US20150191426A1 - Methods and compositions for oral delivery of fts - Google Patents

Methods and compositions for oral delivery of fts Download PDF

Info

Publication number
US20150191426A1
US20150191426A1 US14/662,420 US201514662420A US2015191426A1 US 20150191426 A1 US20150191426 A1 US 20150191426A1 US 201514662420 A US201514662420 A US 201514662420A US 2015191426 A1 US2015191426 A1 US 2015191426A1
Authority
US
United States
Prior art keywords
dosage form
oral dosage
fts
disease
disorder
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/662,420
Inventor
Victor J. Bauer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ramot at Tel Aviv University Ltd
Original Assignee
Kadmon Corp LLC
Concordia Pharmaceuticals LLC
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 Kadmon Corp LLC, Concordia Pharmaceuticals LLC filed Critical Kadmon Corp LLC
Priority to US14/662,420 priority Critical patent/US20150191426A1/en
Publication of US20150191426A1 publication Critical patent/US20150191426A1/en
Assigned to CONCORDIA PHARMACEUTICALS, INC. reassignment CONCORDIA PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAUER, VICTOR J.
Assigned to KADMON CORPORATION, LLC reassignment KADMON CORPORATION, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONCORDIA PHARMACEUTICALS, INC.
Assigned to PERCEPTIVE CREDIT OPPORTUNITIES FUND, LP reassignment PERCEPTIVE CREDIT OPPORTUNITIES FUND, LP SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KADMON CORPORATION, LLC
Assigned to MACQUARIE US TRADING LLC reassignment MACQUARIE US TRADING LLC SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KADMON CORPORATION, LLC
Priority to US15/170,416 priority patent/US20160271089A1/en
Assigned to KADMON CORPORATION, LLC reassignment KADMON CORPORATION, LLC NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: CONCORDIA PHARMACEUTICALS, INC.
Assigned to RAMOT AT TEL-AVIV UNIVERSITY LTD. reassignment RAMOT AT TEL-AVIV UNIVERSITY LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KADMON CORPORATION, LLC
Assigned to KADMON CORPORATION, LLC reassignment KADMON CORPORATION, LLC CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME PREVIOUSLY RECORDED ON REEL 039166 FRAME 0644. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: CONCORDIA PHARMACEUTICALS, LLC
Assigned to CONCORDIA PHARMACEUTICALS, LLC reassignment CONCORDIA PHARMACEUTICALS, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CONCORDIA PHARMACEUTICALS, INC.
Assigned to KADMON CORPORATION, LLC reassignment KADMON CORPORATION, LLC TERMINATION OF PATENT AND TRADEMARK SECURITY AGREEMENT Assignors: PERCEPTIVE CREDIT OPPORTUNITIES FUND, LP
Assigned to KADMON CORPORATION, LLC reassignment KADMON CORPORATION, LLC RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: MACQUARIE US TRADING LLC
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4816Wall or shell material
    • A61K9/4825Proteins, e.g. gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C321/00Thiols, sulfides, hydropolysulfides or polysulfides
    • C07C321/24Thiols, sulfides, hydropolysulfides, or polysulfides having thio groups bound to carbon atoms of six-membered aromatic rings
    • C07C321/28Sulfides, hydropolysulfides, or polysulfides having thio groups bound to carbon atoms of six-membered aromatic rings

Definitions

  • U.S. Pat. No. 5,705,528 teaches farnesylthiosalicyclic acid (FTS) and analogs thereof and their utility as anti-cancer agents.
  • U.S. Pat. No. 6,462,086 teaches additional therapeutic utilities of these compounds, namely in connection with treatment of non-malignant diseases, pathological states or other disorders that feature or otherwise include Ras-induced proliferation of cells. The patent also teaches that these compounds are inactive when administered orally, but that this shortcoming can be overcome by making a salt of the compound (i.e., salification), formulating the salt in cyclodextrin, and then preparing a buccal tablet (which will dissolve in the mouth when held against the mucous membrane).
  • a salt of the compound i.e., salification
  • FTS and its analogs do not require salification or formulation in cyclodextrin to be active upon oral administration.
  • a first aspect of the present invention is directed to an oral dosage form comprising an amount of a Ras antagonist effective to treat a responsive disease or disorder involving abnormal cell proliferation, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula (I)
  • R 1 represents farnesyl, geranyl or geranyl-geranyl
  • R 2 is COOR 7 , or CONR 7 R 8 , wherein R 7 and R 8 are each independently hydrogen, alkyl or alkenyl;
  • R 3 , R 4 , R 5 and R 6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and X represents S.
  • the oral dosage forms are tablets or capsules.
  • the Ras antagonist is not in the form of a salt.
  • a second aspect of the invention is directed to method of treating a treating a disease or disorder involving abnormal cell proliferation, comprising administering to a human subject in need thereof an oral dosage form comprising an amount of a Ras antagonist effective to treat the disease or disorder, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula
  • R 4 represents farnesyl, geranyl or geranyl-geranyl
  • R 2 is COOR 7 , or CONR 7 R 8 , wherein R 7 and R 8 are each independently hydrogen, alkyl or alkenyl
  • R 3 , R 4 , R 5 and R 6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto
  • X represents S.
  • FIG. 1 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (e.g., gavage) administration of FTS in a corn oil carrier.
  • FIG. 2 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (gavage) administration of FTS in aqueous carboxymethylcellulose (CMC) carrier.
  • CMC carboxymethylcellulose
  • FIG. 3 is a bar graph showing average tumor weight in mice (in grams) following oral (gavage) administration of different amounts of FTS in carboxymethylcellulose, as compared to a control.
  • FIG. 4 is a bar graph showing FTS plasma concentration (ng/ml) over 1-24 hours after oral administration of 200 mg/kg of FTS to rats.
  • Ras antagonists useful in the present invention are represented by formula I:
  • R 4 represents farnesyl, geranyl or geranyl-geranyl
  • R 2 is COOR 7 , or CONR 7 R 8 , wherein R 7 and R 8 are each independently hydrogen, alkyl or alkenyl
  • R 3 , R 4 , R 5 and R 6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto
  • X represents S.
  • FTS farnesyl-thiosalicylic acid
  • FTS analogs embraced by formula I include 5-fluoro-FTS, 5-chloro-FTS, 4-chloro-FTS and S-farnesyl-thiosalicylic acid methyl ester (FTSME). Structures of these compounds are set forth below.
  • Ras antagonists of formula I may be useful. In preferred embodiments, however, the Ras antagonist is not in the form of a salt (i.e., non-salified).
  • Oral dosage forms useful in the present invention include tablets (e.g., including film-coated, sugar-coated, controlled or sustained release), and capsules, e.g., hard gelatin capsules (including controlled or sustained release), and soft gelatin capsules.
  • Oral dosage forms may be prepared by mixing the active pharmaceutical ingredient, which in this case are the Ras antagonists of formula I, with one or more appropriate carriers (excipients), and then formulating the composition into the desired dosage form e.g., compressing the composition into a tablet or filling into a capsule.
  • the proviso is that the oral dosage forms do not contain a cyclodextrin.
  • Typical excipients useful as bulking agents or diluents, binders, buffers or pH adjusting agents, disintegrants (including crosslinked and super disintegrants such as croscarmellose), glidants, and/or lubricants include lactose, starch, mannitol, microcrystalline cellulose, ethyl cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, dibasic calcium phosphate, acacia, gelatin, stearic acid, magnesium stearate, corn oil, vegetable oils, and polyethylene glycols, and others known to the pharmaceutical practitioner. Coating agents such as sugar, shellac, and synthetic polymers may be employed. Dyes and other colorants may be added as well. See, Remington's Pharmaceutical Sciences , The Science and Practice of Pharmacy, 20th Edition, (2000).
  • the oral dosage forms of the present invention are useful in treating diseases and disorders responsive to the Ras antagonists of formula I, e.g., diseases and disorders characterized or mediated, at least in part, by abnormal (e.g., uncontrolled) cell proliferation, such as Ras-induced cell proliferation.
  • the proliferating cells may be malignant or non-malignant in nature.
  • responsive does not require that a therapeutic response would be achieved in each and every patient, but rather what a skilled practitioner would reasonably expect based on existing data from patient populations.
  • cancers including breast cancer, colon cancer, glioblastoma, lung cancer (small cell and non-small cell lung cancer), melanoma, Merkel cell carcinoma, neuroblastoma, neurofibromatosis, ovarian cancer, pancreatic cancer and prostate cancer.
  • non-malignant diseases and disorders characterized by or involving abnormal (e.g., uncontrolled) cell proliferation include liver cirrhosis, restenosis after angioplasty (post-angioplasty restenosis), atherosclerosis, and graft rejection (e.g., graft-versus-host disease (GVHD)).
  • graft rejection e.g., graft-versus-host disease (GVHD)
  • Other examples include autoimmune diseases such as type-1 diabetes mellitus, systemic lupus erythematosis, rheumatoid arthritis, psoriasis, multiple sclerosis, Guillain-Barre syndrome and primary antiphospholipid syndrome.
  • the term “effective amount” refers to a dosage of the Ras antagonist that inhibits disease onset or progression or ameliorates symptom(s) of the disease.
  • the typical daily dose is 5 mg to 1000 mg, e.g., 100 mg to 1,000 mg per day, which may be administered once daily or as divided doses two or three times daily.
  • the oral dosage forms of the present invention e.g., tablet, hard gelatin capsule, or soft gelatin capsule, typically contains from 5 mg to 500 mg of the Ras antagonist.
  • the oral dosage forms contain from 10 mg to 250 mg of active pharmaceutical ingredient.
  • the oral dosage forms are typically administered to an individual diagnosed with the disease or disorder, although prophylactic administration may be useful to inhibit onset of a disease or disorder e.g., administration prior to an angioplasty procedure to inhibit onset of restenosis.
  • oral dosage forms may be suitably packaged for distribution and sale, including printed instructions for administering them as described herein.
  • FTS (40 mg) was dispersed in corn oil (10 ml). Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO 2 /O 2 anesthesia at 1, 2, 4, 8, 12 and 24 hours (3 animals at each time point) after dosing. Aliquots (100 ⁇ L) of plasma were mixed with water, pH 11 buffer solution and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was resuspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by liquid chromatography/mass spectrometry (LC/MS).
  • LC/MS liquid chromatography/mass spectrometry
  • mean plasma FTS concentrations were 2159 ng/ml at 1 hour and 1326 ng/ml after 2 hours. There was a rapid decrease to less than 3 ng/ml after 24 hours. These values indicated 55% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.
  • FTS (40 mg) was dispersed in 0.5% aqueous carboxymethylcellulose (10 ml). Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO 2 /0 2 anesthesia at 1, 2, 4, 8, 12, and 24 hours (3 animals at each time point) after dosing. Aliquots (100 ⁇ L) of plasma were mixed with water, pH 11 buffer solution, and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was re-suspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by LC/MS.
  • mean plasma FTS concentrations were 3381 ng/ml at 1 hour, 1593 ng/ml after 2 hours, and decreased rapidly to less than 3 ng/ml after 24 hours. These values indicated 69% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.
  • FTS was dispersed in 0.5% aqueous carboxymethylcellulose at a concentration of 5 mg/ml.
  • mice were segregated into three groups and were given vehicle or FTS suspension by gavage.
  • Group 1 Controls, 6 mice received 0.2 ml vehicle daily.
  • Group 2 (20 mg/kg FTS, 7 mice) received 0.1 ml FTS suspension daily.
  • Group 3 (40 mg/kg FTS, 6 mice) received 0.2 ml FTS suspension daily.
  • mice were treated for ten days, and were sacrificed. Tumors were excised and weighed.
  • average tumor weight for Group 1 was 1.02 g; average tumor weight for Group 2 (20 mg/kg FTS) was 0.68 g (32% lower than control); and average tumor weight for Group 3 (40 mg/kg FTS) was 0.47 g (53% lower than control).
  • FTS active pharmaceutical ingredient 2000 g
  • microcrystalline cellulose 2000 g
  • croscarmellose sodium 200 g
  • magnesium stearate 100 g
  • FTS active pharmaceutical ingredient 1000 g
  • lactose 1000 g
  • microcrystalline cellulose 1000 g
  • amorphous colloidal silicon dioxide 15 g
  • FTS active pharmaceutical ingredient 500 g
  • a mixture of corn oil (3000 g), lecithin (30 g), and Tween-80 (150 g) is uniformly dispersed in a mixture of corn oil (3000 g), lecithin (30 g), and Tween-80 (150 g) and filled into soft gelatin capsules. Assuming a 5% loss on material transfers and soft gelatin encapsulating machine start-up, adjustment, and shut down, approximately 9,500 FTS 50 mg soft gelatin capsules are yielded.
  • FTS active pharmaceutical ingredient 2841 g
  • microcrystalline cellulose 1421 g
  • starch 710 g
  • magnesium stearate 29 g
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #1 hard-shell gelatin capsules, with a fill weight of 0.225 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 4,750 FTS 200 mg capsules are yielded.
  • EXAMPLE 9 FTS Capsules (100 mg)
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #2 hard-shell gelatin capsules, with a fill weight of 0.112.5 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 9,500 FTS 100 mg capsules are yielded.
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #3 hard-shell gelatin capsules, with a fill weight of 0.056 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 18,000 FTS 50 mg capsules are yielded.
  • FTS was dissolved in a mixture of corn oil (95%) and ethanol (5%). Aliquots of the resulting solution were administered to Crl:CD(SD)(IGS)BR rats (twenty-four) at a dose of 200 mg/kg. Blood samples were collected in lithium-heparin-containing tubes via the retro-orbital plexus while under CO 2 /O 2 anesthesia at 1, 2, 4, 8, 12 and 24 hours after dosing. Concentration of FTS at each time point was determined by LC/MS. As shown in FIG.
  • mean plasma FTS concentrations were 4454 ng/ml at 1 hour, 4550 ng/ml at 2 hours, 3047 ng/ml at 4 hours, 2216 ng/ml at 8 hours, 685 ng/ml at 12 hours, and 140 ng/ml at 24 hours. These values indicate a high level of oral bioavailability.

Abstract

Disclosed are oral dosage forms containing a Ras antagonist including FTS and structural analogs thereof, and at least one pharmaceutically acceptable excipient other than a cyclodextrin, and methods of orally administering same to treat diseases and disorders responsive to the Ras antagonists.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation of U.S. patent application Ser. No. 13/326,598, filed on Dec. 15, 2011, which application is a divisional of U.S. patent application Ser. No. 11/659,582, filed on Feb. 7, 2007, which application is a national phase entry under 35 U.S.C. §371 of International Application No. PCT/US2005/029389, filed Aug. 18, 2005, which claims priority from U.S. Provisional Application No. 60/602,361, filed Aug. 18, 2004, all of which are incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • U.S. Pat. No. 5,705,528 teaches farnesylthiosalicyclic acid (FTS) and analogs thereof and their utility as anti-cancer agents. U.S. Pat. No. 6,462,086 teaches additional therapeutic utilities of these compounds, namely in connection with treatment of non-malignant diseases, pathological states or other disorders that feature or otherwise include Ras-induced proliferation of cells. The patent also teaches that these compounds are inactive when administered orally, but that this shortcoming can be overcome by making a salt of the compound (i.e., salification), formulating the salt in cyclodextrin, and then preparing a buccal tablet (which will dissolve in the mouth when held against the mucous membrane).
  • SUMMARY OF THE INVENTION
  • Applicants have discovered that FTS and its analogs do not require salification or formulation in cyclodextrin to be active upon oral administration.
  • A first aspect of the present invention is directed to an oral dosage form comprising an amount of a Ras antagonist effective to treat a responsive disease or disorder involving abnormal cell proliferation, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula (I)
  • Figure US20150191426A1-20150709-C00001
  • wherein:
    R1 represents farnesyl, geranyl or geranyl-geranyl;
  • R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;
  • R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and
    X represents S. The oral dosage forms are tablets or capsules.
    In preferred embodiments, the Ras antagonist is not in the form of a salt.
  • A second aspect of the invention is directed to method of treating a treating a disease or disorder involving abnormal cell proliferation, comprising administering to a human subject in need thereof an oral dosage form comprising an amount of a Ras antagonist effective to treat the disease or disorder, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula
  • Figure US20150191426A1-20150709-C00002
  • wherein:
    R4 represents farnesyl, geranyl or geranyl-geranyl;
    R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;
    R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and
    X represents S.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (e.g., gavage) administration of FTS in a corn oil carrier.
  • FIG. 2 is a graph showing concentrations (ng/mL) of FTS in plasma of mice over time following oral (gavage) administration of FTS in aqueous carboxymethylcellulose (CMC) carrier.
  • FIG. 3 is a bar graph showing average tumor weight in mice (in grams) following oral (gavage) administration of different amounts of FTS in carboxymethylcellulose, as compared to a control.
  • FIG. 4 is a bar graph showing FTS plasma concentration (ng/ml) over 1-24 hours after oral administration of 200 mg/kg of FTS to rats.
  • DETAILED DESCRIPTION
  • Ras antagonists useful in the present invention are represented by formula I:
  • Figure US20150191426A1-20150709-C00003
  • wherein
    R4 represents farnesyl, geranyl or geranyl-geranyl;
    R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;
    R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and
    X represents S.
  • These compounds represent farnesyl-thiosalicylic acid (FTS) (i.e., S-trans, trans-FTS) and its analogs. The structure of FTS is as follows:
  • FTS:
  • Figure US20150191426A1-20150709-C00004
  • FTS analogs embraced by formula I include 5-fluoro-FTS, 5-chloro-FTS, 4-chloro-FTS and S-farnesyl-thiosalicylic acid methyl ester (FTSME). Structures of these compounds are set forth below.
  • Figure US20150191426A1-20150709-C00005
  • Pharmaceutically acceptable salts of the Ras antagonists of formula I may be useful. In preferred embodiments, however, the Ras antagonist is not in the form of a salt (i.e., non-salified).
  • Methods of preparing the compounds of formula I are disclosed in U.S. Pat. Nos. 5,705,528 and 6,462,086. Those disclosures (e.g., Example 7 in the '086 patent) are incorporated herein by reference.
  • Oral dosage forms useful in the present invention include tablets (e.g., including film-coated, sugar-coated, controlled or sustained release), and capsules, e.g., hard gelatin capsules (including controlled or sustained release), and soft gelatin capsules.
  • Oral dosage forms may be prepared by mixing the active pharmaceutical ingredient, which in this case are the Ras antagonists of formula I, with one or more appropriate carriers (excipients), and then formulating the composition into the desired dosage form e.g., compressing the composition into a tablet or filling into a capsule. The proviso is that the oral dosage forms do not contain a cyclodextrin. Typical excipients useful as bulking agents or diluents, binders, buffers or pH adjusting agents, disintegrants (including crosslinked and super disintegrants such as croscarmellose), glidants, and/or lubricants include lactose, starch, mannitol, microcrystalline cellulose, ethyl cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, dibasic calcium phosphate, acacia, gelatin, stearic acid, magnesium stearate, corn oil, vegetable oils, and polyethylene glycols, and others known to the pharmaceutical practitioner. Coating agents such as sugar, shellac, and synthetic polymers may be employed. Dyes and other colorants may be added as well. See, Remington's Pharmaceutical Sciences, The Science and Practice of Pharmacy, 20th Edition, (2000).
  • The oral dosage forms of the present invention are useful in treating diseases and disorders responsive to the Ras antagonists of formula I, e.g., diseases and disorders characterized or mediated, at least in part, by abnormal (e.g., uncontrolled) cell proliferation, such as Ras-induced cell proliferation. The proliferating cells may be malignant or non-malignant in nature. Of course, the term “responsive” does not require that a therapeutic response would be achieved in each and every patient, but rather what a skilled practitioner would reasonably expect based on existing data from patient populations. These diseases and disorders include cancers including breast cancer, colon cancer, glioblastoma, lung cancer (small cell and non-small cell lung cancer), melanoma, Merkel cell carcinoma, neuroblastoma, neurofibromatosis, ovarian cancer, pancreatic cancer and prostate cancer.
  • Examples of non-malignant diseases and disorders characterized by or involving abnormal (e.g., uncontrolled) cell proliferation include liver cirrhosis, restenosis after angioplasty (post-angioplasty restenosis), atherosclerosis, and graft rejection (e.g., graft-versus-host disease (GVHD)). Other examples include autoimmune diseases such as type-1 diabetes mellitus, systemic lupus erythematosis, rheumatoid arthritis, psoriasis, multiple sclerosis, Guillain-Barre syndrome and primary antiphospholipid syndrome.
  • As used herein, the term “effective amount” refers to a dosage of the Ras antagonist that inhibits disease onset or progression or ameliorates symptom(s) of the disease. The typical daily dose is 5 mg to 1000 mg, e.g., 100 mg to 1,000 mg per day, which may be administered once daily or as divided doses two or three times daily. Thus, the oral dosage forms of the present invention e.g., tablet, hard gelatin capsule, or soft gelatin capsule, typically contains from 5 mg to 500 mg of the Ras antagonist. In preferred embodiments, the oral dosage forms contain from 10 mg to 250 mg of active pharmaceutical ingredient. The oral dosage forms are typically administered to an individual diagnosed with the disease or disorder, although prophylactic administration may be useful to inhibit onset of a disease or disorder e.g., administration prior to an angioplasty procedure to inhibit onset of restenosis.
  • The oral dosage forms may be suitably packaged for distribution and sale, including printed instructions for administering them as described herein.
  • The present invention will now be described by way of the following non-limiting examples. They are presented solely for purposes of illustration, and are not intended to limit the invention in any way.
  • Example 1 Bioavailability of Orally Administered FTS
  • a. FTS formulated in corn oil.
  • FTS (40 mg) was dispersed in corn oil (10 ml). Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO2/O2 anesthesia at 1, 2, 4, 8, 12 and 24 hours (3 animals at each time point) after dosing. Aliquots (100 μL) of plasma were mixed with water, pH 11 buffer solution and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was resuspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by liquid chromatography/mass spectrometry (LC/MS).
  • As shown in FIG. 1, mean plasma FTS concentrations were 2159 ng/ml at 1 hour and 1326 ng/ml after 2 hours. There was a rapid decrease to less than 3 ng/ml after 24 hours. These values indicated 55% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.
  • b. FTS formulated in 0.5% aqueous carboxymethylcellulose.
  • FTS (40 mg) was dispersed in 0.5% aqueous carboxymethylcellulose (10 ml). Aliquots of the resulting suspension were administered by gavage to CD-1 mice (eighteen) at a dose of 40 mg/ml (at a volume of 10 ml/kg of FTS suspension calculated for actual animal weight). Blood samples were collected in lithium heparin tubes via the retro-orbital plexus while under CO2/02 anesthesia at 1, 2, 4, 8, 12, and 24 hours (3 animals at each time point) after dosing. Aliquots (100 μL) of plasma were mixed with water, pH 11 buffer solution, and acetonitrile. After centrifugation, the supernatant was evaporated to dryness. The residue was re-suspended in water/acetonitrile and centrifuged, and the supernatant was transferred to auto-sampler vials for FTS determination by LC/MS.
  • As shown in FIG. 2, mean plasma FTS concentrations were 3381 ng/ml at 1 hour, 1593 ng/ml after 2 hours, and decreased rapidly to less than 3 ng/ml after 24 hours. These values indicated 69% oral bioavailabilty compared to intra-peritoneal injection of FTS in a control group of animals.
  • These results, as illustrated in FIGS. 1 and 2, were unexpected, and in addition, show that therapeutic levels may be readily obtained upon oral administration.
  • Example 2 Inhibition of Pancreatic Tumor Growth with Orally Administered FTS
  • Nude CD1-Nu Mice were implanted subcutaneously with 5.0×106 Panc-1 cells in 0.2 ml of PBS just above the right femoral joint. Eleven days after implantation, palpable tumors were observed.
  • FTS was dispersed in 0.5% aqueous carboxymethylcellulose at a concentration of 5 mg/ml. On the eleventh day after implantation, mice were segregated into three groups and were given vehicle or FTS suspension by gavage.
  • Group 1 (Controls, 6 mice) received 0.2 ml vehicle daily.
  • Group 2 (20 mg/kg FTS, 7 mice) received 0.1 ml FTS suspension daily.
  • Group 3 (40 mg/kg FTS, 6 mice) received 0.2 ml FTS suspension daily.
  • Mice were treated for ten days, and were sacrificed. Tumors were excised and weighed.
  • As shown in FIG. 3, average tumor weight for Group 1 (controls) was 1.02 g; average tumor weight for Group 2 (20 mg/kg FTS) was 0.68 g (32% lower than control); and average tumor weight for Group 3 (40 mg/kg FTS) was 0.47 g (53% lower than control).
  • Example 3 Compatibility of FTS with Pharmaceutical Excipients
  • Physical mixtures (1:1 w/w) of FTS and representative pharmaceutical excipients commonly used in oral formulations were stored at 40° C./75% RH accelerated stress conditions for four weeks in loosely capped vials. Samples were assayed by HPLC at the 0, 2-week and 4-week time points. The results are shown in the table below.
  • TABLE
    Excipient
    0 weeks 2 weeks 4 weeks Observation
    Lactose 97.94 97.23 98.09 Compatible
    Dibasic calcium 97.95 95.75 97.08 Compatible
    phosphate
    Starch 97.88 97.30 98.11 Compatible
    Hydroxypropylmethyl 97.93 98.06 98.12 Compatible
    Cellulose
    Microcrystalline 97.93 97.72 98.12 Compatible
    Cellulose
    Polyvinylpyrrolidone 97.71 92.44 92.56 Potential
    incompat-
    ibility
    Croscarmellose 97.76 98.14 98.13 Compatible
    Sodium
    Sodium Carboxymethyl 97.76 98.12 98.17 Compatible
    Cellulose
    FTS Substance 97.75 97.10 97.76
  • Results of the accelerated stability evaluation demonstrated that many conventional pharmaceutical excipients may be employed in FTS tablets and capsules.
  • Example 4 FTS Tablets (200 mg)
  • FTS active pharmaceutical ingredient (2000 g), microcrystalline cellulose (2000 g), croscarmellose sodium (200 g), and magnesium stearate (100 g) are blended to uniformity and compressed into tablets weighing 430 mg. Assuming a 5% loss on material transfers and tablet press start-up, adjustment, and shut down, approximately 9,500 FTS 200 mg tablets are yielded.
  • Example 5 FTS Hard Gelatin Capsules (100 mg.)
  • FTS active pharmaceutical ingredient (1000 g), lactose (1000 g), microcrystalline cellulose (1000 g), and amorphous colloidal silicon dioxide (15 g) are blended to uniformity and filled into hard gelatin capsules. Assuming a 5% loss on material transfers and encapsulating machine start-up, adjustment, and shut down, approximately 9,500 FTS 100 mg capsules are yielded.
  • Example 6 FTS Soft Gelatin Capsules (50 mg)
  • FTS active pharmaceutical ingredient (500 g), is uniformly dispersed in a mixture of corn oil (3000 g), lecithin (30 g), and Tween-80 (150 g) and filled into soft gelatin capsules. Assuming a 5% loss on material transfers and soft gelatin encapsulating machine start-up, adjustment, and shut down, approximately 9,500 FTS 50 mg soft gelatin capsules are yielded.
  • Example 7 FTS Capsules (200 mg)
  • FTS active pharmaceutical ingredient (2841 g), microcrystalline cellulose (1421 g), starch (710 g), and magnesium stearate (29 g) are blended to uniformity and filled into size #0 hard-shell gelatin capsules, with a fill weight of 0.357 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 13,000 FTS 200 mg capsules are yielded.
  • Example 8 FTS Capsules (200 mg)
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #1 hard-shell gelatin capsules, with a fill weight of 0.225 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 4,750 FTS 200 mg capsules are yielded. EXAMPLE 9: FTS Capsules (100 mg)
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #2 hard-shell gelatin capsules, with a fill weight of 0.112.5 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 9,500 FTS 100 mg capsules are yielded.
  • Example 10 FTS Capsules (50 mg)
  • FTS active pharmaceutical ingredient (1000 g) and microcrystalline cellulose (125 g) are blended to uniformity and filled into size #3 hard-shell gelatin capsules, with a fill weight of 0.056 g per capsule. Assuming a 5% loss on material transfers, encapsulation equipment startup and adjustment, approximately 18,000 FTS 50 mg capsules are yielded.
  • Example 11 Bioavailability of Orally Administered FTS to Rats
  • FTS was dissolved in a mixture of corn oil (95%) and ethanol (5%). Aliquots of the resulting solution were administered to Crl:CD(SD)(IGS)BR rats (twenty-four) at a dose of 200 mg/kg. Blood samples were collected in lithium-heparin-containing tubes via the retro-orbital plexus while under CO2/O2 anesthesia at 1, 2, 4, 8, 12 and 24 hours after dosing. Concentration of FTS at each time point was determined by LC/MS. As shown in FIG. 4, mean plasma FTS concentrations were 4454 ng/ml at 1 hour, 4550 ng/ml at 2 hours, 3047 ng/ml at 4 hours, 2216 ng/ml at 8 hours, 685 ng/ml at 12 hours, and 140 ng/ml at 24 hours. These values indicate a high level of oral bioavailability.
  • All patent and non-patent publications cited in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All these publications are herein incorporated by reference to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.
  • Although the invention herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principles and applications of the present invention. It is therefore to be understood that numerous modifications may be made to the illustrative embodiments and that other arrangements may be devised without departing from the spirit and scope of the present invention as defined by the appended claims.

Claims (21)

1. An oral dosage form comprising an amount of a Ras antagonist effective to treat a disease or disorder involving abnormal cell proliferation, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula
Figure US20150191426A1-20150709-C00006
wherein:
R1 represents farnesyl, geranyl or geranyl-geranyl;
R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;
R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and
X represents S, wherein said dosage form is a tablet or capsule.
2. The oral dosage form of claim 1, wherein the Ras antagonist is farnesyl-thiosalicyclic acid (FTS).
3. The oral dosage form of claim 1, wherein the Ras antagonist is 5-fluoro-FTS.
4. The oral dosage form of claim 1, wherein the Ras antagonist is 5-chloro-FTS.
5. The oral dosage form of claim 1, wherein the Ras antagonist is 4-chloro-FTS.
6. The oral dosage form of claim 1, wherein the Ras antagonist is S-farnesyl-thiosalicylic acid methyl ester.
7. The oral dosage form of claim 1, wherein the effective amount is about 5 mg to about 500 mg.
8. The oral dosage form of claim 1, wherein the effective amount is about 10 mg to about 250 mg.
9. The oral dosage form of claim 1, wherein said carrier comprises one or more of a bulking agent, binder, disintegrant, glidant or lubricant.
10. The oral dosage form of claim 1, wherein said tablet has a coating on an outer surface thereof.
11. The oral dosage form of claim 1, which is in the form of a soft or hard gelatin capsule.
12. A method of treating a disease or disorder involving abnormal cell proliferation, comprising administering to a human in need thereof an oral dosage form comprising an amount of a Ras antagonist effective to treat the disease or disorder, and a pharmaceutically acceptable carrier other than a cyclodextrin, wherein said Ras antagonist is represented by the formula I
Figure US20150191426A1-20150709-C00007
wherein:
R1 represents farnesyl, geranyl or geranyl-geranyl;
R2 is COOR7, or CONR7R8, wherein R7 and R8 are each independently hydrogen, alkyl or alkenyl;
R3, R4, R5 and R6 are each independently hydrogen, alkyl, alkenyl, alkoxy, halo, trifluoromethyl, trifluoromethoxy, or alkylmercapto; and
X represents S, wherein the oral dosage form is a tablet or capsule.
13. The method of claim 12, wherein the disease is cancer.
14. The method of claim 13, wherein the cancer is breast cancer.
15. The method of claim 13, wherein the cancer is pancreatic cancer.
16. The method of claim 12, wherein the disease or disorder is cirrohsis of the liver.
17. The method of claim 12, wherein the disease or disorder is post-angioplasty restenosis.
18. The method of claim 17, wherein the oral dosage form is administered prophylactically.
19. The method of claim 12, wherein the disease or disorder is atherosclerosis.
20. The method of claim 12, wherein the disease or disorder is graft rejection.
21. The method of claim 12, wherein the disease or disorder is an autoimmune disease.
US14/662,420 2004-08-18 2015-03-19 Methods and compositions for oral delivery of fts Abandoned US20150191426A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/662,420 US20150191426A1 (en) 2004-08-18 2015-03-19 Methods and compositions for oral delivery of fts
US15/170,416 US20160271089A1 (en) 2004-08-18 2016-06-01 Methods and compositions for oral delivery of fts

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60236104P 2004-08-18 2004-08-18
PCT/US2005/029389 WO2006023639A1 (en) 2004-08-18 2005-08-18 Methods and compositions for oral delivery of fts
US65958207A 2007-02-07 2007-02-07
US13/326,598 US20120082722A1 (en) 2004-08-18 2011-12-15 Methods and compositions for oral delivery of fts
US14/662,420 US20150191426A1 (en) 2004-08-18 2015-03-19 Methods and compositions for oral delivery of fts

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/326,598 Continuation US20120082722A1 (en) 2004-08-18 2011-12-15 Methods and compositions for oral delivery of fts

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/170,416 Continuation US20160271089A1 (en) 2004-08-18 2016-06-01 Methods and compositions for oral delivery of fts

Publications (1)

Publication Number Publication Date
US20150191426A1 true US20150191426A1 (en) 2015-07-09

Family

ID=35447506

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/659,582 Expired - Fee Related US8088756B2 (en) 2004-08-18 2005-08-18 Methods and compositions for oral delivery of FTS
US13/326,598 Abandoned US20120082722A1 (en) 2004-08-18 2011-12-15 Methods and compositions for oral delivery of fts
US14/662,420 Abandoned US20150191426A1 (en) 2004-08-18 2015-03-19 Methods and compositions for oral delivery of fts
US15/170,416 Abandoned US20160271089A1 (en) 2004-08-18 2016-06-01 Methods and compositions for oral delivery of fts

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/659,582 Expired - Fee Related US8088756B2 (en) 2004-08-18 2005-08-18 Methods and compositions for oral delivery of FTS
US13/326,598 Abandoned US20120082722A1 (en) 2004-08-18 2011-12-15 Methods and compositions for oral delivery of fts

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/170,416 Abandoned US20160271089A1 (en) 2004-08-18 2016-06-01 Methods and compositions for oral delivery of fts

Country Status (9)

Country Link
US (4) US8088756B2 (en)
EP (3) EP2301528B1 (en)
AT (2) ATE466574T1 (en)
CA (1) CA2577310C (en)
DE (1) DE602005021115D1 (en)
ES (2) ES2343737T3 (en)
MX (1) MX2007001929A (en)
PL (1) PL1778209T3 (en)
WO (1) WO2006023639A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2301528B1 (en) 2004-08-18 2013-04-03 Kadmon Corporation, LLC Use of FTS for treating malignant disorders
DE602006017728D1 (en) * 2005-11-28 2010-12-02 Univ Ramot CANCER TREATMENT BY AGENT AND 2-DEOXYGLUCOSE
WO2007091241A1 (en) * 2006-02-10 2007-08-16 Ramot At Tel Aviv University Ltd. Treatment of ovarian cancer
WO2007144889A2 (en) * 2006-06-14 2007-12-21 Ramot At Tel Aviv University Ltd. Treatment of neurofibromatosis
ATE543492T1 (en) 2006-12-19 2012-02-15 Univ Ramot TREATMENT OF LUNG CANCER
JP2013508458A (en) * 2009-10-26 2013-03-07 ラモット・アット・テル−アヴィヴ・ユニヴァーシティ・リミテッド Cancer treatment using a combination of FTS and HDAC inhibitor
US9738614B2 (en) 2011-10-07 2017-08-22 Pisces Therapeutics, Llc Malignant and non-malignant disease treatment with Ras antagonists
EP2763681B1 (en) 2011-10-07 2017-08-16 Pisces Therapeutics LLC Malignant and non-malignant disease treatment with ras antagonists

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US20020115696A1 (en) * 1999-06-18 2002-08-22 Yoel Kloog Treatment of post-angioplasty restenosis and atherosclerosis with ras antagonists
US6462086B1 (en) * 1999-06-18 2002-10-08 Ramot University Authority For Applied Research And Industrial Development Ltd. Non-malignant disease treatment with Ras antagonists

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL107587A (en) 1993-11-12 1998-08-16 Univ Ramot Farnesyl geranyl or geranyl-geranyl derivatives pharmaceutical compositions containing them and methods for their preparation
AU2859499A (en) * 1998-03-27 1999-10-18 Chong Kun Dang Corporation (streptomyces) sp. producing tautomycetin and immunosuppressant comprising tautomycetin as active ingredient
US20040072258A1 (en) 2000-10-04 2004-04-15 Yoel Kloog Isoprenoid-dependent ras anchorage (idra) proteins
US20020169183A1 (en) * 2001-03-08 2002-11-14 Villar Hugo O. Acridines as stimulators for Fas-mediated apoptosis
EP1390472A4 (en) * 2001-05-29 2004-11-17 Sirna Therapeutics Inc Nucleic acid treatment of diseases or conditions related to levels of ras, her2 and hiv
EP1604037A1 (en) 2003-03-20 2005-12-14 Universite Catholique De Louvain Medical use of ras antagonists for the treatment of capillary malformation
WO2004103352A1 (en) 2003-05-23 2004-12-02 Ramot At Tel Aviv University, Ltd. Ras antagonists for treating neurodegenerative disorders
WO2005018562A2 (en) * 2003-08-22 2005-03-03 University Of Virginia Patent Foundation Blockade of mtor to prevent a hormonal adaptive response
EP2301528B1 (en) * 2004-08-18 2013-04-03 Kadmon Corporation, LLC Use of FTS for treating malignant disorders
DE602006017728D1 (en) 2005-11-28 2010-12-02 Univ Ramot CANCER TREATMENT BY AGENT AND 2-DEOXYGLUCOSE

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US20020115696A1 (en) * 1999-06-18 2002-08-22 Yoel Kloog Treatment of post-angioplasty restenosis and atherosclerosis with ras antagonists
US6462086B1 (en) * 1999-06-18 2002-10-08 Ramot University Authority For Applied Research And Industrial Development Ltd. Non-malignant disease treatment with Ras antagonists

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Ansel et al. (Pharmaceutical Dosage Forms and Drug Delivery Systems 1999, 7th Ed, Lippincott Williams & Wilkins pages:90, 91, 179, 180, 201) *
CAS MSDS FTS. *
Fromowitz et al. (Abstract of: Hum Pathol. 1987;18(12):1268-75) *

Also Published As

Publication number Publication date
EP1778209B8 (en) 2010-06-16
EP1778209A1 (en) 2007-05-02
EP1778209B1 (en) 2010-05-05
DE602005021115D1 (en) 2010-06-17
ES2414861T3 (en) 2013-07-23
CA2577310A1 (en) 2006-03-02
ES2343737T3 (en) 2010-08-09
ATE535237T1 (en) 2011-12-15
US20160271089A1 (en) 2016-09-22
US8088756B2 (en) 2012-01-03
EP2301528B1 (en) 2013-04-03
CA2577310C (en) 2011-01-11
EP2218451B1 (en) 2011-11-30
EP2301528A1 (en) 2011-03-30
US20090226512A1 (en) 2009-09-10
WO2006023639A1 (en) 2006-03-02
US20120082722A1 (en) 2012-04-05
ATE466574T1 (en) 2010-05-15
PL1778209T3 (en) 2010-10-29
EP2218451A1 (en) 2010-08-18
MX2007001929A (en) 2007-04-17

Similar Documents

Publication Publication Date Title
US20160271089A1 (en) Methods and compositions for oral delivery of fts
KR100858464B1 (en) Combretastatin a-4 phosphate prodrug mono- and di-organic amine salts, mono- and di-amino acid salts, and mono- and di-amino acid ester salts
JP2018035160A (en) Organic compound
KR20020075801A (en) Partial fatty acid oxidation inhibitors in the treatment of congestive heart failure
TW201249800A (en) Method of treating cancer and bone cancer pain
US11731941B2 (en) Crystalline solid forms of salts of N-{4-[(6,7-dimethoxyquinolin-4-yl)oxy]phenyl}-N′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide, processes for making, and methods of use
US6462237B1 (en) Cyclodextrin stabilized pharmaceutical compositions of bupropion hydrochloride
US8022104B2 (en) Formulations of ladostigil tartrate
KR20090067210A (en) Phenylalkyl carbamate compositions
US20140199382A1 (en) Stable pharmaceutical compositions of an s1p receptor agonist
US20080161404A1 (en) Bicalutamide for Delivering Increasing Steady State Plasma Levels
US6482417B2 (en) Stable pharmaceutical formulation comprising torsemide modification II
EA037149B1 (en) Bioavailable polyamines
WO2015104666A2 (en) Pharmaceutical composition of fingolimod
US6187345B1 (en) Flutamide compositions and preparations
CN111683659B (en) Composition of amino pyran derivative
US11701362B2 (en) Compositions and methods of use of cis-4-[2-{[(3S,4R)-3-fluorooxan-4-yl]amino}-8-(2,4,6-trichloroanilino)-9H-purin-9-yl]-1- methylcyclohexane-1-carboxamide
CN109381458A (en) Fomepizole and its salt are preparing the purposes in antiepileptic
WO2020049588A1 (en) Technology for enhancing bioavalaibility of selective estrogen receptor modulator (serm)
AU2016225785A1 (en) Crystalline forms of fingolimod HCL

Legal Events

Date Code Title Description
AS Assignment

Owner name: CONCORDIA PHARMACEUTICALS, INC., FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BAUER, VICTOR J.;REEL/FRAME:036236/0285

Effective date: 20070125

AS Assignment

Owner name: KADMON CORPORATION, LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CONCORDIA PHARMACEUTICALS, INC.;REEL/FRAME:036247/0782

Effective date: 20111215

AS Assignment

Owner name: PERCEPTIVE CREDIT OPPORTUNITIES FUND, LP, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNOR:KADMON CORPORATION, LLC;REEL/FRAME:036515/0084

Effective date: 20150828

AS Assignment

Owner name: MACQUARIE US TRADING LLC, ILLINOIS

Free format text: SECURITY INTEREST;ASSIGNOR:KADMON CORPORATION, LLC;REEL/FRAME:036573/0001

Effective date: 20150828

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: KADMON CORPORATION, LLC, NEW YORK

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:CONCORDIA PHARMACEUTICALS, INC.;REEL/FRAME:039166/0644

Effective date: 20160629

AS Assignment

Owner name: RAMOT AT TEL-AVIV UNIVERSITY LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KADMON CORPORATION, LLC;REEL/FRAME:039225/0829

Effective date: 20160630

AS Assignment

Owner name: KADMON CORPORATION, LLC, NEW YORK

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME PREVIOUSLY RECORDED ON REEL 039166 FRAME 0644. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:CONCORDIA PHARMACEUTICALS, LLC;REEL/FRAME:039696/0734

Effective date: 20160629

AS Assignment

Owner name: CONCORDIA PHARMACEUTICALS, LLC, FLORIDA

Free format text: CHANGE OF NAME;ASSIGNOR:CONCORDIA PHARMACEUTICALS, INC.;REEL/FRAME:040357/0897

Effective date: 20120315

AS Assignment

Owner name: KADMON CORPORATION, LLC, NEW YORK

Free format text: TERMINATION OF PATENT AND TRADEMARK SECURITY AGREEMENT;ASSIGNOR:PERCEPTIVE CREDIT OPPORTUNITIES FUND, LP;REEL/FRAME:051106/0677

Effective date: 20191125

AS Assignment

Owner name: KADMON CORPORATION, LLC, NEW YORK

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:MACQUARIE US TRADING LLC;REEL/FRAME:057973/0751

Effective date: 20191125