bristol-Myers Squibb Company Enhanced performance of amorphous solid and solubilized formulations for achieving therapeutic plasma concentrations
WO2020211781A1 (zh) 2019-04-16 2020-10-22 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
WO2021013209A1 (zh) 2019-07-23 2021-01-28 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
US10913716B2 (en) 2016-03-31 2021-02-09 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11053196B2 (en) 2017-05-22 2021-07-06 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11083707B2 (en) 2017-03-31 2021-08-10 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
WO2021207659A1 (en) * 2020-04-10 2021-10-14 Bristol-Myers Squibb Company Crystalline forms of (9r, 135s)-13- {4-[5-chloro-2-(4-chloro-1h,2,3- triazol- 1 -yl)phenyl] -6-oxo- 1,6-dihydropyrimidin- 1-yl}-3-(difluoromethyl)-9-methyl-3,4,7,15- tetraazatricyclo [ 12.3.1.02·6] octadeca- 1(18), 2(6), 4, 14, 16-pentaen-8-one
US11179368B2 (en) 2017-03-31 2021-11-23 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
CN114375290A (zh) * 2019-04-11 2022-04-19 百时美施贵宝公司 制备(6r,10s)-10-{4-[5-氯-2-(4-氯-1h-1,2,3-三唑-1-基)苯基]-6-氧代基-1(6h)-嘧啶基}-1-(二氧甲基)-6-甲基-1,4,7,8,9,10-六氢-11,15-(亚甲桥基)吡唑并[4,3-b][1,7]二氮杂环十四炔-5(6h)-酮的新颖合成选项
WO2022081473A1 (en) * 2020-10-12 2022-04-21 Bristol-Myers Squibb Company A process toward the manufacture of (6r,10s)-10-{4-[5-chloro-2-(4-chloro-1h-1,2,3-triazol-1-yl)phenyl]-6-oxo-1(6h)-pyrimidinyl}-1-(difluoromethyl)-6-methyl-1,4,7,8,9,10-hexahydro-11,15-(metheno)pyrazolo[4,3-b] [1,7]diazacyclotetradecin-5(6h)-one
US11407715B2 (en) 2017-05-22 2022-08-09 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
WO2022222960A1 (zh) * 2021-04-21 2022-10-27 上海美悦生物科技发展有限公司 FXIa抑制剂及其药物组合物、制备方法和用途

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NO2721243T3 (US07585860-20090908-C00124.png) * 2014-10-01 2018-10-20

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0028489A1 (en) 1979-11-05 1981-05-13 Beecham Group Plc Enzyme derivatives, and their preparation
WO1998037075A1 (de) 1997-02-18 1998-08-27 Boehringer Ingelheim Pharma Kg Disubstituierte bicyclische heterocyclen, ihre herstellung und ihre verwendung als arzneimittel
WO1998057951A1 (en) 1997-06-19 1998-12-23 Du Pont Pharmaceuticals Company NOVEL GUANIDINE MIMICS AS FACTOR Xa INHIBITORS
WO2000076970A2 (en) 1999-06-14 2000-12-21 Eli Lilly And Company Serine protease inhibitors
WO2001047919A1 (de) 1999-12-24 2001-07-05 Bayer Aktiengesellschaft Substituierte oxazolidinone und ihre verwendung im gebiet der blutgerinnung
WO2002044145A1 (en) 2000-12-01 2002-06-06 Astrazeneca Ab New mandelic acid derivatives and their use as throbin inhibitors
WO2003026652A1 (en) 2001-09-21 2003-04-03 Bristol-Myers Squibb Company Lactam-containing compounds and derivatives thereof as factor xa inhibitors
US20040180855A1 (en) 2003-02-19 2004-09-16 Schumacher William A. Methods of treating thrombosis with reduced risk of increased bleeding times
WO2013022814A1 (en) 2011-08-05 2013-02-14 Bristol-Myers Squibb Company Cyclic p1 linkers as factor xia inhibitors
WO2013093484A1 (en) * 2011-12-21 2013-06-27 Ono Pharmaceutical Co., Ltd. Pyridinone and pyrimidinone derivatives as factor xia inhibitors
WO2014022767A1 (en) * 2012-08-03 2014-02-06 Bristol-Myers Squibb Company Dihydropyridone p1 as factor xia inhibitors
WO2014022766A1 (en) 2012-08-03 2014-02-06 Bristol-Myers Squibb Company Dihydropyridone p1 as factor xia inhibitors
WO2015116886A1 (en) * 2014-01-31 2015-08-06 Bristol-Myers Squibb Company Macrocycles with hetrocyclic p2' groups as factor xia inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011215898B2 (en) * 2010-02-11 2016-08-11 Bristol-Myers Squibb Company Macrocycles as Factor XIa inhibitors
TW201311689A (zh) * 2011-08-05 2013-03-16 必治妥美雅史谷比公司 作為因子xia抑制劑之新穎巨環化合物
US9327839B2 (en) 2011-08-05 2016-05-03 General Atomics Method and apparatus for inhibiting formation of and/or removing ice from aircraft components
NO2760821T3 (US07585860-20090908-C00124.png) * 2014-01-31 2018-03-10
NO2721243T3 (US07585860-20090908-C00124.png) * 2014-10-01 2018-10-20

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0028489A1 (en) 1979-11-05 1981-05-13 Beecham Group Plc Enzyme derivatives, and their preparation
WO1998037075A1 (de) 1997-02-18 1998-08-27 Boehringer Ingelheim Pharma Kg Disubstituierte bicyclische heterocyclen, ihre herstellung und ihre verwendung als arzneimittel
WO1998057951A1 (en) 1997-06-19 1998-12-23 Du Pont Pharmaceuticals Company NOVEL GUANIDINE MIMICS AS FACTOR Xa INHIBITORS
WO2000076970A2 (en) 1999-06-14 2000-12-21 Eli Lilly And Company Serine protease inhibitors
WO2001047919A1 (de) 1999-12-24 2001-07-05 Bayer Aktiengesellschaft Substituierte oxazolidinone und ihre verwendung im gebiet der blutgerinnung
WO2002044145A1 (en) 2000-12-01 2002-06-06 Astrazeneca Ab New mandelic acid derivatives and their use as throbin inhibitors
WO2003026652A1 (en) 2001-09-21 2003-04-03 Bristol-Myers Squibb Company Lactam-containing compounds and derivatives thereof as factor xa inhibitors
US20040180855A1 (en) 2003-02-19 2004-09-16 Schumacher William A. Methods of treating thrombosis with reduced risk of increased bleeding times
WO2013022814A1 (en) 2011-08-05 2013-02-14 Bristol-Myers Squibb Company Cyclic p1 linkers as factor xia inhibitors
WO2013093484A1 (en) * 2011-12-21 2013-06-27 Ono Pharmaceutical Co., Ltd. Pyridinone and pyrimidinone derivatives as factor xia inhibitors
WO2014022767A1 (en) * 2012-08-03 2014-02-06 Bristol-Myers Squibb Company Dihydropyridone p1 as factor xia inhibitors
WO2014022766A1 (en) 2012-08-03 2014-02-06 Bristol-Myers Squibb Company Dihydropyridone p1 as factor xia inhibitors
WO2015116886A1 (en) * 2014-01-31 2015-08-06 Bristol-Myers Squibb Company Macrocycles with hetrocyclic p2' groups as factor xia inhibitors

Non-Patent Citations (61)

* Cited by examiner, † Cited by third party
Title
"Comprehensive Heterocyclic Chemistry", vol. 2, 1984, PERGAMON PRESS, pages: 165 - 524
"Comprehensive Heterocyclic Chemistry", vol. 5, 1996, PERGAMON PRESS, pages: 1 - 300
"Design of Prodrugs", 1985, ELSEVIER
"Hawley's Condensed Chemical Dictionary", 1997, JOHN WILEY & SONS, INC.
"Hemostasis and Thrombosis, Basic Principles and Clinical Practice", 2006, LIPPINCOTT WILLIAMS & WILKINS, pages: 853
"IUPAC Recommendations", PURE AND APPLIED CHEMISTRY,, vol. 68, 1996, pages 2193 - 2222
"Medicinal Chemistry: Principles and Practice", 1994, THE ROYAL SOCIETY OF CHEMISTRY
"Methods in Enzymology", vol. 112, 1985, ACADEMIC PRESS, pages: 309 - 396
"Remington's Pharmaceutical Sciences", 1990
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
"The Chemistry of Heterocyclic Compounds", vol. 14, 1974, JOHN WILEY & SONS, article "Pyridine and Its Derivatives"
"The Practice of Medicinal Chemistry", 1999, ACADEMIC PRESS
BLOM, J.W. ET AL., JAMA, vol. 293, no. 6, 2005, pages 715 - 722
BOUMA, B.N ET AL., THROMB. RES., vol. 101, 2001, pages 329 - 354
BUNDGAARD, H. ET AL., J. PHARM. SCI., vol. 77, 1988, pages 285
BUNDGAARD, H. ET AL.: "A Textbook of Drug Design and Development", 1991, HARWOOD ACADEMIC PUBLISHERS, article "Design and Application of Prodrugs", pages: 113 - 191
BUNDGAARD, H., ADV. DRUG DELIV. REV., vol. 8, 1992, pages 1 - 38
CHAN ET AL., AMER. J. PATHOLOGY, vol. 158, 2001, pages 469 - 479
CLERMONT, A ET AL.: "Plasma kallikrein mediates retinal vascular dysfunction and induces retinal thickening in diabetic rats", DIABETES, vol. 60, 2011, pages 1590 - 1598
COLEMAN, R.: "Hemostasis and Thrombosis", 2001, LIPPINCOTT WILLIAMS & WILKINS, article "Contact Activation Pathway", pages: 103 - 122
DI, L. ET AL.: "Impact of Recovery on Fraction Unbound Using Equilibrium Dialysis", J. PHARM. SCI., vol. 101, no. 3, 2011, pages 1327 - 1335
ELLMAN, J., J. ORG. CHEM., vol. 64, 1999, pages 1278
GAILANI, D. ET AL., ARTERIOSCLER. THROMB. VASE. BIOL., vol. 27, 2007, pages 2507 - 2513
GAILANI, D. ET AL., BLOOD COAGULATION AND FIBRINOLYSIS, vol. 8, 1997, pages 134 - 144
GAILANI, D., FRONTIERS IN BIOSCIENCE, vol. 6, 2001, pages 201 - 207
GALIANI, D., TRENDS CARDIOVASC. MED., vol. 10, 2000, pages 198 - 204
GOODNIGHT, S.H. ET AL.: "Screening Tests of Hemostasis'', Disorders of Thrombosis and Hemostasis: A Clinical Guide", 2001, MCGRAW-HILL, pages: 41 - 51
GREENE ET AL.: "Protective Groups in Organic Synthesis", 2006, WILEY-INTERSCIENCE
GRUBER ET AL., BLOOD, vol. 102, 2003, pages 953 - 955
HIRSH, J. ET AL., BLOOD, vol. 105, 2005, pages 453 - 463
HOFFMAN, M., BLOOD REVIEWS, vol. 17, 2003, pages S1 - S5
KAKEYA, N. ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
KLEINSCHMITZ ET AL., J. EXP. MED., vol. 203, 2006, pages 513 - 518
KROEHNKE, F., SYNTHESIS, vol. 1, 1976
KUDUK, TETRAHEDRON LETTERS, vol. 45, 2004, pages 6641
LEHMANN, A.: "Ecallantide (DX-88), a plasma kallikrein inhibitor for the treatment of hereditary angioedema and the prevention of blood loss in on-pump cardiothoracic surgery", EXPERT OPIN. BIOL. THER., vol. 8, 2008, pages 187 - 199
LEVINE M. ET AL., N. ENGL. J. MED., vol. 334, no. 11, 1996, pages 677 - 681
LEVITAN, N. ET AL., MEDICINE (BALTIMORE, vol. 78, no. 5, 1999, pages 285 - 291
MAFFRAND, J.P ET AL., HETEROCYCLES, vol. 16, no. L, 1981, pages 35 - 37
MEIJERS, J.C.M. ET AL., N. ENGL. J. MED., vol. 342, 2000, pages 696 - 701
MINNEMA, M.C. ET AL., ARTERIOSCLER. THROMB. VASE. BIOL., vol. 20, 2000, pages 2489 - 2493
MISMETTI, P. ET AL., BR. J. SURG., vol. 88, 2001, pages 913 - 930
MURAKAMI, T. ET AL., ARTERIOSCLER. THROMB. VASE. BIOL., vol. 15, 1995, pages 1107 - 1113
NEGI, SYNTHESIS, vol. 991, 1996
PLISE, E.G ET AL.: "Semi-automated protein binding methodology using equilibrium dialysis and a novel mixed-matrix cassette approach", J. PHARM. SCI., vol. 99, no. 12, 2010, pages 5070 - 5078
RENNE ET AL., J. EXP. MED., vol. 202, 2005, pages 271 - 281
ROSEN ET AL., THROMB. HAEMOST., vol. 87, 2002, pages 774 - 777
SAMES, J. AM. CHEM. SOC., vol. 131, 2009, pages 3042
SCHMAIER, A.H.: "Contact Activation", THROMBOSIS AND HEMORRHAGE, 1998, pages 105 - 128
SCHUMACHER, W. ET AL., J. THROMB. HAEMOST., vol. 3, no. 1, 2005, pages 1228
SCHUMACHER, W.A. ET AL., EUR. J. PHARMACOL., 2007, pages 167 - 174
SHARIAT-MADAR ET AL., BLOOD, vol. 108, 2006, pages 192 - 199
TESTA, B. ET AL.: "Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology", 2003, VCHA AND WILEY-VCH, ZURICH, SWITZERLAND
VAN LIEMPD, S. ET AL.: "Development and Validation of a Higher-Throughput Equilibrium Dialysis Assay for Plasma Protein Binding", J. LAB. AUTOM., vol. 16, 2011, pages 56 - 67, XP028132867, DOI: doi:10.1016/j.jala.2010.06.002
WANG ET AL., J. THROMB. HAEMOST., vol. 3, 2005, pages 695 - 702
WATERS, N.J. ET AL.: "Validation of a rapid equilibrium dialysis approach for the measurement of plasma protein binding", J. PHARM. SCI., vol. 97, no. 10, 2008, pages 4586 - 4595
WONG ET AL., J. PHARMACOL. EXP. THER., vol. 295, 2000, pages 212 - 218
WONG P.C. ET AL., AMERICAN HEART ASSOCIATION SCIENTIFIC SESSIONS, 12 November 2006 (2006-11-12)
WONG, P.C ET AL., J. PHARMACOL. EXP. THER., vol. 292, 2000, pages 351 - 357
XIAO, ORG. LETT., vol. 11, 2009, pages 1421
XU, M.-H., ORG. LETT., vol. 10, no. 6, 2008, pages 1259

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10550123B2 (en) 2011-09-26 2020-02-04 Katholieke Universiteit Leuven, K.U. Leuven R&D Viral replication inhibitors
US11279704B2 (en) 2011-09-26 2022-03-22 Katholieke Universiteit Leuven Viral replication inhibitors
US10273236B2 (en) 2014-01-31 2019-04-30 Bristol-Myers Squibb Macrocyclic factor XIa inhibitors bearing heterocyclic groups
US9777001B2 (en) 2014-01-31 2017-10-03 Bristol-Myers Squibb Company Macrocycles with aromatic P2′ groups as factor xia inhibitors
US20170096429A1 (en) * 2014-01-31 2017-04-06 Bristol-Myers Squibb Company Macrocycles with aromatic p2' groups as factor xia inhibitors
US10081623B2 (en) 2014-09-04 2018-09-25 Bristol-Myers Squibb Company Diamide macrocycles that are FXIa inhibitors
US9453018B2 (en) 2014-10-01 2016-09-27 Bristol-Myers Squibb Company Pyrimidinones as factor XIa inhibitors
US10336754B2 (en) 2014-10-01 2019-07-02 Bristol-Myers Squibb Company Pyrimidinones as factor XIa inhibitors
US10669289B2 (en) 2014-10-01 2020-06-02 Merck Patent Gmbh Boronic acid derivatives
US11053247B2 (en) 2014-10-01 2021-07-06 Bristol-Myers Squibb Company Pyrimidinones as factor XIA inhibitors
US10919854B2 (en) 2015-05-08 2021-02-16 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10696632B2 (en) 2015-05-08 2020-06-30 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US11827602B2 (en) 2015-05-08 2023-11-28 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10160750B2 (en) 2015-06-19 2018-12-25 Bristol-Myers Squibb Company Diamide macrocycles as factor XIa inhibitors
US10676477B2 (en) 2015-07-29 2020-06-09 Bristol-Myers Squibb Company Factor XIa macrocycle inhibitors bearing a non-aromatic P2' group
US10287288B2 (en) 2015-07-29 2019-05-14 Bristol-Myers Squibb Factor XIa macrocyclic inhibitors bearing alkyl or cycloalkyl P2' moieties
US10765662B2 (en) 2015-09-16 2020-09-08 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10786484B2 (en) 2015-09-16 2020-09-29 Janssen Pharmaceuticals, Inc. Mono- or di-substituted indole derivatives as dengue viral replication inhibitors
US10752641B2 (en) 2016-03-02 2020-08-25 Bristol-Myers Squibb Company Diamide macrocycles having factor XIa inhibiting activity
US10646469B2 (en) 2016-03-31 2020-05-12 Janssen Pharmaceuticals, Inc. Substituted indole derivatives as dengue viral replication inhibitors
US10913716B2 (en) 2016-03-31 2021-02-09 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US10730884B2 (en) 2016-04-01 2020-08-04 Janssen Pharmaceuticals, Inc. Substituted indole compound derivatives as dengue viral replication inhibitors
US10689340B2 (en) 2016-04-01 2020-06-23 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11180450B2 (en) 2016-04-01 2021-11-23 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
WO2018133793A1 (zh) * 2017-01-18 2018-07-26 广东东阳光药业有限公司 凝血因子XIa抑制剂及其用途
US11179368B2 (en) 2017-03-31 2021-11-23 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11083707B2 (en) 2017-03-31 2021-08-10 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11795149B2 (en) 2017-05-22 2023-10-24 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11702387B2 (en) 2017-05-22 2023-07-18 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11053196B2 (en) 2017-05-22 2021-07-06 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
US11407715B2 (en) 2017-05-22 2022-08-09 Janssen Pharmaceuticals, Inc. Substituted indoline derivatives as dengue viral replication inhibitors
CN114375290A (zh) * 2019-04-11 2022-04-19 百时美施贵宝公司 制备(6r,10s)-10-{4-[5-氯-2-(4-氯-1h-1,2,3-三唑-1-基)苯基]-6-氧代基-1(6h)-嘧啶基}-1-(二氧甲基)-6-甲基-1,4,7,8,9,10-六氢-11,15-(亚甲桥基)吡唑并[4,3-b][1,7]二氮杂环十四炔-5(6h)-酮的新颖合成选项
WO2020210629A1 (en) * 2019-04-11 2020-10-15 Bristol-Myers Squibb Company Enhanced performance of amorphous solid and solubilized formulations for achieving therapeutic plasma concentrations
WO2020211781A1 (zh) 2019-04-16 2020-10-22 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
CN113677682B (zh) * 2019-04-16 2023-05-30 华润生物医药有限公司 作为XIa因子抑制剂的大环衍生物
CN113677682A (zh) * 2019-04-16 2021-11-19 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
WO2021013209A1 (zh) 2019-07-23 2021-01-28 南京明德新药研发有限公司 作为XIa因子抑制剂的大环衍生物
WO2021207659A1 (en) * 2020-04-10 2021-10-14 Bristol-Myers Squibb Company Crystalline forms of (9r, 135s)-13- {4-[5-chloro-2-(4-chloro-1h,2,3- triazol- 1 -yl)phenyl] -6-oxo- 1,6-dihydropyrimidin- 1-yl}-3-(difluoromethyl)-9-methyl-3,4,7,15- tetraazatricyclo [ 12.3.1.02·6] octadeca- 1(18), 2(6), 4, 14, 16-pentaen-8-one
WO2022081473A1 (en) * 2020-10-12 2022-04-21 Bristol-Myers Squibb Company A process toward the manufacture of (6r,10s)-10-{4-[5-chloro-2-(4-chloro-1h-1,2,3-triazol-1-yl)phenyl]-6-oxo-1(6h)-pyrimidinyl}-1-(difluoromethyl)-6-methyl-1,4,7,8,9,10-hexahydro-11,15-(metheno)pyrazolo[4,3-b] [1,7]diazacyclotetradecin-5(6h)-one
WO2022222960A1 (zh) * 2021-04-21 2022-10-27 上海美悦生物科技发展有限公司 FXIa抑制剂及其药物组合物、制备方法和用途

Also Published As

Publication number Publication date
NZ731416A (en) 2024-01-26
TW202043228A (zh) 2020-12-01
ES2655884T3 (es) 2018-02-22
CN110734435B (zh) 2022-06-07
MY183987A (en) 2021-03-17
TW201613923A (en) 2016-04-16
JP2017530157A (ja) 2017-10-12
IL276470B (en) 2021-04-29
AU2020200376A1 (en) 2020-02-06
AU2021245098B2 (en) 2023-11-23
PH12017500580A1 (en) 2017-08-30
CA2963395C (en) 2023-03-14
JP2023134734A (ja) 2023-09-27
AU2021245098A1 (en) 2021-10-28
JP2019069989A (ja) 2019-05-09
JP6937734B2 (ja) 2021-09-22
PE20170939A1 (es) 2017-07-13
TN2017000112A1 (en) 2018-07-04
NZ766570A (en) 2024-01-26
KR20180126612A (ko) 2018-11-27
CO2017003833A2 (es) 2017-07-11
CY1119678T1 (el) 2018-04-04
PT3089979T (pt) 2018-01-16
JP2021185182A (ja) 2021-12-09
PH12020500195A1 (en) 2021-02-08
EA031590B1 (ru) 2019-01-31
UY36244A (es) 2016-01-29
MX2017003695A (es) 2017-05-30
CN110734435A (zh) 2020-01-31
MX2020010840A (es) 2020-11-06
AU2015324530B2 (en) 2019-10-24
SG11201702576QA (en) 2017-04-27
AU2015324530A1 (en) 2017-05-18
EP3828186B1 (en) 2023-08-23
SI3293186T1 (sl) 2021-01-29
JP7317905B2 (ja) 2023-07-31
AR101367A1 (es) 2016-12-14
IL251434A0 (en) 2017-05-29
LT3293186T (lt) 2020-12-28
SI3089979T1 (sl) 2017-12-29
KR102269999B1 (ko) 2021-06-25
ES2963267T3 (es) 2024-03-26
MA40123A1 (fr) 2017-09-29
KR101921436B1 (ko) 2018-11-22
LT3089979T (lt) 2017-12-27
EP3293186A1 (en) 2018-03-14
KR20170057431A (ko) 2017-05-24
DK3089979T3 (en) 2018-01-15
PT3293186T (pt) 2020-12-09
JP6462865B2 (ja) 2019-01-30
RS56786B1 (sr) 2018-04-30
ES2836270T3 (es) 2021-06-24
EP3828186A2 (en) 2021-06-02
ZA201702478B (en) 2019-06-26
TW202310844A (zh) 2023-03-16
EA201790595A1 (ru) 2017-07-31
PL3089979T3 (pl) 2018-02-28
CN114957255A (zh) 2022-08-30
CL2017000712A1 (es) 2017-11-03
CN106795161B (zh) 2019-10-15
RS61183B1 (sr) 2021-01-29
HUE038061T2 (hu) 2018-09-28
BR112017006702A2 (pt) 2017-12-26
EP4286372A2 (en) 2023-12-06
EP3828186A3 (en) 2021-07-28
CY1123663T1 (el) 2022-03-24
CN106795161A (zh) 2017-05-31
CA2963395A1 (en) 2016-04-07
IL276470A (en) 2020-09-30
PH12017500580B1 (en) 2017-08-30
SG10201911652TA (en) 2020-02-27
EP4286372A3 (en) 2024-02-21
EP3293186B1 (en) 2020-09-23
IL251434B (en) 2020-08-31
NO2721243T3 (US07585860-20090908-C00124.png) 2018-10-20
EP3089979B1 (en) 2017-10-18
TN2018000229A1 (en) 2019-10-04
HUE052812T2 (hu) 2021-05-28
HRP20201927T1 (hr) 2021-02-05
AU2020200376B2 (en) 2021-07-08
EP3089979A1 (en) 2016-11-09
DK3293186T3 (da) 2020-12-14
HRP20171950T1 (hr) 2018-02-23
TWI692478B (zh) 2020-05-01
TWI769442B (zh) 2022-07-01
PE20210922A1 (es) 2021-05-19

Similar Documents

Publication Publication Date Title
AU2021245098B2 (en) Pyrimidinones as factor XIa inhibitors
WO2015116882A1 (en) Macrocyclic factor xia inhibitors condensed with heterocycles
EP3189047B1 (en) Diamide macrocycles that are fxia inhibitors
WO2015116885A1 (en) Macrocycles with aromatic p2' groups as factor xia inhibitors
CA2880898A1 (en) Dihydropyridone p1 as factor xia inhibitors
EP3328851B1 (en) Factor xia macrocyclic inhibitors bearing alkyl or cycloalkyl p2' moieties
US10752641B2 (en) Diamide macrocycles having factor XIa inhibiting activity
EP3328852B1 (en) Factor xia macrocyclic inhibitors bearing a non-aromatic p2' group
BR112017006702B1 (pt) Pirimidinonas inibidoras de fator xia, composição farmacêutica e seu uso

Legal Events

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

Ref document number: 15747721

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015747721

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015747721

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/A/2017/003695

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 251434

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 000527-2017

Country of ref document: PE

Ref document number: 12017500580

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2017517766

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2963395

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 40123

Country of ref document: MA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 201790595

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017006702

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: NC2017/0003833

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 20177011233

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015324530

Country of ref document: AU

Date of ref document: 20150729

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112017006702

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20170331