WO2007012180A1 - Papain family cysteine protease inhibitors for the treatment of parasitic diseases - Google Patents

Papain family cysteine protease inhibitors for the treatment of parasitic diseases Download PDF

Info

Publication number
WO2007012180A1
WO2007012180A1 PCT/CA2006/001216 CA2006001216W WO2007012180A1 WO 2007012180 A1 WO2007012180 A1 WO 2007012180A1 CA 2006001216 W CA2006001216 W CA 2006001216W WO 2007012180 A1 WO2007012180 A1 WO 2007012180A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
rlo
aryl
halo
cycloalkyl
Prior art date
Application number
PCT/CA2006/001216
Other languages
French (fr)
Inventor
Cameron Black
Christophe Mellon
Deborah Anne Nicoll-Griffith
Renata Oballa
Original Assignee
Merck Frosst Canada Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Frosst Canada Ltd. filed Critical Merck Frosst Canada Ltd.
Priority to US11/989,437 priority Critical patent/US20090264479A1/en
Priority to EP06761177A priority patent/EP1909784A4/en
Priority to MX2008001207A priority patent/MX2008001207A/en
Priority to BRPI0614169A priority patent/BRPI0614169A2/en
Publication of WO2007012180A1 publication Critical patent/WO2007012180A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Cruzipain is a cysteine protease enzyme present in Trypanosoma cruzi and is thought to play an important role in all stages of the parasite's life cycle.
  • the enzyme is highly expressed in the epimastigote stage where it is primarily a lysosomal enzyme and may be involved in protein digestion during differentiation to the infective metacyclic trypomastigote stage. Idenfication of cruzipain in the membrane of the trypomastigote implicates this enzyme in the penetration of the parasite into the host cell.
  • Cruzipain is also found in the membranes of the amastigote form of the parasite, see Cazzulo, J. J., et al, Current Pharmaceutical Design, 7, 1143-1156, 2001.
  • cruzipain efficiently degrades human IgG, which may play a protective role for the parasite by preventing antigen presentation and thus reducing the host immune response. Based on these observations, it has been proposed that cruzipain is a valid drug target for chemotherapy of Chagas disease. Cruzipain has been reported to exist in at least two polymorphic sequences, known as cruzipain 1 and cruzipain 2, both of which may be involved in the viability of Trypanosoma cruzi (Lima, et al, Molecular & Parasitology 114, 41-52, 2001).
  • T. congolense A similar parasite, T. congolense, is responsible for the bovine disease trypanosomiasis.
  • Congopain is the analogous cysteine protease to cruzipain in this parasite.
  • Falcipain is an important cysteine protease in Plasmodium falicparum. This enzyme is reported to be important in the degradation of host hemoglobin in parasite food vacuoles. The processing of hemoglobin is essential to the growth of the parasite, thus an inhibitor of falcipain should be useful as a treatment for malaria.
  • SmCLl cysteine proteases
  • SmCL2 may play a role in the degradation of host hemoglobin
  • SmCL2 may be important to the reproductive system of the parasite (Brady, C. P., et al, Archives of Biochemistry and Biophysics, 380, 46-55, 2000). Inhibition of one or both of these proteases may provide an effective treatment for human schistosomiasis.
  • LmajcatB and CP2.8 ⁇ CTE are important cysteine proteases of the parasitic protazoa Leishmania major and Leishmania mexicanus respectively, see Alves, L.C., et al, Eur. J. Biochem, 268, 1206-1212, 2001. Inhibition of these enzymes may provide a useful treatment for leishmaniasis.
  • the present invention relates to compounds that are capable of treating and preventing mammalian parasitic diseases in which the parasite utilizes a critical cysteine protease from the papain family.
  • the present invention relates to a method of treating a parasitic disease with a papain family cysteine protease inhibitor.
  • parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
  • Papain family cysteine protease inhibitors of present invention is illustrated by a compound of Formula I, and the pharmaceutically acceptable salts, stereoisomers and N- oxide derivatives thereof:
  • the present invention relates to a method of treating a parasitic disease with a papain family cysteine protease inhibitor.
  • examples or parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
  • the present invention also relates to a method of preventing a parasitic disease with a papain family cysteine protease inhibitor.
  • Examples or parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
  • Papain family cysteine protease inhibitors of present invention is illustrated by a compound of the following formula, and the pharmaceutically acceptable salts, stereoisomers and N-oxide derivatives thereof.
  • Rl is hydrogen, Ci.6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3.6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R 9 , -SO2R 12 , - SO2CH(Rl2)(Rl l), -ORl2, -OR9, -N(Rl2)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from C 1-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
  • R2 is hydrogen, Ci_6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3.6 cycloalkyl, -SR.9, -SRl2, -SOR9, -SOR12, -SO2R 9 , -SO2R 12 , - S ⁇ 2CH(Rl2)(Rl l), -ORl2, -OR9, -N(Rl2) 2 , aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from Ci-6 alkyl, C ⁇ - ⁇ cycloalkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto; or Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring or heterocyclyl ring wherein said
  • R3 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo;
  • R4 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo; or R3 and R ⁇ can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring, C5-8 cycloalkenyl ring, or five to seven membered heterocyclyl ring wherein said cycloalkyl, cycloalkenyl and heterocyclyl rings are optionally substituted with C 1-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
  • R5 is hydrogen or Ci -6 haloalkyl
  • R6 is aryl, heteroaryl, Ci-6 haloalkyl, arylalkyl or heteroarylalkyl, wherein said aryl, heteroaryl, arylalkyl and heteroarylalkyl groups are optionally substituted with halo, Ci-6 alkyl, Ci-6 haloalkyl, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO 2 R9, -SO 2 Rl2, -S ⁇ 2CH(Rl2)(Rl l), -ORl2, -
  • D is C 1-3 alkyl, C2-3 alkenyl, C2-3 alkenyl, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl wherein said aryl, heteroaryl, cycloalkyl and heterocyclyl groups, which may be monocyclic or bicyclic, are optionally substituted on either the carbon or the heteroatom with one to five substituents selected from C 1-6 alkyl, halo or keto;
  • R7 is hydrogen, Cl -6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci_6 alkyl oxy, halo, nitro, cyano, aryl, heteroaryl, C3-8 cycloalkyl, heterocyclyl, -C(O)ORlO, -C(O)OSi[CH(CH3)2]3, -ORlO, -C(O)RlO, - R10C(0)R9, -C(O)R9, -C(O)N(Rl2)(Rl2), -C(O) N(RlO)(RlI), -C(RlO)(Rl I)OH, -SRl2, -SR9, - RlOSR9, .R9, -C(R9)3, -C(RlO)(Rl 1)N(R9) 2 , -NR10C(0)NR10S(0)2R 9 , -SO2R 12 , -
  • R9 is hydrogen, aryl, aryl(Ci-4) alkyl, heteroaryl, heteroaryl(Ci-4)alkyl, C3-gcycloalkyl, C3- 8cycloalkyl(Ci-4)alkyl, and heterocyclyl(Ci-4)alkyl wherein said groups can be optionally substituted with halo or alkoxy;
  • RlO is hydrogen or Ci_6 alkyl
  • Rl 1 is hydrogen or Ci -6 alkyl
  • Rl2 is hydrogen or Cl -6 alkyl which is optionally substituted with halo, alkoxy, cyano, -NRlO or -SRlO;
  • Ra is hydrogen, Cl -6 alkyl, (Ci -6 alkyl)aryl, (Ci -6 alkyl)hydroxyl, -O(Ci_6 alkyl), hydroxyl, halo, aryl, heteroaryl, C3.8 cycloalkyl or heterocyclyl, wherein said alkyl, aryl, heteroaryl, C3-8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with C 1-6 alkyl or halo;
  • Rb is hydrogen, Ci -6 alkyl, (Ci .5 alkyl)aryl, (Ci -6 alkyl)hydroxyl, alkoxyl, hydroxyl, halo, aryl, heteroaryl, C3.8 cycloalkyl or heterocyclyl,wherein said alkyl, aryl, heteroaryl, C3.8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with C 1-6 alkyl or halo; or R a and Rb can be taken together with the carbon atom to which they are attached or are between them to form a C3-8 cycloalkyl ring or C3-8 heterocyclyl ring wherein said 3-8 membered ring system may be optionally substituted with Ci-6 alkyl and halo; n is an integer from zero to three.
  • Rl is hydrogen.
  • R 2 is hydrogen.
  • Rl and R 2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring wherein said cycloalkyl ring is optionally substituted with one or two substituents independently selected from Cl -6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo.
  • R ⁇ is hydrogen
  • R4 is Ci-6 alkyl wherein said alkyl group is optionally substituted with C3-6 cycloalkyl, aryl or halo.
  • R ⁇ is hydrogen
  • R6 is CI -6 haloalkyl.
  • papain family cysteine protease inhibitors of the present invention include, but are not limited to:
  • the papain family cysteine protease inhibitors of the present invention are administered once weekly, biweekly, twice monthly or once monthly.
  • compositions which is comprised of a compound of Formula I as described above and a pharmaceutically acceptable carrier.
  • the invention is also contemplated to encompass a pharmaceutical composition which is comprised of a pharmaceutically acceptable carrier and any of the compounds specifically disclosed in the present application.
  • cysteine protease inhibitors for the treatment of Chagas disease and African trypanosomaisis has been discussed in the art. Substantiation of this hypothesis has been provided by the observation that irreversible inhibitors of cruzipain can cure Chagas disease in mouse models, see Engel, J., et al, J. Exp. Chem., 188, 725-734, 1998. Cruzipain has been reported to exist in at least two polymorphic sequences, known as cruzipain 1 and cruzipain 2, both of which may be involved in the viability of Trypanosoma cruzi (Lima, et al, Molecular & Parasitology 114, 41-52, 2001). A similar role for the cysteine protease trypanopain-Tb has been proposed in the life-cycle of Trypanosoma brucei, the parasite responsible for African trypanosomaisis, or sleeping sickness.
  • cysteine protease inhibitors for the treatment of malaria has been discussed in the art. Anti-malarial activity has been found with irreversible falcipain inhibitors in a mouse model of malaria (P. vinckei infection), see Olson, J. E., et al, Biorg. Med. Chem., 7, 633-638, 1999.
  • SmCLl cysteine proteases
  • SmCL2 may play a role in the degradation of host hemoglobin
  • SmCL2 may be important to the reproductive system of the parasite, see Brady, C. P., et al, Archives of Biochemistry and Biophysics, 380, 46-55, 2000.
  • inhibition of one or both of these proteases may provide an effective treatment for human schistosomiasis.
  • LmajcatB and CP2.8 ⁇ CTE are important cysteine proteases of the parasitic protazoa Leishmania major and Leishmania mexicanus respectively, see Alves, L.C., et al, Eur. J. Biochem, 268, 1206-1212, 2001. Thus, inhibition of these enzymes may provide a useful treatment for leishmaniasis.
  • Exemplifying the invention is the use of any of the compounds described above in the preparation of a medicament for the treatment and prevention of Chagas disease, toxoplasmosis, malaria, African trypanosomiasis, leishmaniasis or schistosomiasis in a mammal in need thereof.
  • the compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice.
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • a therapeutic compound in the case of tablets for oral use, carriers which are commonly used include lactose and corn starch, and lubricating agents, such as magnesium stearate, are commonly added.
  • useful diluents include lactose and dried corn starch.
  • the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension.
  • 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. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • 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, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents.
  • sweetening and/or flavoring agents may be added.
  • sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered.
  • the total concentration of solutes should be controlled in order to render the preparation isotonic.
  • 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 delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled.
  • the compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxy-ethylaspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • 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 polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
  • a drug for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
  • the instant compounds are also useful in combination with known agents useful for treating or preventing parasitic diseases, including toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis. Combinations of the presently disclosed compounds with other agents useful in treating or preventing parasitic diseases are within the scope of the invention. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the disease involved.
  • Existing therapies for Chagas Disease include, but are not limited to: nifurtimox, benznidazole, allopurinol.
  • Drugs that may have an effect on the parasite include but are not limited to: terbinafine, lovastatin, ketoconazole, itraconazole, posaconazole, miltefosine, ilmofosine, pamidronate, alendronate, and risedronate.
  • Chagas Disease Other mechanisms being explored for the treatment of Chagas Disease include, but are not limited to: inhibitors of trypanothione reductase and inhibitors of hypoxanthine- guanine phosphoribosyl transferase (HGPRT), See, Urbina, Current Pharmaceutical Design, 8, 287-295, 2002)
  • HGPRT hypoxanthine- guanine phosphoribosyl transferase
  • Existing therapies for malaria include, but are not limited to: chloroquine, proguanil, mefloquine, quinine, pyrimethamine-sulphadoxine, doxocycline, berberine, halofantrine, primaquine, atovaquone, pyrimethamine-dapsone, artemisinin and quinhaosu.
  • Existing therapies for leishmaniasis include, but are not limited to: meglumine antimonite, sodium stibogluconate and amphotericin B.
  • Existing therapies for schistosomiasis include, but are not limited to: praziquantel and oxamniquine.
  • Existing therapies for African trypanosomiasis include, but are not limited to: pentamidine, melarsoprol, suramin and eflornithine.
  • Such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range.
  • Compounds of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate.
  • administration means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment.
  • a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.)
  • administration and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
  • the present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound.
  • the term "administering” shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs,” ed. H. Bundgaard, Elsevier, 1985, which is incorporated by reference herein in its entirety. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • therapeutically effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • treating includes: preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease; inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
  • once weekly and "once-weekly dosing,” as used herein, means that a unit dosage, for example a unit dosage of a cruzipain inhibitor, is administered once a week, i.e., once during a seven-day period, preferably on the same day of each week.
  • the unit dosage is generally administered about every seven days.
  • a non-limiting example of a once-weekly dosing regimen would entail the administration of a unit dosage of the cruzipaininhibitor every Sunday. It is customarily recommended that a unit dosage for once-weekly administration is not administered on consecutive days, but the once-weekly dosing regimen can include a dosing regimen in which unit dosages are administered on two consecutive days falling within two different weekly periods.
  • biweekly dosing is meant that a unit dosage of the cruzipain inhibitor is administered once during a two week period, i.e. one time during a fourteen day period, preferably on the same day during each two week period.
  • each unit dosage is generally administered about every fourteen days.
  • a nonlimiting example of a biweekly dosing regimen would entail the administration of a unit dosage of the cruzipain inhibitor every other Sunday. It is preferred that the unit dosage is not administered on consecutive days, but the biweekly dosing regimen can include a dosing regimen in which the unit dosage is administered on two consecutive days within two different biweekly periods.
  • twice monthly dosing is meant that a unit dosage of the cruzipaininhibitor is administered twice, i.e. two times, during a monthly calendar period. With the twice monthly regimen, the doses are preferably given on the same two dates of each month. In the twice monthly dosing regimen, each unit dosage is generally administered about every fourteen to sixteen days.
  • a nonlimiting example of a twice monthly dosing regimen would entail dosing on or about the first of the month and on or about the fifteenth, i.e. the midway point, of the month.
  • the twice-monthly dosing regimen can include a dosing regimen in which the unit dosages are administered on two consecutive days within a monthly period, or different monthly periods.
  • the twice monthly regimen is defined herein as being distinct from, and not encompassing, the biweekly dosing regimen because the two regimens have a different periodicity and result in the administration of different numbers of dosages over long periods of time. For example, over a one year period, a total of about twenty four dosages would be administered according to the twice monthly regimen (because there are twelve calendar months in a year), whereas a total of about twenty six dosages would be administered according to the biweekly dosing regimen (because there are about fifty-two weeks in a year).
  • once monthly is used in accordance with the generally accepted meaning as a measure of time amounting to approximately four weeks, approximately 30 days or 1/12 of a calendar year.
  • the present invention also encompasses a pharmaceutical composition useful in the treatment of parasitic diseases, comprising the administration of a therapeutically effective amount of the compounds of this invention, with or without pharmaceutically acceptable carriers or diluents.
  • suitable compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacologically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's bloodstream by local bolus injection.
  • the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
  • a suitable amount of compound is administered to a mammal undergoing treatment for a parasitic disease.
  • Oral dosages of the present invention when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day.
  • the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably, from about 1 mg to about 100 mg of active ingredient.
  • the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
  • compounds of the present 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.
  • preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittant throughout the dosage regimen.
  • oral dosages of the present invention when used for the indicated effects, will range between about 0.01 mg per kg of body weight per week (mg/kg/week) to about 10 mg/kg/week, preferably 0.1 to 10 mg/kg/week, and most preferably 0.1 to 5.0 mg/kg/week.
  • the compositions are preferably provided in the form of tablets containing 2.5 mg, 3.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 35 mg, 40 mg, 50 mg, 80 mg, 100 mg, 200 mg, 400 mg, and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 2.5 mg to about 200 mg of the active ingredient, specifically, 2.5 mg, 3.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 35 mg, 40 mg, 50 mg, 80 mg, 100 mg, 200 mg, 400 mg and 500 mg of active ingredient.
  • the papain family cysteine protease inhibitor may be administered in a single weekly dose.
  • the papain family cysteine protease inhibitor may be administered in a biweekly, twice monthly or monthly dose.
  • the compounds of the present invention can be used in combination with other agents useful for treating parasitic diseases.
  • the individual components of such combinations can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
  • the instant invention is therefore to be understood as embracing all such regimes of simultaneous or alternating treatment and the term "administering" is to be interpreted accordingly.
  • the compounds of the present invention may have asymmetric centers, chiral axes, and chiral planes (as described in: E.L. Eliel and S.H. Wilen, Stereochemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention.
  • the compounds disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the scope of the invention, even though only one tautomeric structure is depicted. For example, any claim to compound A below is understood to include tautomeric structure B, and vice versa, as well as mixtures thereof.
  • any variable e.g. R.1, R2, R a etc.
  • its definition on each occurrence is independent at every other occurrence.
  • combinations of substituents and variables are permissible only if such combinations result in stable compounds.
  • Lines drawn into the ring systems from substituents indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms. If the ring system is polycyclic, it is intended that the bond be attached to any of the suitable carbon atoms on the proximal ring only.
  • substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
  • the phrase "optionally substituted with one or more substituents” should be taken to be equivalent to the phrase “optionally substituted with at least one substituent” and in such cases the preferred embodiment will have from zero to three substituents.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • Ci-Cio as in “Ci-Cio alkyl” is defined to include groups having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbons in a linear, branched, or cyclic arrangement.
  • Ci-Cio alkyl specifically includes methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and so on.
  • Alkoxy represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
  • cycloalkyl or “carbocycle” shall mean cyclic rings of alkanes of three to eight total carbon atoms, or any number within this range (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl).
  • alkenyl refers to a non-aromatic hydrocarbon radical, straight or branched, containing from 2 to 10 carbon atoms and at least 1 carbon to carbon double bond. Preferably 1 carbon to carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present.
  • C2-C6 alkenyl means an alkenyl radical having from 2 to 6 carbon atoms.
  • Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated.
  • cycloalkenyl shall mean cyclic rings of 3 to 10 carbon atoms and at least 1 carbon to carbon double bond (i.e., cycloprenpyl, cyclobutenyl, cyclopenentyl, cyclohexenyl, cycloheptenyl or cycloocentyl).
  • alkynyl refers to a hydrocarbon radical straight or branched, containing from 2 to 10 carbon atoms and at least 1 carbon to carbon triple bond. Up to 3 carbon-carbon triple bonds may be present.
  • C2-C6 alkynyl means an alkynyl radical having from 2 to 6 carbon atoms.
  • Alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
  • substituents may be defined with a range of carbons that includes zero, such as (Co-C6)alkylene-aryl. If aryl is taken to be phenyl, this definition would include phenyl itself as well as -CH2PI1, -CH2CH2PI1, CH(CH3) CH2CH(CH3)Ph, and so on.
  • aryl is intended to mean any stable monocyclic or bicyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic.
  • aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
  • the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
  • heteroaryl represents a stable monocyclic, bicyclic or tricyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S.
  • Heteroaryl groups within the scope of this definition include but are not limited to benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimi
  • heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. If the heteroaryl contains nitrogen atoms, it is understood that the corresponding N-oxides thereof are also encompassed by this definition.
  • halo or halogen as used herein is intended to include chloro, fluoro, bromo and iodo.
  • alkoxy as used herein means an alkyl portion, where alkyl is as defined above, connected to the remainder of the molecule via an oxygen atom. Examples of alkoxy include methoxy, ethoxy and the like.
  • haloalkyl includes an alkyl portion, where alkyl is as defined above, which is substituted with one to five halo.
  • arylalkyl includes an alkyl portion where alkyl is as defined above and to include an aryl portion where aryl is as defined above.
  • arylalkyl include, but are not limited to, benzyl, fluorobenzyl, chlorobenzyl, phenylethyl, phenylpropyl, fluorophenylethyl, and chlorophenylethyl.
  • alkylaryl include, but are not limited to, toluyl, ethylphenyl, and propylphenyl.
  • heteroarylalkyl shall refer to a system that includes a heteroaryl portion, where heteroaryl is as defined above, and contains an alkyl portion.
  • heteroarylalkyl include, but are not limited to, thienylmethyl, thienylethyl, thienylpropyl, pyridylmethyl, pyridylethyl and imidazoylmethyl.
  • hydroxyalkyl means a linear monovalent hydrocarbon raidcal of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2- hydroxypropyl, 3- hydroxypropyl, and the like.
  • heterocycle or “heterocyclyl” as used herein is intended to mean a 5- to 10- membered nonaromatic ring containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups.
  • Heterocyclyl therefore includes, but is not limited to the following: imidazolyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl, dihydropiperidinyl, tetrahydrothiophenyl and the like.
  • heterocycle contains a nitrogen atom, it is understood that the corresponding N-oxides thereof are also emcompassed by this definition.
  • the present invention also includes N-oxide derivatives and protected derivatives of compounds of Formula I.
  • compounds of Formula I when compounds of Formula I contain an oxidizable nitrogen atom, the nitrogen atom can be converted to an N-oxide by methods well known in the art.
  • compounds of Formula I when compounds of Formula I contain groups such as hydroxy, carboxy, thiol or any group containing a nitrogen atom(s), these groups can be protected with a suitable protecting groups.
  • a comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, Inc. 1981, the disclosure of which is incorporated herein by reference in its entirety.
  • the protected derivatives of compounds of Formula I can be prepared by methods well known in the art.
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituents may be unsubstituted or unsubstituted, unless specifically defined otherwise.
  • a (Ci -C ⁇ )alkyl may be substituted with one or more substituents selected from OH, oxo, halogen, alkoxy, dialkylamino, or heterocyclyl, such as morpholinyl, piperidinyl, and so on.
  • alkyl or aryl or either of their prefix roots appear in a name of a substituent (e.g., aryl C ⁇ -8 alkyl) it shall be interpreted as including those limitations given above for "alkyl” and "aryl.”
  • Designated numbers of carbon atoms e.g., Ci-io shall refer independently to the number of carbon atoms in an alkyl or cyclic alkyl moiety or to the alkyl portion of a larger substituent in which alkyl appears as its prefix root.
  • the pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed inorganic or organic acids.
  • conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like, as well as 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- acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
  • the preparation of the pharmaceutically acceptable salts described above and other typical pharmaceutically acceptable salts is more fully described by Berg et al., "Pharmaceutical Salts," J. Pharm. ScL, 1977:66: 1-19, hereby incorporated by reference.
  • the pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts of the basic compounds are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents. Similarly, the salts of the acidic compounds are formed by reactions with the appropriate inorganic or organic base.
  • EDCI 1 -(3 -dimethylarninopropyl)-3-ethy lcarbodiimide hydrochloride
  • HATU o-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • KOBu' potassium te/t-butoxide LiOH lithium hydroxide mCPBA metachloroperbenzoic acid
  • PdCl 2 (dppf) [ 1 , 1 ' -bis(diphenylphosphino)ferrocene]dichloropalladium(II)
  • a haloalkylketone or aldehyde may be condensed with an amino alcohol to give a cyclic aminal. Treatment with 3 equivalents of a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol. Oxidation of the alcohol with a chromium system such as a Jones oxidation, or alternatively by a two-step oxidation (eg oxalyl chloride/ DMS0/Et 3 N followed by NaClO) will provide the corresponding carboxylic acid. Peptide coupling and Suzuki reaction as described in Scheme 1 will provide compounds of the current invention.
  • Compounds of the present invention may also be prepared according to Scheme 3, as indicated below.
  • a haloalkylketone or aldehyde may be condensed with an amino alcohol to give an acyclic aminal.
  • Treatment with multiple equivalents of a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol.
  • This alcohol can be converted into compounds of the current invention by the method described in Scheme 2.
  • a hemiacetal may be condensed with an amino alcohol in which the alcohol moiety is protected with a suitable protecting group.
  • Treatment of the resulting imine with a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol.
  • the alcohol protecting group can then be removed and the alcohol can be converted into compounds of the current invention either by the method described in Scheme 2 or by first conducting the Suzuki reaction, followed by oxidizing the alcohol with H 5 IO 6 /CrO 3 and then peptide coupling.
  • Step 1 Preparation of (2SV1-I ftert-but ⁇ l(dimethvl)silvlloxvl-4-methvlpentan-2-amine
  • Step 3 Preparation of (2S)-2- ⁇ [(lS)-l-(4-bromophenyl)-2,2,2-trifluoroethyl]amino ⁇ -4- methylpentan- 1 -ol n-BuLi (2.5 M in hexanes, 42 mL) was added to a -70 0 C THF (400 mL) solution of 1,4- dibromobenzene (25.8 g) and the mixture was stirred for 25 minutes.
  • Step 4 Preparation of (2S)-4-methyl-2-( ⁇ (lS)-2,2,2-trifluoro-H4Xmethylthio)-l,l'- biphenyl-4-vH ethyl ) amino)pentan- 1 -ol
  • the ethyl acetate layer was separated and the aqueous further extracted with ethyl acetate (200 mL).
  • the combined ethyl acetate extracts were washed with 0.5 N NaOH (2 X 200 mL), with aqueous NH 4 Cl, brine and dried with magnesium sulfate. Removal of the solvent left a residue that was purified by chromatography on SiO 2 using a gradient of ethyl acetate and hexanes (1:4 to 1:3) and again with acetone and toluene (1: 10). The residue was dissolve in hot hexanes (200 mL) and the solution was allowed to cool to O 0 C under stirring. The obtained solid was filtered and dried to yield the title compound.
  • Step 5 Preparation of (2S)-4-methyl-2-( ⁇ (lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)-l,r- biphenyl-4-ynethyl ⁇ amino)pentan- 1 -ol
  • Step 6 Preparation of N- ⁇ ( 1 S)-2,2,2-trifluoro- 1 - [4'-(methyl sulf onyl)- 1 , 1 '-biphenyl-4-y 1] ethyl )-L-leucine
  • the crude acid from above (10 g) was dissolved in isopropyl acetate (250 mL) and extracted into cold 0.1 N NaOH (3 X 250 mL). The combined extracts were washed with diethyl ether (250 mL) and then slowly acidified with 6 N HCl to pH 4. The carboxylic acid was extracted with isopropyl acetate (2 X 250 mL) and the isopropyl acetate layer dried and concentrated to yield the product essentially pure and used as such in the next step.
  • the oxidizing reagent (H 5 IO 6 /CrO 3 ) was prepared as described in Tetrahedron Letters 39 (1998) 5323-5326 but using HPLC grade CH 3 CN (contains 0.5% water); no water was added.
  • Step 7 Preparation of N 1 (cyanomethyl)-N 2 ⁇ (lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)
  • Step 1 Preparation of methyl N-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L- leucinate
  • Methyl N-[(lS)-l-(4-bromophenyl)-2,2-difluoroethyl]-L-leucinate was prepared according to methods described in International Publication No. WO 03/075836, which published on
  • Step 2 Preparation of N-r(lS)-l-(2 ⁇ 6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl1-L-leucine
  • Step 3 Preparation of tert-butyl r(lS)-l-cyano-2-phenylethy ⁇ carbamate
  • Step 5 Preparation of N 1 -[(15)-l-cyano-2-phenylethyl]-N 2 -[(15)-l-(2',6'-difluorobiphenyl -4-yl)-2,2-difluoroethvn-L-leucinamide
  • N-[(lS)-l-(2 ⁇ 6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L-leucine 339 mg, 0.88 mmol
  • (25)-2-amino-3-phenylpropanenitrile methanesulfonate 214 mg, 0.88 mmol
  • HATU 400 mg, 1.0 mmol
  • triethylamine (0.40 mL, 2.9 mmol
  • 100 mg of N ; -(cyanomethyl)-N 2 -[2,2,2- trifluoro-l-(4'-piperazin-l-yl-l,r-biphenyl-4-yl)ethyl] -L-leucinamide is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0, hard-gelatin capsule.
  • the compounds disclosed in the present application exhibited activity in the following assays.
  • the compounds disclosed in the present application have an enhanced pharmacological profile relative to previously disclosed compounds.
  • T. cruzi (Brazilian strain) was initiated in a 25cm 2 flask with a cell density of 2x10 6 epimastigotes per mL and grown in liver infusion tryptose (LIT) broth medium, supplemented with 10% newborn calf serum (Gibco) and antibiotics, at 28°C with agitation (80 rpm) to a cell density of O.5xlO 7 to Ix 10 7 , measured with an electronic particle counter (model ZBI; Coulter Electronics Inc., Hialeah, FIa.) and by direct counting with a hemocytometer.
  • LIT liver infusion tryptose
  • Test compounds in DMSO were added to the flasks when the epimastigotes cell density reached 0.5xl0 7 to IxIO 7 per ml then incubated for 24 to 48h and the epimastigotes harvested during the logarithmic growth phase.
  • the harvested epimastigotes were washed three times with IM phosphate-buffered saline (PBS; pH 7.4) by centrifugation at 850 g for 15 minutes at 4°C.
  • PBS IM phosphate-buffered saline
  • the harvested epimastigotes were reincubated in fresh LIT broth supplemented with 10% newborn calf serum and antibiotics, at 28°C with agitation (80 rpm)and the viability of the epimistagotes evaluated for up to one week using trypan blue exclusion (light microscopy) and [ 3 H] -thymidine incorporation assay (see below).
  • the epimastigote forms of T. cruzi were grown as described above and harvested on day 14 (stationary phase) washed three times in Grace's insect medium pH 6.5 (Invitrogen or Wisent) and induced to the trypomastigote form by metacyclogenesis by the addition of fresh Grace medium supplemented with 10% fetal calf serum (FCS) and haemin (25 ⁇ g/ml) and cultured for up to five days at 28 0 C.
  • FCS fetal calf serum
  • haemin 25 ⁇ g/ml
  • the culture may be used to infect a monolayer of mammalian cells such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cells up to 4 days.
  • Trypomastigotes released to the supernatant were collected by a 3000 g centrifugation for 15 minutes and washed twice in Hank's balanced salt saline supplemented with ImM glucose (HBSS).
  • Test compounds in DMSO were added to the culture of trypomastigotes with a cell density of 10 6 per mL then incubated in RPMI- 10% at 37°C for 24 to 48h.
  • the trypomastigotes were harvested and reduction in number (parasite lysis) was determined using a Neubauer chamber and the LD 50 value (drug concentration that resulted in a 50% reduction in trypomastigotes when compared to an untreated control) was estimated by plotting percentage of reduction against the logarithm of drug concentration.
  • the viability of the harvested trypomastigotes was evaluated by their ability to infect macrophages and grow in fresh media as determined by a 3 H-thymidine incorporation assay (see below).
  • the epimastigotes form of T. cruzi was cultured in Grace's insect medium supplemented with 10% FCS and haemin (25 ⁇ g/ml) for up to fourteen days at 28°C to induce the formation of the metacyclic form, so that about 30% of the parasite cells were in the metacyclic form.
  • These parasite cells were harvested and used to infect confluent mammalian cells such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cell cultures grown in 24 wells microplates in MEM at 37°C and 5% CO 2 .
  • the culture media was removed and the test compounds in MEM culture medium were added to the wells and the microplates incubated for 48 h. At the end of the incubation period the media was removed and the macrogphages were fixed and stained with May Griinwald Giemsa stain. The number of amastigotes/100 macrophages (No. A/100 M ⁇ ) was determined and the anti-amastigote activity expressed as (%AA):
  • %AA [1- (No. A/100 M ⁇ )p/(No. A/100 M ⁇ )c] x 100 3 H-thymidine Inco ⁇ oration Assay
  • a 200 /xL MEM suspension containing a mammalian cell line such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cells was added to each well in 96 well flat-bottom microtitre plates and incubated for 24 to 48h at 37°C in 5% CO 2 . The medium was removed and the cells washed three times in PBS.
  • a 200 /LiL mixture of MEM containing lxl0 7 /mL stationary phase T. cruzi trypomastigotes was added to each well then incubated for 24 or 48h under the same conditions. After the incubation period the media was removed and the cells washed three times in PBS.
  • test compounds in MEM were added to the appropriate wells and incubated for up to three days. At the end of the incubation period the media was removed and the cells washed three times in PBS and the macrophages were lysed with 0.01% sodium dodecyl sulphate (SDS) and the parasitic cells harvested. The harvested parasitic cells were suspended in Grace's insect media and incubated at 28°C for 48 h. At the end of the incubation period 1 ⁇ Ci of 3 H-thymidine in Grace's insect media was added to each well and incubated for an additional 20 h; this was harvested and 3 H-thymidine incorporation was measured.
  • SDS sodium dodecyl sulphate

Abstract

The invention relates to the treatment of parasitic disease with inhibitors of the papain family cysteine proteases The parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Changas disease, leishmanasis and schistosomiasis The invention also relate to the pharmaceutical compositions comprising a papain family cysteine protease inhibitor and another agent in the treatment for parasitic disease.

Description

TITLE OF THE INVENTION
PAPAIN FAMILY CYSTEINE PROTEASE INHIB ITORS FOR THE TREATMENT OF PARASITIC
DISEASES
BACKGROUND OF THE INVENTION
Several parasites responsible for mammalian diseases are dependent on cysteine protease for various life-cycle functions. Inhibition of these proteases can be useful in the treatment of these parasitic diseases, see Lecaille, F., et al, Chem. Rev., 102, 4459-4488, 2002.
Cruzipain is a cysteine protease enzyme present in Trypanosoma cruzi and is thought to play an important role in all stages of the parasite's life cycle. The enzyme is highly expressed in the epimastigote stage where it is primarily a lysosomal enzyme and may be involved in protein digestion during differentiation to the infective metacyclic trypomastigote stage. Idenfication of cruzipain in the membrane of the trypomastigote implicates this enzyme in the penetration of the parasite into the host cell. Cruzipain is also found in the membranes of the amastigote form of the parasite, see Cazzulo, J. J., et al, Current Pharmaceutical Design, 7, 1143-1156, 2001. Cruzipain efficiently degrades human IgG, which may play a protective role for the parasite by preventing antigen presentation and thus reducing the host immune response. Based on these observations, it has been proposed that cruzipain is a valid drug target for chemotherapy of Chagas disease. Cruzipain has been reported to exist in at least two polymorphic sequences, known as cruzipain 1 and cruzipain 2, both of which may be involved in the viability of Trypanosoma cruzi (Lima, et al, Molecular & Parasitology 114, 41-52, 2001).
A similar role for the cysteine protease trypanopain-Tb has been proposed in the life- cycle of Trypanosoma brucei, the parasite responsible for African trypanosomaisis, or sleeping sickness.
A similar parasite, T. congolense, is responsible for the bovine disease trypanosomiasis. Congopain is the analogous cysteine protease to cruzipain in this parasite.
Falcipain is an important cysteine protease in Plasmodium falicparum. This enzyme is reported to be important in the degradation of host hemoglobin in parasite food vacuoles. The processing of hemoglobin is essential to the growth of the parasite, thus an inhibitor of falcipain should be useful as a treatment for malaria.
Two cysteine proteases, SmCLl and SmCL2, are present in the human blood fluke Schistosoma mansoni. SmCLl may play a role in the degradation of host hemoglobin, while SmCL2 may be important to the reproductive system of the parasite (Brady, C. P., et al, Archives of Biochemistry and Biophysics, 380, 46-55, 2000). Inhibition of one or both of these proteases may provide an effective treatment for human schistosomiasis.
LmajcatB and CP2.8ΔCTE are important cysteine proteases of the parasitic protazoa Leishmania major and Leishmania mexicanus respectively, see Alves, L.C., et al, Eur. J. Biochem, 268, 1206-1212, 2001. Inhibition of these enzymes may provide a useful treatment for leishmaniasis. The present invention relates to compounds that are capable of treating and preventing mammalian parasitic diseases in which the parasite utilizes a critical cysteine protease from the papain family.
SUMMARY OF THE INVENTION
The present invention relates to a method of treating a parasitic disease with a papain family cysteine protease inhibitor. Examples or parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
One embodiment of the papain family cysteine protease inhibitors of present invention is illustrated by a compound of Formula I, and the pharmaceutically acceptable salts, stereoisomers and N- oxide derivatives thereof:
Figure imgf000003_0001
I.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to a method of treating a parasitic disease with a papain family cysteine protease inhibitor. Examples or parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis. The present invention also relates to a method of preventing a parasitic disease with a papain family cysteine protease inhibitor. Examples or parasitic diseases include toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
One embodiment of the papain family cysteine protease inhibitors of present invention is illustrated by a compound of the following formula, and the pharmaceutically acceptable salts, stereoisomers and N-oxide derivatives thereof.
RexPs R' R3 H .N
O
wherein Rl is hydrogen, Ci.6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3.6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2R12, - SO2CH(Rl2)(Rl l), -ORl2, -OR9, -N(Rl2)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from C 1-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R2 is hydrogen, Ci_6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3.6 cycloalkyl, -SR.9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2R12, - Sθ2CH(Rl2)(Rl l), -ORl2, -OR9, -N(Rl2)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from Ci-6 alkyl, C^-β cycloalkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto; or Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring or heterocyclyl ring wherein said cycloalkyl and heterocycl rings are optionally substituted with one or two substituents independently selected from C 1-6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo;
R3 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo;
R4 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo; or R3 and R^ can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring, C5-8 cycloalkenyl ring, or five to seven membered heterocyclyl ring wherein said cycloalkyl, cycloalkenyl and heterocyclyl rings are optionally substituted with C 1-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R5 is hydrogen or Ci -6 haloalkyl;
R6 is aryl, heteroaryl, Ci-6 haloalkyl, arylalkyl or heteroarylalkyl, wherein said aryl, heteroaryl, arylalkyl and heteroarylalkyl groups are optionally substituted with halo, Ci-6 alkyl, Ci-6 haloalkyl, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2Rl2, -Sθ2CH(Rl2)(Rl l), -ORl2, -
N(RlO)(Rl I), or cyano;
D is C 1-3 alkyl, C2-3 alkenyl, C2-3 alkenyl, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl wherein said aryl, heteroaryl, cycloalkyl and heterocyclyl groups, which may be monocyclic or bicyclic, are optionally substituted on either the carbon or the heteroatom with one to five substituents selected from C 1-6 alkyl, halo or keto;
R7 is hydrogen, Cl -6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci_6 alkyl oxy, halo, nitro, cyano, aryl, heteroaryl, C3-8 cycloalkyl, heterocyclyl, -C(O)ORlO, -C(O)OSi[CH(CH3)2]3, -ORlO, -C(O)RlO, - R10C(0)R9, -C(O)R9, -C(O)N(Rl2)(Rl2), -C(O) N(RlO)(RlI), -C(RlO)(Rl I)OH, -SRl2, -SR9, - RlOSR9, .R9, -C(R9)3, -C(RlO)(Rl 1)N(R9)2, -NR10C(0)NR10S(0)2R9, -SO2R12, -SO(Rl2), -SO2R9, -Sθ2N(Rc)(Rd), -SO2CH(RlO)(Rl I), -Sθ2N(Rl0)C(O)(Rl2), -Sθ2(R10)C(O)N(Rl2)2, -OSO2RlO, . N(RlO)(RlI), -N(R10)C(O)N(R10)(R9), -N(RlO)C(O)RlO, -N(RlO)C(O)ORlO, -N(RlO)SO2(R10), - C(RlO)(Rl I)NRIOC(RlO)(Rl 1)R9, -C(RlO)(Rl 1)N(R10)R9, -C(RlO)(Rl I)N(RlO)(Rl 1), - C(RlO)(Rl I)SC(RlO)(Rl 1)(R9), RIOS-, -C(Ra)(Rb)NRaC(Ra)(Rb)(R9), -C(Ra)(Rb)N(Ra)(Rb), - C(Ra)(Rb)C(Ra)(Rb)N(Ra)(Rb), -C(O)C(Ra)(Rb)N(Ra)(Rb), -C(Ra)(Rb)N(Ra)C(O) R9, - C(O)C(Ra)(Rb)S(Ra) or C(Ra)(Rb)C(O)N(Ra)(Rb); wherein said alkyl, alkenyl, alkynyl, alkoxy, aryl, heteroaryl, cycloalkyl and heterocyclyl groups are optionally substituted on either the carbon or the heteroatom with one to five substituents independently selected from Ci -6 alkyl, halo, keto, cyano, haloalkyl, hydroxyalkyl, -OR9, -NO2, -NH2, -NHS(O)2R8, -R^SO2R12, -SO2Rl2, -SO(Rl2), - Sθ2N(RC)(Rd), -Sθ2N(Rl0)C(O)(Rl2), -C(RlO)(Rl I)N(RlO)(RlI), -C(RlO)(RlI)OH, -COOH, - C(Ra)(Rb)C(O)N(Ra)(Rb), -N(RlO)C(RlO)(Rl l)(R9χ -NH(CH2)2θH, -NHC(O)ORlO, -Si(CH3)3, heterocycyl, aryl or heteroaryl; R8 is hydrogen or Ci.6 alkyl; or R4 and R^ or can be taken together with any of the atoms to which they may be attached or are between them to form a 4-10 membered heterocyclyl ring system wherein said ring system, which may be monocyclic or bicyclic, is optionally substituted with Ci-6 alkyl, halo, hydroxyalkyl, hydroxy, keto, - ORlO, -SRlO or -N(RlO)2;
R9 is hydrogen, aryl, aryl(Ci-4) alkyl, heteroaryl, heteroaryl(Ci-4)alkyl, C3-gcycloalkyl, C3- 8cycloalkyl(Ci-4)alkyl, and heterocyclyl(Ci-4)alkyl wherein said groups can be optionally substituted with halo or alkoxy;
RlO is hydrogen or Ci_6 alkyl;
Rl 1 is hydrogen or Ci -6 alkyl;
Rl2 is hydrogen or Cl -6 alkyl which is optionally substituted with halo, alkoxy, cyano, -NRlO or -SRlO;
Ra is hydrogen, Cl -6 alkyl, (Ci -6 alkyl)aryl, (Ci -6 alkyl)hydroxyl, -O(Ci_6 alkyl), hydroxyl, halo, aryl, heteroaryl, C3.8 cycloalkyl or heterocyclyl, wherein said alkyl, aryl, heteroaryl, C3-8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with C 1-6 alkyl or halo;
Rb is hydrogen, Ci -6 alkyl, (Ci .5 alkyl)aryl, (Ci -6 alkyl)hydroxyl, alkoxyl, hydroxyl, halo, aryl, heteroaryl, C3.8 cycloalkyl or heterocyclyl,wherein said alkyl, aryl, heteroaryl, C3.8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with C 1-6 alkyl or halo; or Ra and Rb can be taken together with the carbon atom to which they are attached or are between them to form a C3-8 cycloalkyl ring or C3-8 heterocyclyl ring wherein said 3-8 membered ring system may be optionally substituted with Ci-6 alkyl and halo; n is an integer from zero to three.
In a class of the invention, Rl is hydrogen. In another class of the invention, R2 is hydrogen. In another class of the invention, Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring wherein said cycloalkyl ring is optionally substituted with one or two substituents independently selected from Cl -6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo.
In a class of the invention, R^ is hydrogen.
In a class of the invention, R4 is Ci-6 alkyl wherein said alkyl group is optionally substituted with C3-6 cycloalkyl, aryl or halo.
In a class of the invention, R^ is hydrogen.
In a class of the invention, R6 is CI -6 haloalkyl. Reference to the preferred embodiments set forth above is meant to include all combinations of particular and preferred groups unless stated otherwise.
Specific embodiments of the papain family cysteine protease inhibitors of the present invention include, but are not limited to:
N1 -(cyanomethyl)-N2-(2,2,2-trifluoro- 1 -phenylethyl)-L-leucinamide;
N1(cyanomethyl)-N2{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)-l,r-biphenyl-4-yl]ethyl}-L-leucinamide; N1-[(lS)-l-cyano-2-phenylethyl]-N2-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L- leucinamide;
N1-[(lR(2/?)-l-cyano-2-phenylcyclopropyl]-N2-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide;
N1-[(15,25)-l-cyano-2-phenylcyclopropyl]-N2-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide;
N1-[(lS,25)-l-cyano-2-phenylcyclopropyl]-N2-[(lS)-l-(2l,6'-difluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide;
N1-[(15)-l-cyano-2-phenylethyl]-N2-{(15)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4-yl]ethyl}-L- leucinamide;
N1 -[( IR)- 1 -cyano-2-(methylsulfonyl)ethyl]-N2-{ ( 1 S)-2,2,2-trifluoro- 1 -[4'-(methylsulfonyl)biphenyl-4- yljethyl } -L-leucinamide;
N1 -( 1 -cyanocyclopropyl)-N2-{ ( 15)-2,2,2-trifluoro- 1 -[4-(6-methylpyridin-2-yl)phenyl]ethyl } -L- leucinamide;
N1-[(15)-l-cyano-3-(methylthio)propyl]-N2-{(15)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4- yljethyl } -L-leucinamide;
(25)-4,4-dichloro-N-( 1 -cyanocyclopropyl)-2-( { ( 15)-2,2,2-trifluoro- 1 -[4'-(methylsulfonyl)biphenyl-4- yl]ethyl}amino)butanamide;
N'-tCl^-l-cyano-S-CmethylsulfonyOpropylJ-N^Kl^^^^-trifluoro-l-^'-CmethylsulfonyObiphenyM- yl]ethyl } -L-leucinamide;
N1-(l-cyanocyclopropyl)-N2-[(15)-l-(4'-{ l-[(cyclopropylamino)carbonyl]cyclopropyl}biphenyl-4-yl)- 2,2,2-trifluoroethyl]-L-leucinamide;
N1-(l-cyanocyclopropyl)-4-fluoro-N2-{(lS)-2,2,2-trifluoro-l-[4'-methoxy-3'-(methylsulfonyl)biphenyl-4- yl]ethyl } -L-leucinamide;
^-[cyanoCphenyOmethy^-^-iCl^^^^-trifluoro-l-^'-CmethylsulfonyDbiphenyl^-ylJethylJ-L- leucinamide; or a pharmaceutically acceptable salt, stereoisomer or Ν-oxide derivative thereof.
In a class of the invention, the papain family cysteine protease inhibitors of the present invention are administered once weekly, biweekly, twice monthly or once monthly.
Also included within the scope of the present invention is a pharmaceutical composition which is comprised of a compound of Formula I as described above and a pharmaceutically acceptable carrier. The invention is also contemplated to encompass a pharmaceutical composition which is comprised of a pharmaceutically acceptable carrier and any of the compounds specifically disclosed in the present application. These and other aspects of the invention will be apparent from the teachings contained herein.
Utilities
The use of cysteine protease inhibitors for the treatment of Chagas disease and African trypanosomaisis has been discussed in the art. Substantiation of this hypothesis has been provided by the observation that irreversible inhibitors of cruzipain can cure Chagas disease in mouse models, see Engel, J., et al, J. Exp. Chem., 188, 725-734, 1998. Cruzipain has been reported to exist in at least two polymorphic sequences, known as cruzipain 1 and cruzipain 2, both of which may be involved in the viability of Trypanosoma cruzi (Lima, et al, Molecular & Parasitology 114, 41-52, 2001). A similar role for the cysteine protease trypanopain-Tb has been proposed in the life-cycle of Trypanosoma brucei, the parasite responsible for African trypanosomaisis, or sleeping sickness.
The use of cysteine protease inhibitors for the treatment of malaria has been discussed in the art. Anti-malarial activity has been found with irreversible falcipain inhibitors in a mouse model of malaria (P. vinckei infection), see Olson, J. E., et al, Biorg. Med. Chem., 7, 633-638, 1999.
Two cysteine proteases, SmCLl and SmCL2, are present in the human blood fluke Schistosoma mansoni. SmCLl may play a role in the degradation of host hemoglobin, while SmCL2 may be important to the reproductive system of the parasite, see Brady, C. P., et al, Archives of Biochemistry and Biophysics, 380, 46-55, 2000. Thus, inhibition of one or both of these proteases may provide an effective treatment for human schistosomiasis.
LmajcatB and CP2.8ΔCTE are important cysteine proteases of the parasitic protazoa Leishmania major and Leishmania mexicanus respectively, see Alves, L.C., et al, Eur. J. Biochem, 268, 1206-1212, 2001. Thus, inhibition of these enzymes may provide a useful treatment for leishmaniasis.
Exemplifying the invention is the use of any of the compounds described above in the preparation of a medicament for the treatment and prevention of Chagas disease, toxoplasmosis, malaria, African trypanosomiasis, leishmaniasis or schistosomiasis in a mammal in need thereof.
The compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch, and lubricating agents, such as magnesium stearate, are commonly added. For oral administration in capsule form, useful diluents include lactose and dried corn starch. For oral use of a therapeutic compound according to this invention, the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension. For oral administration in the form of a tablet or capsule, 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. Moreover, when desired or necessary, 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, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents may be added. For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered. For intravenous use, the total concentration of solutes should be controlled in order to render the preparation isotonic.
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 delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxy-ethylaspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, 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 polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.
The instant compounds are also useful in combination with known agents useful for treating or preventing parasitic diseases, including toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis. Combinations of the presently disclosed compounds with other agents useful in treating or preventing parasitic diseases are within the scope of the invention. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the disease involved.
Existing therapies for Chagas Disease include, but are not limited to: nifurtimox, benznidazole, allopurinol. Drugs that may have an effect on the parasite include but are not limited to: terbinafine, lovastatin, ketoconazole, itraconazole, posaconazole, miltefosine, ilmofosine, pamidronate, alendronate, and risedronate. Other mechanisms being explored for the treatment of Chagas Disease include, but are not limited to: inhibitors of trypanothione reductase and inhibitors of hypoxanthine- guanine phosphoribosyl transferase (HGPRT), See, Urbina, Current Pharmaceutical Design, 8, 287-295, 2002)
Existing therapies for malaria include, but are not limited to: chloroquine, proguanil, mefloquine, quinine, pyrimethamine-sulphadoxine, doxocycline, berberine, halofantrine, primaquine, atovaquone, pyrimethamine-dapsone, artemisinin and quinhaosu.
Existing therapies for leishmaniasis include, but are not limited to: meglumine antimonite, sodium stibogluconate and amphotericin B.
Existing therapies for schistosomiasis include, but are not limited to: praziquantel and oxamniquine.
Existing therapies for African trypanosomiasis include, but are not limited to: pentamidine, melarsoprol, suramin and eflornithine.
If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range. Compounds of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate.
The term "administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.), "administration" and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents. The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs," ed. H. Bundgaard, Elsevier, 1985, which is incorporated by reference herein in its entirety. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The term "therapeutically effective amount" as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
The terms "treating" or "treatment" of a disease as used herein includes: preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease; inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
The terms "once weekly" and "once-weekly dosing," as used herein, means that a unit dosage, for example a unit dosage of a cruzipain inhibitor, is administered once a week, i.e., once during a seven-day period, preferably on the same day of each week. In the once-weekly dosing regimen, the unit dosage is generally administered about every seven days. A non-limiting example of a once-weekly dosing regimen would entail the administration of a unit dosage of the cruzipaininhibitor every Sunday. It is customarily recommended that a unit dosage for once-weekly administration is not administered on consecutive days, but the once-weekly dosing regimen can include a dosing regimen in which unit dosages are administered on two consecutive days falling within two different weekly periods.
By "biweekly" dosing is meant that a unit dosage of the cruzipain inhibitor is administered once during a two week period, i.e. one time during a fourteen day period, preferably on the same day during each two week period. In the twice-weekly dosing regimen, each unit dosage is generally administered about every fourteen days. A nonlimiting example of a biweekly dosing regimen would entail the administration of a unit dosage of the cruzipain inhibitor every other Sunday. It is preferred that the unit dosage is not administered on consecutive days, but the biweekly dosing regimen can include a dosing regimen in which the unit dosage is administered on two consecutive days within two different biweekly periods.
By "twice monthly" dosing is meant that a unit dosage of the cruzipaininhibitor is administered twice, i.e. two times, during a monthly calendar period. With the twice monthly regimen, the doses are preferably given on the same two dates of each month. In the twice monthly dosing regimen, each unit dosage is generally administered about every fourteen to sixteen days. A nonlimiting example of a twice monthly dosing regimen would entail dosing on or about the first of the month and on or about the fifteenth, i.e. the midway point, of the month. It is preferred that the unit dosages are not administered on the same or consecutive days but the twice-monthly dosing regimen can include a dosing regimen in which the unit dosages are administered on two consecutive days within a monthly period, or different monthly periods. The twice monthly regimen is defined herein as being distinct from, and not encompassing, the biweekly dosing regimen because the two regimens have a different periodicity and result in the administration of different numbers of dosages over long periods of time. For example, over a one year period, a total of about twenty four dosages would be administered according to the twice monthly regimen (because there are twelve calendar months in a year), whereas a total of about twenty six dosages would be administered according to the biweekly dosing regimen (because there are about fifty-two weeks in a year).
The term "once monthly" is used in accordance with the generally accepted meaning as a measure of time amounting to approximately four weeks, approximately 30 days or 1/12 of a calendar year.
The present invention also encompasses a pharmaceutical composition useful in the treatment of parasitic diseases, comprising the administration of a therapeutically effective amount of the compounds of this invention, with or without pharmaceutically acceptable carriers or diluents. Suitable compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacologically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's bloodstream by local bolus injection.
When a compound according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for a parasitic disease. Oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per day (mg/kg/day) to about 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day, and most preferably 0.1 to 5.0 mg/kg/day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably, from about 1 mg to about 100 mg of active ingredient. Intravenously, the most preferred doses will range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion. Advantageously, compounds of the present 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. Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittant throughout the dosage regimen.
In another exemplary application, oral dosages of the present invention, when used for the indicated effects, will range between about 0.01 mg per kg of body weight per week (mg/kg/week) to about 10 mg/kg/week, preferably 0.1 to 10 mg/kg/week, and most preferably 0.1 to 5.0 mg/kg/week. For oral administration, the compositions are preferably provided in the form of tablets containing 2.5 mg, 3.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 35 mg, 40 mg, 50 mg, 80 mg, 100 mg, 200 mg, 400 mg, and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 2.5 mg to about 200 mg of the active ingredient, specifically, 2.5 mg, 3.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 35 mg, 40 mg, 50 mg, 80 mg, 100 mg, 200 mg, 400 mg and 500 mg of active ingredient. Advantageously, the papain family cysteine protease inhibitor may be administered in a single weekly dose. Alternatively, the papain family cysteine protease inhibitor may be administered in a biweekly, twice monthly or monthly dose.
The compounds of the present invention can be used in combination with other agents useful for treating parasitic diseases. The individual components of such combinations can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms. The instant invention is therefore to be understood as embracing all such regimes of simultaneous or alternating treatment and the term "administering" is to be interpreted accordingly.
These and other aspects of the invention will be apparent from the teachings contained herein.
Definitions
The compounds of the present invention may have asymmetric centers, chiral axes, and chiral planes (as described in: E.L. Eliel and S.H. Wilen, Stereochemistry of Carbon Compounds, John Wiley & Sons, New York, 1994, pages 1119-1190), and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers and mixtures thereof, including optical isomers, being included in the present invention. In addition, the compounds disclosed herein may exist as tautomers and both tautomeric forms are intended to be encompassed by the scope of the invention, even though only one tautomeric structure is depicted. For example, any claim to compound A below is understood to include tautomeric structure B, and vice versa, as well as mixtures thereof.
Figure imgf000012_0001
A B
When any variable (e.g. R.1, R2, Ra etc.) occurs more than one time in any constituent, its definition on each occurrence is independent at every other occurrence. Also, combinations of substituents and variables are permissible only if such combinations result in stable compounds. Lines drawn into the ring systems from substituents indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms. If the ring system is polycyclic, it is intended that the bond be attached to any of the suitable carbon atoms on the proximal ring only.
It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results. The phrase "optionally substituted with one or more substituents" should be taken to be equivalent to the phrase "optionally substituted with at least one substituent" and in such cases the preferred embodiment will have from zero to three substituents.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, Ci-Cio, as in "Ci-Cio alkyl" is defined to include groups having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbons in a linear, branched, or cyclic arrangement. For example, "Ci-Cio alkyl" specifically includes methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, and so on. "Alkoxy" represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
The term "cycloalkyl" or "carbocycle" shall mean cyclic rings of alkanes of three to eight total carbon atoms, or any number within this range (i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl).
If no number of carbon atoms is specified, the term "alkenyl" refers to a non-aromatic hydrocarbon radical, straight or branched, containing from 2 to 10 carbon atoms and at least 1 carbon to carbon double bond. Preferably 1 carbon to carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present. Thus, "C2-C6 alkenyl" means an alkenyl radical having from 2 to 6 carbon atoms. Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted if a substituted alkenyl group is indicated.
The term "cycloalkenyl" shall mean cyclic rings of 3 to 10 carbon atoms and at least 1 carbon to carbon double bond (i.e., cycloprenpyl, cyclobutenyl, cyclopenentyl, cyclohexenyl, cycloheptenyl or cycloocentyl).
The term "alkynyl" refers to a hydrocarbon radical straight or branched, containing from 2 to 10 carbon atoms and at least 1 carbon to carbon triple bond. Up to 3 carbon-carbon triple bonds may be present. Thus, "C2-C6 alkynyl" means an alkynyl radical having from 2 to 6 carbon atoms. Alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted if a substituted alkynyl group is indicated.
In certain instances, substituents may be defined with a range of carbons that includes zero, such as (Co-C6)alkylene-aryl. If aryl is taken to be phenyl, this definition would include phenyl itself as well as -CH2PI1, -CH2CH2PI1, CH(CH3) CH2CH(CH3)Ph, and so on.
As used herein, "aryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl. In cases where the aryl substituent is bicyclic and one ring is non-aromatic, it is understood that attachment is via the aromatic ring.
The term "heteroaryl", as used herein, represents a stable monocyclic, bicyclic or tricyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S. Heteroaryl groups within the scope of this definition include but are not limited to benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, aziridinyl, 1,4-dioxanyl, hexahydroazepinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, tetrahydrothienyl, acridinyl,carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, isoxazolyl, isothiazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetra-hydroquinoline. In cases where the heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. If the heteroaryl contains nitrogen atoms, it is understood that the corresponding N-oxides thereof are also encompassed by this definition.
As appreciated by those of skill in the art, "halo" or "halogen" as used herein is intended to include chloro, fluoro, bromo and iodo. The term "keto" means carbonyl (C=O). The term "alkoxy" as used herein means an alkyl portion, where alkyl is as defined above, connected to the remainder of the molecule via an oxygen atom. Examples of alkoxy include methoxy, ethoxy and the like.
The term "haloalkyl" includes an alkyl portion, where alkyl is as defined above, which is substituted with one to five halo.
The term "arylalkyl" includes an alkyl portion where alkyl is as defined above and to include an aryl portion where aryl is as defined above. Examples of arylalkyl include, but are not limited to, benzyl, fluorobenzyl, chlorobenzyl, phenylethyl, phenylpropyl, fluorophenylethyl, and chlorophenylethyl. Examples of alkylaryl include, but are not limited to, toluyl, ethylphenyl, and propylphenyl.
The term "heteroarylalkyl" as used herein, shall refer to a system that includes a heteroaryl portion, where heteroaryl is as defined above, and contains an alkyl portion. Examples of heteroarylalkyl include, but are not limited to, thienylmethyl, thienylethyl, thienylpropyl, pyridylmethyl, pyridylethyl and imidazoylmethyl.
The term "hydroxyalkyl" means a linear monovalent hydrocarbon raidcal of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2- hydroxypropyl, 3- hydroxypropyl, and the like.
The term "heterocycle" or "heterocyclyl" as used herein is intended to mean a 5- to 10- membered nonaromatic ring containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes, but is not limited to the following: imidazolyl, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, tetrahydropyranyl, dihydropiperidinyl, tetrahydrothiophenyl and the like.
If the heterocycle contains a nitrogen atom, it is understood that the corresponding N-oxides thereof are also emcompassed by this definition.
The present invention also includes N-oxide derivatives and protected derivatives of compounds of Formula I. For example, when compounds of Formula I contain an oxidizable nitrogen atom, the nitrogen atom can be converted to an N-oxide by methods well known in the art. Also when compounds of Formula I contain groups such as hydroxy, carboxy, thiol or any group containing a nitrogen atom(s), these groups can be protected with a suitable protecting groups. A comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, Inc. 1981, the disclosure of which is incorporated herein by reference in its entirety. The protected derivatives of compounds of Formula I can be prepared by methods well known in the art.
The alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituents may be unsubstituted or unsubstituted, unless specifically defined otherwise. For example, a (Ci -Cό)alkyl may be substituted with one or more substituents selected from OH, oxo, halogen, alkoxy, dialkylamino, or heterocyclyl, such as morpholinyl, piperidinyl, and so on. In the case of a disubstituted alkyl, for instance, wherein the substituents are oxo and OH, the following are included in the definition: - (C=O)CH2CH(OH)CH3, -(C=O)OH, -CH2(OH)CH2CH(O), and so on.
Whenever the term "alkyl" or "aryl" or either of their prefix roots appear in a name of a substituent (e.g., aryl Cθ-8 alkyl) it shall be interpreted as including those limitations given above for "alkyl" and "aryl." Designated numbers of carbon atoms (e.g., Ci-io) shall refer independently to the number of carbon atoms in an alkyl or cyclic alkyl moiety or to the alkyl portion of a larger substituent in which alkyl appears as its prefix root.
The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed inorganic or organic acids. For example, conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like, as well as 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- acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like. The preparation of the pharmaceutically acceptable salts described above and other typical pharmaceutically acceptable salts is more fully described by Berg et al., "Pharmaceutical Salts," J. Pharm. ScL, 1977:66: 1-19, hereby incorporated by reference. The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts of the basic compounds are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents. Similarly, the salts of the acidic compounds are formed by reactions with the appropriate inorganic or organic base.
For purposes of this specification, the following abbreviations have the indicated meanings:
AcOH = acetic acid
B oc = t-butyloxycarbonyl
BoC2O = di-tert-butyl dicarbonate
BuLi = butyl lithium
CCl4 = carbon tetrachloride
CH2Cl2 = methylene chloride
CH3CN = acetonitrile
CHCl3 = chloroform
Cs2CO3 = cesium carbonate
CuI = copper iodide
DMA = N,N-dimethyl acetamide
DMAP = 4-(dimethylamino)pyridine
DMF = Ν,Ν-dimethylformamide
DMSO = dimethylsulfoxide
EDCI = 1 -(3 -dimethylarninopropyl)-3-ethy lcarbodiimide hydrochloride
Et2O = diethyl ether
Et3N = triethylamine
EtOAc = ethyl acetate
EtOH = ethanol
HATU = o-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
HOAc = acetic acid
K2CO3 = potassium carbonate
KOBu' = potassium te/t-butoxide LiOH lithium hydroxide mCPBA metachloroperbenzoic acid
MeOH methanol
MeSO3H methane sulfonic acid
MgSO4 magnesium sulfate
Ms methanesulfonyl = mesyl
MsCl methanesulfonyl chloride
NaBH4 sodium borohydride
NaH sodium hydride
Na2CO3 sodium carbonate
NaHCO3 sodium hydrogencarbonate
NaOH sodium hydroxide
Na2SO4 sodium sulfate
NBS N-bromosuccinimide
NH3 ammonia
NH4Cl ammonium chloride
Pd/C palladium on carbon
PdCl2(dppf) = [ 1 , 1 ' -bis(diphenylphosphino)ferrocene]dichloropalladium(II)
Pd2(dba)3 tris(dibenzylideneacetone)dipalladium(0)
PG protecting group
PPh3 triphenylphosphine
PPTS pyridinium p-toluenesulfonate
/Pr2NIi lithium diisopropyl amide
PyBOP benzotriazol-l-yloxytris(pyrrolidino)phosphonium-hexafluorophosphate rt room temperature sat. aq. = saturated aqueous
TFA trifluoroacetic acid
THF tetrahydrofuran tic thin layer chromatography
Me methyl
Et ethyl n-Pr normal propyl i-Pr isopropyl n-Bu normal butyl i-Bu isobutyl s-Bu secondary butyl t-Bu = tertiary butyl The compounds of the present invention can be prepared according to the following general procedures using appropriate materials and are further exemplified by the following specific examples. The compounds illustrated in the examples are not, however, to be construed as forming the only genus that is considered as the invention. The following examples further illustrate details for the preparation of the compounds of the present invention. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. All temperatures are degrees Celsius unless otherwise noted.
SCHEMES
Compounds of the present invention can be prepared according to Scheme 1, as indicated below. Thus an α-amino ester may be added to a haloalkyl ketone to form an aminal which may be dehydrated to an imine in the presence of a dehydrating agent such as TiCLi, MgSC>4 or isopropyl trifluoroacetate. Reduction of the imine with a reducing agent such as sodium cyanoborohydride or sodium borohydride provides the amine. Ester hydrolysis and amide formation with an appropriately substituted aminoacetonitrile provides compounds of the current invention. If the substituent on D system is a halogen, a palladium-catalyzed Suzuki coupling with an appropriate boronic acid provides additional compounds of the current invention.
Scheme 1
Figure imgf000019_0001
R7— B(OH)2 aq. Na2CO3, DMF, PdCI2(dppf), Δ
Figure imgf000019_0002
Compounds of the present invention may also be prepared according to Scheme 2, as indicated below. A haloalkylketone or aldehyde may be condensed with an amino alcohol to give a cyclic aminal. Treatment with 3 equivalents of a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol. Oxidation of the alcohol with a chromium system such as a Jones oxidation, or alternatively by a two-step oxidation (eg oxalyl chloride/ DMS0/Et3N followed by NaClO) will provide the corresponding carboxylic acid. Peptide coupling and Suzuki reaction as described in Scheme 1 will provide compounds of the current invention.
Scheme 2
HaIo-D-MgBr
Figure imgf000020_0001
Figure imgf000020_0002
Compounds of the present invention may also be prepared according to Scheme 3, as indicated below. A haloalkylketone or aldehyde may be condensed with an amino alcohol to give an acyclic aminal. Treatment with multiple equivalents of a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol. This alcohol can be converted into compounds of the current invention by the method described in Scheme 2.
Scheme 3
Figure imgf000020_0003
Compounds of the current invention may also be prepared according to Scheme 4, as indicated below. A hemiacetal may be condensed with an amino alcohol in which the alcohol moiety is protected with a suitable protecting group. Treatment of the resulting imine with a Grignard reagent or organolithium reagent will provide the appropriate alkylated amino alcohol. The alcohol protecting group can then be removed and the alcohol can be converted into compounds of the current invention either by the method described in Scheme 2 or by first conducting the Suzuki reaction, followed by oxidizing the alcohol with H5IO6 /CrO3 and then peptide coupling.
Scheme 4
MF,
Figure imgf000021_0001
The following examples describe the synthesis of selected compounds of the present invention.
EXAMPLE 1 Synthesis of N'-(cvanomethyl')-N2-('2,2.2-trifluoro-l-phenylethyl)-L-leucinamide
Figure imgf000021_0002
To a solution of L-leucine methyl ester hydrochloride (975 mg, 5.37 mmol) in dichloromethane (30 mL) was added 2,2,2-trifluoroacetophenone (0.75 mL, 5.34 mmol) and diisopropylethylamine (3.5 mL, 20 mmol). TiCl4 (0.55 mL, 5.0 mmol) in 0.45 mL dichloromethane was added dropwise, and the mixture was stirred overnight. Additional TiCl4 (0.4 mL, 3.6 mmol) was then added and the mixture was stirred 3h. A solution of NaCNBH3 (1050 mg, 16.7 mmol) in MeOH (20 mL) was added and the mixture was stirred 2h. Poured into IN NaOH and extracted with ethyl acetate (2x). The organic phase was washed with IN NaOH and brine, then dried over MgSO4 and evaporated. Purification by ISCO column chromatography (gradient 30% to 90% ethyl acetate/hexanes) provided methyl N-(2,2,2-trifluoro- 1 -phenylethyl)-L-leucinate. To a room temperature solution of methyl N-(2,2,2-trifluoro-l-phenylethyl)-L-leucinate (150 mg, 0.50 mmol) in 2:1 THF/MeOH was added IM LiOH. The mixture was stirred overnight and concentrated. The residue was partitioned between ethyl acetate and pH 3.5 phosphate buffer. The organic phase was washed with brine, dried over MgSO4 and concentrated to give N-(2,2,2-trifmoro-l- phenylethyl)-L-leucine.
A mixture of N-(2,2,2-trifluoro-l-phenylethyl)-L-leucine (149 mg, 0.50 mmol), aminoacetonitrile hydrochloride (102 mg, 1.1 mmol) and PyBOP (260 mg, 0.50 mmol) was dissolved in DMF (5 mL). Triethylamine (0.3 mL, 2.1 mmol) was added and the mixture was stirred overnight, then poured into pH 3 phosphate buffer and extracted with 3: 1 ether/ethyl acetate. The organic phase was washed with saturated aqueous NaHCO3 and brine, dried over MgSO4 and evaporated. Purification by ISCO column chromatography (gradient 20% to 50% ethyl acetate/hexanes) provided ^-(cyanomethyl)- N2-(2,2,2-trifluoro-l-phenylethyl)-L-leucinamide as a 1:1 mixture of diastereomers. MS (+APCI): 313.9 [M+l].
EXAMPLE 2
Synthesis of N1(cyanomethyl)-N2{(lS)-2,2 ,2-trifluoro- 1 -[4'-(methylsulfonyl)- 1 , 1 '-biphenyl-4-yl] ethvl I -L-leucinamide
Figure imgf000022_0001
Step 1: Preparation of (2SV1-I ftert-butγl(dimethvl)silvlloxvl-4-methvlpentan-2-amine
To a room temperature dichloromethane (100 mL) solution of L-leucinol (6.0 g) was added triethylamine (11 mL), DMAP (0.1 g) and t-butyldimethylsilyl chloride (8.5 g). The mixture was stirred at room temperature for 2 hours and then water was added. The organic layer was separated and the aqueous further extracted with dichloromethane. The combined organic layers were washed with brine, dried with magnesium sulfate and the solvent was removed in vacuo to yield the title compound, a residue which was used as such in the next reaction.
1H NMR (CD3COCD3) δ 3.48(m, 2H), 3.32(m, IH), 2.76(m, IH), 1.78(m, IH), 1.22-1.02(m, 2H), 0.88(m, 15H), 0.06(s, 6H).
Preparation of (25)-l-{ [te7t-butyl(dimethyl)silyl]oxy }-4-methyl-N-[(l£)-2,2,2- trifluoroethylideneipentan-2-amine
A toluene (300 mL) solution of (2S)-l-{ [rerf-butyl(dimethyl)silyl]oxy }-4-methylpentan- 2-amine from Step 1 (50 g) and tifluoroacetaldehyde methyl hemiacetal (35 mL) was heated to reflux for 16 hours during which time water was collected in a Dean-Stark trap. The solvent was evaporated in vacuum and the residue was purified on SiO2 using hexanes and ethyl acetate (9:1) as eluant to yield the title compound.
1H NMR (CD3COCD3) δ 7.88(m, IH), 3.76-3.45(m, 3H), 1.60-1.25(m, 3H), 0.88(m, 15H), 0.06(s, 3H), 0.04(s, 3H).
Step 3: Preparation of (2S)-2-{ [(lS)-l-(4-bromophenyl)-2,2,2-trifluoroethyl]amino}-4- methylpentan- 1 -ol n-BuLi (2.5 M in hexanes, 42 mL) was added to a -700C THF (400 mL) solution of 1,4- dibromobenzene (25.8 g) and the mixture was stirred for 25 minutes. A THF (30 mL) solution of (2S)-I- { [tert-butyl(dimethyl)silyl]oxy }-4-methyl-N-[(l£)-2,2,2-trifluoroethylidene]pentan-2-amine (31 g) was then added dropwise and the mixture was stirred for 1.5 hour. It was then poured slowly into a mixture of ethyl acetate (500 mL), water (2 L), ice (300 g) and ammonium chloride (100 g) under vigorous stirring. The organic layer was separated and the aqueous further extracted with ethyl acetate (2 X 500 mL). The combined organic layers were washed with brine, dried with magnesium sulfate and the solvent was removed in vacuo to yield a residue, which was used as such. The residue from above was dissolved in THF (250 mL) and the solution was cooled to 00C. A I M THF solution of t- butylammonium fluoride (110 mL) was added dropwise and the mixture was reacted for 4 hours. It was poured into ethyl acetate (300 mL), water (2 L) and ammonium chloride (100 g) under vigorous stirring. The organic layer was separated and the aqueous further extracted with ethyl acetate (2 X 100 mL). The combined organic layers were washed with brine, dried with magnesium sulfate and the solvent was removed in vacuo to yield a residue which was purified purified on SiO2 using a gradient of ethyl acetate and hexanes (1:5 to 1:4) as eluant to yield the title compound.
1H ΝMR (CD3COCD3) δ 7.6(2H, d), 7.45(2H, d), 4.55(1H, m), 3.65-3.7(1H, m), 3.5-3.55(1H, m), 3.25- 3.35(1H, m), 2.6-2.7(1H, m), 2.25-2.35(1H, m), 1.65-1.75(1H, m), 1.3-1.4(1H, m), 1.2-1.3(1H, m), 0.75- 0.9(6H, dd).
Step 4: Preparation of (2S)-4-methyl-2-({(lS)-2,2,2-trifluoro-H4Xmethylthio)-l,l'- biphenyl-4-vH ethyl ) amino)pentan- 1 -ol
A stream of nitrogen was passed through a suspension made of the bromide from Step 3 (27.7 g), 4-(methylthio)phenylboronic acid (15.7 g), 2 M Na2CO3 (100 mL) and n-propanol (500 mL) for 15 minutes. A 1:3 mixture (3.5 g) Of Pd(OAc)2 and PPh3 was then added and the reaction was warmed to 700C and stirred under nitrogen for 8 hours. The mixture was cooled to room temperature, diluted with ethylacetate (500 mL) and poured over water (2 L) and ice (500 g). The ethyl acetate layer was separated and the aqueous further extracted with ethyl acetate (200 mL). The combined ethyl acetate extracts were washed with 0.5 N NaOH (2 X 200 mL), with aqueous NH4Cl, brine and dried with magnesium sulfate. Removal of the solvent left a residue that was purified by chromatography on SiO2 using a gradient of ethyl acetate and hexanes (1:4 to 1:3) and again with acetone and toluene (1: 10). The residue was dissolve in hot hexanes (200 mL) and the solution was allowed to cool to O0C under stirring. The obtained solid was filtered and dried to yield the title compound.
1H NMR (CD3COCD3) δ 7.7(2H, d), 7.65(2H, d), 7.6(2H, d), 7.35(2H, d), 4.5-4.6(1H, m), 3.7(1H(OH), m), 3.5-3.6(1H, m), 3.3-3.4(1H, m), 2.7(1H, m), 2.5(3H, s), 2.3-2.4(1H(NH), m), 1.65-1.75(1H, m), 1.2- 1.4(3H, m), 0.8-0.9(6H, dd).
Step 5: Preparation of (2S)-4-methyl-2-({(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)-l,r- biphenyl-4-ynethyl } amino)pentan- 1 -ol
To a 00C solution of the sulfide (19 g) from Step 4 in toluene (400 mL) was added Na2WO4^H2O (0.16 g) and Bu4NHSO4 (0.81 g). Then 30 % hydrogen peroxide (12.2 mL) was slowly added and the mixture was stirred at room temperature for 4.5 hours. The mixture was poured slowly on a mixture of ice, dilute aqueous sodium thiosulfate and ethyl acetate. The organic layer was separated and the aqueous further extracted with ethyl acetate (2 X 100 mL). The combined organic layers were washed with brine, dried with magnesium sulfate and the solvent were removed in vacuo to yield a residue which was purified on SiO2 using ethyl acetate and hexanes (1:1) as eluant to yield the product.
1H NMR (CD3COCD3) δ 8.05(2H, d), 8.0(2H, d), 7.85(2H, d), 7.7(2H, d), 4.6-4.7(1H, m), 3.75(1H, m), 3.6(1H, m), 3.35-3.45(1H, m), 3.2(3H, s), 2.7-2.8(1H, m), 2.35-2.45(1H, m), 1.7-1.8(1H, m), 1.2-1.5(2H, m), 0.8-0.95(6H, dd).
Step 6: Preparation of N- { ( 1 S)-2,2,2-trifluoro- 1 - [4'-(methyl sulf onyl)- 1 , 1 '-biphenyl-4-y 1] ethyl )-L-leucine
A suspension of H5IO6 /CrO3 (529 mL of 0.44 M in CH3CN; see Note below) was cooled to 00C and a solution of the alcohol from Step 5 (20 g) in CH3CN (230 mL) was added dropwise. The mixture was stirred at 0-50C for 3.5 hours. It was poured into pH 4 Na2HPO4 (1.5 L) under vigorous stirring and the mixture was extracted with diethyl ether (3 X 250 mL). The combined ether extracts were washed with water and brine (1:1), with dilute aqueous NaHSO3 and brine. The organic extract was dried with sodium sulfate, filtered and the solvents were evaporated to dryness to yield a residue that was split into two batches for the following purification.
The crude acid from above (10 g) was dissolved in isopropyl acetate (250 mL) and extracted into cold 0.1 N NaOH (3 X 250 mL). The combined extracts were washed with diethyl ether (250 mL) and then slowly acidified with 6 N HCl to pH 4. The carboxylic acid was extracted with isopropyl acetate (2 X 250 mL) and the isopropyl acetate layer dried and concentrated to yield the product essentially pure and used as such in the next step.
Note: The oxidizing reagent (H5IO6 /CrO3) was prepared as described in Tetrahedron Letters 39 (1998) 5323-5326 but using HPLC grade CH3CN (contains 0.5% water); no water was added.
1H NMR (CD3COCD3) δ 8.05(2H, d), 7.95(2H, d), 7.8(2H, d), 7.65(2H, d), 4.45-4.55(1H, m), 3.55- 3.6(1H, m), 3.2(3H, s), 2.8-3.0(broad m, NH/OH)1.95-2.05(1H, m), 1.55-1.6(2H, m), 0.9-1.0(6H, m).
Step 7: Preparation of N1(cyanomethyl)-N2{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)
-1,1 '-biphenyl-4-yll ethyl 1 -L-leucinamide
To a DMF (200 mL) solution of the acid from Step 7 (9 g) was added benzotriazol-1- yloxytris(dimethylamino)phosphonium hexafluorophosphate (11.6 g), aminoacetonitrile hydrochloride (3.94 g) and the mixture was cooled to 00C. Triethylamine (9.9 mL) was added dropwise and the mixture warmed to room temperature and stirred for 16 hours. It was poured into ice and saturated aqueous sodium bicarbonate and extracted with diethyl ether (3 X 100 mL). The combined extracts were washed with brine, dried with magnesium sulfate and the solvent removed in vacuo. The residue was purified by chromatography on SiO2 using ethyl acetate and hexanes (1:1). The title compound was then stirred in diethyl ether for 16 hours, filtered and dried (mp 140.50C).
1H NMR (CD3COCD3) δ 8.0(2H, d), 7.95(2H, d), 7.8(2H, d), 7.65(2H, d), 4.35-4.45(1H, m), 4.1-4.2(2H, m), 3.45-3.55(lH, m), 3.15(3H, s), 2.65-2.7(1H, m), 1.85-1.95(1H, m), 1.4-1.6(2H, m), 0.85-0.95(6H, m).
EXAMPLE 3
Synthesis of N1-[(15)-l-cyano-2-phenylethyl]-N2-[(15)-l-(2',6'-difluorobiphenyl-4-yl)- 2.2-difluoroethvn -L-leucinamide
Figure imgf000025_0001
Step 1: Preparation of methyl N-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L- leucinate
Methyl N-[(lS)-l-(4-bromophenyl)-2,2-difluoroethyl]-L-leucinate was prepared according to methods described in International Publication No. WO 03/075836, which published on
September 18, 2003. To a solution of methyl N-[(lS)-l-(4-bromophenyl)-2,2-difluoroethyl]-L-leucinate (164 mg, 0.45 mmol) in DMF (4.5 mL) was added bis(pinacolato)diboron (137 mg, 0.54 mmol), PdCl2dppf ( 11 mg, 0.013 mmol) and KOAc (132 mg, 1.35 mmol). The mixture was placed under a nitrogen atmosphere, then heated with microwave irradiation to 1200C for 10 min. The mixture was cooled and partitioned between EtOAc and aq. NaHCO3. The aqueous layer was extracted 3x with EtOAc and the combined organic layers were washed with brine, dried over MgSO4 and concentrated to provide the crude product. To a solution of this material in DMF (4 mL) was added l-bromo-2,6-difluorobenzene (260 mg, 1.35 mmol), PdCl2dppf (11 mg, 0.013 mmol) and aq. Na2CO3 (2M solution, 0.68 mL). The mixture was heated with microwave irradiation to 120°C for 8 min. The mixture was cooled and partitioned between EtOAc and aq. NaHCO3. The aqueous layer was extracted 3x with EtOAc and the combined organic layers were washed with brine, dried over MgSO4 and concentrated. Purification by silica gel chromatography (gradient: hexanes to 15% EtOAc :hexanes) provided 150 mg of the title compound.
Step 2: Preparation of N-r(lS)-l-(2\6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl1-L-leucine
To a solution of methyl N-[(lS)-l-(2\6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L- leucinate in a mixture of THF (10 mL), MeOH (2 mL) and water (4 mL) was added lithium hydroxide hydrate (24 mg, 0.565 mmol). The mixture was stirred overnight, then diluted with pH 3 phosphate buffer and concentrated by one third. The residue was extracted twice with dichloromethane which was dried over MgSO4 and concentrated to provide the title compound.
Step 3: Preparation of tert-butyl r(lS)-l-cyano-2-phenylethyπcarbamate
To a solution of N-(teτt-butoxycarbonyl)-L-phenylalaninamide (1.86 g, 7.0 mmol) in dioxane (30 mL) was added pyridine (3 mL, 35 mmol) and trifluoroacetic anhydride (2 mL, 14 mmol). The mixture was stirred at room temperature overnight, then diluted with 1 : 1 EtO Ac/ether and washed sequentially with aq NaHCO3, water, IM HCl (2x) and brine. The organic phase was dried over MgSO4 and concentrated to provide the title compound.
Step 4: Preparation of (2S)-2-amino-3-phenylpropanenitrile
To a solution of tert-butyl [(I S)- l-cyano-2-phenylethyl] carbamate (1.8 g, 7.0 mmol) in dichloromethane (10 mL) was added thioanisole (1.65 mL, 14 mmol) followed by a solution of methanesulfonic acid (0.68 mL, 10.5 mmol) in dichloromethane (3 mL). The mixture was stirred for 1.5h, giving a suspension. Diethyl ether (50 mL) was added and the title compound was collected by filtration as its mesylate salt.
Step 5: Preparation of N1-[(15)-l-cyano-2-phenylethyl]-N2-[(15)-l-(2',6'-difluorobiphenyl -4-yl)-2,2-difluoroethvn-L-leucinamide To a solution of N-[(lS)-l-(2\6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L-leucine (339 mg, 0.88 mmol) and (25)-2-amino-3-phenylpropanenitrile methanesulfonate (214 mg, 0.88 mmol) in DMF (5 mL) was added HATU (400 mg, 1.0 mmol) followed by triethylamine (0.40 mL, 2.9 mmol). The mixture was stirred at room temperature overnight, then diluted with aq NaHCO3 and extracted with 1:1 EtO Ac/ether. The organic phase was washed with aq NaH2PO4 (3x) and brine, then dried over MgSO4 and concentrated. Purification by flash chromatography (30% EtOAc:hexanes) provided the title compound.
IH NMR (d6-acetone, 500 MHz) δ 8.12 (d, IH), 7.45 (m, 5H), 7.34 (m, 4H), 7.28 (m, IH), 7.10 (m, 2H), 6.05 (dt, IH), 5.08 (m, IH), 3.82 (m, IH), 3.40 (m, IH), 3.17 (m, IH), 3.08 (m, IH), 2.32 (m, IH), 1.80 (m, IH), 1.42 (m, 2H), 0.88 (m, 6H).
EXAMPLE 4
Using the methods described above, the following compounds were prepared:
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Pharmaceutical Composition
As a specific embodiment of this invention, 100 mg of N;-(cyanomethyl)-N2-[2,2,2- trifluoro-l-(4'-piperazin-l-yl-l,r-biphenyl-4-yl)ethyl] -L-leucinamide, is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0, hard-gelatin capsule.
The compounds disclosed in the present application exhibited activity in the following assays. In addition, the compounds disclosed in the present application have an enhanced pharmacological profile relative to previously disclosed compounds.
Cruzipain Assay
Serial dilutions (1/3) from 500 μM down to 0.0085 μM of test compounds were prepared in dimethyl sulfoxide (DMSO). Then 2 μL of DMSO from each dilution were added to 100 μL of cruzipain (500 ng/mL) in assay buffer solution (NaOAc, 50 mM (pH 5.5); DTT, 5 mM; and DMSO 10% v/v). The assay solutions were mixed for 5-10 seconds on a shaker plate and incubated for 15 minutes at room temperature. Z-Phe-Arg-AMC (20 μM) in 10 μL of assay buffer was added to the assay solutions. Hydrolysis of the coumarin leaving group (AMC) was followed by spectrofluorometry (Exλ =350 nm; Emλ = 460 nm) for 10 minutes. Percent of inhibition were calculated by fitting experimental values to standard mathematical model for dose response curve.
T. cruzi Epimastigotes Assay
The epimastigote form of T. cruzi (Brazilian strain) was initiated in a 25cm2 flask with a cell density of 2x106 epimastigotes per mL and grown in liver infusion tryptose (LIT) broth medium, supplemented with 10% newborn calf serum (Gibco) and antibiotics, at 28°C with agitation (80 rpm) to a cell density of O.5xlO7 to Ix 107, measured with an electronic particle counter (model ZBI; Coulter Electronics Inc., Hialeah, FIa.) and by direct counting with a hemocytometer. Test compounds in DMSO were added to the flasks when the epimastigotes cell density reached 0.5xl07 to IxIO7 per ml then incubated for 24 to 48h and the epimastigotes harvested during the logarithmic growth phase. The harvested epimastigotes were washed three times with IM phosphate-buffered saline (PBS; pH 7.4) by centrifugation at 850 g for 15 minutes at 4°C. The harvested epimastigotes were reincubated in fresh LIT broth supplemented with 10% newborn calf serum and antibiotics, at 28°C with agitation (80 rpm)and the viability of the epimistagotes evaluated for up to one week using trypan blue exclusion (light microscopy) and [3H] -thymidine incorporation assay (see below).
T. cruzi Trypomastigote Assay
The epimastigote forms of T. cruzi were grown as described above and harvested on day 14 (stationary phase) washed three times in Grace's insect medium pH 6.5 (Invitrogen or Wisent) and induced to the trypomastigote form by metacyclogenesis by the addition of fresh Grace medium supplemented with 10% fetal calf serum (FCS) and haemin (25 μg/ml) and cultured for up to five days at 280C. To produce more trypomastigotes the culture may be used to infect a monolayer of mammalian cells such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cells up to 4 days. Trypomastigotes released to the supernatant were collected by a 3000 g centrifugation for 15 minutes and washed twice in Hank's balanced salt saline supplemented with ImM glucose (HBSS). Test compounds in DMSO were added to the culture of trypomastigotes with a cell density of 106per mL then incubated in RPMI- 10% at 37°C for 24 to 48h. The trypomastigotes were harvested and reduction in number (parasite lysis) was determined using a Neubauer chamber and the LD50 value (drug concentration that resulted in a 50% reduction in trypomastigotes when compared to an untreated control) was estimated by plotting percentage of reduction against the logarithm of drug concentration. The viability of the harvested trypomastigotes was evaluated by their ability to infect macrophages and grow in fresh media as determined by a 3H-thymidine incorporation assay (see below).
T. cruzi Amastigote Activity (intracellular) Assay
The epimastigotes form of T. cruzi was cultured in Grace's insect medium supplemented with 10% FCS and haemin (25 μg/ml) for up to fourteen days at 28°C to induce the formation of the metacyclic form, so that about 30% of the parasite cells were in the metacyclic form. These parasite cells were harvested and used to infect confluent mammalian cells such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cell cultures grown in 24 wells microplates in MEM at 37°C and 5% CO2. After the parasitic cells were allowed to infect the macrophages, the culture media was removed and the test compounds in MEM culture medium were added to the wells and the microplates incubated for 48 h. At the end of the incubation period the media was removed and the macrogphages were fixed and stained with May Griinwald Giemsa stain. The number of amastigotes/100 macrophages (No. A/100 Mø) was determined and the anti-amastigote activity expressed as (%AA):
%AA = [1- (No. A/100 Mø)p/(No. A/100 Mø)c] x 100 3H-thymidine Incoφoration Assay
A 200 /xL MEM suspension containing a mammalian cell line such as U937 (human macrophage), J774 (mouse macrophage) or Vero (African green monkey kidney) cells was added to each well in 96 well flat-bottom microtitre plates and incubated for 24 to 48h at 37°C in 5% CO2. The medium was removed and the cells washed three times in PBS. A 200 /LiL mixture of MEM containing lxl07/mL stationary phase T. cruzi trypomastigotes was added to each well then incubated for 24 or 48h under the same conditions. After the incubation period the media was removed and the cells washed three times in PBS. The test compounds in MEM were added to the appropriate wells and incubated for up to three days. At the end of the incubation period the media was removed and the cells washed three times in PBS and the macrophages were lysed with 0.01% sodium dodecyl sulphate (SDS) and the parasitic cells harvested. The harvested parasitic cells were suspended in Grace's insect media and incubated at 28°C for 48 h. At the end of the incubation period 1 μCi of 3H-thymidine in Grace's insect media was added to each well and incubated for an additional 20 h; this was harvested and 3H-thymidine incorporation was measured.

Claims

WHAT IS CLAIMED IS:
1. A method of treating a parasitic disease with a papain family cysteine protease inhibitor.
2. The method of Claim 1 wherein the parasitic disease is toxoplasmosis, malaria, African trypanosomiasis, Chagas disease, leishmaniasis or schistosomiasis.
3. The method of Claim 1 wherein the papain family cysteine protease inhibitor is a compound of formula I:
Figure imgf000034_0001
wherein Rl is hydrogen, C 1-6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SORl2, -SO2R9, -SO2R12, - SO2CH(Rl2)(Rl l), -ORl2, -OR9, -N(Rl2)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from C\ -6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R2 is hydrogen, C 1-6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2R12, - Sθ2CH(Rl2)(Rl 1), -ORl2, -OR9, -N(R12)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from Ci-6 alkyl, C3-6 cycloalkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto; or Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring or heterocyclyl ring wherein said cycloalkyl and heterocycl rings are optionally substituted with one or two substituents independently selected from C 1-6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo;
R3 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo;
R4 is hydrogen, Ci_6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo; or R3 and R4 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring, C5-8 cycloalkenyl ring, or five to seven membered heterocyclyl ring wherein said cycloalkyl, cycloalkenyl and heterocyclyl rings are optionally substituted with C 1-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R5 is hydrogen or C 1-6 haloalkyl;
R6 is aryl, heteroaryl, Ci -6 haloalkyl, arylalkyl or heteroarylalkyl, wherein said aryl, heteroaryl, arylalkyl and heteroarylalkyl groups are optionally substituted with halo, C 1-6 alkyl, C 1-6 haloalkyl, C3-6 cycloalkyl, -SR9, -SR12, -SOR9, -SOR12, -SO2R9, -SO2R12, -Sθ2CH(Rl2)(Rll), -ORl2, -
N(RlO)(Rl I), or cyano;
D is C 1-3 alkyl, C2-3 alkenyl, C2-3 alkenyl, aryl, heteroaryl, C3_8 cycloalkyl or heterocyclyl wherein said aryl, heteroaryl, cycloalkyl and heterocyclyl groups, which may be monocyclic or bicyclic, are optionally substituted on either the carbon or the heteroatom with one to five substituents selected from Ci-6 alkyl, halo or keto;
R7 is hydrogen, Cl -6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 1-6 alkyloxy, halo, nitro, cyano, aryl, heteroaryl, C3-8 cycloalkyl, heterocyclyl, -C(O)ORlO, -C(O)OSi[CH(CH3)2]3, -ORlO, -C(O)RlO, - R10C(0)R9, -C(O)R9, -C(O)N(R 12)(R 12), -C(O) N(RlO)(Rl I), -C(RlO)(Rl I)OH, -SRl2, -SR9, - RlOSR9, -R9, -C(R9)3, -C(RlO)(Rl 1)N(R9)2, -NRIOC(O)NRIOS(O^R9, -SO2R12, -SO(Rl2), -SO2R9, -Sθ2N(RC)(Rd), -802CH(RlO)(Rl I), -SO2N(Rl0)C(O)(Rl2), -Sθ2(R10)C(O)N(Rl2)2, -OSO2RIO, - N(RlO)(Rl I), -N(R10)C(O)N(R10)(R9), -N(RlO)C(O)RlO, -N(RlO)C(O)ORlO, -N(RlO)Sθ2(R10), - C(RlO)(Rl 1)NR10C(R10)(R11)R9, -C(RlO)(Rl 1)N(R1O)R9, -C(RlO)(Rl I)N(RlO)(Rl I), - C(RlO)(Rl I)SC(RlO)(Rl 1)(R9), RIOS-, -C(Ra)(Rb)NRaC(Ra)(Rb)(R9), -C(Ra)(Rb)N(Ra)(Rb), . C(Ra)(Rb)C(Ra)(Rb)N(Ra)(Rb), -C(O)C(Ra)(Rb)N(Ra)(Rb), -C(Ra)(Rb)N(Ra)C(O) R9, - C(O)C(Ra)(Rb)S(Ra) or C(Ra)(Rb)C(O)N(Ra)(Rb); wherein said alkyl, alkenyl, alkynyl, alkoxy, aryl, heteroaryl, cycloalkyl and heterocyclyl groups are optionally substituted on either the carbon or the heteroatom with one to five substituents independently selected from Ci -6 alkyl, halo, keto, cyano, haloalkyl, hydroxyalkyl, -OR9, -NO2, -NH2, -NHS(O)2R8, -R9SO2R12, -SO2R12, -SO(Rl2), - Sθ2N(RC)(Rd), -Sθ2N(Rl0)C(O)(Rl2), -C(RlO)(Rl I)N(RlO)(RlI), -C(RlO)(RlI)OH, -COOH, - C(Ra)(Rb)C(O)N(Ra)(Rb), -N(RlO)C(RlO)(Rl 1)(R9), -NH(CH2)2θH, -NHC(O)ORlO, -Si(CH3)3, heterocycyl, aryl or heteroaryl; R8 is hydrogen or Ci -6 alkyl; or R4 and R^ or can be taken together with any of the atoms to which they may be attached or are between them to form a 4-10 membered heterocyclyl ring system wherein said ring system, which may be monocyclic or bicyclic, is optionally substituted with Ci -6 alkyl, halo, hydroxyalkyl, hydroxy, keto, - ORlO, -SRlO or -N(RlO)2;
R9 is hydrogen, aryl, aryl(Ci_4) alkyl, heteroaryl, heteroaryl(Ci-4)alkyl, C3-8cycloalkyl, C3- 8cycloalkyl(Ci-4)alkyl, and heterocyclyl(Ci-4)alkyl wherein said groups can be optionally substituted with halo or alkoxy;
RlO is hydrogen or Ci -6 alkyl;
Rl 1 is hydrogen or Ci-6 alkyl;
Rl2 is hydrogen or Ci-6 alkyl which is optionally substituted with halo, alkoxy, cyano, -NRlO Or -SRlO; Ra is hydrogen, Ci_6 alkyl, (Ci -6 alkyl)aryl, (Q -6 alkyl)hydroxyl, -O(Ci-6 alkyl), hydroxyl, halo, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl, wherein said alkyl, aryl, heteroaryl, C3-8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with Ci_6 alkyl or halo; Rb is hydrogen, Ci_6 alkyl, (Ci -6 alkyl)aryl, (Ci -6 alkyl)hydroxyl, alkoxyl, hydroxyl, halo, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl,wherein said alkyl, aryl, heteroaryl, C3.8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with Ci_6 alkyl or halo; or Ra and Rb can be taken together with the carbon atom to which they are attached or are between them to form a C3_8 cycloalkyl ring or C3-8 heterocyclyl ring wherein said 3-8 membered ring system may be optionally substituted with C 1-6 alkyl and halo; n is an integer from zero to three; or a pharmaceutically acceptable salt, stereoisomer or N-oxide derivative thereof.
4. The method of Claim 3 wherein Rl is hydrogen; R2 is hydrogen; or Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring wherein said cycloalkyl ring is optionally substituted with one or two substituents independently selected from C 1-6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo; or a pharmaceutically acceptable salt, stereoisomer or N-oxide derivative thereof.
5. The method of Claim 4 wherein R3 is hydrogen; R4 is Cl-6 alkyl wherein said alkyl group is optionally substituted with C3-6 cycloalkyl, aryl or halo; or a pharmaceutically acceptable salt, stereoisomer or N-oxide derivative thereof.
6. The method of Claim 5 wherein R5 is hydrogen and R6 is C 1-6 haloalkyl; or a pharmaceutically acceptable salt, stereoisomer or N-oxide derivative thereof.
7. The method of Claim 1 wherein the papain family cysteine protease inhibitor is selected from
N1-(cyanomethyl)-N2-(2,2,2-trifluoro-l-phenylethyl)-L-leucinamide;
N1(cyanomethyl)-N2{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)-l,r-biphenyl-4-yl]ethyl}-L-leucinamide; N1-[(lS)-l-cyano-2-phenylethyl]-N2-[(lS)-l-(2',6'-difluorobiphenyl-4-yl)-2,2-difluoroethyl]-L- leucinamide;
N1-[(l/?,2R)-l-cyano-2-phenylcyclopropyl]-N2-[(lS)-l-(2',6'-<iifluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide;
N1-[(15,25)-l-cyano-2-phenylcyclopropyl]-N2-[(15)-l-(2',6'-difluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide;
N1-[(15,25)-l-cyano-2-phenylcyclopropyl]-N2-[(lS)-l-(2',6'-difluorobiphenyl-4-yl)-2,2,2-trifluoroethyl]- L-leucinamide; N1-[(15)-l-cyano-2-phenylethyl]-N2-{(15)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4-yl]ethyl}-L- leucinamide;
N1-[(lR)-l-cyano-2-(methylsulfonyl)ethyl]-N2-{(15)-2,2,2-trifluoro-l-t4'-(methylsulfonyl)biphenyl-4- yl]ethyl }-L-leucinamide;
N1-(l-cyanocyclopropyl)-N2-{(15)-2,2,2-trifluoro-l-[4-(6-methylpyridin-2-yl)phenyl]ethyl}-L- leucinamide;
N1-[(15)-l-cyano-3-(methylthio)propyl]-N2-{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4- yl]ethyl }-L-leucinamide;
(25)-4,4-dichloro-N-(l-cyanocyclopropyl)-2-({(lS)-2,2,2-trifluoro-l-[41-(methylsulfonyl)biphenyl-4- yl]ethyl } amino)butanamide;
N1-[(lS)-l-cyano-3-(methylsulfonyl)propyl]-N2-{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4- yl]ethyl } -L-leucinamide;
N1 -( 1-cy anocyclopropy l)-N2-[( 1 S)- 1 -(41- { 1 -[(cyclopropy lamino)carbony 1] cy clopropy 1 }bipheny 1-4-yl)-
2,2,2-trifluoroethyl]-L-leucinarnide;
N1 -( 1 -cyanocyclopropy l)-4-fluoro-Λ/2- { ( 1 S)-2,2,2-trifluoro- 1 - [4'-methoxy-3'-(methylsulfonyl)bipheny 1-4- yl]ethyl } -L-leucinamide;
N1-[cyano(phenyl)methyl]-N2-{(lS)-2,2,2-trifluoro-l-[4'-(methylsulfonyl)biphenyl-4-yl]ethyl}-L- leucinamide; or a pharmaceutically acceptable salt, stereoisomer or Ν-oxide derivative thereof.
8. The method of Claim 1 comprising another agent selected from the group consisting of nifurtimox, benznidazole, allopurinol, terbinafine, lovastatin, ketoconazole, itraconazole, posaconazole, miltefosine, ilmofosine, pamidronate, alendronate, risedronate, chloroquine, proguanil, mefloquine, quinine, pyrimethamine-sulphadoxine, doxocycline, berberine, halofantrine, primaquine, atovaquone, pyrimethamine-dapsone, artemisinin, quinhaosu. meglumine antimonite, sodium stibogluconate, amphotericin B, praziquantel, oxamniquine, pentamidine, melarsoprol, suramin and eflomithine.
9. A pharmaceutical composition comprising a papain family cysteine protease inhibitor of formula I:
Figure imgf000037_0001
wherein Rl is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3.6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2R12, - SO2CH(Rl2)(Rl l), -OR12, -OR9, -N(R 12)2, aryU heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from Cl -6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R2 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with one to six halo, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SC^R12, - SO2CH(Rl2)(Rl l), -ORl2, -OR9, -N(R 12)2, aryl, heteroaryl or heterocyclyl wherein said aryl, heteroaryl and heterocyclyl groups are optionally substituted with one or two substitutents independently selected from Ci-6 alkyl, C3-6 cycloalkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto; or Rl and R2 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring or heterocyclyl ring wherein said cycloalkyl and heterocycl rings are optionally substituted with one or two substituents independently selected from Cl -6 alkyl, hydroxyalkyl, haloalkyl, aryl, heteroaryl, heterocyclyl or halo;
R3 is hydrogen, Cl -6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo;
R4 is hydrogen, Ci-6 alkyl or C2-6 alkenyl wherein said alkyl and alkenyl groups are optionally substituted with C3-6 cycloalkyl, aryl or halo; or R3 and R4 can be taken together with the carbon atom to which they are attached to form a C3-8 cycloalkyl ring, C5.8 cycloalkenyl ring, or five to seven membered heterocyclyl ring wherein said cycloalkyl, cycloalkenyl and heterocyclyl rings are optionally substituted with Ci-6 alkyl, halo, hydroxyalkyl, hydroxy, alkoxy or keto;
R5 is hydrogen or Q -6 haloalkyl;
R6 is aryl, heteroaryl, Cl -6 haloalkyl, arylalkyl or heteroarylalkyl, wherein said aryl, heteroaryl, arylalkyl and heteroarylalkyl groups are optionally substituted with halo, Ci-6 alkyl, Ci-6 haloalkyl, C3-6 cycloalkyl, -SR9, -SRl2, -SOR9, -SOR12, -SO2R9, -SO2RI2, -Sθ2CH(Rl2)(Rll), -ORl2, -
N(RlO)(Rl 1), or cyano;
D is C 1-3 alkyl, C2-3 alkenyl, C2-3 alkenyl, aryl, heteroaryl, C3.8 cycloalkyl or heterocyclyl wherein said aryl, heteroaryl, cycloalkyl and heterocyclyl groups, which may be monocyclic or bicyclic, are optionally substituted on either the carbon or the heteroatom with one to five substituents selected from C 1-6 alkyl, halo or keto;
R7 is hydrogen, Ci -6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 1-6 alkyloxy, halo, nitro, cyano, aryl, heteroaryl, C3.8 cycloalkyl, heterocyclyl, -C(O)ORlO, -C(O)OSi[CH(CH3)2]3, -ORlO, -C(O)RlO, . RlOC(0)R9, -C(O)R9, -C(O)N(Rl2)(Rl2), _C(O) N(RlO)(Rl I), -C(RlO)(Rl I)OH, -SRl2, -SR9, - RlOSR9, -R9, -C(R9)3, -C(RlO)(Rl 1)N(R9)2, -NR10C(O)NR10S(O)2R9, -SO2R12, -SO(Rl2), -SO2R9, -Sθ2N(RC)(Rd), -Sθ2CH(RlO)(Rl l), -Sθ2N(Rl0)C(O)(Rl2), -Sθ2(Rl°)C(O)N(Rl2)2, -OSO2RIO, - N(RlO)(Rl I), -N(Rl0)C(O)N(Rl0)(R9), -N(RlO)C(O)RlO, -N(RlO)C(O)ORlO, .N(RlO)SO2(RlO), - C(RlO)(Rl I)NRIOC(RlO)(Rl 1)R9, -C(RlO)(Rl l)N(RlO)R9, -C(R 1O)(R H)N(R 1O)(R 11), - C(RlO)(Rl I)SC(RlO)(Rl 1)(R9), RIOS-, -C(Ra)(Rb)NRaC(Ra)(Rb)(R9), -C(Ra)(Rb)N(Ra)(Rb), . C(Ra)(Rb)C(Ra)(Rb)N(Ra)(Rb), -C(O)C(Ra)(Rb)N(Ra)(Rb), -C(Ra)(Rb)N(Ra)C(O) R9, - C(O)C(Ra)(Rb)S(Ra) or C(Ra)(Rb)C(O)N(Ra)(Rb); wherein said alkyl, alkenyl, alkynyl, alkoxy, aryl, heteroaryl, cycloalkyl and heterocyclyl groups are optionally substituted on either the carbon or the heteroatom with one to five substituents independently selected from Ci -6 alkyl, halo, keto, cyano, haloalkyl, hydroxyalkyl, -OR9, -NO2, -NH2, -NHS(O)2R8, -R9SO2R12, -802R12, -S0(Rl2), - Sθ2N(RC)(Rd), -SO2N(Rl0)C(O)(Rl2), -C(RlO)(Rl I)N(RlO)(RlI), -C(RlO)(RlI)OH, -COOH, - C(Ra)(Rb)C(O)N(Ra)(Rb), -N(RlO)C(RlO)(Rl 1)(R9), -NH(CH2)2θH, -NHC(O)ORlO, -Si(CH3)3, heterocycyl, aryl or heteroaryl; RB is hydrogen or Ci _6 alkyl; or R4 and R8 or can be taken together with any of the atoms to which they may be attached or are between them to form a 4-10 membered heterocyclyl ring system wherein said ring system, which may be monocyclic or bicyclic, is optionally substituted with Ci-6 alkyl, halo, hydroxyalkyl, hydroxy, keto, - ORlO, -SRlO or -N(RlO)2;
R9 is hydrogen, aryl, aryl(Ci_4) alkyl, heteroaryl, heteroaryl(Ci -4)alkyl, C3-8cycloalkyl, C3- 8cycloalkyl(Ci -4)alkyl, and heterocyclyl(Ci-4)alkyl wherein said groups can be optionally substituted with halo or alkoxy;
RlO is hydrogen or Ci _6 alkyl;
Rl 1 is hydrogen or C\.β alkyl;
Rl2 is hydrogen or Ci-6 alkyl which is optionally substituted with halo, alkoxy, cyano, -NRlO or -SRlO;
Ra is hydrogen, C\.β alkyl, (Q -6 alkyl)aryl, (Ci -6 alkyl)hydroxyl, -O(Ci-6 alkyl), hydroxyl, halo, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl, wherein said alkyl, aryl, heteroaryl, C3-8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with C] -6 alkyl or halo;
Rb is hydrogen, Cl -6 alkyl, (C 1-6 alkyl)aryl, (Ci -β alkyl)hydroxyl, alkoxyl, hydroxyl, halo, aryl, heteroaryl, C3-8 cycloalkyl or heterocyclyl,wherein said alkyl, aryl, heteroaryl, C3-8 cycloalkyl and heterocyclyl are optionally substituted on either the carbon or the heteroatom with Ci_6 alkyl or halo; or Ra and Rb can be taken together with the carbon atom to which they are attached or are between them to form a C3-8 cycloalkyl ring or C3-8 heterocyclyl ring wherein said 3-8 membered ring system may be optionally substituted with Ci -6 alkyl and halo; n is an integer from zero to three; and another agent selected from the group consisting of nifurtimox, benznidazole, allopurinol, terbinafine, lovastatin, ketoconazole, itraconazole, posaconazole, miltefosine, ilmofosine, pamidronate, alendronate, risedronate, chloroquine, proguanil, mefloquine, quinine, pyrimethamine-sulphadoxine, doxocycline, berberine, halofantrine, primaquine, atovaquone, pyrimethamine-dapsone, artemisinin, quinhaosu. meglumine antimonite, sodium stibogluconate, amphotericin B, praziquantel, oxamniquine, pentamidine, melarsoprol, suramin and eflornithine.
PCT/CA2006/001216 2005-07-26 2006-07-24 Papain family cysteine protease inhibitors for the treatment of parasitic diseases WO2007012180A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/989,437 US20090264479A1 (en) 2005-07-26 2006-07-24 Papain Family Cysteine Protease Inhibitors for the Treatment of Parasitic Diseases
EP06761177A EP1909784A4 (en) 2005-07-26 2006-07-24 Papain family cysteine protease inhibitors for the treatment of parasitic diseases
MX2008001207A MX2008001207A (en) 2005-07-26 2006-07-24 Papain family cysteine protease inhibitors for the treatment of parasitic diseases.
BRPI0614169A BRPI0614169A2 (en) 2005-07-26 2006-07-24 method for treating a parasitic disease and pharmaceutical composition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US70245505P 2005-07-26 2005-07-26
US60/702,455 2005-07-26

Publications (1)

Publication Number Publication Date
WO2007012180A1 true WO2007012180A1 (en) 2007-02-01

Family

ID=37682953

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2006/001216 WO2007012180A1 (en) 2005-07-26 2006-07-24 Papain family cysteine protease inhibitors for the treatment of parasitic diseases

Country Status (5)

Country Link
US (1) US20090264479A1 (en)
EP (1) EP1909784A4 (en)
BR (1) BRPI0614169A2 (en)
MX (1) MX2008001207A (en)
WO (1) WO2007012180A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009067797A1 (en) * 2007-11-29 2009-06-04 Merck Frosst Canada Ltd. Cysteine protease inhibitors for the treatment of parasitic diseases
WO2010107791A2 (en) 2009-03-17 2010-09-23 Concert Pharmaceuticals, Inc. Pyrazinoisoquinoline compounds
EP2445872A1 (en) * 2009-06-22 2012-05-02 Merck Canada Inc. Cathepsin cysteine protease inhibitors for the treatment of various diseases
EP2633855A1 (en) 2012-03-01 2013-09-04 Veterinärmedizinische Universität Wien Protease inhibitors for treating Trichomonas gallinae infections
WO2017089389A1 (en) * 2015-11-26 2017-06-01 F. Hoffmann-La Roche Ag Trypanosomes inhibitors
CN106866502A (en) * 2015-12-10 2017-06-20 广东东阳光药业有限公司 Cathepsin K inhibitor and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8957865B2 (en) * 2009-01-05 2015-02-17 Apple Inc. Device, method, and graphical user interface for manipulating a user interface object

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999053039A1 (en) * 1998-04-09 1999-10-21 Smithkline Beecham Corporation Treatment of parasitic diseases by inhibition of cysteine proteases of the papain superfamily
WO2000009653A2 (en) * 1998-08-14 2000-02-24 Smithkline Beecham Corporation Method of inhibiting cathepsin k
EP1200457A2 (en) * 1999-07-31 2002-05-02 Naeja Pharmaceutical Inc. Cysteine protease inhibitors
WO2003075836A2 (en) * 2002-03-05 2003-09-18 Merck Frosst Canada & Co. Cathepsin cysteine protease inhibitors
WO2004110988A1 (en) * 2003-06-18 2004-12-23 Prozymex A/S Protease inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL112759A0 (en) * 1994-02-25 1995-05-26 Khepri Pharmaceuticals Inc Novel cysteine protease inhibitors
US6586466B2 (en) * 1995-10-30 2003-07-01 Smithkline Beecham Corporation Carbohydrazide-protease inhibitors
KR100630986B1 (en) * 1998-12-23 2006-10-09 스미스클라인 비참 코포레이션 Protease Inhibitors
CA2535359A1 (en) * 2003-08-21 2005-03-03 Merck Frosst Canada Ltd. Cathepsin cysteine protease inhibitors
JP2007503401A (en) * 2003-08-27 2007-02-22 メルク フロスト カナダ リミテツド Cathepsin inhibitor
AU2004296905A1 (en) * 2003-12-12 2005-06-23 Merck Frosst Canada Ltd Cathepsin cysteine protease inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999053039A1 (en) * 1998-04-09 1999-10-21 Smithkline Beecham Corporation Treatment of parasitic diseases by inhibition of cysteine proteases of the papain superfamily
WO2000009653A2 (en) * 1998-08-14 2000-02-24 Smithkline Beecham Corporation Method of inhibiting cathepsin k
EP1200457A2 (en) * 1999-07-31 2002-05-02 Naeja Pharmaceutical Inc. Cysteine protease inhibitors
WO2003075836A2 (en) * 2002-03-05 2003-09-18 Merck Frosst Canada & Co. Cathepsin cysteine protease inhibitors
WO2004110988A1 (en) * 2003-06-18 2004-12-23 Prozymex A/S Protease inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1909784A4 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2008329515B2 (en) * 2007-11-29 2013-09-05 Merck Canada Inc. Cysteine protease inhibitors for the treatment of parasitic diseases
US8642799B2 (en) 2007-11-29 2014-02-04 Merck Canada Inc. Cysteine protease inhibitors for the treatment of parasitic diseases
EP2225196A4 (en) * 2007-11-29 2012-02-29 Merck Canada Inc Cysteine protease inhibitors for the treatment of parasitic diseases
JP2011504883A (en) * 2007-11-29 2011-02-17 メルク フロスト カナダ リミテツド Cysteine protease inhibitors for the treatment of parasitic diseases
WO2009067797A1 (en) * 2007-11-29 2009-06-04 Merck Frosst Canada Ltd. Cysteine protease inhibitors for the treatment of parasitic diseases
EP2225196A1 (en) * 2007-11-29 2010-09-08 Merck Frosst Canada Ltd. Cysteine protease inhibitors for the treatment of parasitic diseases
WO2010107791A2 (en) 2009-03-17 2010-09-23 Concert Pharmaceuticals, Inc. Pyrazinoisoquinoline compounds
EP2445872A1 (en) * 2009-06-22 2012-05-02 Merck Canada Inc. Cathepsin cysteine protease inhibitors for the treatment of various diseases
EP2445872A4 (en) * 2009-06-22 2012-12-05 Merck Canada Inc Cathepsin cysteine protease inhibitors for the treatment of various diseases
WO2013127981A1 (en) 2012-03-01 2013-09-06 Veterinärmedizinische Universität Wien Protease inhibitors for treating trichomonas gallinae infections
EP2633855A1 (en) 2012-03-01 2013-09-04 Veterinärmedizinische Universität Wien Protease inhibitors for treating Trichomonas gallinae infections
WO2017089389A1 (en) * 2015-11-26 2017-06-01 F. Hoffmann-La Roche Ag Trypanosomes inhibitors
CN108368056A (en) * 2015-11-26 2018-08-03 豪夫迈·罗氏有限公司 Trypanosome inhibitor
US10377719B2 (en) 2015-11-26 2019-08-13 Hoffmann-La Roche Inc. Trypanosomes inhibitors
CN106866502A (en) * 2015-12-10 2017-06-20 广东东阳光药业有限公司 Cathepsin K inhibitor and application thereof
CN106866502B (en) * 2015-12-10 2020-10-09 广东东阳光药业有限公司 Cathepsin K inhibitors and uses thereof

Also Published As

Publication number Publication date
BRPI0614169A2 (en) 2016-11-22
MX2008001207A (en) 2008-03-24
EP1909784A1 (en) 2008-04-16
US20090264479A1 (en) 2009-10-22
EP1909784A4 (en) 2010-04-21

Similar Documents

Publication Publication Date Title
US10179781B2 (en) Sodium channel modulators for the treatment of pain
ES2390227T3 (en) New phenylaminopyrimidine derivatives as inhibitors of BCR-ABL kinase
US9284295B2 (en) Tetrahydrobenzothiophene compound
WO2017219931A1 (en) Dihydro pteridinone derivative, preparation method therefor, and application thereof
WO2007012180A1 (en) Papain family cysteine protease inhibitors for the treatment of parasitic diseases
US20070219239A1 (en) Nitrogen-containing heterocycle derivatives, pharmaceutical compositions, and methods of use thereof as antiviral agents
CA2870666A1 (en) Compositions and methods of modulating 15-pgdh activity
JP5898230B2 (en) Novel substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US11147819B2 (en) EZH2 inhibitors for treating cancer
US8642799B2 (en) Cysteine protease inhibitors for the treatment of parasitic diseases
BR112013006594B1 (en) OREXIN RECEPTOR ANTAGONIST CYCLOPROPAN COMPOUND AND PHARMACEUTICAL COMPOSITION
JP2021528470A (en) TAIRE Family Kinase Inhibitors and Their Use
CA3012846A1 (en) Max binders as myc modulators and uses thereof
KR20170102504A (en) New calcium modifiers
CA2325201A1 (en) Methods of administering apo b-secretion/mtp inhibitors
CA3008485C (en) Alkynyl dihydroquinoline sulfonamide compounds
JP5673676B2 (en) Imidazo [1,2-a] pyridine derivatives
US20120101053A1 (en) Cathepsin cysteine protease inhibitors for the treatment of various diseases
US10532987B2 (en) Compounds and methods for inducing browning of white adipose tissue
US20240092802A1 (en) Mu-opioid receptor agonists and uses therefor
JP2014532656A (en) Compounds for inflammation and immune related applications
KR100721087B1 (en) Novel use of phenylheteroalkylamine derivatives
US20230312481A1 (en) Substituted (phthalazin-1-ylmethyl)ureas, substituted n-(phthalazin-1-ylmethyl)amides, and analogues thereof
WO2011150682A1 (en) 4-hydroxyquinoline-3-amide derivatives and preparation methods and uses thereof
CA3148310A1 (en) Kdm inhibitors and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 11989437

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/001207

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006761177

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2006761177

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0614169

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080125