US20030220297A1 - Phosphorus-containing compounds and uses thereof - Google Patents

Phosphorus-containing compounds and uses thereof Download PDF

Info

Publication number
US20030220297A1
US20030220297A1 US10/357,152 US35715203A US2003220297A1 US 20030220297 A1 US20030220297 A1 US 20030220297A1 US 35715203 A US35715203 A US 35715203A US 2003220297 A1 US2003220297 A1 US 2003220297A1
Authority
US
United States
Prior art keywords
compound
independently
moiety
occurrence
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/357,152
Other languages
English (en)
Inventor
David Berstein
Chester Metcalf
Leonard Rozamus
Yihan Wang
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.)
Ariad Gene Therapeutics Inc
Original Assignee
Ariad Gene Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ariad Gene Therapeutics Inc filed Critical Ariad Gene Therapeutics Inc
Priority to US10/357,152 priority Critical patent/US20030220297A1/en
Assigned to ARIAD GENE THERAPEUTICS, INC. reassignment ARIAD GENE THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: METCALF, III, CHESTER A., ROZAMUS, LEONARD W., WANG, YIHAN, BERSTEIN, DAVID L.
Priority to US10/635,054 priority patent/US20040073024A1/en
Publication of US20030220297A1 publication Critical patent/US20030220297A1/en
Priority to US10/862,149 priority patent/US7091213B2/en
Priority to US11/429,582 priority patent/US7432277B2/en
Priority to US11/494,418 priority patent/US7186826B2/en
Priority to US11/650,017 priority patent/US7709020B2/en
Priority to US12/798,501 priority patent/US8058426B2/en
Priority to US13/295,826 priority patent/US9024014B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • Rapamycin is a macrolide antibiotic produced by Streptomyces hygroscopicus . It binds to a FK506-binding protein, FKBP12, with high affinity to form a rapamycin:FKBP complex. Reported Kd values for that interaction are as low as 200 pM.
  • the rapamycin:FKBP complex binds with high affinity to the large cellular protein, FRAP, to form a tripartite, [FKBP:rapamycin]:[FRAP], complex.
  • complex rapamycin can be viewed as a dimerizer or adapter to join FKBP to FRAP. Formation of the complex is associated with rapamycin's various biological activities.
  • Rapamycin is a potent immunosuppressive agent and is used clinically to prevent rejection of transplanted organs. Rapamycin and/or its analogs, CCI 779 (Wyeth) and SDZ Rad (“RAD001”, Novartis) are promising agents for treating certain cancers, for immune suppression and/or for helping to decrease the incidence of restenosis following interventional cardiology.
  • Rapamycin has also been shown to have activity as an antifungal agent, in the experimental allergic encephalomyelitis model (a model for multiple sclerosis), in the adjuvant arthritis model (for rheumatoid arthritis), in inhibiting the formation of IgE-like antibodies, and for treating or preventing lupus erythematosus, pulmonary inflammation, insulin dependent diabetes mellitus, adult T-cell leukemia/lymphoma, and smooth muscle cell proliferation and intimal thickening following vascular injury. See e.g. published U.S. patent application 2001/0010920.
  • rapamycin is also capable of multimerizing appropriately designed chimeric proteins incorporating domains derived from FKBP and FRAP, respectively. Because of that activity, rapamycin and various derivatives or analogs thereof have also been used as multimerizing agents for activating biological switches based on such chimeric proteins. See e.g., WO96/41865; WO 99/36553; WO 01/14387; Rivera et al, Proc Natl Acad Sci U S A 96, 8657-8662; and Ye, X. et al (1999) Science 283, 88-91.
  • Rapamycin's potential for providing relief from such an important swath of wasted diseases has stimulated the search for rapamycin analogs with improved therapeutic index, pharmacokinetics, formulatability, ease or economy of production, etc.
  • the resulting investigation by the pharmaceutical industry and academic researchers has been a sustained one over the past few decades. This has led to the exploration of materials and methods for effecting chemical transformations of rapamycin, including reductions of ketones, demethylations, epimerizations, various acylations and alkylations of hydroxyls, etc.
  • rapalogs include, among others, variants of rapamycin having one or more of the following modifications relative to rapamycin: demethylation, elimination or replacement of the methoxy at C7, C42 and/or C29; elimination, derivatization or replacement of the hydroxy at C13, C43 and/or C28; reduction, elimination or derivatization of the ketone at C14, C24 and/or C30; replacement of the 6-membered pipecolate ring with a 5-membered prolyl ring; and alternative substitution on the cyclohexyl ring or replacement of the cyclohexyl ring with a substituted cyclopentyl ring.
  • New rapalogs with reduced immunosuppressive activity and/or interesting pharmacokinetic or bioavailability profiles would be very desirable for use as multimerizing agents or antifungal agents.
  • New rapalogs with attractive physicochemical or functional characteristics relative to rapamycin, e.g., in therapeutic index, bioavailability, pharmacokinetics, stability, etc., would also be of interest for a variety of pharmaceutical uses such as are mentioned above, including among others use as immunosuppressants, as anticancer agents and in reducing the incidence of restenosis following interventional cardiology (e.g. on drug-bearing stents).
  • rapalogs thought to be in clinical development as immunosuppressants at present are those with rather modest, conventional structural modifications, i.e., acylation or alkylation at C-43 (CCI 779 and SDZ RAD, respectively; see e.g., Yu, K. et al., Endocrine-Related Cancer (2001) 8, 249-258; Geoerger, B. et al., Cancer Res. (2001) 61 1527-1532) and Dancey, Hematol Oncol Clin N Am 16 (2002):1101-1114.
  • compositions containing such compounds and uses thereof are also provided.
  • A is —O—, —S— or —NR 2 —, or is absent (i.e., is a covalent bond linking JQ—to carbon 43);
  • Q is absent (i.e., is a covalent bond linking J to A or to carbon 43) or, if A is —O—, —S— or —NR 2 —, Q may be —V—, —OV—, —SV—, or —NR 2 V—, where V is an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, such that J is linked to the cyclohexyl ring directly, through A or through VA, OVA, SVA or NR 2 VA;
  • K is O or S
  • each occurrence of Y is independently —O—, —S—, —NR 2 —, or a chemical bond linking a R 5 moiety to P;
  • each occurrence of R 2 and R 5 is independently an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, or H; and each occurrence of R 6 is independently R 5 , —PK(YR 5 )(YR 5 ), —SO 2 (YR 5 ) or —C(O)(YR 5 ); so long as any R 2 , R 5 or R 6 moiety linked directly to P is not H (e.g., —PR 2 , —PR 5 and —PR 6 cannot be —PH);
  • R 2 , R 5 and/or R 6 moieties may be chemically linked to one another to form a ring
  • each occurrence of G is independently —O—, —S—, —NR 2 —, (M) x or a chemical bond linking R 6 to P;
  • each occurrence of M is independently a substituted or unsubstituted methylene moiety, and any M-M′ moiety may be saturated or unsaturated;
  • each occurrence of x is independently an integer from 0-6;
  • R 7a and R 7b are H and the other is H, halo, —R A , —OR A , —SR A , —OC(O)R A , —OC(O)NR A R B , —NR A R B , —NR B C(O)R A , —NR B C(O)OR A , —NR B SO2R A or —NR B SO2NR A R B ′; or R 7a and R 7b , taken together, are H in the tetraene moiety:
  • R A is R 2 and where R B is OH or R 2 . In some cases one or both of R A and R B is H;
  • R 28 is hydrogen; J; or an aliphatic, heteroaliphatic, aryl, heteroaryl, acyl, aroyl or heteroaroyl moiety; and n is 1 or 2;
  • each of the foregoing aliphatic and heteroaliphatic moieties is independently linear or branched, or cyclic or acyclic, and substituted or unsubstituted, and each of the aryl, heteoraryl, acyl, aroyl or heteroaroyl moieties is independently substituted or unsubstituted;
  • W comprises a substituted or unsubstituted heterocycle comprising
  • J-Q-A is not (HO) 2 (PO)—O— or the dimethyl phosphate ester thereof (and preferably not another di-lower alkyl ester thereof);
  • K, R 2 , R 5 and R 6 are as defined above.
  • J moieties currently of special interest are those in which K is oxygen, as are illustrated in numerous exemplary compounds depicted below, including among others, any of the following:
  • each occurrence of R 5 is an independently chosen lower aliphatic or aryl moiety, which may be substituted or unsubstituted, or in the case of —OR 5 moieties, may alternatively be H.
  • -Q-A— is O, especially in cases in which J is one of the currently preferred J moieties noted just above (although preferably not —PO 3 H 2 ).
  • JQA— is (R 2 Y)(Me)(P ⁇ O)O— in which R 2 Y— contains 15 or fewer carbon atoms, preferably 10 or fewer carbon atoms, and in some embodiments 6 or fewer carbon atoms.
  • This new family of compounds includes a number of classes of compounds of particular interest.
  • each R 5 is an independently selected, aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially a lower (i.e. from 1 to 6 carbons) aliphatic moiety, e.g., a lower alkyl, which may be optionally substituted (e.g. with a halo, hydroxyl, -0-acyl (i.e., acyloxy) , alkoxyl, haloalkyl-, hydroxyalkoxyl, aryl, or heteroaryl moiety, etc.).
  • the compounds of formula (a) comprise a moiety, J, selected from the following:
  • each R 5 is an independently selected, aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially a lower aliphatic moiety, e.g. a lower alkyl, which may be optionally substituted (e.g. with a hydroxyl, alkoxyl, hydroxyalkoxyl, acyloxy-, aryl, or heteroaryl moiety, etc.).
  • the R 5 moiety may additionally be H.
  • Illustrative examples include compounds of formula (b) in which J is selected from the following:
  • J-Q-A— is (R 5 )(R 5 O)PO—O—.
  • each R 5 is independently selected, and is H or an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially lower aliphatic moiety, including lower alkyl, which may be optionally substituted (e.g. with a hydroxyl, alkoxyl, hydroxyalkoxyl, acyloxy-, aryl, or heteroaryl moiety, etc.).
  • Illustrative examples include compounds of formula (c) in which J is selected from the following:
  • J-Q-A— is (R 5 O)(R 5 O)PO—O—.
  • each R 5 is an independently selected, aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially lower (i.e. from 1 to 6 carbons) aliphatic moiety including lower alkyl, which may be optionally substituted (e.g. with a hydroxyl, alkoxyl, hydroxyalkoxyl, acyloxy-, aryl, or heteroaryl moiety, etc.).
  • —NHR 5 is —NH 2 .
  • Illustrative examples include compounds of formula (d) in which J is selected from the following:
  • J-Q-A— is (R 5 )(R 5 N)PO—O—.
  • each R 5 is independently selected and is H or an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially lower (i.e. from 1 to 6 carbons) aliphatic moiety including lower alkyl, which may be optionally substituted (e.g. with a hydroxyl, alkoxyl, hydroxyalkoxyl, acyloxy-, aryl, or heteroaryl moiety, etc.).
  • Illustrative examples include compounds of formula (e) in which J is selected from the following:
  • J-Q-A— is (R 5 N)(R 5 N)PO—O—.
  • each R 5 is independently selected and is H or an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety (which moiety may be substituted or unsubstituted), especially lower (i.e. from 1 to 6 carbons) aliphatic moiety including lower alkyl, which may be optionally substituted (e.g. with a hydroxyl, alkoxyl, hydroxyalkoxyl, acyloxy-, aryl, or heteroaryl moiety, etc.).
  • Illustrative examples include compounds of formula (f) in which J is selected from the following:
  • QA is preferably —O— or —OVO—.
  • J, Q, n and the various R groups are as previously defined, and with the proviso noted previously.
  • This class encompasses a number of subclasses of interest, including the following-.
  • J is a moiety other than —PO 3 H 2 , a salt thereof, or —PO 3 Me 2 .
  • Those choices for the subsituent, J are permitted only in combination with one or more additional structural changes relative to rapamycin, e.g. altered stereochemistry at one or more sites including C43 or C28, modification in the substituent or stereochemistry at C7, reduction of one or more of the ketone functionalities, demethylation at one or more sites, etc.
  • additional structural changes relative to rapamycin e.g. altered stereochemistry at one or more sites including C43 or C28, modification in the substituent or stereochemistry at C7, reduction of one or more of the ketone functionalities, demethylation at one or more sites, etc.
  • R 5 is H or lower alkyl, including, among others, methyl.
  • subclass of compounds which differs from subclass (g)(i)(a) in one or more of the following respects: (a) the substituent at position 28 is epimerized (relative to the orientation of rapamycin's C28—OH), (b) one or both ketones at positions 24 and 30 are reduced to hydroxyl groups, (c) the methoxyl group at position 7 is replaced by H or by one of the various C7 substituents listed elsewhere, and (d) the substituent J-O— at position 43 is in the epimeric orientation (relative to the orientation of rapamycin's C43—OH).
  • J is any of the phosphorus-containing moieties as previously described.
  • This class includes the subclass in which Q is absent, i.e., in which J is linked (i.e., covalently bonded) to the cyclohexyl ring via a nitrogen as illustrated below:
  • rapamycin derivatives in which Q is present and comprises an aliphatic or heteroaliphatic moiety, V, which may be substituted or unsubstituted, where each of the variable moieties are as previously defined or otherwise exemplified herein:
  • J, Q, n and the various R groups are as previously defined.
  • This class encompasses a number of subclasses of interest, including the following:
  • (k) Compounds as in class (j), but with one or more additional structural modifications relative to rapamycin. Numerous such modifications are known in the art and are alluded to elsewhere herein, including replacement of the —OMe substituent at C7, or alteration of its stereochemistry; epimerization at one or both of C28 and C43; reduction of one or more of the ketone functionalities e.g. at one or both of ring positions 24 and 30; desmethylation at one or more sites; reduction of one or more of the double bonds between C1 and C6; and/or use of the prolyl analog instead of the pipicolate structure of rapamycin.
  • Compounds of this invention may be prepared by starting with the appropriate rapamycin analog in place of rapamycin itself.
  • A is —O—, —S— or —NR 2 — or is absent (i.e., or is a covalent bond linking JQ to C-43);
  • Q is absent (i.e., is a covalent bond) or (if A is —O—, —S— or —NR 2 —)
  • Q may be —V—, —OV—, —SV—, or —NR 2 V—, where V is an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, such that J is linked to the cyclohexyl ring directly, through A or through VA, OVA, SVA or NR 2 VA; K is O or S;
  • each occurrence of Y is independently —O—, —S—, —NR 2 —, or a bond linking a R 5 moiety to P;
  • each occurrence of R 2 and R 5 is independently an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, or H; and each occurrence of R 6 is independently R 5 , —PK(YR 5 )(YR 5 ), —SO 2 (YR 5 ) or —C(O)(YR 5 ); so long as any R 2 , R 5 or R 6 moiety linked directly to P is not H; wherein two R 2 , R 5 and/or R 6 moieties may be chemically linked to one another to form a ring; each occurrence of G is independently —O—, —S—, —NR 2 —, (M) x or a chemical bond linking R 6 to P; each occurrence of M is independently a substituted or unsubstituted methylene moiety, and any M-M′ moiety may be saturated or unsaturated; each occurrence of x is independently an integer from 0-6;
  • each of the foregoing aliphatic and heteroaliphatic moieties is independently linear or branched, or cyclic or acyclic, and substituted or unsubstituted, and each of the aryl, heteoraryl, acyl, aroyl or heteroaroyl moieties is independently substituted or unsubstituted;
  • J-Q-A— is not (HO) 2 (P ⁇ O)O— or (MeO) 2 (P ⁇ O)O—, or (HO) 2 (P ⁇ O)—W—O— (or a desmethyl or reduced analog of such (HO) 2 (P ⁇ O)—W—O-containing rapamycin derivative, where W comprises a substituted or unsubstituted heterocycle comprising
  • variable groups are as otherwise defined above in (p) and (q) except that each occurrence of R 2 and R 5 is an independently chosen lower aliphatic or aryl moiety, which may be substituted or unsubstituted (except that in addition, —OR 5 and —NR 2 R 5 , may be —OH and —NHR 5 );
  • J-Q-A— is (R 2 Y)(Me)(P ⁇ O)O—, then (R 2 Y) is not an immunogenic carrier material, detector carrier material or a solid matrix, or a salt thereof.
  • A is absent or is —O—, —S— or —NR 2 —;
  • Q is absent or (if A is —O—, —S— or —NR 2 —)
  • Q may be —V—, —OV—, —SV—, or —NR 2 V—, where V is an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, such that J is linked to the cyclohexyl ring directly, through A or through VA, OVA, SVA or NR 2 VA;
  • K is O or S; each occurrence of Y is independently —O—, —S—, —NR 2 —, or a chemical bond linking a R 5 moiety to P; each occurrence of R 2 and R 5 is independently an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, or H; and each occurrence of R 6 is independently R 5 , —PK(YR 5 )(YR 5 ), —SO 2 (YR 5 ) or —C(O)(YR 5 ); so long as any R 2 , R 5 or R 6 moiety linked directly to P is not H;
  • R 2 , R 5 and/or R 6 moieties may be chemically linked to one another to form a ring; each occurrence of G is independently —O—, —S—, —NR 2 —, (M) x or a chemical bond linking R 6 to P; each occurrence of M is independently a substituted or unsubstituted methylene moiety, and any M-M′ moiety may be saturated or unsaturated; each occurrence of x is independently an integer from 0-6;
  • each of the foregoing aliphatic and heteroaliphatic moieties is independently linear or branched, or cyclic or acyclic, and substituted or unsubstituted, and each of the aryl, heteoraryl, acyl, aroyl or heteroaroyl moieties is independently substituted or unsubstituted;
  • each occurrence of R 2 and R 5 is an independently chosen lower aliphatic or aryl moiety, which may be substituted or unsubstituted, except that in addition, —OR 5 and —NR 2 R 5 , may be —OH and —NHR 5 ; with the proviso that if J-Q-A—is (R 2 Y)(Me)(P ⁇ O)O—, then (R 2 Y) contains 15 or fewer carbon atoms.
  • each occurrence of R 2 and R 5 is an independently chosen C1-C6 alkyl group optionally bearing one or more halo, —OH, alkoxyl-, alkyloxyalkyloxy-, haloalkyl-, hydroxyalkoxyl-, acyl-, acyloxy-, hetrocyclic, aryl or heteroaryl substituents, except that in addition, —OR 5 and —NR 2 R 5 , may be —OH and —NHR 5 .
  • each occurrence of R 2 and R 5 is independently chosen from methyl, ethyl, n-propyl, --propyl, n-butyl, 2-butyl, t-butyl, phenyl, or heteroaryl, each of which optionally bearing one or more halo, —OH, alkoxyl-, alkoxylalkoxyl-, haloalkyl-, hydroxyalkoxyl-, acyl-, acyloxy-, heterocyclic, aryl or heteroaryl substituents, and in addition, —OR 5 and —NR 2 R 5 , may be —OH and —NHR 5 .
  • each occurrence of Y is independently —O—, —S—, —NR 2 —, or a bond linking a R 5 moiety to P;
  • each occurrence of R 2 and R 5 is independently an aliphatic, heteroaliphatic, aryl, or heteroaryl moiety, or H; and each occurrence of R 6 is independently R 5 , —PK(YR )(YR 5 ), —SO 2 (YR 5 ) or —C(O)(YR 5 ); so long as any R 2 , R 5 or R 6 moiety linked directly to P is not H; wherein two R 2 , R 5 and/or R 6 moieties may be chemically linked to one another to form a ring;
  • each occurrence of G is independently —O—, —S—, —NR 2 —, (M) x or a chemical bond linking R 6 to P;
  • each occurrence of M is independently a substituted or unsubstituted methylene moiety, and any M-M′ moiety may be saturated or unsaturated;
  • each occurrence of x is independently an integer from 0-6;
  • each of the foregoing aliphatic and heteroaliphatic moieties is independently linear or branched, or cyclic or acyclic, and substituted or unsubstituted, and each of the aryl, heteoraryl, acyl, aroyl or heteroaroyl moieties is independently substituted or unsubstituted;
  • each occurrence of R 2 and R 5 is an independently chosen C1-C6 alkyl group optionally bearing one or more halo, —OH, alkoxyl-, alkyloxyalkyloxy-, haloalkyl-, hydroxyalkoxyl-, acyl-, acyloxy-, hetrocyclic, aryl or heteroaryl substituents, except that in addition, —OR 5 and —NR 2 R 5 , may be —OH and —NHR 5 .
  • each occurrence of R 2 and R 5 is independently chosen from methyl, ethyl, n-propyl, -propyl, n-butyl, 2-butyl, t-butyl, phenyl or heteroaryl, each of which optionally bearing one or more of the foregoing types of substituents or other substituents as disclosed herein.
  • One class of compounds that are not within the invention are conjugates or rapamycin, or derivatives thereof, comprising a substituent on the oxygen at position 43 which comprises —P(O)(Me)(Z) where Z is a immunogenic carrier material, detector carrier material or a solid matrix, or a salt thereof, linked to the P via a carbonyl, —NH—, —S—, —O— or certain aliphatic groups such as disclosed in US 2001/0010920 A1. That document discloses conjugates of rapamycin with such carriers or matrices for use in generating and detecting antibodies, for measuring levels of rapamycin iand for isolating rapamycin binding proteins.
  • the immunogenic carrier material can be selected from any conventionally known immunogenic carrier material, and will usually a protein or polypeptide, and in some cases could be a sufficiently large and immunogenic carbohydrate, polysaccharide, lipopolysaccharide, or nucleic acid; immunogenic proteins and polypeptides will have molecular weights between 5,000 and 10,000,000, preferably greater than 15,000 and more usually greater than 40,000; examples included albumins, globulins, enzymes, hemocyanins, glutelins or proteins having significant non-proteinaceous constituents, e.g., glycoproteins; the detector carrier material can be an enzyme such as horseradish peroxidase, alkaline phosphatase, luciferase, a fluorescent moiety such as fluorescein, Texas Red, or rhodamine, a chemiluminescent moiety, and the like; the solid matrix carrier material can be resin beads, an ELISA plate, glass beads as commonly used in a radioi
  • compositions comprising a compound of the invention, including any of the various types of compounds noted above, together with a pharmaceutically acceptable vehicle and optionally containing one or more pharmaceutically acceptable excipients.
  • the composition may be one which is suitable for oral or parenteral administration to a subject, e.g. a mammalian subject, including a human patient.
  • Compositions may be prepared using conventional materials such that they are suitable for administration by any of the routes of administration noted in this document.
  • a method for suppressing the immune response of a subject by administering to the subject an immunosuppressive amount i.e., an immunosuppressive treatment course involving periodic administration of an immunosuppressive dose
  • an immunosuppressive amount i.e., an immunosuppressive treatment course involving periodic administration of an immunosuppressive dose
  • a method for suppressing the rejection of transplanted tissues in a recipient for instance as a method treating or suppressing the rejection of transplanted tissues in a recipient.
  • a method for treating coronary artery disease, cerebrovascular disease, arteriosclerosis, atherosclerosis, nonatheromatous arteriosclerosis, vascular wall damage from cellular events leading toward immune mediated vascular damage, stroke or multiinfarct dementia in a subject in need thereof comprising administering to the subject a composition containing a compound of this invention, alone or in combination with treatment with one or more other therapeutic agents as are noted elsewhere herein, including among others an ACE inhibitor (such as quinapril, perindopril, ramipril, captopril, trandolapril, fosinopril, lisinopril, moexipril, and enalapril); angiotensin II receptor antagonist (such as candesartan, irbesartan, losartan, valsartan, and telmisartan); fibric acid derivative (such as clofibrate, and gemfibrozil); HMG Co-A reductase
  • a method for treating cancer in a subject in need thereof which comprises administering to the subject a treatment effective amount of a composition containing a compound of this invention.
  • This treatment may be provided in combination with one or more other cancer therapies, such as in combination with the administration to the subject of one or more of an anti-cancer alkylating or intercalating agent; an antiestrogen; an inhibitor of a kinase (e.g., Src, BRC/Abl, kdr, aurora-2, glycogen synthase kinase 3 (“GSK-3”)); an antibody to a receptor or hormone implicated in a cancer (e.g.
  • EGFR EGFR
  • PDGFR EGFR
  • IGF-R IGF-R
  • IL-2 a soluble receptor or other receptor antagonist to such receptor
  • proteasome inhibitor or other NF-kB inhibitor or radiation.
  • examples of other thereapeutic agents include among others, Zyloprim, alemtuzmab, altretamine, amifostine, nastrozole, antibodies against prostate-specific membrane antigen (such as MLN-591, MLN591 RL and MLN2704), arsenic trioxide, Avastin® (or other anti-VEGF antibody), bexarotene, bleomycin, busulfan, capecitabine, carboplatin, Gliadel Wafer, celecoxib, chlorambucil, cisplatin, cisplatin-epinephrine gel, cladribine, cytarabine liposomal, daunorubicin liposomal, daunorubicin,
  • a drug eluting stent comprising a vascular stent containing a compound this invention, dispersed in a matrix or disposed in channels, reservoirs or other chambers on or in said stent.
  • vascular stent containing a compound this invention dispersed in a matrix or disposed in channels, reservoirs or other chambers on or in said stent.
  • Various types of stents and means and materials for loading such stents with drug are noted elsewhere herein and in references cited herein.
  • Various matrices, polymers and other materials are also noted herein or in the cited references.
  • Illustrative stents include the following stents: Angiomed (Bard), Cardiocoil (In-Stent Medtronic), CORINTHIAN (BSC), Radius (Scimed), Wallstent (Schneider), Act-one (ACT), Angiostent (angioynamics), be-Stent (In-Stent Medtronic), BiodivYsio (Biocompatibles), Cordis, Cross-flex (Cordis), Crown (JJIS), Freedom (Global therapeutics), Gianturco-Roubin II (Cook), Jo-med, Jostent flex (Jomed), Microstent GFX (AVE), Multilink (Guidant-ACS), NIR (Medinol), NIR Royal (Medinol), NIRflex (Medinol), NIRSIDE flex (Medinol), Palmaz-Scatz (JJIS), STS (De Scheerder), Tensum (Biotronic), Wiktor-GX (Medtronic), Wiktor-I (
  • the stent may be any of the foregoing, or may be another example of any of the types of stents noted herein and in the cited references, and may contain other materials (e.g. polymers which may be degradable or erodable or not) as noted elsewhere.
  • compositions containing a compound of the invention and a diluent suitable for applying the compound to a stent are provided.
  • This invention provides a family of new rapalogs, many illustrative types and specific examples of which are disclosed herein. Those compounds, rapamycin analogs modified relative tto rapamycin at position 43, may also be further derivatized relative to rapamycin, e.g. at one or more of C7, C28, C13, C24 and C30 and elsewhere, by adapting chemical transformations such as those disclosed in U.S. Pat. No. 6,258,823, WO 96/41865, WO 98/02441, WO 99/36553 and WO 01/14387and in the other patent documents and scientific references cited therein or within this document.
  • Compounds of interest include among others, those which bind to human FKBP12, or inhibit its rotamase activity, within two, and more preferably within one order of magnitude of results obtained with rapamycin in any conventional FKBP binding or rotamase assay.
  • pharmaceutically acceptable derivatives of the foregoing compounds, where the phrase “pharmaceutically acceptable derivative” denotes any pharmaceutically acceptable salt, ester, carbamate, or salt of such ester or carbamate of such compound, or any other adduct or derivative which, upon administration to a patient, is capable of providing (directly or indirectly) a JQA-containing rapalog as described herein, or a biologically active metabolite or residue thereof.
  • Pharmaceutically acceptable derivatives thus include among others pro-drugs of the rapalogs.
  • a pro-drug is a derivative of a compound, usually with significantly reduced pharmacological activity, which contains an additional moiety which is susceptible to removal in vivo yielding the parent molecule as the pharmacologically active species.
  • An example of a pro-drug is an ester which is cleaved in vivo to yield a compound of interest.
  • Various pro-drugs of rapamycin and of other compounds, and materials and methods for derivatizing the parent compounds to create the pro-drugs are known and may be adapted to the present invention.
  • Compounds of this invention may be provided in substantially pure form (relative to side products, residual reactants or other unwanted materials), for example at least 50% pure, suitably at least 60% pure, advantageously at least 75% pure, preferably at least 85% pure, more preferably at least 95% pure, especially at least 98% pure, all percentages being calculated on a weight/weight basis.
  • An impure or less pure form of a compound of the invention may be useful in the preparation of a more pure form of the same compound or of a related compound (for example a corresponding derivative) suitable for pharmaceutical use.
  • the compounds of this invention may be used for multimerizing chimeric proteins in cells (in vitro, ex vivo or in vivo, i.e. in organisms harboring them) for a variety of important purposes, as described in detail for rapamycin and other rapalogs in WO 96/41865, WO 99/36553 and WO 01/14387. See also Rivera V M, Ye X, Courage N L, Sachar J, Cerasoli F, Wilson J M and Gilman M. (1999) Long-term regulated expression of growth hormone in mice following intramuscular gene transfer.
  • Compounds of this invention having antifungal activity are useful for the prophylaxis and treatment of fungal infections in animals, especially mammals, including humans, in particular humans and domesticated animals (including farm animals).
  • the compounds may be used, for example, in the treatment of topical fungal infections caused by, among other organisms, species of Candida (e.g. C. albicans ), Trichophyton (e.g. Trichophyton mentagrophytes ), Microsporum (e.g. Microsporum gypseum ) or Epidermophyton or in mucosal infections caused by Candida albicans (e.g. thrush and vaginal candidiasis).
  • Candida e.g. C. albicans
  • Trichophyton e.g. Trichophyton mentagrophytes
  • Microsporum e.g. Microsporum gypseum
  • Epidermophyton e.g. thrush and vaginal candidiasis
  • antifungal rapalogs of this invention may retain the methoxyl substituent at C7 or may contain any of a variety of replacement substituents, including H and bulky or non-bulky substituents.
  • Certain compounds of this invention have been found to inhibit T cell proliferation with observed EC50 values ranging to potency levels comparable to rapamycin. Potent activity has also been observed against human tumors in a nude mouse xenograft model. These rapalogs typically retain the methoxyl substituent at C7 or replace it with H or a replacement substituent which is not so much bulkier than methoxyl that it unduly reduces inhibition of T cell proliferation.
  • rapalogs may be used as immunosuppressant, antiproliferative, anti-tumor, and anti-restenotic agents, as well as for other uses described herein or in the litertaure for rapamycin and analogs such as CCI 779 and SDZ RAD (“RAD 001”) in the scientific and patent literature, examples of which are cited herein.
  • CCI 779 and SDZ RAD (“RAD 001”) in the scientific and patent literature, examples of which are cited herein.
  • Compounds with an unmodified C7 substituent relative to rapamycin, or which contain a replacement C7 substituent (i.e., in place of —OMe) that does not unduly reduce potency in a T cell inhibition assay, are of particular interest for such uses.
  • certain compounds of this invention have immunomodulatory activity, meaning that the compounds are capable of inducing immune suppression by inhibiting immune cell responses or proliferation in vitro or in vivo and/or by producing a statistically significant decrease in the inflammatory response as determined by any scientifically acceptable cellular, tissue or animal model.
  • Such compounds may be administered in a treatment effective amount and dosing regimen for treating, among others, conditions such as rheumatoid arthritis, osteoarthritis, systemic lupus erythematosis, multiple sclerosis, acute transplantation/graft rejection, myasthenia gravis, progressive systemic sclerosis, tuberous sclerosis, multiple myeloma, atopic dermatitis, hyperimmunoglobulin E, hepatitis B antigen negative chronic active hepatitis, Hashimoto's thyroiditis, Familial Mediterranean fever, Grave's disease, autoimmune hemolytic anemia, primary biliary cirrhosis, inflammatory bowel disease, and insulin dependent diabetes mellitus.
  • conditions such as rheumatoid arthritis, osteoarthritis, systemic lupus erythematosis, multiple sclerosis, acute transplantation/graft rejection, myasthenia gravis, progressive systemic sclerosis, tuberous sclerosis, multiple
  • Compounds of this invention also have activity against primary and/or metastatic cancers. They should be useful for reducing tumor size, inhibiting tumor growth or metastasis; treating various leukemias and/or prolonging the survival time of animals or patients with those diseases.
  • the invention provides compounds for use in medical therapy, in particular for use as antifungal, anticancer, immunosuppressive or anti-restenotic agents, or as agents against the other diseases and conditions disclosed herein.
  • the invention further provides a method of treating a human or non-human animal suffering from any of those diseases or conditions by the administration of an effective amount of the rapalog, and further provides pharmaceutical compositions comprising a compound of the invention together with a pharmaceutically acceptable diluent or carrier, as well as medical devices, such as drug-bearing stents, containing a compound of this invention.
  • compositions of this invention may be formulated as disclosed below and elsewhere herein (or using formulations based on those reported for rapamycin or rapamycin derivatives such as CCI-779 or RAD001), and may then be administered in treatment effective amounts to patients in need thereof for the treatment of a variety of diseases as noted herein.
  • Such compositions may be administered in any manner useful in directing the active compounds to the recipient's bloodstream or site of action, including orally, parenterally (including intravenous, intraperitoneal and subcutaneous injections as well as injection into joints or other tissues), via stents or other implants, rectally, intranasally, vaginally, and transdermally.
  • transdermal administrations are understood to include all administrations across the surface of the body and the inner linings of bodily passages including epithelial and mucosal tissues. Such administration may be carried out using the present compounds, or pharmaceutically acceptable salts or prodrugs thereof, in lotions, creams, foams, patches, suspensions, solutions, and suppositories (rectal and vaginal).
  • solutions or suspensions of these active compounds or a pharmacologically acceptable salt thereof can be prepared in water suitably mixed with a surfactant such as hydroxy-propylcellulose or by adaptation of formulations used for rapamycin, CC1779 or RAD001.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • compositions which contain a compound of this invention and which are suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the composition to be injected should be sterile and should be sufficiently fluid to permit transfer via syringe. It should be stable under the conditions of manufacture and storage and will preferably be protected from the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Parenteral formulations which may be adapted for use with rapalogs of this invention are disclosed in U.S. Pat. Nos. 5,530,006; 5,516,770; and 5,616,588, which are hereby incorporated by reference.
  • Formulation, routes of administration and dosing may be selected from, or based upon, those used for rapamycin and other rapamycin derivatives used for the same or analogous indications.
  • a preferred approach may be to determine through genotype analysis and/or in vitro culture and study of biopsied tumor samples, those patients with tumors in which the phosphatidyl-inositol 3 kinase/Akt-mTOR signaling pathway is particular important to cell growth, and then to selectively treat those patients with rapalog.
  • cancers involving abnormalities in the phosphatidyl-inositol 3 kinase/Akt-mTOR pathway include glioma, lymphoma and tumors of the lung, bladder, ovary, endometrium, prostate or cervix which are associated with abnormal growth factor receptors (e.g.
  • EGFR, PDGFR, IGF-R and IL-2 ovarian tumors which are associated with abnormalities in P13 kinase; melanoma and tumors of the breast, prostate or endometrium which are associated with abnormalities in PTEN; breast, gastric, ovarian, pancreatic, and prostate cancers associated with abnormalities with Akt; lymphoma, cancers of the breast or bladder and head and neck carcinoma associated with abnormalities in elF-4E; mantle cell lymphoma; breast cancer and head and neck carcinomas associated with abnormalities in Cyclin D; and, familial melanoma and pancreas carcinomas associated with abnormalities in P16.
  • a patient being treated with an anti-cancer compound of this invention may (before, during or after such treatment) also be treated one or more other anti-cancer agents such as cisplatin; an antiestrogen (e.g., raloxifene, droloxifene, idoxifine, nafoxidine, toremifene, TAT-59, levomeloxifene, LY-353381, CP-3361656, MDL-103323, EM-800 and ICI-182,780; see e.g.
  • an antiestrogen e.g., raloxifene, droloxifene, idoxifine, nafoxidine, toremifene, TAT-59, levomeloxifene, LY-353381, CP-3361656, MDL-103323, EM-800 and ICI-182,780; see e.g.
  • WO 02/13802 which may be adapted to the present invention
  • an inhibitor of a kinase such as Src, BRC/Abl, kdr, aurora-2, glycogen synthase kinase 3 (“GSK-3”), an epidermal growth factor receptor (“EGF-R”), or platelet derived growth factor receptor (“PDGF-R”) for example, including inhibitors such as Gleevec, Iressa, CP-358 7 7 4 (Tarceva), ZD-1839, SU-5416, or NSC-649890; an antibody (such as Herceptin) to a receptor or hormone (e.g.
  • VEGF vascular endothelial growth factor
  • a soluble receptor or other receptor antagonist to such receptor a proteasome inhibitor such as Velcade
  • an IKK inhibitor or other NF-kB inhibitor or radiation.
  • Each component of the combination may be administered as it would be if given alone, although in some cases reduced dosing of one or more components may be possible or beneficial in view of the combined action of the different drugs.
  • Compounds of this invention can also be used to help prevent restenosis or other complications following the introduction into the patient's body of a graft, stent, or other device. See e.g. Sousa et al, above, and Marx and Marks, 2001, Circulation 104:852-855.
  • the rapalogs of this invention may be applied to stents, grafts, shunts or other devices or scaffolds (including cardiac pacemaker leads or lead tips, cardiac defibrillator leads or lead tips, heart valves, pacemakers, orthopedic devices, etc.) to provide drug-eluting devices for implantation into patients in need thereof.
  • Stents and such devices are typically inserted into a patient's vasculature (e.g. veins, arteries, aorta, etc., including both coronary and peripheral arteries) but can also be used in many other organs, glands, ducts, etc.
  • vasculature e.g. veins, arteries, aorta, etc., including both coronary and peripheral arteries
  • a stent is an expandable tube, typically an expandable wire mesh tube, small enough to be inserted into a blood vessel. They are typically used to prevent closure of the vessel after a procedure such as angioplasty.
  • stents made from nitinol (a nickel-titanium alloy), cobalt alloy on a platinum core, platinum-iridium, stainless steel, gold plated stainless steel, silicon carbide, tantalum, coated tantalum, and other metals or non-metals, in a variety of designs including wire braid, spiral coil, slotted tube (zig zag design, serpentine mesh with rotating junctions, sinusoidal slot, cellular mesh, spiral articulation, etc.), wire mesh, sinusoidal single wire coil, single helical coil, fishbone, flexible coil, connected zig-zag wires, multiple rings, multicellular, etc.
  • re-closure An all too frequent complication of the use of stents is re-closure (“restenosis”) of the vessel after insertion of the stent.
  • restenosis One of the primary causes of restenosis is thought to be rapid proliferation of blood vessel cells in the region of the stent (“neointimal hyperplasia”), eventually blocking the vessel.
  • rapamycin-eluting stents One approach for reducing the incidence of restenosis has been the use of rapamycin-eluting stents. Other benefits of rapamycin-eluting stents have also been noted in the literature.
  • a compound of this invention may be disposed on or in such a device in place of rapamycin or other drug such that the compound is released (“elutes”) from the device after it is implanted in the recipient.
  • Drug eluting stents are generally prepared by coating at least part of the stent with a carrier material (typically a polymer) containing the drug or by filling one or more chambers or channels in, or on the surface of, the stent with the drug or a composition containing the drug. Coatings may be applied in more than one layer, some of which might not contain the drug. Sometimes an additional coating is provided on top of the drug-containing layer or reservoir, which additional coating permits gradual release of drug to the recipient's tissues.
  • a carrier material typically a polymer
  • a variety of methods and materials for applying drugs to stents and for using such stents are available to the practitioner and may be adapted to use with compounds of this invention.
  • methods and materials for releasing drugs from implantable and other devices are described in U.S. Pat. Nos. 6,471,980; 6,096,070; 5,824,049; 5,624,411; 5,609,629; 5,569,463; 5,447,724; and 5,464,650 as well as WO02066092.
  • the use of stents for drug delivery within the vasculature are described in PCT Publication No. WO 01/01957 and U.S. Pat. Nos.
  • patent application Pub. No. 2002/0082680 See also e.g., U.S. Pat. No. 6,471,979 which discloses methods and materials for loading a stent which may be applied to compounds of this invention.
  • Exemplary coatings disclosed therein include phosphorylcholine, polyurethane, segmented polyurethane, poly-L-lactic acid, cellulose ester, polyethylene glycol and polyphosphate esters, as well as naturally occurring vehicles or carriers like collagens, laminens, heparins, fibrins, and other naturally occurring substances that absorb to cellulose.
  • Using such a coating is advantageous in that it allows the compound to slowly release from the device. This extends the time that the affected portion of the body sustains the efficacious effects of the compounds.
  • the manner in which these coatings interact with the device material as well as the inherent structure of the coating provide a diffusion barrier, thereby controlling the release of the entrapped compound(s).
  • the matrix or coating with which the compounds are loaded on the stent or delivery device can control slow or fast delivery of the compound.
  • a coating such as a Phosphorylcholine-based coating like Biocompatibles' (LO) PC polymer is used.
  • Such coatings contain a hydrophobic component which aids in the initial adhesion and film-formation of the polymer on to the stainless steel stent substrate, while other groups allow cross-linking both within the polymer and with the stent surface to achieve firm anchorage.
  • the coating is thus tightly adhered to the stent and can survive balloon expansion without damage.
  • the coating can absorb a variety of molecules of different size and physical characteristics into the PC coating, and to release those in a controlled manner.
  • the PC coating may be as thin as ⁇ 0.1 ⁇ m, although thicker layers may also be used.
  • the stent, coated with the LO matrix may be immersed in a solution of the compound in an organic solvent for as little as a few minutes.
  • the loading level can be controlled by the concentration of the solution of the compound. After removal from the solution, the coating is allowed to dry briefly before it is ready for use. See e.g., WO 01/00109, 01/01957, 01/52915, 02/55121 and 02/55122.
  • rapalog of this invention in conjunction with a stent may be achieved, e.g., by adapting methods and materials used for the delivery of other drugs, especially rapamycin, using such devises, e.g. as disclosed in the foregoing documents as well as in U.S. Pat. Nos. 5,516,781; 6,153,252; 5,665,728; 5,646,160; 5,563,146; and 5,516,781 as well as in published international patent applications WO 01/01957, 01/49338, 01/87263, 01/87342, 01/87372, 01/87373, 01/87374, 01/87375, and 01/87376.
  • Rapalogs of this invention are broadly compatible across the range of stent designs and of methods and materials for coating, depositing, layering or otherwise loading them with drug. Loading such medical devices with a rapalog of this invention, medical devices loaded with a rapalog of this invention and the insertion into a blood vessel or other lumen in a recipient of such a device loaded with such a rapalog are all encompassed by this invention, including the various matrices, polymers, barriers, and other options available to the practitioner. In practice, the choice of materials (e.g. solvents, polymers, barrier layers, matrices, etc.) and precise methods may be optimized, depending on the choice of compound, using routine experimentation.
  • materials e.g. solvents, polymers, barrier layers, matrices, etc.
  • aliphatic as used herein includes both saturated and unsaturated (but non-aromatic), straight chain (i.e., unbranched), branched, cyclic, or polycyclic non-aromatic hydrocarbon moieties, which are optionally substituted with one or more functional groups.
  • alkyl, other aliphatic, alkoxy and acyl groups preferably contain 1-8, and in many cases 1-6, contiguous aliphatic carbon atoms.
  • Illustrative aliphatic groups thus include, for example, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, —CH 2 -cyclopropyl, allyl, n-butyl, sec-butyl, isobutyl, tert-butyl, cyclobutyl, —CH 2 -cyclobutyl, n-pentyl, sec-pentyl, isopentyl, tert-pentyl, cyclopentyl, —CH 2 -cyclopentyl, n-hexyl, sec-hexyl, cyclohexyl, —CH 2 -cyclohexyl moieties and the like, which again, may bear one or more substituents.
  • aliphatic is thus intended to include alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties.
  • alkyl includes both straight, branched and cyclic alkyl groups. An analogous convention applies to other generic terms such as “alkenyl”, “alkynyl” and the like. Furthermore, as used herein, the language “alkyl”, “alkenyl”, “alkynyl” and the like encompasses both substituted and unsubstituted groups.
  • alkyl refers to groups usually having one to eight, preferably one to six carbon atoms.
  • “alkyl” may refer to methyl, ethyl, n-propyl, isopropyl, cyclopropyl, butyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, isopentyl tert-pentyl, cyclopentyl, hexyl, isohexyl, cyclohexyl, and the like.
  • Suitable substituted alkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 3-fluoropropyl, hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, benzyl, substituted benzyl and the like.
  • alkenyl refers to groups usually having two to eight, preferably two to six carbon atoms.
  • alkenyl may refer to prop-2-enyl, but-2-enyl, but-3-enyl, 2-methylprop-2-enyl, hex-2-enyl, hex-5-enyl, 2,3-dimethylbut-2-enyl, and the like.
  • alkynyl which also refers to groups having two to eight, preferably two to six carbons, includes, but is not limited to, prop-2-ynyl, but-2-ynyl, but-3-ynyl, pent-2-ynyl, 3-methylpent-4-ynyl, hex-2-ynyl, hex-5-ynyl, and the like.
  • cycloalkyl refers specifically to groups having three to seven, preferably three to ten carbon atoms. Suitable cycloalkyls include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like, which, as in the case of other aliphatic or heteroaliphatic or heterocyclic moieties, may optionally be substituted.
  • heteroaliphatic refers to aliphatic moieties which contain one or more oxygen, sulfur, nitrogen, phosphorous or silicon atoms, e.g., in place of carbon atoms. Heteroaliphatic moieties may be branched, unbranched or cyclic and include heterocycles such as morpholino, pyrrolidinyl, etc.
  • heterocycle refers to non-aromatic ring systems having five to fourteen members, preferably five to ten, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, or S.
  • heterocyclic rings include 3-1H-benzimidazol-2-one, (1-substituted)-2-oxo-benzimidazol-3-yl, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-thiomorpholinyl, 3-thiomorpholinyl, 4-thiomorpholinyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-piperazinyl, 2-piperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substituted diazolonyl, 1-phthalimidinyl, benzoxanyl, benzopyrrolidinyl, benzopiperidin
  • heterocyclyl or “heterocyclic”, as it is used herein, is a group in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic or non-aromatic rings, such as in an indolinyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring.
  • heterocycle “heterocyclyl”, or “heterocyclic” whether saturated or partially unsaturated, also refers to rings that are optionally substituted.
  • aryl used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to aromatic ring groups having five to fourteen members, such as phenyl, 1-naphthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl.
  • aryl also refers to rings that are optionally substituted.
  • aryl may be used interchangeably with the term “aryl ring”.
  • Aryl also includes fused polycyclic aromatic ring systems in which an aromatic ring is fused to one or more rings.
  • Non-limiting examples of useful aryl ring groups include phenyl, halophenyl, alkoxyphenyl, dialkoxyphenyl, trialkoxyphenyl, alkylenedioxyphenyl, naphthyl, phenanthryl, anthryl, phenanthro and the like, as well as 1-naphthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as in a indanyl, phenanthridinyl, or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic ring.
  • heteroaryl refers to stable heterocyclic, and polyheterocyclic aromatic moieties having 3-14, usually 5-14, carbon atoms, which moieties may be substituted or unsubstituted and may comprise one or more rings. Substituents include any of the previously mentioned substituents.
  • heteroaryl rings include 5-membered monocyclic ring groups such as thienyl, pyrrolyl, imidazolyl, pyrazolyl, furyl, isothiazolyl, furazanyl, isoxazolyl, thiazolyl and the like; 6-membered monocyclic groups such as pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like; and polycyclic heterocyclic ring groups such as benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathienyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridin
  • heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxadiazolyl, 5-oxadiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 3-pyridazinyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 5-tetrazolyl, 2-triazolyl, 5-triazolyl, 2-thienyl, 3-thienyl, carbazolyl, benzimidazolyl,
  • Heteroaryl groups further include a group in which a heteroaromatic ring is fused to one or more aromatic or nonaromatic rings where the radical or point of attachment is on the heteroaromatic ring. Examples include tetrahydroquinoline, tetrahydroisoquinoline, and pyrido[3,4-d]pyrimidinyl.
  • heteroaryl also refers to rings that are optionally substituted.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic”.
  • An aryl group including the aryl portion of an aralkyl, aralkoxy, or aryloxyalkyl moiety and the like
  • heteroaryl group including the heteroaryl portion of a heteroaralkyl or heteroarylalkoxy moiety and the like
  • Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group include halogen, —YR 2 (i.e., including —R 2 , —OR 2 , —SR 2 and —NR 2 R 5 ), —Y—C( ⁇ O)R 2 , —Y—C( ⁇ O)OR 2 , —Y—C( ⁇ O)NR 2 R 5 , —Y—C( ⁇ NR 2′ )NR 2 R 5 , —COCOR 2 , —COMCOR 2 ), J, —CN, —S( ⁇ O)R 2 , —SO 2 R 2 , —SO 2 NR 2 R 5 , —NO 2 , —NR 5 SO 2 R 2 and —NR 5 SO 2 NR 2 R 5 .
  • halogen i.e., including —R 2 , —OR 2 , —SR 2 and —NR 2 R 5
  • —Y—C( ⁇ O)R 2
  • substituents in which Y is NR 2 thus include among others, —NR 2 C( ⁇ O)R 5 , —NR 2 C( ⁇ O)NR 5 , —NR 2 C( ⁇ O)OR 5 , and —NR 2 C( ⁇ NH)NR 5 .
  • R 2 and R 5 substituents may themselves be substituted or unsubstituted (e.g.
  • R 5 moiety examples include -alkylhalo such as chloromethyl or trichloromethyl; -alkoxyalkyl such as methoxyethyl-; mono-, di- and tri-alkoxyphenyl; methylenedioxyphenyl or ethylenedioxyphenyl; halophenyl; and alkylamino).
  • Additional illustrative examples include 1,2-methylene-dioxy, 1,2-ethylenedioxy, protected OH (such as acyloxy)), phenyl, substituted phenyl, —O-phenyl, —O-(substituted) phenyl, -benzyl, substituted benzyl, —O-phenethyl (i.e., —OCH 2 CH 2 C 6 H 5 ), —O-(substituted)phenethyl, —C(O)CH 2 C(O)R 2 , —CO 2 R 2 , —C( ⁇ O)R 2 (i.e., acyl in cases in which R 2 is aliphatic, aroyl in cases in which R 2 is aryl and heteroaroyl in cases in which R 2 is heteroaryl), —C( ⁇ O)NR 2 R 5 , —OC( ⁇ O)NR 2 R 5 , —C( ⁇ NH)NR 2 R 5 , and
  • substituents include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, and haloalkyl groups.
  • An aliphatic, heteroaliphatic or non-aromatic heterocyclic group may also contain one or more substituents.
  • suitable substituents on such groups include those listed above for the carbon atoms of an aryl or heteroaryl group and in addition include the following substituents for a saturated carbon atom: ⁇ O, ⁇ S, ⁇ NR 2 , ⁇ NNR 2 R 5 , ⁇ NNHC(O)R 2 , ⁇ NNHCO 2 R 2 , or ⁇ NNHSO 2 R 2 .
  • substituents on an aliphatic, heteroaliphatic or heterocyclic group include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl groups.
  • Illustrative substituents on the nitrogen of an aromatic or non-aromatic heterocyclic ring include —R 2 , —NR 2 R 5 , —C( ⁇ O)R 2 , —C( ⁇ O)OR 2 , —C( ⁇ O)NR 2 R 5 , —C( ⁇ NR 2′ )NR 2 R 5 , —COCOR 2 , —COMCOR 2 ), —CN, —NR 5 SO 2 R 2 and —NR 5 SO 2 NR 2 R 5 .
  • substituents on the aliphatic group or the phenyl ring include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • a combination of substituents or variables is permissible only if such a combination results in a stable or chemically feasible compound.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40° C. or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • Certain compounds of this invention may exist in tautomeric forms, and this invention includes all such tautomeric forms of those compounds unless otherwise specified.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • a JQA-containing rapalog as described herein may differ from the corresponding 43-JQA-containing derivative of rapamycin with respect to zero, one, two, three, four, five, six or seven (or more) substituent moieties or functional groups at positions other than position 43.
  • One class of rapalogs of this invention includes JQA-containing rapalogs with no other modifications, relative to rapamycin, i.e., other than the JQA modification at position 43.
  • Another class includes among others JQA-containing rapalogs with additional modification(s) at any one, two, three, four, five or all six of positions C7, C13, C14, C24, C28 and C30.
  • Modifications in rapalog structure are known for a number of previously known rapalogs (see e.g. WO 99/36553, Table III and Liberles et al, 1997, Proc Natl Acad Sci USA 94:7825-7830 and infra) and may be readily adapted to the present invention. See also WO 01/14387, already incorporated herein by reference, including among others pages 24-30, for information on known modifications and combinations of modifications known for rapamycin which may be used in the design of JQA-containing rapalogs.
  • JQA-containing rapalogs of special interest for practicing the methods of this invention are those those (or pharmaceutically acceptable derivatives thereof) in which R C7a is a moiety other than OMe.
  • This subset (“C7 JQA-containing rapalogs”) includes compounds in which one of R 7a and R 7b is H and the other is selected from —R A , -Z-R A , -Z-(CO)R A , -Z-(CO)ZR A , —NRASO 2 R A and —NSO 2 R A , where each Z is independently O, S or NR A .
  • JQA-containing rapalogs bearing a C7 substituent selected from the following group: aryl; heteroaryl; aryl, heteroaryl or benzyl ether;and —NH(CO)OR A , —NH(CO)R A , —NH(SO 2 )R A or —NH(SO 2 )NHR A (where R A is a substituted or unsubstituted lower alkyl, e.g., methyl, ethyl, iPr, butyl, benzyl, etc.
  • R 7a and R 7b are independently selected from the following groups: H; a substituted or unsubstituted two to eight carbon straightchain, branched or cyclic alkenyl, alkoxyl or alkylmercapto; and a substituted or unsubstituted aryl, heteroaryl, aryloxy or heteroaryloxy, arylmercapto or heteroarylmercapto.
  • R 7a is H; (together with R 7b ) ⁇ O; alkoxy; alkylmercapto; amino (primary, secondary, tertiary or quaternary); amido; carbamate; aryl or substituted aryl; phenyl or substituted phenyl; substituted or unsubstituted heteroaryl such as substituted or unsubstituted thiophenyl, furyl, indolyl, etc.; or benzyloxy or substituted benzyloxy.
  • C7 JQA-containing rapalogs which may be used in practicing the methods of this invention include those in which one of R 7a and R 7b is H and the other is selected from —OEt, —O-propyl, —O-butyl, —OCH 2 CH 2 —OH, —O-benzyl, —O-substituted benzyl (including e.g., 3-nitro-, 4-chloro-, 3-iodo-4-diazo-, 3,4-dimethoxy-, and 2-methoxy-), —S—Me, —S-phenyl, —O(CO)Me, -allyl, —CH 2 C(Me) ⁇ CH 2 , —OCH 2 —CCH, —OCH 2 —CC—Me, —OCH 2 —CC-Et, —OCH 2 —CC—CH 2 OH, or -2,4-dimethoxyphenyl, 2,4,
  • C7-modified JQA-containing rapalogs of particular interest are those bearing a substituted or unsubstituted aromatic ether, a substituted or unsubstituted benzyl ether or a carbamate moiety at C7.
  • the substituent at C43 may be present in either stereochemical orientation (or as a mixture of isomers).
  • C7 JQA-containing rapalogs may further vary from the corresponding C7-modified rapamycin at one, two, three, four, five or more other positions as well.
  • JQA-containing rapalogs of special interest in the practice of the various methods of the invention are those in which the substituents at C24 and C30 are both other than ( ⁇ O).
  • substituents at C24 and C30 are both other than ( ⁇ O).
  • C30 and C24 substituents disclosed in WO 99/36553 are those C30 and C24 substituents disclosed in WO 99/36553.
  • This subset includes among others all 43-JQA-containing rapalogs in which R C30 and R C24 are OH and one of R C7a and R C7b comprises any of the replacement substituents at that position specified herein, including any of the C7 substituents identified in WO 01/14387.
  • R C7a and R C7b are cyclic aliphatic, aryl, heterocyclic or heteroaryl, which may be optionally substituted.
  • Other compounds within this subset include those in which one, two, three, four or five of the hydroxyl groups is epimerized, fluorinated, alkylated, acylated or otherwise modified via other ester, carbamate, carbonate or urea formation.
  • An illustrative compound for example is the JQA-containing rapalog in which the hydroxyl groups at C28 and C30 are alkylated, acylated or linked via carbonate formation.
  • JQA-containing rapalogs of special interest are the mono- and difluoro-JQA-containing rapalogs which contain an F at one or both of C13 and C28, as disclosed in WO 99/36553, with or without additional changes elsewhere in the JQA-containing rapalog molecule.
  • Another subset of JQA-containing rapalogs of interest have an R C24 which is other than ⁇ O, again, with or without one or more other modifications at other positions relative to rapamycin.
  • JQA-containing rapalogs of interest include those in which R C14 is OH.
  • this invention encompasses JQA-containing rapalogs in which one or more of the carbon-carbon double bonds at the 1, 2, 3, 4 or 5, 6 positions in rapamycin are saturated, alone or in combination with a modification elsewhere in the molecule, e.g. at one or more of C7, C13, C24 C28 and/or C30.
  • the C3,C4 double bond may be epoxidized; that the C6 methyl group may be replaced with —CH 2 OH or —CH 2 OMe; that the C42 methoxy moiety may be demethylated, in any of the compounds disclosed herein, using methods known in the art.
  • rapamycin by fermentation and by total synthesis is known.
  • the production of a number of rapalogs as fermentation products is also known. These include among others rapalogs bearing alternative moieties in place of the characteristic cyclohexyl ring or pipecolate ring of rapamycin, as well as C7-desmethyl-rapamycin, C29-desmethyl-rapamycin and C29-desmethoxyrapamycin, among others.
  • rapamycin converted to 28-epirapamycin can be readily carried out as described e.g. in WO/014387. That document also identifies materials and methods for effecting many other known chemical transformations of rapamycin. See also references cited therein and U.S. 2001/0010920. 28-epi rapamycin may be used in place of rapamycin in the practice of the present invention to afford the corresponding 28-epi rapalog modified at position 43 in accordance with the present disclosure.
  • ketones at one or both of positions 24 and 30 may be effected using known methods, e.g. methods previously used with rapamycin itself, but now applied to the C-43 rapalogs disclosed herein to afford the corresponding 24-hydroxyl-, 30-hydroxyl- or 24, 30-tetrahydro-rapalogs modified at position 43 in accordance with the present disclosure.
  • rapalogs for use as intermediates in the production of 43-JQA-rapalogs may be prepared by directed biosynthesis, e.g. as described by Katz et al, WO 93/13663 and by Cane et al, WO 9702358. See also Khaw et al, 1998, J. Bacteriology 180 (4):809-814 for additional biological methods.
  • Rapalogs of this invention may be prepared by one of ordinary skill in this art relying upon methods and materials known in the art as guided by the disclosure presented herein. For instance, methods and materials may be adapted from known methods set forth or referenced in the documents cited above, the full contents of which are incorporated herein by reference. Additional guidance and examples are provided herein by way of illustration and further guidance to the practitioner. It should be understood that the chemist of ordinary skill in this art would be readily able to make modifications to the foregoing, e.g. to add appropriate protecting groups to sensitive moieties during synthesis, followed by removal of the protecting groups when no longer needed or desired, and would be readily capable of determining other synthetic approaches.
  • the synthesis of compounds of this invention often involves preparation of an activated form of the desired moiety “J”, such as a phosphoryl chloride as shown above (e.g. (R )(RO)P—Cl or RR′P( ⁇ O)—Cl, etc), and reaction of that reagent with rapamycin (or the appropriate rapalog) under conditions yielding the desired product, which may then be recovered from residual reactants and any undesired side products.
  • Protecting groups may be chosen, added and removed as appropriate using conventional methods and materials.
  • the identity, purity and chemical/physical properties of the rapalogs may be determined or confirmed using known methods and materials, including HPLC, mass spectral analysis, X ray crystallography and NMR spectroscopy. High resolution 1D 1 H and 31 P NMR spectra acquired using a typical relaxation delay of 3 seconds have proved useful, as has reverse phase HPLC analysis (analytical column, 3 micron particle size, 120 ansgstrom pore size, thermostatted to 50° C. with a mobile phase of 50% acetonitrile, 5% methanol and 45% water (all %s by volume), for example, in an isocratic elution system, with elution of product and impurity peaks followed by UV detection at 280 nanometers).
  • Normal phase HPLC may also be used, especially to evaluate the level of residual rapamycin or rapalog by-products. The presence of residual solvent, heavy metals, moisture and bioburden may be assessed using conventional methods.
  • the biological properties of the rapalogs may be determined using known methods and materials, including e.g. assays measuring binding to FKBP12, inhibition of T cell proliferation, anti-fungal activity, antitumor activity In vitro or in vivo (e.g. against one or more cancer cell lines in vitro and/or in vivo), immunosuppressive activity, and activity in a 3-hybrid assay based on FKBP- and FRAP-containing fusion proteins. Examples of many such assays are disclosed or referenced-in Sorbera et al, Drugs of the Future 2002, 27(1): 7-13, which is incorporated herein by reference.
  • Rapamycin is known to bind to the human protein, FKBP12 and to form a tripartite complex with hFKBP12 and FRAP, a human counterpart to the yeast proteins TOR1 and TOR2. Rapalogs may be characterized and compared to rapamycin with respect to their ability to bind to human FKBP12 and/or to form tripartite complexes with human FKBP12 and human FRAP (or fusion proteins or fragments containing its FRB domain). See WO 96/41865 (Clackson et al).
  • That application discloses various materials and methods which can be used to quantify the ability of a compound to bind to human FKBP12 or to form a tripartite complex with (i.e., “heterodimerize”) proteins comprising human FKBP12 and the FRB domain of human FRAP, respectively.
  • Such assays include fluorescence polarization assays to measure binding.
  • Other useful assays include cell based transcription assays in which the ability of a rapalog to form the tripartite complex is measured indirectly by correlation with the observed level of reporter gene product produced by engineered mammalian cells in the presence of the compound. Corresponding cell-based assays may also be conducted in engineered yeast cells. See e.g. WO 95/33052 (Berlin et al).
  • the rapalogs of this invention be physiologically acceptable (i.e., lack undue toxicity toward the cell or organism with which it is to be used), can be administered orally or parenterally to animals and/or can cross cellular and other membranes, as necessary for a particular application.
  • preferred rapalogs are those which bind to mutant or fungal binding proteins preferentially over the human counterpart binding proteins.
  • a non-limiting example of a mutant binding protein is a human FKBP in which Phe36 is replaced with a different amino acid, preferably an amino acid with a less bulky side chain such as valine or alanine).
  • such compounds may bind preferentially to mutant FKBPs at least an order of magnitude better than they bind to human FKBP12, and in some cases may bind to mutant FKBPs greater than 2 or even 3 or more orders of magnitude better than they do to human FKBP12, as determined by any scientifically valid or art-accepted assay methodology.
  • Binding affinities of various rapalogs of this invention with respect to human FKBP12, variants thereof or other immunophilin proteins may be determined by adaptation of known methods used in the case of FKBP. For instance, the practitioner may measure the ability of a compound of this invention to compete with the binding of a known ligand to the protein of interest. See e.g. Sierkierka et al, 1989, Nature 341, 755-757 (test compound competes with binding of labeled FK506 derivative to FKBP).
  • Certain rapalogs of this invention which are of particular interest bind to human FKBP12, to a mutant thereof as discussed above, or to a fusion protein containing such FKBP domains, with a Kd value below about 200 nM, more preferably below about 50 nM , even more preferably below about 10 nM, and even more preferably below about 1 nM, as measured by direct binding measurement (e.g. fluorescence quenching), competition binding measurement (e.g. versus FK506), inhibition of FKBP enzyme activity (rotamase), or other assay methodology.
  • direct binding measurement e.g. fluorescence quenching
  • competition binding measurement e.g. versus FK506
  • inhibition of FKBP enzyme activity rotamase
  • a known Competitive Binding FP Assay is described in detail in WO99/36553 and WO96/41865. That assay permits the in vitro measurement of an IC50 value for a given compound which reflects its ability to bind to an FKBP protein in competition with a labeled FKBP ligand, such as, for example, FK506.
  • One interesting class of compounds of this invention have an IC50 value in the Competitive Binding FP Assay (e.g., using a flouresceinated FK506 standard) of better than 1000 nM, preferably better than 300 nM, more preferably better than 100 nM, and even more preferably better than 10 nM with respect to a given FKBP domain and ligand pair, e.g. human FKBP12 or a variant thereof with up to 10 amino acid replacements, preferably 1-5 amino acid replacements.
  • IC50 value in the Competitive Binding FP Assay e.g., using a flouresceinated FK506 standard
  • 1000 nM preferably better than 300 nM, more preferably better than 100 nM, and even more preferably better than 10 nM with respect to a given FKBP domain and ligand pair, e.g. human FKBP12 or a variant thereof with up to 10 amino acid replacements, preferably 1-5 amino acid replacements.
  • the ability of the rapalogs to multimerize chimeric proteins may be measured in cell-based assays by measuring the occurrence of an event triggered by such multimerization. For instance, one may use cells containing and capable of expressing DNA encoding a first chimeric protein comprising one or more FKBP-domains and one or more effector domains as well as DNA encoding a second chimeric protein containing an FRB domain and one or more effector domains capable, upon multimerization, of actuating a biological response.
  • cells which further contain a reporter gene under the transcriptional control of a regulatory element (i.e., promoter) which is responsive to the multimerization of the chimeric proteins.
  • the reporter gene product may be a conveniently detectable protein (e.g. by ELISA) or may catalyze the production of a conveniently detectable product (e.g. colored).
  • ELISA ELISA-activated bacterium-catalyzes the production of a conveniently detectable product
  • Materials and methods for producing appropriate cell lines for conducting such assays are disclosed in the international patent applications cited above in this section.
  • target genes include by way of example SEAP, hGH, beta-galactosidase, Green Fluorescent Protein and luciferase, for which convenient assays are commercially available.
  • Conducting such assays permits the practitioner to select rapalogs possessing the desired IC50 values and/or binding characteristics.
  • the Competitive Binding FP Assay permits one to select rapalogs which possess the desired IC50 values and/or binding preference for a mutant FKBP or wild-type FKBP relative to a control, such as FK506.
  • the rapalogs of this invention can be used as described in WO94/18317, WO95/02684, WO96/20951, WO95/41865, WO99/36553 and WO/01/14387, e.g. to regulatably activate the transcription of a desired gene, delete a target gene, actuate apoptosis, or trigger other biological events in engineered cells growing in culture or in whole organisms, including in gene therapy applications. Additionally, certain compounds of this invention possess immunosuppressive and/or anti-cancer and/or antiinflammatory activity and/or anti-proliferative and/or antifungal activity, and/or inhibit thymocyte proliferation in vitro, as can be quantified and compared using conventional assay methods.
  • autoimmune diseases such as lupus, rheumatoid arthritis, diabetes mellitus and multiple sclerosis; fungal infection; inflammatory diseases (such as psoriasis, exzema, seborrhea, inflammatory bowel disease and pulmonary inflammation such as asthma, chronic obstructive disease, empheysema, bronchitis, etc.; hyperproliferative vascular disease (e.g., restenosis following the introduction of a vascular stent)(see e.g. Sousa et al, Circulation, 2001, 103:192-195); syndromes such as tuberous sclerosis (see e.g.
  • cancers e.g., tumors of the breast, prostate, ovarian, lung, pancreatic, colon, head and-neck, glioblastoma or other cancers of the brain, melanoma, and cervix
  • certain cancers e.g., tumors of the breast, prostate, ovarian, lung, pancreatic, colon, head and-neck, glioblastoma or other cancers of the brain, melanoma, and cervix
  • PTEN-deficient tumors see e.g.
  • Certain compounds of the invention will be of interest for their ability to inhibit osteoclast function, and may be useful in treating patients with debilitating bone disorders such as osteoporosis, particularly osteoporosis associated with the peri and post menopausal conditions.
  • osteoporotic diseases and related disorders including but not limited to involutional osteoporosis, Type I or postmenopausal osteoporosis, Type II or senile osteoporosis, juvenile osteoporosis, idiopathic osteoporosis, endocrine abnormality, hyperthyroidism, hypogonadism, ovarian agensis or Turner's syndrome, hyperadrenocorticism or Cushing's syndrome, hyperparathyroidism, bone marrow abnormalities, multiple myeloma and related disorders, systemic mastocytosis, disseminated carcinoma, Gaucher's disease, connective tissue abnormalities, osteogenesis imperfecta, homocystinuria, Ehlers-Danlos syndrome, Marfan's syndrome, Menke's syndrome, immobil
  • the ability to switch a therapeutic gene on and off at will or to titrate its expression level is important for therapeutic efficacy.
  • This invention is particularly well suited for achieving regulated expression of a therapeutic target gene in the context of human gene therapy.
  • One example uses a pair of fusion proteins (one containing at least one FKBP:rapamycin binding domain (an “FRB” domain) of the protein, FRAP, the other containing at least one FKBP domain), a rapalog of this invention capable of dimerizing the fusion proteins, and a target gene construct.
  • One of the fusion proteins comprises a DNA-binding domain, preferably a composite DNA-binding domain as described in Pomerantz et al, supra, as the heterologous effector domain.
  • the second fusion protein comprises a transcriptional activating domain as the heterologous effector domain.
  • the rapalog is capable of binding to both fusion proteins and thus of effectively cross-linking them.
  • DNA molecules encoding and capable of directing the expression of these chimeric proteins are introduced into the cells to be engineered. Also introduced into the cells is a target gene linked to a DNA sequence to which the DNA-binding domain is capable of binding.
  • the level of target gene expression should be a function of the number or concentration of chimeric transcription factor complexes, which should in turn be a function of the concentration of the rapalog. Dose (of rapalog)-responsive gene expression is typically observed.
  • the rapalog may be administered to the recpient as desired to activate transcription of the target gene. Depending upon the binding affinity of the rapalog, the response desired, the manner of administration, the biological half-life of the rapalog and/or target gene mRNA, the number of engineered cells present, various protocols may be employed.
  • the rapalog may be administered by various routes, including parenterally or orally. The number of administrations will depend upon the factors described above.
  • the rapalog may be administered orally as a pill, powder, or dispersion; buccally; sublingually; by inhalation; or by intravascular, intraperitoneal, intramuscular, subcutaneous or intra-articular injection.
  • rapalog and monomeric antagonist compound
  • the rapalog may be formulated using conventional methods and materials well known in the art for the various routes of administration.
  • the precise dose and particular method of administration will depend upon the above factors and be determined by the attending physician or human or animal healthcare provider. For the most part, the manner of administration will be determined empirically.
  • rapalog In the event that transcriptional activation by the rapalog is to be reversed or terminated, adminstration of the rapalog is terminated. Furthermore, if desired, a monomeric compound which can compete with the rapalog may be administered. Thus, in the case of an adverse reaction or the desire to terminate the therapeutic effect, an antagonist to the dimerizing agent can be administered in any convenient way, particularly intravascularly, if a rapid reversal is desired. Alternatively, one may provide for the presence of an inactivation domain (or transcriptional silencer) with a ligand binding domain.
  • cells are engineered to express a pair of chimeric proteins containing FRB and FKBP domains as discussed above, but containing a cellular signaling domain in place of a DNA binding domain or transcription activation domain.
  • signaling domains are known which upon their clustering or dimerization or oligomerization, trigger cell death, proliferation or idfferentiation.
  • This approach permits rapalog-mediated regulation of cell signaling (i.e., of cell death, proliferation or differentiation) in genetically engineered cells or organisms harboring them, as described elsewhere, which may be adapted for use of rapalogs of this invention. See International Patent Applications PCT/US94/01617 and PCT/US94/08008.
  • the particular dosage of the rapalog for any application may be determined in accordance with the procedures used for therapeutic dosage monitoring, where maintenance of a particular level of expression is desired over an extended period of times, for example, greater than about two weeks, or where there is repetitive therapy, with individual or repeated doses of rapalog over short periods of time, with extended intervals, for example, two weeks or more.
  • a dose of the rapalog within a predetermined range would be given and monitored for response, so as to obtain a time-expression level relationship, as well as observing therapeutic response. Depending on the levels observed during the time period and the therapeutic response, one could provide a larger or smaller dose the next time, following the response. This process would be iteratively repeated until one obtained a dosage within the therapeutic range.
  • the rapalog is chronically administered, once the maintenance dosage of the rapalog is determined, one could then do assays at extended intervals to be assured that the cellular system is providing the appropriate response and level of the expression product.
  • the system is subject to a number of variables, such as the cellular response to the rapalog, the efficiency of expression and, as appropriate, the level of secretion, the activity of the expression product, the particular need of the patient, which may vary with time and circumstances, the rate of loss of the cellular activity as a result of loss of cells or expression activity of individual cells, and the like.
  • a similar problem is encountered in the construction and use of “packaging lines” for the production of recombinant viruses for commercial (e.g., gene therapy) and experimental use. These cell lines are engineered to produce viral proteins required for the assembly of infectious viral particles harboring defective recombinant genomes. Viral vectors that are dependent on such packaging lines include retrovirus, adenovirus, and adeno-associated virus. In the latter case, the titer of the virus stock obtained from a packaging line is directly related to the level of production of the viral rep and cap proteins. But these proteins are highly toxic to the host cells. Therefore, it has proven difficult to generate high-titer recombinant AAV viruses.
  • This invention provides a solution to this problem, by allowing the construction of packaging lines in which the rep and core genes are placed under the control of regulatable transcription factors of the design described here.
  • the packaging cell line can be grown to high density, infected with helper virus, and transfected with the recombinant viral genome. Then, expression of the viral proteins encoded by the packaging cells is induced by the addition of dimerizing agent to allow the production of virus at high titer.
  • This invention is applicable to a wide range of biological experiments in which precise control over the expression of a target gene is desired. These include, among others: (1) expression of a protein or RNA of interest for biochemical purification; (2) regulated expression of a protein or RNA of interest in tissue culture cells (or in vivo, via engineered cells) for the purposes of evaluating its biological function; (3) regulated expression of a protein or RNA of interest in transgenic animals for the purposes of evaluating its biological function; (4) regulating the expression of a gene encoding another regulatory protein, ribozyme or antisense molecule that acts on an endogenous gene for the purposes of evaluating the biological function of that gene.
  • Transgenic animal models and other applications in which the components of this invention may be adapted include those disclosed in PCT/US95/10591.
  • kits useful for the foregoing applications contain DNA constructs encoding and capable of directing the expression of chimeric proteins of this invention (and may contain additional domains as discussed above) and, in embodiments involving regulated gene transcription, a target gene construct containing a target gene linked to one or more transcriptional control elements which are activated by the multimerization of the chimeric proteins.
  • the target gene construct may contain a cloning site for insertion of a desired target gene by the practitioner.
  • kits may also contain a sample of a dimerizing agent capable of dimerizing the two recombinant proteins and activating transcription of the target gene. Kits can also be provided for genetic engineering of cells or organisms to permit drug-regulated cellular signaling (e.g., leading to cell proliferation, differentiation or death) using rapalogs of this invention.
  • Compounds of this invention which were tested for activity against a variety of cancer cell lines were found to inhibit cancer cell growth and are therefore useful as antineoplastic agents.
  • the compounds of this invention may be used alone or in combination with other drugs and/or radiation therapy in treating or inhibiting the growth of various cancers, including leukemias and solid tumors, including sarcomas and carcinomas, such as astrocytomas, prostate cancer, breast cancer, small cell lung cancer, and ovarian cancer.
  • Their use is analogous to that of rapamycin or CCI779 as disclosed in Sorbera et al, “CCI-779” Drugs of the Future 2002, 27(1):7-13; WO 02/4000 and WO 02/13802, for example.
  • Examples of other drugs that can be used to treat cancer patients in conjunction with (i.e., before, during or after administration of a compound of this invention) a compound of this invention include, among others, Zyloprim, alemtuzmab, altretamine, amifostine, nastrozole, antibodies against prostate-specific membrane antigen (such as MLN-591, MLN591RL and MLN2704), arsenic trioxide, Avastin® (or other anti-VEGF antibody), bexarotene, bleomycin, busulfan, capecitabine, carboplatin, Gliadel Wafer, celecoxib, chlorambucil, cisplatin, cisplatin-epinephrine gel, cladribine, cytarabine liposomal, daunorubicin liposomal, daunorubicin, daunomycin, dexrazoxane, docetaxel, doxorubicin, Elliott'
  • the compounds of this invention may also be used for the treatment or inhibition of rejection of transplanted tissues including, e.g., kidney, heart, liver, lung, bone marrow, pancreas (islet cells), cornea, small bowel, and skin allografts, and heart valve xenografts; in the treatment or inhibition of graft vs.
  • transplanted tissues including, e.g., kidney, heart, liver, lung, bone marrow, pancreas (islet cells), cornea, small bowel, and skin allografts, and heart valve xenografts; in the treatment or inhibition of graft vs.
  • autoimmune diseases such as lupus, rheumatoid arthritis, diabetes mellitus, myasthenia gravis, and multiple sclerosis
  • diseases of inflammation such as psoriasis, dermatitis, eczema, seborrhea, inflammatory bowel disease, pulmonary inflammation (including asthma, chronic obstructive pulmonary disease, emphysema, acute respiratory distress syndrome, bronchitis, and the like) and ocular uveitis; adult T-cell leukemia/lymphoma; fungal infections; hyperproliferative vascular diseases including restenosis; graft vascular atherosclerosis; and cardiovascular disease, cerebral vascular disease, and peripheral vascular disease, such as coronary artery disease, cerebrovacular disease, arteriosclerosis, atherosclerosis, nonatheromatous arteriosclerosis, or vascular wall damage from cellular events leading toward immune mediated vascular damage, and inhibiting stroke or multiinfarct
  • a rapalog of this invention can be administered in conjunction with one or more other immunoregulatory agents.
  • immunoregulatory agents include, but are not limited to azathioprine, corticosteroids, such as prednisone and methylprednisolone, cyclophosphamide, rapamycin, cyclosporin A, FK-506, OKT-3, mycophenolate, and ATG.
  • Rapalogs of this invention may be used for treating cardiac inflammatory disease, e.g., by adaptation of the methods and materials disclosed in U.S. Pat. No. 5,496,832; for treating ocular inflammation, e.g., by adaptation of the methods and materials disclosed in U.S. Pat. No. 5,387,589; and for treating immunoinflammatory bowel disease and other immunoinflammatory diseases, e.g., by adaptation of the methods and materials disclosed in U.S. Pat. Nos. 5,286,731 and 5,286,730.
  • Rapalogs of this invention may also be used in the treatment or inhibition of vascular disease (including among others, coronary artery disease, cerebrovascular disease, arteriosclerosis, atherosclerosis, nonatheromatous arteriosclerosis, vascular wall damage from cellular events leading toward immune mediated vascular damage, stroke and multiinfarct dementia) to provide cardiovascular, cerebral, or peripheral vascular benefits as the sole active ingredient, or may be administered in combination with other agents which provide beneficial cardiovascular, cerebral, or peripheral vascular effects.
  • vascular disease including among others, coronary artery disease, cerebrovascular disease, arteriosclerosis, atherosclerosis, nonatheromatous arteriosclerosis, vascular wall damage from cellular events leading toward immune mediated vascular damage, stroke and multiinfarct dementia
  • Such agents include ACE inhibitors, such as quinapril, perindopril, ramipril, captopril, trandolapril, fosinopril, lisinopril, moexipril, and enalapril; angiotensin 11 receptor antagonists, such as candesartan, irbesartan, losartan, valsartan, and telmisartan; fibric acid derivatives, such as clofibrate, and gemfibrozil; HMG Co-A reductase inhibitors, such as cerivastatin, fluvastatin, atorvastatin, lovastatin, pravastatin, simvastatin; beta adrenergic blocking agents, such as sotalol, timolol, esmolol, carteolol, propranolol, betaxolol, penbutolol, nadolol, acebutolo
  • the compounds of this invention may also be used as neurotrophic agents, and may be particularly useful in promoting neuronal regeneration and functional recovery and in stimulating neurite outgrowth and thus treatment of various neuropathological states, including damage to peripheral nerves and the central nervous system caused by physical injury (e.g., spinal cord injury and trauma, sciatic or facial nerve lesion or injury), disease (e.g., diabetic neuropathy), cancer chemotherapy (e.g., by vinca alkaloids and doxorubicin), brain damage associated with stroke and ischemia associated with stroke, and neurological disorders including, but not limited to, various peripheral neuropathic and neurological disorders related to neurodegeneration including, but not limited to: trigeminal neuralgia, glossopharyngeal neuralgia, Bell's palsy, myasthenia gravis, muscular dystrophy, amyotrophic lateral sclerosis, progressive muscular atrophy, progressive bulbar inherited muscular atrophy, herniated, ruptured or prolapsed vertebral disk syndromes, cervical spondylos
  • a rapalog of this invention may be used by itself or in conjunction with mycophenolic acid, e.g. by adaptation of the methods and materials disclosed in U.S. Pat. No. 5,665,728, in treating or reducing the risk or severity of intimal smooth muscle cell hyperplasia, restenosis, and vascular occlusion in a mammal, particularly following either biologically or mechanically mediated vascular injury, or under conditions that would predispose a mammal to suffering such a vascular injury.
  • Biologically mediated vascular injury includes injury attributed to infectious disorders including endotoxins and herpes viruses such as cytomegalovirus; metabolic disorders such as atherosclerosis; and vascular injury resulting from hypothermia, and irradiation, among others.
  • Mechanically mediated vascular injury includes, among others, vascular injury caused by catheterization procedures or vascular scraping procedures such as percutaneous transluminal coronary angioplasty; vascular surgery; transplantation surgery; laser treatment; and other invasive procedures which disrupt the integrity of the vascular intima or endothelium.
  • a rapalog of this invention may be used in preventing restenosis following invasive procedures that disrupt the vascular endothelial lining, such as percutaneous transluminal coronary angioplasty, vascular catheterization, vascular scraping, vascular surgery, or laser treatment procedures.
  • a rapalog of this invention for treating or reducing the likelihood of restenosis such as occurs in a portion of patients following an angioplasty procedure.
  • the compounds of this invention can be administered prior to the angioplasty procedure, during the procedure, subsequent to the procedure, or any combination of the above.
  • a rapalog of this invention delivered on or in a stent (or other inserted or implanted device) in order to reduce the chances of restenosis following introduction of the device.
  • rapalog of this invention in conjunction with a stent may be achieved by adapting the methods and materials used for the delivery of other drugs, especially rapamycin, using such devises, e.g. as disclosed in U.S. Pat. Nos. 5,516,781; 6,153,252; 5,665,728; 5,646,160; 5,563,146; and 5,516,781 as well as in published international patent applications WO 01/01957, 01/49338, 01/87263, 01/87342, 01/87372, 01/87373, 01/87374, 01/87375, and 01/87376 and other patent documents relating to stents and other drug-bearing devices noted elsewhere herein.
  • Rapalogs of this invention are broadly compatible across the range of stent designs and of methods and materials for coating or otherwise loading them with drug. Loading such medical devices with a rapalog of this invention, medical devices loaded with a rapalog of this invention and the insertion of such a device loaded with such a rapalog are all encompassed by this invention.
  • a rapalog-bearing device of this invention comprises an expansible structure which is implantable within a body lumen and a means on or within the structure for releasing a compound of this invention, in some cases at a pre-selected rate.
  • Release of the compound may be at rates in a range from 5 ⁇ g/day to 200 ⁇ g/day, preferably in a range from 10 ⁇ g/day to 60 ⁇ g/day.
  • the total amount of compound released will in some cases be in a range from 100 ⁇ g to 10 mg, preferably in a range from 300 ⁇ g to 2 mg, more preferably in a range from 500 ⁇ g to 1.5 mg.
  • the expansible structure may be in the form of a stent, which additionally maintains luminal patency, or may be in the form of a graft, which additionally protects or enhances the strength of a luminal wall.
  • the expansible structure may be radially expansible and/or self-expanding and is preferably suitable for luminal placement in a body lumen.
  • the site of implantation may be any blood vessel in the patient's vasculature, including veins, arteries, aorta, and particularly including coronary and peripheral arteries, as well as previously implanted grafts, shunts, fistulas, and the like, or other body lumens, such as the biliary duct, or other organs, such as organs, nerves, glands, ducts, and the like.
  • An exemplary stent for use in the present invention is the commercially available Duraflex Coronary Stent (Avantec).
  • radially expansible it is meant that the device or segment thereof can be converted from a small diameter configuration to a radially expanded, usually cylindrical, configuration which is achieved when the device is implanted at a desired target site.
  • the device may be minimally resilient, e.g., malleable, thus requiring the application of an internal force to expand and set it at the target site.
  • the expansive force can be provided by a balloon, such as the balloon of an angioplasty catheter for vascular procedures.
  • the device may provide sigmoidal links between successive unit segments which are particularly useful to enhance flexibility and crimpability of the stent.
  • the device can be self-expanding.
  • Such self-expanding structures are provided by using a resilient material, such as a tempered stainless steel or a superelastic alloy such as a NitinolTM alloy, and forming the body segment so that it possesses its desired, radially-expanded diameter when it is unconstrained, i.e. released from the radially constraining forces of a sheath.
  • a resilient material such as a tempered stainless steel or a superelastic alloy such as a NitinolTM alloy
  • the dimensions of the device will depend on its intended use. Typically, the device will have a length in a range from about 5 mm to 100 mm, usually being from about 8 mm to 50 mm, for vascular applications.
  • the small (radially collapsed) diameter of cylindrical device will usually be in a range from about 0.5 mm to 10 mm, more usually being in a range from 0.8 mm to 8 mm for vascular applications.
  • the expanded diameter will usually be in a range from about 1.0 mm to 100 mm, preferably being in a range from about 2.0 mm to 30 mm for vascular applications.
  • the device will have a thickness in a range from 0.025 mm to 2.0 mm, preferably being in a range from 0.05 mm to 0.5 mm.
  • the segments of the device may be formed from conventional materials used for body lumen stents and grafts, typically being formed from malleable metals, such as 300 series stainless steel, or from resilient metals, such as superelastic and shape memory alloys, e.g., NitinolTM alloys, spring stainless steels, and the like. It is possible that the body segments could be formed from combinations of these metals, or combinations of these types of metals and other non-metallic materials. Additional structures for the body or unit segments of the present invention are illustrated in U.S. Pat. Nos. 5,195,417; 5,102,417; and 4,776,337, the full disclosures of which are incorporated herein by reference. Examplary stents and delayed or staged release of drug, including at preselected rates, are described in U.S. Pat. No. 6,471,980, the full disclosure of which is also incorporated herein by reference.
  • the means for releasing the compound comprises a matrix formed over at least a portion of the structure.
  • the matrix may be composed of a material which is degradable, partially degradable, nondegradable polymer, synthetic, or natural material.
  • the compound may be disposed within the matrix or adjacent to the matrix in a pattern that provides a desired release rate. Alternatively, compound may be disposed on or within the expansible structure adjacent to the matrix to provide the desired release rate.
  • Suitable biodegradable or bioerodible matrix materials include polyanhydrides, polyorthoesters, polycaprolactone, poly vinly acetate, polyhydroxybutyrate-polyhyroxyvalerate, polyglycolic acid, polyactic/polyglycolic acid copolymers and other aliphatic polyesters, among a wide variety of polymeric substrates employed for this purpose.
  • An often preferred biodegradable matrix material is a copolymer of poly-1-lactic acid and poly-e-caprolactone.
  • Suitable nondegradable matrix materials include polyurethane, polyethylene imine, cellulose acetate butyrate, ethylene vinyl alcohol copolymer, or the like.
  • the polymer matrix may degrade by bulk degradation (i.e., in which the matrix degrades throughout) or by surface degradation (i.e., in which a surface of the matrix degrades over time while maintaining bulk integrity).
  • Hydrophobic matrices are sometimes preferred as they tend to release compound at a predetermined release rate.
  • a nondegradable matrix may be used to release the substance by diffusion.
  • the matrix may comprise multiple adjacent layers of same or different matrix material, wherein at least one layer contains compound and another layer contains compound, at least one substance other than compound, or no substance.
  • compound disposed within a top degradable layer of the matrix is released as the top matrix layer degrades and a second substance disposed within an adjacent nondegradable matrix layer is released primarily by diffusion.
  • multiple substances may be disposed within a single matrix layer.
  • the optional substance used in addition to the compound of this invention may, for example, be selected form those listed in U.S. Pat. No. 6,471,980.
  • a rate limiting barrier may be formed adjacent to the structure and/or the matrix.
  • Such rate limiting barriers may be nonerodible or nondegradable, such as silicone, polytetrafluorethylene (PTFE), parylene, and PARYLASTTM, and control the flow rate of release passing through the rate limiting barrier.
  • the compound may be released by diffusion through the rate limiting barrier.
  • a biocompatible or blood compatible layer such as polyethylene glycol (PEG), may be formed over the matrix or rate limiting barrier to make the delivery prosthesis more biocompatible.
  • the means for releasing the compound may comprise a rate limiting barrier formed over at least a portion of the structure.
  • Compound may be disposed within the barrier or adjacent to the barrier.
  • the rate limiting barrier may have a sufficient thickness so as to provide the desired release rate of compound.
  • Rate limiting barriers will typically have a total thickness in a range from 0.01 micron to 100 microns, preferably in a range from 0.1 micron to 10 microns, to provide compound release at the desired release rate.
  • the rate limiting barrier is typically nonerodible such as silicone, PTFE, PARYLASTTM, polyurethane, parylene, or a combination thereof and compound release through such rate limiting barriers is usually accomplished by diffusion.
  • the rate limiting barrier may comprise multiple adjacent layers of same or different barrier material, wherein at least one layer contains compound and another layer contains compound, at least one substance other than compound, or no substance. Multiple substances may also be contained within a single barrier layer.
  • the means for releasing the substance comprises a reservoir on or within the structure containing compound and a cover over the reservoir.
  • the cover may be degradable or partially degradable over a preselected time period so as to provide the desired compound release rate.
  • the cover may comprise a polymer matrix, as described above, which contains compound within the reservoir.
  • a rate limiting barrier, such as silicone, may additionally be formed adjacent to the reservoir and/or the cover, thus allowing compound to be released by diffusion through the rate limiting barrier.
  • the cover may be a nondegradable matrix or a rate limiting barrier.
  • Another device of this invention comprises an expansible structure which is implantable within a body lumen with a rate limiting barrier on the structure permitting release of compound at a pre-selected rate, e.g., a rate selected to inhibit smooth muscle cell proliferation.
  • the barrier comprises multiple layers, wherein each layer comprises PARYLASTTM or parylene and has a thickness in a range of 50 nm-10 microns. At least one layer contains compound, and another layer contains compound, at least one substance other than compound, or no substance.
  • vascular prosthesis which comprises an expansible structure, a source of compound on or within the structure, and optionally a source of at least one other substance in addition to compound on or within the structure.
  • the compound is released from the source when the expansible structure is implanted in a blood vessel.
  • the optional additional substance is also released from its source when the expansible structure is implanted in a blood vessel.
  • Each source may comprise a matrix, rate limiting membrane, reservoir, or other rate controlling means as described herein.
  • the optional additional substance include an immunosuppressive substance (e.g., mycophenolic acid or an analog thereof, anti-platelet agent, anti-thrombotic agent, or IIb/IIIa agent such as disclosed in U.S. Pat. No. 6,471,980.
  • This invention thus provides methods for inhibiting restenosis in a blood vessel following recanalization of the blood vessel.
  • one method includes implanting a vascular prosthesis, bearing a compound of this invention, in the body lumen such as a coronary or peripheral blood vessel, for instance, to prevent reclosure of the vessel.
  • the compound is then released, in some cases at a rate selected to inhibit smooth muscle cell proliferation.
  • the release of the compound may occur immediately upon introduction or expansion of the stent, or the release may be delayed.
  • substantial release of compound may be delayed for at least one hour following implantation of the prosthesis.
  • the delayed release from its reservoir may be provided using with a material that at least partially degrades in a vascular environment over that one hour.
  • release may be slowed by the use of a matrix that at least partially degrades in a vascular environment over that one hour.
  • release may be slowed with a nondegradable matrix or rate limiting barrier that allows diffusion of compound through said nondegradable matrix or barrier after that hour.
  • the release of compound will typically be at rates in a range from 5 ⁇ g/day to 200 ⁇ g/day, preferably in a range from 10 ⁇ g/day to 60 ⁇ g/day.
  • compound is released within a time period of 1 day to 45 days in a vascular environment, preferably in a time period of 7 day to 21 days in a vascular environment.
  • the device may be coated with a matrix or barrier by spraying, dipping, deposition, or painting. Such coatings may be non-uniform. For example, the coating may be applied to only one side of the prosthesis or the coating may be thicker on one side. Likewise, the device may also incorporate the compound by virtue of coating, spraying, dipping, deposition, chemical bonding, or painting compound on all or partial surfaces of the device.
  • the compound may be released within a time period of 1 day to 45 days and the additional substance or substances may be released within a time period of 2 days to 3 months.
  • release of the compound and the additional substance(s) may be simultaneous or sequential.
  • the total amount of the compound released depends in part on the level and amount of vessel injury.
  • the release rate will be in a range from 100 ⁇ g to 10 mg, preferably in a range from 300 ⁇ g to 2 mg, more preferably in a range from 500 ⁇ g to 1.5 mg. It is often preferred that the release rate during the initial phase be from 0 ⁇ g/day to 50 ⁇ g/day, usually from 5 ⁇ g/day to 30 ⁇ g/day.
  • the compound release rate during the subsequent phase will be much higher, typically being in the range from 5 ⁇ g/day to 200 ⁇ g/day, usually from 10 ⁇ g/day to 100 ⁇ g/day.
  • the initial release rate will typically be from 0% to 99% of the subsequent release rates, usually from 0% to 90%, preferably from 0% to 75%.
  • a mammalian tissue concentration of the compound at an initial phase will typically be within a range from 0 ⁇ g/mg of tissue to 100 ⁇ g/mg of tissue, preferably from 0 ⁇ g/mg of tissue to 10 ⁇ g/mg of tissue.
  • a mammalian tissue concentration of the substance at a subsequent phase will typically be within a range from 1 picogram/mg of tissue to 100 ⁇ g/mg of tissue, preferably from 1 nanogram/mg of tissue to 10 ⁇ g/mg of tissue.
  • the duration of the initial, subsequent, and any other additional phases may vary.
  • the initial phase will be sufficiently long to allow initial cellularization or endothelialization of at least part of the stent, usually being less than 12 weeks, more usually from 1 hour to 8 weeks, more preferably from 12 hours to 2 weeks, most preferably from 1 day to 1 week.
  • the durations of the subsequent phases may also vary, typically being from 4 hours to 24 weeks, more usually from 1 day to 12 weeks, more preferably in a time period of 2 days to 8 weeks in a vascular environment, most preferably in a time period of 3 days to 50 days in a vascular environment.
  • the release profile of the compound over a predetermined time may allow for a higher release rate during an initial phase, typically from 40 ⁇ g/day to 300 ⁇ g/day, usually from 40 ⁇ g/day to 200 ⁇ g/day.
  • the compound release during the subsequent phase will be much lower, typically being in the range from 1 ⁇ g/day to 100 ⁇ g/day, usually from 10 ⁇ g/day to 40 ⁇ g/day.
  • the duration of the initial phase period for the higher release rate will be in a range from 1 day to 7 days, with the subsequent phase period for the lower release rate being in a range from 2 days to 45 days.
  • a mammalian tissue concentration of the substance at the initial phase of 1-7 days will typically be within a range from 10 nanogram/mg of tissue to 100 ⁇ g/mg of tissue.
  • a mammalian tissue concentration of the substance at the subsequent phase of 2-45 days will typically be within a range from 0.1 nanogram/mg of tissue to 10 ⁇ g/mg of tissue.
  • the release of the compound may be constant at a rate between 5 ⁇ g/day to 200 ⁇ g/day for a duration of time in the range from 1 day to 45 days.
  • a mammalian tissue concentration over this period of 1-45 days will typically be within a range from 1 nanogram/mg of tissue to 10 ⁇ g/mg of tissue.
  • the means for releasing the compound comprises a matrix or coat formed over at least a portion of the device, wherein the matrix is composed of material which undergoes degradation.
  • the compound may be disposed within the matrix in a pattern that provides the desired release rates.
  • the compound may be disposed within or on the device under the matrix to provide the desired release rates.
  • the device acts as a mechanical support for the delivery matrix, thus allowing a wide variety of materials to be utilized as the delivery matrix.
  • Suitable biodegradable or bioerodible matrix materials include polyanhydrides, polyorthoesters, polycaprolactone, poly vinyl acetate, polyhydroxybutyrate-polyhyroxyvalerate, polyglycolic acid, polyactic/polyglycolic acid copolymers and other aliphatic polyesters, among a wide variety of synthetic or natural polymeric substrates employed for this purpose.
  • An example of a biodegradable matrix material useful in practicing the present invention is a copolymer of poly-1-lactic acid (having an average molecular weight of about 200,000 daltons) and poly-e-caprolactone (having an average molecular weight of about 30,000 daltons).
  • Poly-e-caprolactone (PCL) is a semi crystalline polymer with a melting point in a range from 59° C. to 64° C. and a degradation time of about 2 years.
  • PCL poly-1-lactic acid
  • a preferred ratio of PLLA to PCL is 75:25 (PLLA/PCL).
  • a 75:25 PLLA/PCL copolymer blend exhibits sufficient strength and tensile properties to allow for easier coating of the PLLA/PLA matrix on the scaffold. Additionally, a 75:25 PLLA/PCL copolymer matrix allows for controlled drug delivery over a predetermined time period as a lower PCL content makes the copolymer blend less hydrophobic while a higher PLLA content leads to reduced bulk porosity.
  • the polymer matrix may degrade by bulk degradation, in which the matrix degrades throughout, or preferably by surface degradation, in which only a surface of the matrix degrades over time while maintaining bulk integrity.
  • the matrix may be composed of a nondegradable material which releases the compound by diffusion.
  • Suitable nondegradable matrix materials include polyurethane, polyethylene imine, cellulose acetate butyrate, ethylene vinyl alcohol copolymer, or the like.
  • the matrix may comprise multiple layers, each layer containing the compound, a different substance, or no substance.
  • a top layer may contain no substance while a bottom layer contains the compound.
  • the present invention may employ a rate limiting barrier formed between the device and the matrix, or may optionally be formed over the matrix.
  • rate limiting barriers may be nonerodible and control the flow rate of release by diffusion of the compound through the barrier.
  • Suitable nonerodible rate limiting barriers include silicone, PTFE, PARYLASTTM, and the like.
  • a layer such as polyethylene glycol (PEG), and the like, may be formed over the matrix to make the delivery device more biocompatible.
  • the means for releasing the compound comprises a reservoir on or within the device containing the compound and a cover over the reservoir.
  • the cover is degradable over a preselected time period so that release of the compound from the reservoir begins substantially after the preselected time period.
  • the cover in this example, may comprise a polymer matrix, as described above, which contains the compound within the reservoir so that the matrix is replenished by the compound within the reservoir.
  • a rate limiting barrier may additionally be formed between the reservoir and the cover, or on top of the cover, thus allowing the compound to be released by diffusion through the rate limiting barrier.
  • methods for the compound delivery comprise providing a luminal prosthesis incorporating or bearing the compound.
  • the prosthesis is coated with a matrix which undergoes degradation in a vascular environment.
  • the prosthesis is implanted in a body lumen so that at least a portion of the matrix degrades over a predetermined time period and substantial the compound release begins after the portion has degraded.
  • the prosthesis may be coated with a rate limiting barrier or nondegradable matrix having a sufficient thickness to allow diffusion of the compound through the barrier or nondegradable matrix.
  • the prosthesis is implanted in a body lumen so that substantial release of compound from the barrier or nondegradable matrix begins, preferably after a preselected time period.
  • substantial release of the compound in some embodiments will begin within a time period of 4 hours to 24 weeks in a vascular environment, preferably in a time period of 1 day to 12 weeks in a vascular environment, more preferably in a time period of 2 days to 8 weeks in a vascular environment, most preferably in a time period of 3 days to 50 days in a vascular environment.
  • the compound may be incorporated in a reservoir in or on the device.
  • the reservoir is covered by the matrix so that the compound release begins substantially after the matrix has degraded sufficiently to uncover the reservoir.
  • the compound may be disposed in the matrix with the matrix coating a device.
  • an outer layer of the matrix is substantially free from the compound so that the compound release will not substantially begin until the outer layer has degraded.
  • the compound may be disposed within or on a portion of the device coated by the matrix.
  • the prosthesis may incorporate the compound by coating, spraying, dipping, deposition, or painting the compound on the prosthesis.
  • a solvent e.g., water with pH buffers, pH adjusters, organic salts, and inorganic salts
  • alcohols e.g., methanol, ethanol, propanol, isopropanol, hexanol, and glycols
  • nitrites e.g., acetonitrile, benzonitrile, and butyronitrile
  • amides e.g., formamide and N dimethylformamide
  • ketones esters, ethers, DMSO, gases (e.g., carbon dioxide), and the like.
  • the prosthesis may be sprayed with or dipped in the solution and dried so that the compound crystals are left on a surface of the prosthesis.
  • the prosthesis may be coated with the matrix solution by spraying, dipping, deposition, or painting the polymer solution onto the prosthesis.
  • the polymer is finely sprayed on the prosthesis while the prosthesis is rotating on a mandrel.
  • a thickness of the matrix coating may be controlled by a time period of spraying and a speed of rotation of the mandrel.
  • the thickness of the matrix coating is typically in a range from 0.01 micron to 100 microns, preferably in a range from 0.1 micron to 10 microns.
  • a stainless steel DuraflexTM stent having dimensions of 3.0 mm — 14 mm is sprayed with a solution of 25 mg/ml the compound in a 100% ethanol, methanol, acetone, ethyl acetate, methylene chloride or other solvent.
  • the stent is dried and the solvent is evaporated leaving the compound on a stent surface.
  • a 75:25 PLLA/PCL copolymer (sold commercially by Polysciences) is prepared in 1,4 Dioxane (sold commercially by Aldrich Chemicals).
  • the compound loaded stent is loaded on a mandrel rotating at 200 rpm and a spray gun (sold commercially by Binks Manufacturing) dispenses the copolymer solution in a fine spray on to the compound loaded stent as it rotates for a 10-30 second period.
  • the stent is then placed in a oven at 25-35° C. up to 24 hours to complete evaporation of the solvent.
  • stents bearing a rapalog of this invention may be prepared and used by analogy to the methods of Sousa et al, Circulation, 2001; 103:192; Sousa et al, Circulation, 2001; 104:2007; and Morice et al. N Engl J Med 2002;346(23):1773-1780, but substituting the rapalog for Sirolimus.
  • patients with native coronary artery disease and angina pectoris may be treated with the implantation of a single rapalog-eluting Bx VELOCITY stent.
  • the rapalog is blended in a mixture of nonerodable polymers (see e.g., Kindt-Larsen et al, J Appl Biomater. 1995;6:75-83; Revell et al, Biomaterials. 1998;19:1579-1586), and a 5 - ⁇ m-thick layer of rapalog-polymer matrix is applied onto the surface of the Bx VELOCITY stent (Cordis), a laser-cut 316L stainless-steel balloon-expandable stent. This is referred to as the fast-release [FR] formulation.
  • FR fast-release
  • the drug will be almost completely eluted by 15 days after implantation in the FR formulation.
  • SR slow-release
  • another layer of drug-free polymer is applied on top of the drug-polymer matrix to introduce a diffusion barrier and prolong drug release to >28 days.
  • About 80% of the rapalog should be released from the SR formulation within about 30 days.
  • the stents regardless of the coating composition, are loaded with a fixed amount of drug per unit of metal surface area (140 ⁇ g drug/cm 2 ).
  • the rapalog-bearing Bx VELOCITY stents are implanted according to standard practice, after balloon predilatation and followed by high-pressure (>12 atmospheres) balloon after dilatation.
  • the stents in this embodiment are 18 mm long and 3 to 3.5 mm in diameter.
  • Heparin may be given to maintain the activated clotting time >300 seconds.
  • Patients may also receive aspirin (325 mg/d, indefinitely) starting at least 12 hours before the procedure and a 300 -mg loading dose of clopidogrel immediately after stent implantation and 75 mg/d for 60 days.
  • the means for releasing the compound may comprise a reservoir on or within the scaffold holding the compound and an external energy source for directing energy at the prosthesis after implantation to effect release of the compound.
  • a matrix may be formed over the reservoir to contain the compound within the reservoir.
  • the means for releasing the compound may comprise a matrix formed over at least a portion of the scaffold, wherein the compound is disposed under or within the matrix, and an external energy source for directing energy at the prosthesis after implantation to effect release of the compound.
  • Suitable external energy source include ultrasound, magnetic resonance imaging, magnetic field, radio frequency, temperature change, electromagnetic, x-ray, radiation, heat, gamma, and microwave.
  • an ultrasound external energy source may be used having a frequency in a range from 20 kHz to 100 MHz, preferably in a range from 0.1 MH z to 20 MHz, and an intensity level in a range from 0.05 W/cm 2to 10 W/cm 2, preferably in a range from 0.5 W/cm 2to 5 W/cm 2.
  • the ultrasound energy should be directed at the prosthesis from a distance in a range from 1 mm to 30 cm, preferably in a range from 1 cm to 20 cm.
  • the ultrasound may be continuously applied or pulsed, for a time period in a range from 5 sec to 30 minutes, preferably in a range from 1 minute to 15 minutes.
  • the temperature of the delivery prosthesis during this period will be in a range from 37° C. to 48° C.
  • the ultrasound may be used to increase a porosity of the prosthesis, thereby allowing release of the compound from the prosthesis.
  • means for releasing the compound comprises magnetic particles coupled to the compound and a magnetic source for directing a magnetic field at the prosthesis after implantation to effect release of the compound.
  • the means for releasing the compound may comprise magnetic particles coupled to a matrix formed over the device and a magnetic source for directing a magnetic field at the prosthesis after implantation to effect release of the compound.
  • the compound may be disposed under or within the matrix.
  • the magnetic particles may be formed from magnetic beads and will typically have a size in a range from 1 nm to 100 nm.
  • the magnetic source exposes the prosthesis to its magnetic field at an intensity typically in the range from 0.01 T to 2 T, which will activate the magnetic particles, and thereby effect release of the compound from the prosthesis.
  • This invention thus includes methods for delivering one of the described compounds to an artery.
  • the method is of the type where a prosthesis is implanted in the artery and the prosthesis releases the compound.
  • the method involves implanting a prosthesis that is programmed to begin substantial release of the compound beginning preferably after growth of at least one layer of cells over a part of the prosthesis.
  • the cells will typically comprise inflammation, smooth muscle, or endothelial cells, indicating the onset of restenosis.
  • a method for positioning the delivery prosthesis in a body lumen in order to deliver the compound therein is also provided.
  • a balloon dilation catheter will typically be used to deliver the prosthesis to a region of stenosis in a blood vessel.
  • the prosthesis is initially carried in its radially collapsed diameter configuration on an deflated balloon of the balloon catheter.
  • the balloon catheter is typically introduced over a guidewire under fluoroscopic guidance.
  • the catheters and guidewires may be introduced through conventional access sites to the vascular system, such as through the femoral artery, or brachial, subclavian or radial arteries, for access to the coronary arteries.
  • the balloon is inflated to radially expand the prosthesis within the stenotic region.
  • the balloon may then be deflated, and the catheter withdrawn over the guidewire. After removal of the guidewire, the expanded prosthesis is left in place to provide luminal delivery of the compound as described above to inhibit restenotic effects.
  • a prosthesis having reservoir means for releasing the compound may further incorporate a rate limiting barrier.
  • methods of the present invention may combine balloon angioplasty and/or other interventional treatments to resolve a stenotic site with the presently described luminal the compound delivery treatments.
  • rapalogs of this invention can exist in free form or, where appropriate, in salt form.
  • Pharmaceutically acceptable salts of many types of compounds and their preparation are well-known to those of skill in the art.
  • Pharmaceutically acceptable salts include conventional non-toxic salts including the quaternary ammonium salts of formed by such compounds with inorganic or organic acids of bases.
  • the compounds of the invention may form hydrates or solvates. It is known to those of skill in the art that charged compounds form hydrated species when lyophilized with water, or form solvated species when concentrated in a solution with an appropriate organic solvent.
  • compositions comprising a therapeutically (or prophylactically) effective amount of a compound of the invention, and one or more pharmaceutically acceptable carriers and/or other excipients.
  • Carriers include e.g. saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof, and are discussed in greater detail below.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • the composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Formulation may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation. In another approach, the composition may be formulated into nanoparticles.
  • the pharmaceutical carrier employed may be, for example, either a solid or liquid.
  • Illustrative solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • a solid carrier can include one or more substances which may also act as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aids, binders or tablet-disintegrating agents; it can also be an encapsulating material.
  • the carrier is a finely divided solid which is in admixture with the finely divided active ingredient.
  • the active ingredient is mixed with a carrier having the necessary compression properties in suitable proportions, and compacted in the shape and size desired.
  • the powders and tablets preferably contain up to 99% of the active ingredient.
  • suitable solid carriers include, for example, calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, sodium carboxymethyl cellulose, polyvinylpyrrolidine, low melting waxes and ion exchange resins.
  • Illustrative liquid carriers include syrup, peanut oil, olive oil, water, etc. Liquid carriers are used in preparing solutions, suspensions, emulsions, syrups, elixirs and pressurized compositions.
  • the active ingredient can be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, a mixture of both or pharmaceutically acceptable oils or fats.
  • the liquid carrier can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers or osmo-regulators.
  • liquid carriers for oral and parenteral administration include water (partially containing additives as above, e.g. cellulose derivatives, preferably sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g. glycols) and their derivatives, and oils (e.g. fractionated coconut oil and arachis oil).
  • the carrier can also be an oily ester such as ethyl oleate and isopropyl myristate.
  • Sterile liquid carriers are useful in sterile liquid form compositions for parenteral administration.
  • the liquid carrier for pressurized compositions can be halogenated hydrocarbon or other pharmaceutically acceptable propellant.
  • Liquid pharmaceutical compositions which are sterile solutions or suspensions can be administered by, for example, intravenous, intramuscular, intraperitoneal or subcutaneous injection. Injection may be via a single push or by gradual infusion, e.g. 30 minute intravenous infusion.
  • the compound can also be administered orally either in liquid or solid composition form.
  • the carrier or excipient may include a time delay material, examples of which are well known to the art, such as glyceryl monostearate or glyceryl distearate, and may further include a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate and the like.
  • a time delay material examples of which are well known to the art, such as glyceryl monostearate or glyceryl distearate, and may further include a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate and the like.
  • Tween 80 in PHOSAL PG-50 phospholipid concentrate with 1,2-propylene glycol, A. Nattermann & Cie. GmbH
  • PHOSAL PG-50 phospholipid concentrate with 1,2-propylene glycol, A. Nattermann & Cie. GmbH
  • a wide variety of pharmaceutical forms can thus be employed in administering compounds of this invention.
  • a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge.
  • the amount of solid carrier will vary widely but preferably will be from about 25 mg to about 1 g.
  • a liquid carrier is used, the preparation will be in the form of a syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampule or vial or nonaqueous liquid suspension.
  • the compound, or a pharmaceutically acceptable salt thereof may be dissolved in an aqueous solution of an organic or inorganic acid, such as a 0.3M solution of succinic acid or citric acid.
  • an organic or inorganic acid such as a 0.3M solution of succinic acid or citric acid.
  • acidic derivatives can be dissolved in suitable basic solutions. If a soluble form is not available, the compound is dissolved in a suitable cosolvent or combinations thereof.
  • suitable cosolvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin, polypxyethylated fatty acids, fatty alcohols or glycerin hydroxy fatty acids esters and the like in concentrations ranging from 0-60% of the total volume.
  • Various delivery systems are known and can be used to administer the compound, or the various formulations thereof, including tablets, capsules, injectable solutions, encapsulation in liposomes, microparticles, microcapsules, etc.
  • Methods of introduction include but are not limited to dermal, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, pulmonary, epidural, ocular and (as is usually preferred) oral routes.
  • the compound may be administered by any convenient or otherwise appropriate route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) or via a drug-loaded stent and may be administered together with other biologically active agents. Administration can be systemic or local.
  • preferred routes of administration are oral, nasal or via a bronchial aerosol or nebulizer.
  • the composition is formulated using routine methods as a pharmaceutical composition for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • a solution of a rapalog of this invention for injection may contain 0.1 to 10 mg/ml, e.g. 1-3 mg/ml, of rapalog in a diluant solution containing Phosal 50 PG (phosphatidylcholine, propylene glycol, mono- and di-glycerides, ethanol, soy fatty acids and ascorbyl palmitate) and polysorbate 80, containing 0.5-4% ethanol, e.g. 1.5% -2.5% ethanol.
  • the diluant may contain 2-8%, e.g. 5-6%, each of propylene glycol USP and polysorbate 80 in water for injection. We have found that 5.2% of each works well in some cases.
  • a solution is processed using conventional methods and materials, including e.g. one or more rounds of sterile filteration.
  • Oral formulations containing a compound of this invention may comprise any conventionally used oral forms, including tablets, capsules, buccal forms, troches, lozenges and oral liquids, suspensions or solutions.
  • Capsules may contain mixtures of the active compound(s) with inert fillers and/or diluents such as the pharmaceutically acceptable starches (e.g. corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses, such as crystalline and microcrystalline celluloses, flours, gelatins, gums, etc.
  • Useful tablet formulations may be made by conventional compression, wet granulation or dry granulation methods and utilize pharmaceutically acceptable diluents, binding agents, lubricants, disintegrants, surface modifying agents (including surfactants), suspending or stabilizing agents, including, but not limited to, magnesium stearate, stearic acid, talc, sodium lauryl sulfate, microcrystalline cellulose, carboxymethylcellulose calcium, polyvinylpyrrolidone, gelatin, alginic acid, acacia gum, xanthan gum, sodium citrate, complex silicates, calcium carbonate, glycine, dextrin, sucrose, sorbitol, dicalcium phosphate, calcium sulfate, lactose, kaolin, mannitol, sodium chloride, talc, dry starches and powdered sugar.
  • pharmaceutically acceptable diluents including, but not limited to, magnesium stearate, stearic acid, talc, sodium lau
  • Suitable surface modifying agents include nonionic and anionic surface modifying agents.
  • Representative examples of surface modifying agents include, but are not limited to, poloxamer 188, benzalkonium chloride, calcium stearate, cetostearl alcohol, cetomacrogol emulsifying wax, sorbitan esters, colloidol silicon dioxide, phosphates, sodium dodecylsulfate, magnesium aluminum silicate, and triethanolamine.
  • Oral formulations herein may utilize standard delay or time release formulations to alter the absorption of the active compound(s).
  • the oral formulation may also consist of administering the active ingredient in water or a fruit juice, containing appropriate solubilizers or emulsifiers as needed.
  • Tablets containing a rapalog of this invention may contain conventional inactive ingredients including for example sucrose, lactose, polyethylene glycol 8000, calcium sulfate, microcrystalline cellulose, pharmaceutical grade glaze, talc, titanium dioxide, magnesium stearate, povidone, poloxamer 188, polyethylene glycol 20,000, glyceryl monooleate, carnauba wax, and other ingredients. Nanosized compositions for oral administration may also be used.
  • nanoparticles are formed from compositions containing (on a weight/weight basis) 1-20% rapalog, 70-95% inert material such as sucrose, 0.1 to 4% of materials such as polyvinyl pyrrolidone and benzylconium chloride and 0-1% surfactant such as Tween.
  • An illustrative such composition contains about 15% rapalog, 81% sucrose, 2% polyvinyl pyrrolidone, 2% benzylconium chloride and 0.1% Tween.
  • Administration to an individual of an effective amount of the compound can also be accomplished topically by administering the compound(s) directly to the affected area of the skin of the individual.
  • the compound is administered or applied in a composition including a pharmacologically acceptable topical carrier, such as a gel, an ointment, a lotion, or a cream, which includes, without limitation, such carriers as water, glycerol, alcohol, propylene glycol, fatty alcohols, triglycerides, fatty acid esters, or mineral oils.
  • Topical carriers include liquid petroleum, isopropyl palmitate, polyethylene glycol, ethanol (95%), polyoxyethylene monolaurate (5%) in water, or sodium lauryl sulfate (5%) in water.
  • Other materials such as anti-oxidants, humectants, viscosity stabilizers, and similar agents may be added as necessary.
  • Percutaneous penetration enhancers such as Azone may also be included.
  • transdermal administrations are understood to include all administrations across the surface of the body and the inner linings of bodily passages including epithelial and mucosal tissues. Such administrations may be carried out using the present compounds, or pharmaceutically acceptable salts thereof, in lotions, creams, foams, patches, suspensions, solutions, and suppositories (rectal and vaginal).
  • Transdermal administration may be accomplished through the use of a transdermal patch containing the active compound and a carrier that is inert to the active compound, is non toxic to the skin, and allows delivery of the agent for systemic absorption into the blood stream via the skin.
  • the carrier may take any number of forms such as creams and ointments, pastes, gels, and occlusive devices.
  • the creams and ointments may be viscous liquid or semisolid emulsions of either the oil-in-water or water-in-oil type. Pastes comprised of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the active ingredient may also be suitable.
  • occlusive devices may be used to release the active ingredient into the blood stream such as a semi-permeable membrane covering a reservoir containing the active ingredient with or without a carrier, or a matrix containing the active ingredient.
  • Other occlusive devices are known in the literature.
  • Suppository formulations may be made from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerin.
  • Water soluble suppository bases such as polyethylene glycols of various molecular weights, may also be used.
  • the effective systemic dose of the compound will typically be in the range of about 0.01 to about 100 mg/kgs, preferably about 0.1 to about 10 mg/kg of mammalian body weight, administered in single or multiple doses.
  • the compound may be administered to patients in need of such treatment in a daily dose range of about 1 to about 2000 mg per patient. Administration may be once or multiple times daily, weekly (or at some other multiple-day interval) or on an intermittent schedule. For example, the compound may be administered one or more times per day on a weekly basis (e.g. every Monday) for a period of weeks, e.g. 4-10 weeks. Alternatively, it may be administered daily for a period of days (e.g. 2-10 days) followed by a period of days (e.g.
  • an anti-cancer compound of the invention may be administered daily for 5 days, then discontinued for 9 days, then administered daily for another 5 day period, then discontinued for 9 days, and so on, repeating the cycle a total of 4-10 times.
  • the amount of compound which will be effective in the treatment or prevention of a particular disorder or condition will depend in part on well known factors affecting drug dosage, and in the case of gene and cell therapy applications, will also depend on the characteristics of the fusion proteins to be multimerized, the characteristics and location of the genetically engineered cells, and on the nature of the disorder or condition, which can be determined by standard clinical techniques.
  • in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the precise dosage level should be determined by the attending physician or other health care provider and will depend upon well known factors, including route of administration, and the age, body weight, sex and general health of the individual; the nature, severity and clinical stage of the disease; the use (or not) of concomitant therapies; and the nature and extent of genetic engineering of cells in the patient.
  • the effective dosage of the rapalog of this invention may vary depending upon the particular compound utilized, the mode of administration, the condition, and severity thereof, of the condition being treated, as well as the various physical factors related to the individual being treated. In many cases, satisfactory results may be obtained when the rapalog is administered in a daily dosage of from about 0.01 mg/kg-100 mg/kg, preferably between 0.01-25 mg/kg, and more preferably between 0.01-5 mg/kg.
  • the projected daily dosages are expected to vary with route of administration. Thus, parenteral dosing will often be at levels of roughly 10% to 20% of oral dosing levels.
  • rapalog When the rapalog is used as part of a combination regimen, dosages of each of the components of the combination are administered during a desired treatment period.
  • the components of the combination may administered at the same time; either as, a unitary dosage form containing both components, or as separate dosage units; the components of the combination can also be administered at different times during a treatment period, or one may be administered as a pretreatment for the other.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers containing one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers containing one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the notice or package insert may contain instructions for use of a rapalog of this invention, consistent with the disclosure herein.
  • the cold (0° C.) reaction solution was diluted with ⁇ 20 mL EtOAc then transferred to a separatory funnel containing EtOAc (150 mL) and saturated NaHCO 3 (100 mL). Upon removing the aqueous layer, the organic layer was washed successively with ice cold 1N HCl (1 ⁇ 100 mL), saturated NaHCO 3 (1 ⁇ 100 mL), and brine (1 ⁇ 100 mL), then dried over MgSO4 and concentrated.
  • Rapamycin and dichloromethane are charged into a nitrogen-purged reaction flask.
  • the stirred solution is cooled to approximately 0° C. (an external temperature of ⁇ 5 ⁇ 5° C. is maintained throughout the reaction).
  • a solution of ethyl methylphosphonochloridate in dichloromethane is then added over a period of approximately 8-13 minutes. This is followed immediately by the addition of a solution of 3,5-lutidine in dichloromethane over a period of approximately 15-20 minutes. Throughout both additions, the internal temperature of the reaction is maintained below 0° C.
  • reaction solution is stirred for 3 hours during which time reaction progress is monitored by TLC (1:10:3:3 MeOH/DCM/EtOAc/hexanes) and reverse-phase HPLC analyses.
  • reaction mixture is diluted with ethyl acetate and worked up as above.
  • the organic layer was washed successively with ice cold 1N HCl (1 ⁇ 100 mL), saturated NaHCO 3 (3 ⁇ 100 mL), and brine (1 ⁇ 100 mL), then dried over MgSO 4 and concentrated.
  • the cold (0° C.) reaction solution was diluted with ⁇ 20 mL EtOAc then transferred to a separatory funnel containing EtOAc (150 mL) and saturated NaHCO 3 (100 mL). Upon removing the aqueous layer, the organic layer was washed successively with ice cold 1N HCl (1 ⁇ 100 mL), saturated NaHCO 3 (1 ⁇ 100 mL), and brine (1 ⁇ 100 mL), then dried over MgSO 4 and concentrated.
  • Rapamycin and dichloromethane are charged into a nitrogen-purged reaction flask. The stirred solution is cooled to approximately 0° C. (an external temperature of ⁇ 5 ⁇ 5° C. is maintained throughout the reaction). A solution of dimethylphosphinic chloride (2.0 molar equivalents) in dichloromethane is then added over a period of approximately 8-13 minutes. This is followed immediately by the addition of a solution of 3,5-lutidine (2.2 molar equivalents) in dichloromethane over a period of approximately 15-20 minutes. Throughout both additions, the internal temperature of the reaction sssstays below 0° C.
  • 43-epi compounds of the sort shown above may be prepared by adaptation of the methods of Kay, P. B. et al, J Chem Soc, Perkin Trans. 1, 1987, [8], 1813-1815, using the depicted reagents where R and each R′ is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc. moiety.
  • Phosphorous-linked compounds of the sort shown above may be prepared by adaptation of the methods of Yamashita, M. et al, Bull Chem Soc Japan, 1983, 56, 1871-1872 using the depicted reagents where X is halogen or anhydride for example, R*X generates a C-43 R*O-moiety which acts as a leaving group, and each occurrence of R is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc..moiety.
  • Amine-linked compounds of the sort shown above may be prepared by adaptation of the methods of Cavalla, D. et. al. Tet. Lett., 1983, 24, 295-298; Grinfield, A. et al, WO 98/09972 and Or, Y. S. et al U.S. Pat. No.
  • R*X generates a C-43 R*O-moiety which acts as a leaving group
  • m is a number from 1 to 10
  • each occurrence of R is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc. moiety.
  • Ethers of the sort shown above may be prepared by adaptation of the methods of Cottens, S et al, PCT International. Appln. Publication No. WO 94/09010 and Cheng, D. et al, WO 98/09970 using the depicted reagent (and base) where X is a leaving group, m is a number from 1 to 10, and each occurrence of R is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc..moiety.
  • Thio-ethers of the sort shown above may be prepared by adaptation of the methods of Grinfield et al, PCT International. Appin. Publication No. WO 98/09972 using the depicted reagent reagents where X is halogen or anhydride for example, R*X generates a C-43 R*O-moiety which acts as a leaving group, m is a number from 1 to 10, PG is a thiol protecting group, and each occurrence of R is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc. moiety.
  • Thio-compounds of the sort shown above may be prepared by adaptation of the methods of Yuan et al, Synthesis, 1989, 1, 48-50 (Route A) or Grinfield et al, PCT International. Appin. Publication No.
  • R*X generates a C-43 R*O-moiety which acts as a leaving group
  • m is a number from 1 to 10
  • each occurrence of R is a substituted or unsubstituted aliphatic, aliphatic-O—, aryl, aryloxy, heteroaryl, heteroaryloxy, etc. moiety.
  • the compounds of the foregoing illustrative Examples may be purified using silica gel flash chromatography to remove possible impurities such as residual reactants (including residual rapamycin or rapalog starting material) and undesired byproducts.
  • Suitable flash chromatography systems include commercially available prepacked cartridge systems, such as those of BIOTAGE, Inc. (PO Box 8006), Charlottesville, Va. 22906-8006). Cartridges may be obtained containing —30-70 ⁇ M particle size, 60 ⁇ pore size silica.
  • a typical protocol is provided below for using such flash chromatography systems for purifying compounds of this invention
  • Crude product is dissolved in a minimum amount of an appropriate solvent (e.g. dichloromethane, “DCM”) and loaded onto a FLASH Biotage cartridge.
  • DCM dichloromethane
  • Nonpolar impurities are eluted with DCM, followed by elution of the product with a solvent system such as 0.5:10:3:3 MeOH/DCM/EtOAc/hexanes.
  • a final wash of the column is performed, e.g., with a 1:10:3:3 MeOH/DCM/EtOAc/hexanes solvent system.
  • the collected fractions may be analyzed by TLC, normal-, and reverse-phase HPLC.
  • impure fractions may be repurified on a separate FLASH Biotage system using the same elution solvents and purity criteria for combination.
  • the multiple purified product pools are individually subjected to multiple solvent exchanges, e.g., with acetone (typically 4 to 6 times) and then combined using the same solvent (e.g., acetone) as the transfer solvent. Prior to combining, the pools may be assayed to confirm acceptable purity. Additional solvent exchanges (typically 2) may be performed with the same solvent (acetone, in this example) on the combined product batch, which is then dried in vacuo to constant weight at ambient temperature to provide material that may be sampled if desired for QC analysis.
  • a stainless steel DuraflexTM stent having dimensions of 3.0 mm ⁇ 14 mm is sprayed with a solution of 25 mg/ml a compound of any of Examples 1-12 in a 100% ethanol, acetone or ethyl acetate solvent. The stent is dried and the solvent is evaporated leaving the compound on a stent surface.
  • a 75:25 PLLA/PCL copolymer (sold commercially by Polysciences) is prepared in 1,4 Dioxane (sold commercially by Aldrich Chemicals).
  • the compound-loaded stent is loaded on a mandrel rotating at 200 rpm and a spray gun (sold commercially by Binks Manufacturing) dispenses the copolymer solution in a fine spray on to the compound-loaded stent as it rotates for a 10-30 second period.
  • the stent is then placed in a oven at 25-35° C. up to 24 hours to complete evaporation of the solvent.
  • Stainless steel Duraflex stent (3.0 ⁇ 13 mm) is laser cut from a SS tube.
  • the surface area for loading the drug is increased by increasing the surface roughness of the stent.
  • the surface area and the volume of the stent can be further increased by creating 10 nm wide and 5 nm deep grooves along the links of the stent strut. The grooves are created in areas which experience low stress during expansion so that the stent radial strength is not compromised.
  • a compound of any of Examples 1-12 can then be loaded on the stent and in the groove by dipping or spraying the stent in a solution of the compound prepared in low surface tension solvent such as dichloromethane, isopropyl alcohol, acetone, ethyl acetate, ethanol, or methanol.
  • the stent is then dried and the compound resides on the stent surface and in the grooves, which serve as a drug reservoir.
  • Parylene is then deposited on the stent to serve as a rate limiting barrier.
  • the compound elutes from the stent over a period of time in the range from 1 day to 45 days.
  • a compound of any of Examples 1-12 is dissolved in ethyl acetate, then sprayed on the stent, and left to dry evaporating the solvent with the compound remaining on the stent surface.
  • a matrix or barrier (silicone, polytetrafluorethylene, PARYLASTTM, parylene) is sprayed or deposited on the stent encapsulating the compound. The amount of the compound varies from 100 micrograms to 2 milligrams, with release rates from 1 day to 45 days.
  • a matrix with compound coated on a stent is prepared as described in Example 25, and then coated or sprayed with a top coat of a rate limiting barrier (and/or a matrix without a drug so to act as a rate limiting barrier).
  • the compound may be coated on a stent via a rate limiting barrier, and then covered with a top coat (another barrier or matrix).
  • top coats provide further control of release rate, improved biocompatibility, and/or resistance to scratching and cracking upon stent delivery or expansion.
US10/357,152 2002-02-01 2003-02-03 Phosphorus-containing compounds and uses thereof Abandoned US20030220297A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US10/357,152 US20030220297A1 (en) 2002-02-01 2003-02-03 Phosphorus-containing compounds and uses thereof
US10/635,054 US20040073024A1 (en) 2002-02-01 2003-08-06 Phosphorus-containing compounds and uses thereof
US10/862,149 US7091213B2 (en) 2002-02-01 2004-06-04 Phosphorus-containing compounds and uses thereof
US11/429,582 US7432277B2 (en) 2002-02-01 2006-05-05 Phosphorus-containing macrocycles
US11/494,418 US7186826B2 (en) 2002-02-01 2006-07-27 Phosphorus-containing compounds and uses thereof
US11/650,017 US7709020B2 (en) 2002-02-01 2007-01-05 Implantable device comprising phosphorus-containing macrolides
US12/798,501 US8058426B2 (en) 2002-02-01 2010-04-06 Phosphorus-containing compounds and uses thereof
US13/295,826 US9024014B2 (en) 2002-02-01 2011-11-14 Phosphorus-containing compounds and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US35325202P 2002-02-01 2002-02-01
US42692802P 2002-11-15 2002-11-15
US42838302P 2002-11-22 2002-11-22
US43393002P 2002-12-17 2002-12-17
US10/357,152 US20030220297A1 (en) 2002-02-01 2003-02-03 Phosphorus-containing compounds and uses thereof

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/635,054 Continuation-In-Part US20040073024A1 (en) 2002-02-01 2003-08-06 Phosphorus-containing compounds and uses thereof
US10/862,149 Continuation-In-Part US7091213B2 (en) 2002-02-01 2004-06-04 Phosphorus-containing compounds and uses thereof

Publications (1)

Publication Number Publication Date
US20030220297A1 true US20030220297A1 (en) 2003-11-27

Family

ID=27671080

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/357,152 Abandoned US20030220297A1 (en) 2002-02-01 2003-02-03 Phosphorus-containing compounds and uses thereof
US10/635,054 Abandoned US20040073024A1 (en) 2002-02-01 2003-08-06 Phosphorus-containing compounds and uses thereof
US10/862,149 Expired - Lifetime US7091213B2 (en) 2002-02-01 2004-06-04 Phosphorus-containing compounds and uses thereof
US11/494,418 Expired - Lifetime US7186826B2 (en) 2002-02-01 2006-07-27 Phosphorus-containing compounds and uses thereof
US11/650,017 Expired - Lifetime US7709020B2 (en) 2002-02-01 2007-01-05 Implantable device comprising phosphorus-containing macrolides
US12/798,501 Expired - Fee Related US8058426B2 (en) 2002-02-01 2010-04-06 Phosphorus-containing compounds and uses thereof
US13/295,826 Expired - Fee Related US9024014B2 (en) 2002-02-01 2011-11-14 Phosphorus-containing compounds and uses thereof

Family Applications After (6)

Application Number Title Priority Date Filing Date
US10/635,054 Abandoned US20040073024A1 (en) 2002-02-01 2003-08-06 Phosphorus-containing compounds and uses thereof
US10/862,149 Expired - Lifetime US7091213B2 (en) 2002-02-01 2004-06-04 Phosphorus-containing compounds and uses thereof
US11/494,418 Expired - Lifetime US7186826B2 (en) 2002-02-01 2006-07-27 Phosphorus-containing compounds and uses thereof
US11/650,017 Expired - Lifetime US7709020B2 (en) 2002-02-01 2007-01-05 Implantable device comprising phosphorus-containing macrolides
US12/798,501 Expired - Fee Related US8058426B2 (en) 2002-02-01 2010-04-06 Phosphorus-containing compounds and uses thereof
US13/295,826 Expired - Fee Related US9024014B2 (en) 2002-02-01 2011-11-14 Phosphorus-containing compounds and uses thereof

Country Status (16)

Country Link
US (7) US20030220297A1 (US20030220297A1-20031127-C00082.png)
EP (2) EP1478648B1 (US20030220297A1-20031127-C00082.png)
JP (1) JP4547911B2 (US20030220297A1-20031127-C00082.png)
KR (2) KR100956195B1 (US20030220297A1-20031127-C00082.png)
CN (2) CN101717410B (US20030220297A1-20031127-C00082.png)
AU (2) AU2003210787B2 (US20030220297A1-20031127-C00082.png)
BR (1) BR0307544A (US20030220297A1-20031127-C00082.png)
CA (1) CA2472341C (US20030220297A1-20031127-C00082.png)
DK (1) DK1478648T3 (US20030220297A1-20031127-C00082.png)
EA (2) EA008379B1 (US20030220297A1-20031127-C00082.png)
ES (1) ES2485841T3 (US20030220297A1-20031127-C00082.png)
IL (3) IL162734A0 (US20030220297A1-20031127-C00082.png)
MX (1) MXPA04007402A (US20030220297A1-20031127-C00082.png)
PL (1) PL216224B1 (US20030220297A1-20031127-C00082.png)
PT (1) PT1478648E (US20030220297A1-20031127-C00082.png)
WO (1) WO2003064383A2 (US20030220297A1-20031127-C00082.png)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040110813A1 (en) * 2002-09-24 2004-06-10 Boehringer Ingelheim International Gmbh Solid telmisartan pharmaceutical formulations
US20040202789A1 (en) * 2003-03-31 2004-10-14 Council Of Scientific And Industrila Research Process for preparing thin film solids
US20050049693A1 (en) * 2003-08-25 2005-03-03 Medtronic Vascular Inc. Medical devices and compositions for delivering biophosphonates to anatomical sites at risk for vascular disease
US20050187608A1 (en) * 2004-02-24 2005-08-25 O'hara Michael D. Radioprotective compound coating for medical devices
WO2005110480A2 (en) * 2004-05-17 2005-11-24 Novartis Ag Combination of organic compounds
US20060067983A1 (en) * 2004-09-28 2006-03-30 Atrium Medical Corporation Stand-alone film and methods for making the same
US20060173033A1 (en) * 2003-07-08 2006-08-03 Michaela Kneissel Use of rapamycin and rapamycin derivatives for the treatment of bone loss
US20060188542A1 (en) * 2005-02-22 2006-08-24 Bobyn John D Implant improving local bone formation
US20070190106A1 (en) * 2002-02-01 2007-08-16 Berstein David L Phosphorus-containing compounds & uses thereof
US20080032989A1 (en) * 2006-05-31 2008-02-07 Robinson William H Method of treating inflammatory diseases using tyroskine kinase inhibitors
US8124127B2 (en) * 2005-10-15 2012-02-28 Atrium Medical Corporation Hydrophobic cross-linked gels for bioabsorbable drug carrier coatings
US8226977B2 (en) 2004-06-04 2012-07-24 Teva Pharmaceutical Industries Ltd. Pharmaceutical composition containing irbesartan
US8263102B2 (en) 2004-09-28 2012-09-11 Atrium Medical Corporation Drug delivery coating for use with a stent
US8312836B2 (en) 2004-09-28 2012-11-20 Atrium Medical Corporation Method and apparatus for application of a fresh coating on a medical device
US8367099B2 (en) 2004-09-28 2013-02-05 Atrium Medical Corporation Perforated fatty acid films
US8496967B2 (en) 2006-11-14 2013-07-30 Ariad Pharmaceuticals, Inc. Oral formulations
US8574627B2 (en) 2006-11-06 2013-11-05 Atrium Medical Corporation Coated surgical mesh
US8633178B2 (en) 2011-11-23 2014-01-21 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US20140227254A1 (en) * 2006-11-02 2014-08-14 Acceleron Pharma, Inc. Antagonists of bmp9, bmp10, alk1 and other alk1 ligands, and uses thereof
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9000040B2 (en) 2004-09-28 2015-04-07 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US9012506B2 (en) 2004-09-28 2015-04-21 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US9278161B2 (en) 2005-09-28 2016-03-08 Atrium Medical Corporation Tissue-separating fatty acid adhesion barrier
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9427423B2 (en) 2009-03-10 2016-08-30 Atrium Medical Corporation Fatty-acid based particles
US9492596B2 (en) 2006-11-06 2016-11-15 Atrium Medical Corporation Barrier layer with underlying medical device and one or more reinforcing support structures
US9801982B2 (en) 2004-09-28 2017-10-31 Atrium Medical Corporation Implantable barrier device
US9867880B2 (en) 2012-06-13 2018-01-16 Atrium Medical Corporation Cured oil-hydrogel biomaterial compositions for controlled drug delivery
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US10322213B2 (en) 2010-07-16 2019-06-18 Atrium Medical Corporation Compositions and methods for altering the rate of hydrolysis of cured oil-based materials
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10864304B2 (en) 2009-08-11 2020-12-15 Atrium Medical Corporation Anti-infective antimicrobial-containing biomaterials
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations

Families Citing this family (254)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040073294A1 (en) 2002-09-20 2004-04-15 Conor Medsystems, Inc. Method and apparatus for loading a beneficial agent into an expandable medical device
US7432277B2 (en) 2002-02-01 2008-10-07 Araid Gene Therapeutics, Inc. Phosphorus-containing macrocycles
US7649015B2 (en) 2002-04-26 2010-01-19 Gilead Sciences, Inc. Cellular accumulation of phosphonate analogs of HIV protease inhibitor compounds
CN100569944C (zh) * 2002-07-16 2009-12-16 百奥帝卡技术有限公司 聚酮化合物和其它天然产物的产生
WO2004096286A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Antiviral phosphonate analogs
WO2004096236A2 (en) * 2003-04-25 2004-11-11 Gilead Sciences, Inc. Immunomodulator phosphonate conjugates
US7407965B2 (en) 2003-04-25 2008-08-05 Gilead Sciences, Inc. Phosphonate analogs for treating metabolic diseases
WO2005002626A2 (en) 2003-04-25 2005-01-13 Gilead Sciences, Inc. Therapeutic phosphonate compounds
US7452901B2 (en) 2003-04-25 2008-11-18 Gilead Sciences, Inc. Anti-cancer phosphonate analogs
US7300924B2 (en) 2003-04-25 2007-11-27 Gilead Sciences, Inc. Anti-infective phosphonate analogs
US7432261B2 (en) 2003-04-25 2008-10-07 Gilead Sciences, Inc. Anti-inflammatory phosphonate compounds
US7470724B2 (en) 2003-04-25 2008-12-30 Gilead Sciences, Inc. Phosphonate compounds having immuno-modulatory activity
WO2004096287A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Inosine monophosphate dehydrogenase inhibitory phosphonate compounds
EA200501676A1 (ru) 2003-04-25 2006-04-28 Джилид Сайэнс, Инк. Фосфонатсодержащие ингибиторы киназы (варианты), способ их получения, фармацевтическая композиция, лекарственная форма на их основе и способ ингибирования киназы у млекопитающего (варианты)
WO2005016252A2 (en) * 2003-07-11 2005-02-24 Ariad Gene Therapeutics, Inc. Phosphorus-containing macrocycles
WO2005044279A1 (en) 2003-10-24 2005-05-19 Gilead Sciences, Inc. Purine nucleoside phosphonate conjugates
WO2005042773A1 (en) 2003-10-24 2005-05-12 Gilead Sciences, Inc. Methods and compositions for identifying therapeutic compounds
WO2005044308A1 (en) 2003-10-24 2005-05-19 Gilead Sciences, Inc. Phosphonate analogs of antimetabolites
WO2005049021A1 (en) * 2003-11-03 2005-06-02 Oy Helsinki Transplantation R & D Ltd Materials and methods for inhibiting neointimal hyperplasia
AR046194A1 (es) 2003-11-04 2005-11-30 Mayo Foundation Metodo de tratamiento del linfoma de celulas del manto
GB0327840D0 (en) * 2003-12-01 2003-12-31 Novartis Ag Organic compounds
US7294123B2 (en) * 2003-12-17 2007-11-13 Corris Neurovascular, Inc. Activatable bioactive vascular occlusive device and method of use
US20050137568A1 (en) * 2003-12-17 2005-06-23 Jones Donald K. Activatable bioactive implantable medical device and method of use
EP1557182B1 (en) * 2003-12-17 2010-07-21 Codman & Shurtleff Activatable bioactive implantable medical device
KR20060127906A (ko) 2003-12-22 2006-12-13 길리애드 사이언시즈, 인코포레이티드 4'-치환된 카보버와 아바카비어 유도체 및 hiv와 hcv항바이러스 활성을 갖는 관련 화합물
US7247159B2 (en) 2004-04-08 2007-07-24 Cordis Neurovascular, Inc. Activatable bioactive vascular occlusive device
EP1778251B1 (en) 2004-07-27 2011-04-13 Gilead Sciences, Inc. Nucleoside phosphonate conjugates as anti hiv agents
US8075904B2 (en) * 2004-08-11 2011-12-13 California Institute Of Technology High aspect ratio template and method for producing same for central and peripheral nerve repair
CA2581372A1 (en) * 2004-09-30 2006-04-13 Ariad Gene Therapeutics, Inc. Treatment method
ES2364495T3 (es) * 2005-02-03 2011-09-05 The General Hospital Corporation Método para tratar cáncer resistente a gefitinib.
GB0504995D0 (en) * 2005-03-11 2005-04-20 Biotica Tech Ltd Use of a compound
US20100061994A1 (en) * 2005-03-11 2010-03-11 Rose Mary Sheridan Medical uses of 39-desmethoxyrapamycin and analogues thereof
GB0504994D0 (en) * 2005-03-11 2005-04-20 Biotica Tech Ltd Novel compounds
GB0507918D0 (en) 2005-04-19 2005-05-25 Novartis Ag Organic compounds
AU2006254397A1 (en) * 2005-05-31 2006-12-07 Novartis Ag Combination of HMG-CoA reductase inhibitors and mTOR inhibitors
US20070071754A1 (en) * 2005-09-26 2007-03-29 Peyman Gholam A Method to ameliorate inflammation
US8119655B2 (en) 2005-10-07 2012-02-21 Takeda Pharmaceutical Company Limited Kinase inhibitors
JP2009513645A (ja) 2005-10-26 2009-04-02 ノバルティス アクチエンゲゼルシャフト IL−1β化合物の新規使用
US20070104721A1 (en) 2005-11-04 2007-05-10 Wyeth Antineoplastic combinations with mTOR inhibitor,herceptin, and/or hki-272
EA015922B1 (ru) * 2005-11-14 2011-12-30 Ариад Фармасьютикалз, Инк. ВВЕДЕНИЕ ИНГИБИТОРА mTOR ДЛЯ ЛЕЧЕНИЯ ПАЦИЕНТОВ СО ЗЛОКАЧЕСТВЕННОЙ ОПУХОЛЬЮ
DK2275103T3 (da) 2005-11-21 2014-07-07 Novartis Ag mTor-inhibitorer ved behandling af endokrine tumorer
ES2761180T3 (es) 2005-12-23 2020-05-19 Ariad Pharma Inc Compuestos bicíclicos de heteroarilo
DE102006011507A1 (de) * 2006-03-14 2007-09-20 Lts Lohmann Therapie-Systeme Ag Wirkstoffbeladene Nanopartikel auf Basis hydrophiler Proteine
GB0609963D0 (en) * 2006-05-19 2006-06-28 Biotica Tech Ltd Novel compounds
EP2032168A4 (en) * 2006-06-02 2010-12-29 Ariad Pharma Inc COMBINED THERAPY BASED ON CAPECITABINE
WO2008016633A2 (en) * 2006-08-02 2008-02-07 Ariad Gene Therapeutics, Inc. Combination therapy
US10695327B2 (en) 2006-09-13 2020-06-30 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
US8088789B2 (en) 2006-09-13 2012-01-03 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
EP2083834B1 (en) 2006-09-13 2017-06-21 Elixir Medical Corporation Macrocyclic lactone compounds and methods for their use
JP2010505962A (ja) 2006-10-09 2010-02-25 武田薬品工業株式会社 キナーゼ阻害剤
US8466096B2 (en) * 2007-04-26 2013-06-18 Afton Chemical Corporation 1,3,2-dioxaphosphorinane, 2-sulfide derivatives for use as anti-wear additives in lubricant compositions
KR20230130763A (ko) 2007-05-29 2023-09-12 노파르티스 아게 항-il-1-베타 치료법에 대한 신규 적응증
US8142702B2 (en) * 2007-06-18 2012-03-27 Molecular Imprints, Inc. Solvent-assisted layer formation for imprint lithography
US8022216B2 (en) 2007-10-17 2011-09-20 Wyeth Llc Maleate salts of (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide and crystalline forms thereof
WO2009054001A1 (en) * 2007-10-22 2009-04-30 Biocon Limited A pharmaceutical composition and a process thereof
US20110104052A1 (en) * 2007-12-03 2011-05-05 The Johns Hopkins University Methods of synthesis and use of chemospheres
US9002344B2 (en) * 2007-12-05 2015-04-07 Microsoft Technology Licensing, Llc Phone content service
CA2711765A1 (en) 2008-01-11 2009-07-16 Massachusetts Eye & Ear Infirmary Conditional-stop dimerizable caspase transgenic animals
US20090253733A1 (en) * 2008-04-02 2009-10-08 Biointeractions, Ltd. Rapamycin carbonate esters
WO2009131631A1 (en) * 2008-04-14 2009-10-29 Poniard Pharmaceuticals, Inc. Rapamycin analogs as anti-cancer agents
SI2300013T1 (en) 2008-05-21 2018-03-30 Adriad Pharmacaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
KR20130088908A (ko) 2008-06-17 2013-08-08 와이어쓰 엘엘씨 Hki-272 및 비노렐빈을 함유하는 항신생물성 조합물
PL2307435T3 (pl) 2008-07-08 2012-11-30 Gilead Sciences Inc Sole związków inhibitorów HIV
KR101138294B1 (ko) * 2008-09-24 2012-04-25 김형일 혈관벽 임시 골격용 생분해성 블랜드
JP5590040B2 (ja) 2008-11-12 2014-09-17 アリアド・ファーマシューティカルズ・インコーポレイテッド キナーゼ阻害剤としてのピラジノピラジンおよび誘導体
US20120129809A1 (en) * 2009-03-02 2012-05-24 Merck & Co., Lung cancer treatment
CA2758297A1 (en) 2009-04-16 2010-10-21 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
EP2448942B1 (en) 2009-07-02 2014-09-24 Merck Sharp & Dohme Corp. FUSED TRICYCLIC COMPOUNDS AS mTOR INHIBITORS
EP2470179B1 (en) 2009-08-26 2017-11-29 Yissum Research Development Company of the Hebrew University of Jerusalem, Ltd. Sustained release delivery systems for the prevention and treatment of head and neck cancers
CA2774769A1 (en) 2009-09-30 2011-04-07 Schering Corporation Novel compounds that are erk inhibitors
CA2778291C (en) 2009-10-23 2014-02-11 Eli Lilly And Company Akt inhibitors
WO2011053938A1 (en) 2009-10-30 2011-05-05 Ariad Pharmaceuticals, Inc. Methods and compositions for treating cancer
WO2011120911A1 (en) 2010-03-30 2011-10-06 Novartis Ag Pkc inhibitors for the treatment of b-cell lymphoma having chronic active b-cell-receptor signalling
NZ607337A (en) 2010-08-20 2015-06-26 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
US8883801B2 (en) 2010-08-23 2014-11-11 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as mTOR inhibitors
WO2012061907A2 (en) 2010-11-10 2012-05-18 Katholieke Universiteit Leuven Osteoclast activity
EP2455104B1 (en) 2010-11-19 2013-07-17 Universitätsklinikum Freiburg Bio-functionalized stimulus-responsive dissolvable PEG-hydrogels
WO2012143879A1 (en) 2011-04-21 2012-10-26 Piramal Healthcare Limited A crystalline form of a salt of a morpholino sulfonyl indole derivative and a process for its preparation
US8901142B2 (en) 2011-07-26 2014-12-02 Merck Sharp & Dohme Corp. Fused tricyclic compounds as mTOR inhibitors
US8716363B2 (en) 2011-09-28 2014-05-06 Globus Medical, Inc. Biodegradable putty compositions and implant devices, methods, and kits relating to the same
EP2589383A1 (en) 2011-11-06 2013-05-08 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Berlin FKBP subtype-specific rapamycin analogue for use in treatment of diseases
US20140348848A1 (en) 2011-12-02 2014-11-27 Dhananjay Kaul Anti-il-1beta (interleukin-1beta) antibody-based prophylactic therapy to prevent complications leading to vaso-occlusion in sickle cell disease
CN104159924B (zh) 2011-12-05 2018-03-16 诺华股份有限公司 表皮生长因子受体3(her3)的抗体
AU2012349736A1 (en) 2011-12-05 2014-06-26 Novartis Ag Antibodies for epidermal growth factor receptor 3 (HER3) directed to domain II of HER3
US20140329858A1 (en) 2011-12-05 2014-11-06 Novartis Ag Cyclic Urea Derivatives As Androgen Receptor Antagonists
JP6158833B2 (ja) 2012-01-09 2017-07-05 アローヘッド ファーマシューティカルズ インコーポレイテッド ベータ−カテニン関連疾患を処置するための有機組成物
MY172706A (en) 2012-03-23 2019-12-11 Univ Queensland Immunomodulatory agent and uses therefor
TW201408682A (zh) 2012-08-22 2014-03-01 Chunghwa Chemical Synthesis & Biotech Co Ltd 高轉化率之42-(二甲基亞磷醯)雷帕霉素的製備方法
CA2899030C (en) 2013-02-19 2021-03-09 Novartis Ag Benzothiophene derivatives and compositions thereof as selective estrogen receptor degraders
EP2958943B1 (en) 2013-02-20 2019-09-11 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
US9498532B2 (en) 2013-03-13 2016-11-22 Novartis Ag Antibody drug conjugates
US20160022648A1 (en) 2013-03-13 2016-01-28 Santen Pharmaceutical Co., Ltd. Therapeutic agent for meibomian gland dysfunction
CA2903772A1 (en) 2013-03-15 2014-09-25 Novartis Ag Antibody drug conjugates
UY35468A (es) 2013-03-16 2014-10-31 Novartis Ag Tratamiento de cáncer utilizando un receptor quimérico de antígeno anti-cd19
WO2014184734A1 (en) 2013-05-14 2014-11-20 Novartis Ag Markers associated with mtor inhibition
US10682415B2 (en) 2013-07-22 2020-06-16 Wisconsin Alumni Research Foundation Thermogel formulation for combination drug delivery
CN105764502A (zh) 2013-07-26 2016-07-13 现代化制药公司 改善比生群及其类似物及衍生物的治疗益处的组合方法
WO2015044854A1 (en) 2013-09-24 2015-04-02 Novartis Ag Markers associated with mtor inhibition
US20150140036A1 (en) 2013-11-13 2015-05-21 Novartis Institutes For Biomedical Research, Inc. Low, immune enhancing, dose mtor inhibitors and uses thereof
EP2878312A1 (en) 2013-12-02 2015-06-03 Albert-Ludwigs-Universität Freiburg Reversible PEGylation of nanocarriers
WO2015092634A1 (en) 2013-12-16 2015-06-25 Novartis Ag 1,2,3,4-tetrahydroisoquinoline compounds and compositions as selective estrogen receptor antagonists and degraders
CN116478927A (zh) 2013-12-19 2023-07-25 诺华股份有限公司 人间皮素嵌合抗原受体及其用途
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
DK3089737T3 (da) 2013-12-31 2021-12-13 Rapamycin Holdings Llc Orale rapamycin-nanopartikelpræparater og anvendelse.
JP6523303B2 (ja) 2014-01-17 2019-05-29 ノバルティス アーゲー Shp2の活性を阻害するための1−ピリダジン/トリアジン−3−イル−ピペラジン/ピペリジン/ピロリジン誘導体およびその組成物
ES2699354T3 (es) 2014-01-17 2019-02-08 Novartis Ag Derivados de 1-(triazin-3-il/piridazin-3-il)-piper(-azin)idina y composiciones de las mismas para inhibir la actividad de SHP2
JO3517B1 (ar) 2014-01-17 2020-07-05 Novartis Ag ان-ازاسبيرو الكان حلقي كبديل مركبات اريل-ان مغايرة وتركيبات لتثبيط نشاط shp2
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
JOP20200096A1 (ar) 2014-01-31 2017-06-16 Children’S Medical Center Corp جزيئات جسم مضاد لـ tim-3 واستخداماتها
BR122024001145A2 (pt) 2014-03-14 2024-02-27 Novartis Ag Molécula de anticorpo isolada capaz de se ligar a lag-3, seu método de produção, composição farmacêutica, ácidos nucleicos, vetor de expressão, método para detecção de lag-3 em uma amostra biológica, e uso das referidas molécula de anticorpo e composição
JP2017513818A (ja) 2014-03-15 2017-06-01 ノバルティス アーゲー キメラ抗原受容体を使用する癌の処置
ES2857226T3 (es) 2014-03-15 2021-09-28 Novartis Ag Receptor de antígeno quimérico regulable
PL3129470T3 (pl) 2014-04-07 2021-11-29 Novartis Ag Leczenie nowotworu złośliwego z zastosowaniem chimerycznego receptora antygenowego anty-CD19
CN106659758A (zh) 2014-06-02 2017-05-10 儿童医疗中心有限公司 用于免疫调节的组合物和方法
SG10201913753VA (en) 2014-06-19 2020-03-30 Ariad Pharma Inc Heteroaryl compounds for kinase inhibition
US20170291956A1 (en) 2014-07-16 2017-10-12 David Livingston HER3 Inhibition in Low-grade Serous Ovarian Cancers
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
BR112017001183A2 (pt) 2014-07-21 2017-11-28 Novartis Ag tratamento de câncer usando receptor de antígeno quimérico anti-bcma humanizado
EP3660042B1 (en) 2014-07-31 2023-01-11 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
WO2016020791A1 (en) 2014-08-05 2016-02-11 Novartis Ag Ckit antibody drug conjugates
CN106659790A (zh) 2014-08-12 2017-05-10 诺华股份有限公司 抗cdh6抗体药物缀合物
AU2015301460B2 (en) 2014-08-14 2021-04-08 Novartis Ag Treatment of cancer using GFR alpha-4 chimeric antigen receptor
CN112410363A (zh) 2014-08-19 2021-02-26 诺华股份有限公司 抗cd123嵌合抗原受体(car)用于癌症治疗
EP3191127A1 (en) 2014-09-13 2017-07-19 Novartis AG Combination therapies of egfr inhibitors
DK3194443T3 (da) 2014-09-17 2021-09-27 Novartis Ag Målretning af cytotoksiske celler med kimære receptorer i forbindelse med adoptiv immunterapi
MX2017004360A (es) 2014-10-03 2017-06-26 Novartis Ag Terapias de combinacion.
MA41044A (fr) 2014-10-08 2017-08-15 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer
RU2743657C2 (ru) 2014-10-08 2021-02-20 Новартис Аг Биомаркеры, прогнозирующие способность к терапевтическому ответу на терапию химерным рецептором антигена, и их применение
SG11201702401RA (en) 2014-10-14 2017-04-27 Novartis Ag Antibody molecules to pd-l1 and uses thereof
MA40913A (fr) 2014-11-14 2017-09-20 Novartis Ag Conjugués anticorps-médicament
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
WO2016100882A1 (en) 2014-12-19 2016-06-23 Novartis Ag Combination therapies
US10377818B2 (en) 2015-01-30 2019-08-13 The Board Of Trustees Of The Leland Stanford Junior University Method of treating glioma
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
JP6695353B2 (ja) 2015-03-25 2020-05-20 ノバルティス アーゲー Fgfr4阻害剤としてのホルミル化n−複素環式誘導体
US20180140602A1 (en) 2015-04-07 2018-05-24 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
RU2021121771A (ru) 2015-04-08 2022-01-12 Новартис Аг Cd20 терапия, cd22 терапия и комбинированная терапия клетками, экспрессирующими химерный антигенный рецептор (car) к cd19
AU2016249005B2 (en) 2015-04-17 2022-06-16 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US10576076B2 (en) 2015-05-20 2020-03-03 Novartis Ag Pharmaceutical combination of everolimus with dactolisib
EP3310813A1 (en) 2015-06-17 2018-04-25 Novartis AG Antibody drug conjugates
EP3310771B1 (en) 2015-06-19 2020-07-22 Novartis AG Compounds and compositions for inhibiting the activity of shp2
US10287266B2 (en) 2015-06-19 2019-05-14 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
US10975080B2 (en) 2015-06-19 2021-04-13 Novartis Ag Compounds and compositions for inhibiting the activity of SHP2
AU2016297014B2 (en) 2015-07-21 2021-06-17 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US20180207273A1 (en) 2015-07-29 2018-07-26 Novartis Ag Combination therapies comprising antibody molecules to tim-3
DK3317301T3 (da) 2015-07-29 2021-06-28 Immutep Sas Kombinationsterapier omfattende antistofmolekyler mod lag-3
EP3344996A2 (en) 2015-09-03 2018-07-11 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
MA44334A (fr) 2015-10-29 2018-09-05 Novartis Ag Conjugués d'anticorps comprenant un agoniste du récepteur de type toll
CA3004138A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and tim-3 and their uses
JP2018537447A (ja) 2015-11-11 2018-12-20 ノバルティス アーゲー ミオスタチンアンタゴニストの使用、それを含有する組合せ物およびその使用
AU2016369537B2 (en) 2015-12-17 2024-03-14 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
EP3409499B1 (en) * 2016-02-29 2022-12-07 FUJIFILM Corporation Lithographic printing original plate and plate making method for lithographic printing plates
MX2018010733A (es) 2016-03-04 2019-07-04 Novartis Ag Celulas que expresan multiples moleculas del receptor de antigeno quimerico (car) y usos de las mismas.
SI3452465T1 (sl) 2016-05-04 2021-04-30 Genoscience Pharma Substituirani derivati 2,4-diamino-kinolina za uporabo v zdravljenju proliferativnih bolezni
PT3468972T (pt) 2016-06-14 2020-08-21 Novartis Ag Compostos e composições para inibir a atividade da shp2
KR102565885B1 (ko) 2016-07-20 2023-08-09 유니버시티 오브 유타 리서치 파운데이션 Cd229 car t 세포 및 이의 사용 방법
US11077178B2 (en) 2016-09-21 2021-08-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptor (CAR) that targets chemokine receptor CCR4 and its use
WO2018064076A1 (en) 2016-09-27 2018-04-05 Cero Therapeutics, Inc. Chimeric engulfment receptor molecules
AU2017341047A1 (en) 2016-10-07 2019-05-02 Novartis Ag Chimeric antigen receptors for the treatment of cancer
EP3544608A1 (en) 2016-11-23 2019-10-02 Novartis AG Methods of enhancing immune response with everolimus, dactolisib or both
WO2018163051A1 (en) 2017-03-06 2018-09-13 Novartis Ag Methods of treatment of cancer with reduced ubb expression
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
AR111651A1 (es) 2017-04-28 2019-08-07 Novartis Ag Conjugados de anticuerpos que comprenden agonistas del receptor de tipo toll y terapias de combinación
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018215937A1 (en) 2017-05-24 2018-11-29 Novartis Ag Interleukin-7 antibody cytokine engrafted proteins and methods of use in the treatment of cancer
PE20200303A1 (es) 2017-05-24 2020-02-06 Novartis Ag Proteinas de anticuerpo injertadas con citocina y metodos de uso en el tratamiento del cancer
CN111107868A (zh) 2017-05-24 2020-05-05 诺华股份有限公司 抗体细胞因子移植蛋白及使用方法
CN110719799A (zh) 2017-06-09 2020-01-21 俄勒冈州普罗维登斯健康与服务部 Cd39和cd103在鉴定用于癌症治疗的人肿瘤反应性t细胞中的应用
KR20200021087A (ko) 2017-06-22 2020-02-27 노파르티스 아게 Cd73에 대한 항체 분자 및 이의 용도
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag ANTIBODY MOLECULES DIRECTED AGAINST CD73 AND CORRESPONDING USES
PL3661937T3 (pl) 2017-08-01 2021-12-20 Gilead Sciences, Inc. Formy krystaliczne ((s)-((((2r,5r)-5-(6-amino-9h-puryn-9-ylo)-4-fluoro-2,5-dihydrofuran-2-ylo)oksy)metylo)(fenoksy)fosforylo)-l-alaninianu etylu (gs-9131) do leczenia zakażeń wirusowych
MA50245A (fr) 2017-09-11 2020-07-22 Krouzon Pharmaceuticals Inc Inhibiteurs allostériques octahydrocyclopenta[c]pyrrole de shp2
JP7286658B2 (ja) 2017-09-26 2023-06-05 セロ・セラピューティクス・インコーポレイテッド キメラエンガルフメント受容体分子および使用方法
AU2018375738A1 (en) 2017-11-30 2020-06-11 Novartis Ag BCMA-targeting chimeric antigen receptor, and uses thereof
WO2019120094A1 (zh) 2017-12-21 2019-06-27 深圳市塔吉瑞生物医药有限公司 用于抑制激酶活性的芳基磷氧化物
US11254696B2 (en) 2017-12-21 2022-02-22 Shenzhen Targetrx, Inc. Dianilinopyrimidine compound for inhibiting kinase activity
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
CA3090249A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
US10596165B2 (en) 2018-02-12 2020-03-24 resTORbio, Inc. Combination therapies
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
RU2020135107A (ru) 2018-03-28 2022-04-29 Серо Терапьютикс, Инк. Клеточные иммунотерапевтические композиции и их применения
CN112218886A (zh) 2018-03-28 2021-01-12 森罗治疗公司 嵌合吞噬受体的表达载体、基因修饰的宿主细胞及其用途
EP3774906A1 (en) 2018-03-28 2021-02-17 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
EP3784351A1 (en) 2018-04-27 2021-03-03 Novartis AG Car t cell therapies with enhanced efficacy
EP3788369A1 (en) 2018-05-01 2021-03-10 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
US11746157B2 (en) 2018-05-24 2023-09-05 Janssen Biotech, Inc. PSMA binding agents and uses thereof
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
AR126019A1 (es) 2018-05-30 2023-09-06 Novartis Ag Anticuerpos frente a entpd2, terapias de combinación y métodos de uso de los anticuerpos y las terapias de combinación
WO2019232244A2 (en) 2018-05-31 2019-12-05 Novartis Ag Antibody molecules to cd73 and uses thereof
TW202016136A (zh) 2018-06-01 2020-05-01 瑞士商諾華公司 針對bcma之結合分子及其用途
CN112203725A (zh) 2018-06-13 2021-01-08 诺华股份有限公司 Bcma嵌合抗原受体及其用途
AR116109A1 (es) 2018-07-10 2021-03-31 Novartis Ag Derivados de 3-(5-amino-1-oxoisoindolin-2-il)piperidina-2,6-diona y usos de los mismos
JOP20210001A1 (ar) 2018-07-10 2021-01-05 Novartis Ag مشتقات 3-(5- هيدروكسي -1- أوكسو أيزو إندولين -2- يل) بيبريدين -2، 6- دايون واستخدامها لمعالجة أمراض مرتبطة ببروتين ذات أصبع الزنك من عائلة (ikaros 2 (ikzf2
CA3153630A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications Sa Stable, concentrated radionuclide complex solutions
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
CN112996795A (zh) 2018-09-18 2021-06-18 尼坎医疗公司 作为src同源-2磷酸酶抑制剂的稠合的三环衍生物
CN117122707A (zh) 2018-09-25 2023-11-28 意大利国际先进加速器应用有限公司 联合疗法
WO2020068873A1 (en) 2018-09-25 2020-04-02 Black Diamond Therapeutics, Inc. Tyrosine kinase inhibitor compositions, methods of making and methods of use
WO2020068867A1 (en) 2018-09-25 2020-04-02 Black Diamond Therapeutics, Inc. Quinazoline derivatives as tyrosine kinase inhibitor, compositions, methods of making them and their use
US20220047633A1 (en) 2018-09-28 2022-02-17 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
EP3856345A1 (en) 2018-09-29 2021-08-04 Novartis AG Process of manufacture of a compound for inhibiting the activity of shp2
WO2020089811A1 (en) 2018-10-31 2020-05-07 Novartis Ag Dc-sign antibody drug conjugates
EP3874030A4 (en) 2018-11-01 2022-03-09 Gracell Biotechnologies (Shanghai) Co., Ltd. COMPOSITIONS AND METHODS FOR T-CELL ENGINEERING
CN113271945A (zh) 2018-12-20 2021-08-17 诺华股份有限公司 包含3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物的给药方案和药物组合
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
KR20210129672A (ko) 2019-02-15 2021-10-28 노파르티스 아게 치환된 3-(1-옥소이소인돌린-2-일)피페리딘-2,6-디온 유도체 및 이의 용도
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
CA3145864A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
CN110407872A (zh) * 2019-07-25 2019-11-05 江苏好收成韦恩农化股份有限公司 烷基氯化膦酸单烷酯联产高纯度三氯氧磷的方法
CN114787150A (zh) 2019-08-15 2022-07-22 黑钻治疗公司 炔基喹唑啉化合物
JP2022548881A (ja) 2019-09-18 2022-11-22 ノバルティス アーゲー Entpd2抗体、組合せ療法並びに抗体及び組合せ療法を使用する方法
US20240058446A1 (en) 2019-10-03 2024-02-22 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
AU2020406350A1 (en) 2019-12-20 2022-08-11 Novartis Ag Uses of anti-TGF-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
US20230093147A1 (en) 2020-03-09 2023-03-23 President And Fellows Of Harvard College Methods and compositions relating to improved combination therapies
WO2021195206A1 (en) 2020-03-24 2021-09-30 Black Diamond Therapeutics, Inc. Polymorphic forms and related uses
WO2021217508A1 (en) 2020-04-29 2021-11-04 Novartis Ag COMPOUNDS AND COMPOSITIONS FOR INHIBITING THE ACTIVITY OF HIF2α AND THEIR METHODS OF USE
US20230173095A1 (en) 2020-05-29 2023-06-08 President And Fellows Of Harvard College Living cells engineered with polyphenol-functionalized biologically active nanocomplexes
EP4165169A1 (en) 2020-06-11 2023-04-19 Novartis AG Zbtb32 inhibitors and uses thereof
US20230321067A1 (en) 2020-06-23 2023-10-12 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2021262969A1 (en) 2020-06-24 2021-12-30 The General Hospital Corporation Materials and methods of treating cancer
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022036265A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Chimeric tim receptors and uses thereof
WO2022036285A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Compositions and methods for treating cancer with chimeric tim receptors in combination with inhibitors of poly (adp-ribose) polymerase
WO2022036287A1 (en) 2020-08-14 2022-02-17 Cero Therapeutics, Inc. Anti-cd72 chimeric receptors and uses thereof
EP4204020A1 (en) 2020-08-31 2023-07-05 Advanced Accelerator Applications International S.A. Method of treating psma-expressing cancers
US20230321285A1 (en) 2020-08-31 2023-10-12 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043556A1 (en) 2020-08-31 2022-03-03 Novartis Ag Stable radiopharmaceutical composition
IL302569A (en) 2020-11-06 2023-07-01 Novartis Ag CD19 binding molecules and their uses
TW202237638A (zh) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 烏苷酸環化酶c(gcc)抗原結合劑之組成物及其使用方法
TW202237119A (zh) 2020-12-10 2022-10-01 美商住友製藥腫瘤公司 Alk﹘5抑制劑和彼之用途
EP4267575A1 (en) 2020-12-22 2023-11-01 Nikang Therapeutics, Inc. Compounds for degrading cyclin-dependent kinase 2 via ubiquitin proteosome pathway
AU2022207648A1 (en) 2021-01-13 2023-07-27 Monte Rosa Therapeutics Ag Isoindolinone compounds
US20240059789A1 (en) 2021-01-28 2024-02-22 Janssen Biotech, Inc. Psma binding proteins and uses thereof
WO2022170052A1 (en) 2021-02-05 2022-08-11 Black Diamond Therapeutics, Inc. Quinazoline derivatives, pyridopyrimidine derivatives, pyrimidopyrimidine derivatives, and uses thereof
CA3212006A1 (en) 2021-02-26 2022-09-01 Kelonia Therapeutics, Inc. Lymphocyte targeted lentiviral vectors
TW202304979A (zh) 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
US20220363696A1 (en) 2021-04-13 2022-11-17 Nuvalent, Inc. Amino-substituted heterocycles for treating cancers with egfr mutations
WO2022219407A1 (en) 2021-04-14 2022-10-20 Monte Rosa Therapeutics Ag Isoindolinone compounds
WO2022219412A1 (en) 2021-04-14 2022-10-20 Monte Rosa Therapeutics Ag Isoindolinone amide compounds useful to treat diseases associated with gspt1
AR125874A1 (es) 2021-05-18 2023-08-23 Novartis Ag Terapias de combinación
AU2022288058A1 (en) 2021-06-07 2023-11-16 Agonox, Inc. Cxcr5, pd-1, and icos expressing tumor reactive cd4 t cells and their use
TW202317589A (zh) 2021-07-14 2023-05-01 美商尼坎醫療公司 作為kras抑制劑的伸烷基衍生物
WO2023288046A1 (en) 2021-07-15 2023-01-19 President And Fellows Of Harvard College Compositions and methods relating to cells with adhered particles
WO2023010097A1 (en) 2021-07-28 2023-02-02 Cero Therapeutics, Inc. Chimeric tim4 receptors and uses thereof
TW202346292A (zh) 2022-03-28 2023-12-01 美商尼坎醫療公司 作為週期蛋白依賴性激酶2抑制劑的磺醯胺基衍生物
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023240024A1 (en) 2022-06-08 2023-12-14 Nikang Therapeutics, Inc. Sulfamide derivatives as cyclin-dependent kinase 2 inhibitors
WO2024023666A1 (en) 2022-07-26 2024-02-01 Novartis Ag Crystalline forms of an akr1c3 dependent kars inhibitor

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5068091A (en) * 1988-02-22 1991-11-26 Kabushiki Kaisha Tiyoda Seisakusho Device for dyeing tissues for immune response observation
US5206018A (en) * 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
US5234456A (en) * 1990-02-08 1993-08-10 Pfizer Hospital Products Group, Inc. Hydrophilic stent
US5310903A (en) * 1993-03-05 1994-05-10 Merck & Co., Inc. Imidazolidyl rapamycin derivatives
US5385910A (en) * 1993-11-22 1995-01-31 American Home Products Corporation Gem-distributed esters of rapamycin
US5391730A (en) * 1993-10-08 1995-02-21 American Home Products Corporation Phosphorylcarbamates of rapamycin and oxime derivatives thereof
US5434260A (en) * 1992-10-13 1995-07-18 American Home Products Corporation Carbamates of rapamycin
US5489680A (en) * 1992-10-13 1996-02-06 American Home Products Corporation Carbamates of rapamycin
US5491231A (en) * 1994-11-28 1996-02-13 American Home Products Corporation Hindered N-oxide esters of rapamycin
US5516781A (en) * 1992-01-09 1996-05-14 American Home Products Corporation Method of treating restenosis with rapamycin
US5665591A (en) * 1994-12-06 1997-09-09 Trustees Of Boston University Regulation of smooth muscle cell proliferation
US5851217A (en) * 1990-02-28 1998-12-22 Medtronic, Inc. Intralumenal drug eluting prosthesis
US6146358A (en) * 1989-03-14 2000-11-14 Cordis Corporation Method and apparatus for delivery of therapeutic agent
US6152141A (en) * 1994-07-28 2000-11-28 Heartport, Inc. Method for delivery of therapeutic agents to the heart
US20010010920A1 (en) * 1993-04-23 2001-08-02 Molnar-Kimber Katherine L. Monoclonal antibodies obtained using rapamycin position 27 conjugates as an immunogen
US6585764B2 (en) * 1997-04-18 2003-07-01 Cordis Corporation Stent with therapeutically active dosage of rapamycin coated thereon

Family Cites Families (185)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3976071A (en) 1974-01-07 1976-08-24 Dynatech Corporation Methods of improving control of release rates and products useful in same
US5066493A (en) * 1978-11-03 1991-11-19 American Home Products Corporation Rapamycin in treatment of tumors
US4335094A (en) 1979-01-26 1982-06-15 Mosbach Klaus H Magnetic polymer particles
US4345588A (en) 1979-04-23 1982-08-24 Northwestern University Method of delivering a therapeutic agent to a target capillary bed
US4357925A (en) 1980-12-17 1982-11-09 The Bendix Corporation Distributor injection pump for diesel engines
US4501726A (en) 1981-11-12 1985-02-26 Schroeder Ulf Intravascularly administrable, magnetically responsive nanosphere or nanoparticle, a process for the production thereof, and the use thereof
US5206159A (en) 1984-11-01 1993-04-27 Miles Inc., As Legal Successor By Merger With Technicon Instruments Corp. Polymer particles containing colloidal iron oxide granules for use as a magnetically responsive reagent carrier
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4733665C2 (en) 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5102417A (en) 1985-11-07 1992-04-07 Expandable Grafts Partnership Expandable intraluminal graft, and method and apparatus for implanting an expandable intraluminal graft
GB8601100D0 (en) 1986-01-17 1986-02-19 Cosmas Damian Ltd Drug delivery system
US4871716A (en) 1986-02-04 1989-10-03 University Of Florida Magnetically responsive, hydrophilic microspheres for incorporation of therapeutic substances and methods of preparation thereof
US5000185A (en) 1986-02-28 1991-03-19 Cardiovascular Imaging Systems, Inc. Method for intravascular two-dimensional ultrasonography and recanalization
US4832686A (en) 1986-06-24 1989-05-23 Anderson Mark E Method for administering interleukin-2
US5069216A (en) 1986-07-03 1991-12-03 Advanced Magnetics Inc. Silanized biodegradable super paramagnetic metal oxides as contrast agents for imaging the gastrointestinal tract
JPS6390534A (ja) 1986-10-06 1988-04-21 Hitachi Ltd アルカリ可溶性ラダ−シリコ−ン重合体
JPS63101427A (ja) 1986-10-17 1988-05-06 Hitachi Ltd アルカリ可溶性ラダ−シリコ−ン
US4883666A (en) 1987-04-29 1989-11-28 Massachusetts Institute Of Technology Controlled drug delivery system for treatment of neural disorders
US4837311A (en) 1987-06-22 1989-06-06 Hoffman-La Roche Inc. Anti-retroviral compounds
US4894231A (en) 1987-07-28 1990-01-16 Biomeasure, Inc. Therapeutic agent delivery system
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US4936281A (en) 1989-04-13 1990-06-26 Everest Medical Corporation Ultrasonically enhanced RF ablation catheter
WO1990013332A1 (en) 1989-05-11 1990-11-15 Cedars-Sinai Medical Center Stent with sustained drug delivery
US5182293A (en) 1989-11-13 1993-01-26 Merrell Dow Pharmaceuticals Inc. Treatment of multi-drug resistant tumors with pyridyloxazole-2-ones
US5067491A (en) 1989-12-08 1991-11-26 Becton, Dickinson And Company Barrier coating on blood contacting devices
US6004346A (en) 1990-02-28 1999-12-21 Medtronic, Inc. Intralumenal drug eluting prosthesis
US5195417A (en) 1990-04-26 1993-03-23 Northern Telecom Limited Registration of artwork panels in the manufacture of printed circuit boards
WO1991017724A1 (en) 1990-05-17 1991-11-28 Harbor Medical Devices, Inc. Medical device polymer
CA2081896A1 (en) 1990-06-15 1991-12-16 James E. Shapland Drug delivery apparatus and method
US5112457A (en) 1990-07-23 1992-05-12 Case Western Reserve University Process for producing hydroxylated plasma-polymerized films and the use of the films for enhancing the compatiblity of biomedical implants
US5163952A (en) 1990-09-14 1992-11-17 Michael Froix Expandable polymeric stent with memory and delivery apparatus and method
US5130307A (en) 1990-09-28 1992-07-14 American Home Products Corporation Aminoesters of rapamycin
US5378696A (en) 1990-09-19 1995-01-03 American Home Products Corporation Rapamycin esters
US5484584A (en) 1990-10-02 1996-01-16 Board Of Regents, The University Of Texas System Therapeutic and diagnostic use of modified polymeric microcapsules
CA2093429A1 (en) * 1990-10-09 1992-04-10 Shieh-Shung T. Chen Process for biophosphorylating organic compounds
US5893840A (en) 1991-01-04 1999-04-13 Medtronic, Inc. Releasable microcapsules on balloon catheters
US5368557A (en) 1991-01-11 1994-11-29 Baxter International Inc. Ultrasonic ablation catheter device having multiple ultrasound transmission members
WO1993013663A1 (en) 1992-01-17 1993-07-22 Abbott Laboratories Method of directing biosynthesis of specific polyketides
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5100883A (en) 1991-04-08 1992-03-31 American Home Products Corporation Fluorinated esters of rapamycin
US5194447A (en) 1992-02-18 1993-03-16 American Home Products Corporation Sulfonylcarbamates of rapamycin
US5118678A (en) 1991-04-17 1992-06-02 American Home Products Corporation Carbamates of rapamycin
US5118677A (en) 1991-05-20 1992-06-02 American Home Products Corporation Amide esters of rapamycin
DE4117782C2 (de) 1991-05-28 1997-07-17 Diagnostikforschung Inst Nanokristalline magnetische Eisenoxid-Partikel, Verfahren zu ihrer Herstellung sowie diagnostische und/oder therapeutische Mittel
IL102414A (en) 1991-07-25 1996-08-04 Univ Louisville Res Found Medicinal preparations for the treatment of ocular inflammation, containing rapamycin
US5356433A (en) 1991-08-13 1994-10-18 Cordis Corporation Biocompatible metal surfaces
US5176907A (en) 1991-08-13 1993-01-05 The Johns Hopkins University School Of Medicine Biocompatible and biodegradable poly (phosphoester-urethanes)
US5604294A (en) * 1991-09-05 1997-02-18 Luly; Jay R. Macrocyclic immunomodulators
US5252732A (en) * 1991-09-09 1993-10-12 Merck & Co., Inc. D-heteroaryl, O-alkylheteroaryl, O-alkenylheteroaryl and O-alkynylheteroarylmacrolides having immunosuppressive activity
US5411550A (en) 1991-09-16 1995-05-02 Atrium Medical Corporation Implantable prosthetic device for the delivery of a bioactive material
US5286730A (en) 1991-09-17 1994-02-15 American Home Products Corporation Method of treating immunoinflammatory disease
US5286731A (en) 1991-09-17 1994-02-15 American Home Products Corporation Method of treating immunoinflammatory bowel disease
US5464450A (en) 1991-10-04 1995-11-07 Scimed Lifesystems Inc. Biodegradable drug delivery vascular stent
US5500013A (en) 1991-10-04 1996-03-19 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
WO1993006792A1 (en) 1991-10-04 1993-04-15 Scimed Life Systems, Inc. Biodegradable drug delivery vascular stent
US5225282A (en) 1991-12-13 1993-07-06 Molecular Bioquest, Inc. Biodegradable magnetic microcluster comprising non-magnetic metal or metal oxide particles coated with a functionalized polymer
US5876452A (en) 1992-02-14 1999-03-02 Board Of Regents, University Of Texas System Biodegradable implant
US5177203A (en) 1992-03-05 1993-01-05 American Home Products Corporation Rapamycin 42-sulfonates and 42-(N-carboalkoxy) sulfamates useful as immunosuppressive agents
US5656297A (en) 1992-03-12 1997-08-12 Alkermes Controlled Therapeutics, Incorporated Modulated release from biocompatible polymers
US5599352A (en) 1992-03-19 1997-02-04 Medtronic, Inc. Method of making a drug eluting stent
DK0633783T3 (da) 1992-03-30 2000-09-25 American Home Prod Rapamycin-præparat til intravenøs injektion
US5288711A (en) 1992-04-28 1994-02-22 American Home Products Corporation Method of treating hyperproliferative vascular disease
US5283257A (en) 1992-07-10 1994-02-01 The Board Of Trustees Of The Leland Stanford Junior University Method of treating hyperproliferative vascular disease
MX9304868A (es) * 1992-08-13 1994-05-31 American Home Prod 27-hidroxirapamicina, derivados de la misma y composicion farmaceutica que la contiene.
GB9221220D0 (en) * 1992-10-09 1992-11-25 Sandoz Ag Organic componds
US5342348A (en) 1992-12-04 1994-08-30 Kaplan Aaron V Method and device for treating and enlarging body lumens
US5419760A (en) 1993-01-08 1995-05-30 Pdt Systems, Inc. Medicament dispensing stent for prevention of restenosis of a blood vessel
HUT73101A (en) 1993-02-12 1996-06-28 Harvard College Method for regulating transcription of targeted genes and other biological events
US5464650A (en) 1993-04-26 1995-11-07 Medtronic, Inc. Intravascular stent and method
CA2167282A1 (en) 1993-07-16 1995-01-26 Gerald R. Crabtree Regulated apoptosis
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5387680A (en) 1993-08-10 1995-02-07 American Home Products Corporation C-22 ring stabilized rapamycin derivatives
US5409000A (en) 1993-09-14 1995-04-25 Cardiac Pathways Corporation Endocardial mapping and ablation system utilizing separately controlled steerable ablation catheter with ultrasonic imaging capabilities and method
US5616588A (en) 1993-09-30 1997-04-01 American Home Products Corporation Rapamycin formulation for IV injection
US5516770A (en) 1993-09-30 1996-05-14 American Home Products Corporation Rapamycin formulation for IV injection
IL111004A (en) 1993-09-30 1998-06-15 American Home Prod Oral formulations of rapamycin
AU688782B2 (en) 1993-09-30 1998-03-19 Wyeth Rapamycin formulations for oral administration
US5536729A (en) 1993-09-30 1996-07-16 American Home Products Corporation Rapamycin formulations for oral administration
US5463010A (en) 1993-11-12 1995-10-31 Surface Engineering Technologies, Division Of Innerdyne, Inc. Hydrocyclosiloxane membrane prepared by plasma polymerization process
JP4105761B2 (ja) 1993-11-19 2008-06-25 アボット・ラボラトリーズ ラパミシン(マクロライド)の半合成類似体免疫調節剤
US5527907A (en) 1993-11-19 1996-06-18 Abbott Laboratories Macrolide immunomodulators
WO1995014865A1 (en) 1993-11-23 1995-06-01 Alliedsignal Inc. Frictionless resilient bearing mount
US5525610A (en) 1994-03-31 1996-06-11 American Home Products Corporation 42-Epi-rapamycin and pharmaceutical compositions thereof
US5362718A (en) 1994-04-18 1994-11-08 American Home Products Corporation Rapamycin hydroxyesters
US6150137A (en) 1994-05-27 2000-11-21 Ariad Pharmaceuticals, Inc. Immunosuppressant target proteins
US5660873A (en) 1994-09-09 1997-08-26 Bioseal, Limited Liability Corporaton Coating intraluminal stents
US5891108A (en) 1994-09-12 1999-04-06 Cordis Corporation Drug delivery stent
US5649977A (en) 1994-09-22 1997-07-22 Advanced Cardiovascular Systems, Inc. Metal reinforced polymer stent
JP3942201B2 (ja) 1994-11-18 2007-07-11 株式会社カネカ フェニルポリシルセスキオキサンの製造方法
US5563145A (en) * 1994-12-07 1996-10-08 American Home Products Corporation Rapamycin 42-oximes and hydroxylamines
US5637113A (en) 1994-12-13 1997-06-10 Advanced Cardiovascular Systems, Inc. Polymer film for wrapping a stent structure
ATE544776T1 (de) 1994-12-29 2012-02-15 Massachusetts Inst Technology Chimäre dna-bindeproteine
US5496832A (en) 1995-03-09 1996-03-05 American Home Products Corporation Method of treating cardiac inflammatory disease
CA2178541C (en) 1995-06-07 2009-11-24 Neal E. Fearnot Implantable medical device
US5609629A (en) 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
CA2219080A1 (en) 1995-06-07 1996-12-27 Ariad Gene Therapeutics, Inc. Rapamycin-based regulation of biological events
IL122212A (en) * 1995-06-09 2001-08-26 Novartis Ag Rapamycin derivatives, pharmaceutical compositions comprising them and their preparation
ATE286133T1 (de) 1995-07-06 2005-01-15 Univ Leland Stanford Junior Zellfreie synthese von polyketiden
FR2736550B1 (fr) 1995-07-14 1998-07-24 Sandoz Sa Composition pharmaceutique sous la forme d'une dispersion solide comprenant un macrolide et un vehicule
JP3324360B2 (ja) 1995-09-25 2002-09-17 信越化学工業株式会社 ポリシロキサン化合物及びポジ型レジスト材料
US5879808A (en) 1995-10-27 1999-03-09 Alpha Metals, Inc. Parylene polymer layers
US5725494A (en) 1995-11-30 1998-03-10 Pharmasonics, Inc. Apparatus and methods for ultrasonically enhanced intraluminal therapy
US5728062A (en) 1995-11-30 1998-03-17 Pharmasonics, Inc. Apparatus and methods for vibratory intraluminal therapy employing magnetostrictive transducers
US5735811A (en) 1995-11-30 1998-04-07 Pharmasonics, Inc. Apparatus and methods for ultrasonically enhanced fluid delivery
CA2199890C (en) * 1996-03-26 2002-02-05 Leonard Pinchuk Stents and stent-grafts having enhanced hoop strength and methods of making the same
US5928145A (en) 1996-04-25 1999-07-27 The Johns Hopkins University Method of magnetic resonance imaging and spectroscopic analysis and associated apparatus employing a loopless antenna
US5951586A (en) 1996-05-15 1999-09-14 Medtronic, Inc. Intraluminal stent
AU735648B2 (en) 1996-07-12 2001-07-12 Ariad Pharmaceuticals, Inc. Materials and method for treating or preventing pathogenic fungal infection
US6168695B1 (en) 1999-07-12 2001-01-02 Daniel J. Woodruff Lift and rotate assembly for use in a workpiece processing station and a method of attaching the same
WO1998009970A2 (en) 1996-09-09 1998-03-12 American Home Products Corporation Alkylated rapamycin derivatives
AU4246197A (en) 1996-09-09 1998-03-26 American Home Products Corporation Rapamycin derivatives with unnatural stereochemistries
US6099561A (en) 1996-10-21 2000-08-08 Inflow Dynamics, Inc. Vascular and endoluminal stents with improved coatings
ZA9710342B (en) 1996-11-25 1998-06-10 Alza Corp Directional drug delivery stent and method of use.
US5980551A (en) 1997-02-07 1999-11-09 Endovasc Ltd., Inc. Composition and method for making a biodegradable drug delivery stent
US6051276A (en) 1997-03-14 2000-04-18 Alpha Metals, Inc. Internally heated pyrolysis zone
US5989591A (en) 1997-03-14 1999-11-23 American Home Products Corporation Rapamycin formulations for oral administration
US5843172A (en) 1997-04-15 1998-12-01 Advanced Cardiovascular Systems, Inc. Porous medicated stent
RU2184541C2 (ru) 1997-06-13 2002-07-10 Американ Хоум Продактс Корпорейшн Препараты рапамицина для орального применения
US5985325A (en) 1997-06-13 1999-11-16 American Home Products Corporation Rapamycin formulations for oral administration
WO1999010510A2 (en) 1997-08-26 1999-03-04 Ariad Gene Therapeutics, Inc. Fusion proteins comprising a dimerization, trimerization or tetramerization domain and an additional heterologous transcription activation, transcription repression, dna binding or ligand binding domain
US5780604A (en) * 1997-09-26 1998-07-14 Abbott Laboratories 11,12-cyclic phosphite or phosphate derivatives of erythromycin and related macrolides
US5972027A (en) 1997-09-30 1999-10-26 Scimed Life Systems, Inc Porous stent drug delivery system
US6031375A (en) 1997-11-26 2000-02-29 The Johns Hopkins University Method of magnetic resonance analysis employing cylindrical coordinates and an associated apparatus
WO1999036553A2 (en) 1998-01-15 1999-07-22 Ariad Gene Therapeutics, Inc. Regulation of biological events using multimeric chimeric proteins
US6623521B2 (en) 1998-02-17 2003-09-23 Md3, Inc. Expandable stent with sliding and locking radial elements
US7208010B2 (en) 2000-10-16 2007-04-24 Conor Medsystems, Inc. Expandable medical device for delivery of beneficial agent
US5980566A (en) 1998-04-11 1999-11-09 Alt; Eckhard Vascular and endoluminal stents with iridium oxide coating
US20010029351A1 (en) 1998-04-16 2001-10-11 Robert Falotico Drug combinations and delivery devices for the prevention and treatment of vascular disease
US8029561B1 (en) 2000-05-12 2011-10-04 Cordis Corporation Drug combination useful for prevention of restenosis
EP1085846A2 (en) * 1998-06-08 2001-03-28 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
US6153252A (en) 1998-06-30 2000-11-28 Ethicon, Inc. Process for coating stents
US6087064A (en) 1998-09-03 2000-07-11 International Business Machines Corporation Silsesquioxane polymers, method of synthesis, photoresist composition, and multilayer lithographic method
US6063101A (en) 1998-11-20 2000-05-16 Precision Vascular Systems, Inc. Stent apparatus and method
US7125875B2 (en) 1999-04-15 2006-10-24 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
US6872225B1 (en) 1999-05-27 2005-03-29 Biocompatibles Uk Limited Local drug delivery
CA2374944A1 (en) 1999-06-10 2000-12-21 Nigel Hacker Spin-on-glass anti-reflective coatings for photolithography
US6890448B2 (en) 1999-06-11 2005-05-10 Shipley Company, L.L.C. Antireflective hard mask compositions
DE60009493T2 (de) 1999-06-24 2005-03-17 Abbott Vascular Devices Ltd. Mittels eines ballons expandierbarer stent
JP4187879B2 (ja) 1999-08-06 2008-11-26 東京応化工業株式会社 感放射線レジスト組成物
DE60010098T2 (de) 1999-08-24 2005-03-31 Ariad Gene Therapeutics, Inc., Cambridge 28-epirapaloge
WO2001047572A2 (en) 1999-12-29 2001-07-05 Advanced Cardiovascular Systems, Inc. Device and active component for inhibiting formation of thrombus-inflammatory cell matrix
US6471979B2 (en) 1999-12-29 2002-10-29 Estrogen Vascular Technology, Llc Apparatus and method for delivering compounds to a living organism
AU2623201A (en) 1999-12-30 2001-07-16 Kam W Leong Controlled delivery of therapeutic agents by insertable medical devices
WO2001052915A1 (en) 2000-01-24 2001-07-26 Biocompatibles Limited Coated implants
US6776796B2 (en) 2000-05-12 2004-08-17 Cordis Corportation Antiinflammatory drug and delivery device
CA2408754C (en) 2000-05-12 2011-01-04 Cordis Corporation Delivery devices for treatment of vascular disease
US20020013335A1 (en) 2000-06-16 2002-01-31 American Home Products Corporation Method of treating cardiovascular disease
ATE311892T1 (de) 2000-07-10 2005-12-15 Univ Mississippi Hochaktive immunstimulierende mittel aus mikroalgen
JP4622061B2 (ja) 2000-07-27 2011-02-02 Jsr株式会社 レジスト下層膜用組成物およびその製造方法
TW556047B (en) 2000-07-31 2003-10-01 Shipley Co Llc Coated substrate, method for forming photoresist relief image, and antireflective composition
JP4141625B2 (ja) 2000-08-09 2008-08-27 東京応化工業株式会社 ポジ型レジスト組成物およびそのレジスト層を設けた基材
ES2228932T3 (es) 2000-08-11 2005-04-16 Wyeth Procedimiento de tratamiento del carcinoma positivo de estrogenos.
ATE411321T1 (de) 2000-09-19 2008-10-15 Wyeth Corp Wasserlösliche rapamycin-ester
TWI286074B (en) * 2000-11-15 2007-09-01 Wyeth Corp Pharmaceutical composition containing CCI-779 as an antineoplastic agent
US6471980B2 (en) 2000-12-22 2002-10-29 Avantec Vascular Corporation Intravascular delivery of mycophenolic acid
US20030033007A1 (en) * 2000-12-22 2003-02-13 Avantec Vascular Corporation Methods and devices for delivery of therapeutic capable agents with variable release profile
GB0100760D0 (en) 2001-01-11 2001-02-21 Biocompatibles Ltd Drug delivery from stents
GB0100761D0 (en) 2001-01-11 2001-02-21 Biocompatibles Ltd Drug delivery from stents
US20020119178A1 (en) 2001-02-23 2002-08-29 Luc Levesque Drug eluting device for treating vascular diseases
ES2312568T3 (es) 2001-04-06 2009-03-01 Wyeth Combinaciones antineoplasicas que comprenden cci-779 (derivado de rapamicina) junto con gemcitabina o fluoruracilo.
TW594416B (en) 2001-05-08 2004-06-21 Shipley Co Llc Photoimageable composition
GB0123400D0 (en) 2001-09-28 2001-11-21 Novartis Ag Organic compounds
WO2003057218A1 (en) 2002-01-10 2003-07-17 Novartis Ag Drug delivery systems for the prevention and treatment of vascular diseases comprising rapamycin and derivatives thereof
BR0307544A (pt) * 2002-02-01 2004-12-07 Ariad Gene Therapeutics Inc Compostos contendo fósforos, componentes e composição bem como, sua utilização através de métodos de tratamentos
US7432277B2 (en) * 2002-02-01 2008-10-07 Araid Gene Therapeutics, Inc. Phosphorus-containing macrocycles
JP4557497B2 (ja) 2002-03-03 2010-10-06 ローム・アンド・ハース・エレクトロニック・マテリアルズ,エル.エル.シー. シランモノマー及びポリマーを製造する方法及びそれを含むフォトレジスト組成物
US20060094674A1 (en) * 2002-07-05 2006-05-04 Neel Benjamin G Combination of mtor inhibitor and a tyrosine kinase inhibitor for the treatment of neoplasms
AU2003261100A1 (en) * 2002-07-25 2004-02-16 Avantec Vascular Corporation Devices delivering therapeutic agents and methods regarding the same
AU2003232653A1 (en) * 2002-07-26 2004-02-16 Samsung Electronics Co., Ltd. Upper substrate, liquid crystal display apparatus having the same and method of fabricating the same
US20040077677A1 (en) 2002-09-17 2004-04-22 Wyeth Oral formulations
WO2005016252A2 (en) * 2003-07-11 2005-02-24 Ariad Gene Therapeutics, Inc. Phosphorus-containing macrocycles
EP1658080A1 (en) 2003-08-18 2006-05-24 Pfizer Products Inc. Dosing schedule for a erbb2 anticancer agents
AR045957A1 (es) 2003-10-03 2005-11-16 Novartis Ag Composicion farmaceutica y combinacion
GB0327840D0 (en) 2003-12-01 2003-12-31 Novartis Ag Organic compounds
CA2581372A1 (en) 2004-09-30 2006-04-13 Ariad Gene Therapeutics, Inc. Treatment method
US7528145B2 (en) 2004-10-28 2009-05-05 Wyeth Use of an mTOR inhibitor in treatment of uterine leiomyoma
US20060194829A1 (en) 2004-12-20 2006-08-31 Clackson Timothy P Therapeutic materials and methods
US20060160837A1 (en) 2004-12-29 2006-07-20 The Brigham And Women's Hospital, Inc. Rapamycin compounds in the treatment of neurofibromatosis type 1
JP2008530145A (ja) 2005-02-15 2008-08-07 ワイス 経口投与可能なcci−779製剤
US20070004767A1 (en) * 2005-06-30 2007-01-04 Gutmann David H Methods for treating neurofibromatosis 1
WO2007056117A1 (en) 2005-11-04 2007-05-18 Wyeth Antineoplastic combinations of temsirolimus and sunitinib malate
DE102010042183A1 (de) 2010-10-08 2012-04-12 Robert Bosch Gmbh Hybridantriebseinrichtung
TWM483731U (zh) 2013-12-31 2014-08-11 Yong-Hua Chen 辦公椅結構
US9408008B2 (en) 2014-02-28 2016-08-02 Sonos, Inc. Playback zone representations
US20160120174A1 (en) 2014-10-29 2016-05-05 Ronald Steven Cok Imprinted multi-layer biocidal particle structure

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5206018A (en) * 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
US5068091A (en) * 1988-02-22 1991-11-26 Kabushiki Kaisha Tiyoda Seisakusho Device for dyeing tissues for immune response observation
US6146358A (en) * 1989-03-14 2000-11-14 Cordis Corporation Method and apparatus for delivery of therapeutic agent
US5234456A (en) * 1990-02-08 1993-08-10 Pfizer Hospital Products Group, Inc. Hydrophilic stent
US5851217A (en) * 1990-02-28 1998-12-22 Medtronic, Inc. Intralumenal drug eluting prosthesis
US5516781A (en) * 1992-01-09 1996-05-14 American Home Products Corporation Method of treating restenosis with rapamycin
US5434260A (en) * 1992-10-13 1995-07-18 American Home Products Corporation Carbamates of rapamycin
US5489680A (en) * 1992-10-13 1996-02-06 American Home Products Corporation Carbamates of rapamycin
US5310903A (en) * 1993-03-05 1994-05-10 Merck & Co., Inc. Imidazolidyl rapamycin derivatives
US20010010920A1 (en) * 1993-04-23 2001-08-02 Molnar-Kimber Katherine L. Monoclonal antibodies obtained using rapamycin position 27 conjugates as an immunogen
US5391730A (en) * 1993-10-08 1995-02-21 American Home Products Corporation Phosphorylcarbamates of rapamycin and oxime derivatives thereof
US5385910A (en) * 1993-11-22 1995-01-31 American Home Products Corporation Gem-distributed esters of rapamycin
US6152141A (en) * 1994-07-28 2000-11-28 Heartport, Inc. Method for delivery of therapeutic agents to the heart
US5491231A (en) * 1994-11-28 1996-02-13 American Home Products Corporation Hindered N-oxide esters of rapamycin
US5665591A (en) * 1994-12-06 1997-09-09 Trustees Of Boston University Regulation of smooth muscle cell proliferation
US6585764B2 (en) * 1997-04-18 2003-07-01 Cordis Corporation Stent with therapeutically active dosage of rapamycin coated thereon

Cited By (107)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8058426B2 (en) 2002-02-01 2011-11-15 Ariad Pharmaceuticals, Inc. Phosphorus-containing compounds and uses thereof
US20100197907A1 (en) * 2002-02-01 2010-08-05 Ariad Pharmaceuticals, Inc. Phosphorus-containing compounds & uses thereof
US9024014B2 (en) 2002-02-01 2015-05-05 Ariad Pharmaceuticals, Inc. Phosphorus-containing compounds and uses thereof
US7709020B2 (en) * 2002-02-01 2010-05-04 Ariad Pharmaceuticals, Inc. Implantable device comprising phosphorus-containing macrolides
US20070190106A1 (en) * 2002-02-01 2007-08-16 Berstein David L Phosphorus-containing compounds & uses thereof
US20040110813A1 (en) * 2002-09-24 2004-06-10 Boehringer Ingelheim International Gmbh Solid telmisartan pharmaceutical formulations
US8980870B2 (en) * 2002-09-24 2015-03-17 Boehringer Ingelheim International Gmbh Solid telmisartan pharmaceutical formulations
US20040202789A1 (en) * 2003-03-31 2004-10-14 Council Of Scientific And Industrila Research Process for preparing thin film solids
US20060173033A1 (en) * 2003-07-08 2006-08-03 Michaela Kneissel Use of rapamycin and rapamycin derivatives for the treatment of bone loss
US20050049693A1 (en) * 2003-08-25 2005-03-03 Medtronic Vascular Inc. Medical devices and compositions for delivering biophosphonates to anatomical sites at risk for vascular disease
US20050187608A1 (en) * 2004-02-24 2005-08-25 O'hara Michael D. Radioprotective compound coating for medical devices
WO2005110480A3 (en) * 2004-05-17 2006-07-27 Novartis Ag Combination of organic compounds
WO2005110480A2 (en) * 2004-05-17 2005-11-24 Novartis Ag Combination of organic compounds
US8226977B2 (en) 2004-06-04 2012-07-24 Teva Pharmaceutical Industries Ltd. Pharmaceutical composition containing irbesartan
US8414920B2 (en) 2004-06-04 2013-04-09 Teva Pharmaceutical Industries Ltd. Pharmaceutical composition containing irbesartan
US8858978B2 (en) 2004-09-28 2014-10-14 Atrium Medical Corporation Heat cured gel and method of making
US10814043B2 (en) 2004-09-28 2020-10-27 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US8263102B2 (en) 2004-09-28 2012-09-11 Atrium Medical Corporation Drug delivery coating for use with a stent
US9682175B2 (en) 2004-09-28 2017-06-20 Atrium Medical Corporation Coating material and medical device system including same
US8312836B2 (en) 2004-09-28 2012-11-20 Atrium Medical Corporation Method and apparatus for application of a fresh coating on a medical device
US8367099B2 (en) 2004-09-28 2013-02-05 Atrium Medical Corporation Perforated fatty acid films
US20060067983A1 (en) * 2004-09-28 2006-03-30 Atrium Medical Corporation Stand-alone film and methods for making the same
US11793912B2 (en) 2004-09-28 2023-10-24 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US9801913B2 (en) 2004-09-28 2017-10-31 Atrium Medical Corporation Barrier layer
US9012506B2 (en) 2004-09-28 2015-04-21 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US10869902B2 (en) 2004-09-28 2020-12-22 Atrium Medical Corporation Cured gel and method of making
US8795703B2 (en) 2004-09-28 2014-08-05 Atrium Medical Corporation Stand-alone film and methods for making the same
US9801982B2 (en) 2004-09-28 2017-10-31 Atrium Medical Corporation Implantable barrier device
US9000040B2 (en) 2004-09-28 2015-04-07 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US10792312B2 (en) 2004-09-28 2020-10-06 Atrium Medical Corporation Barrier layer
US9827352B2 (en) 2004-09-28 2017-11-28 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US10772995B2 (en) 2004-09-28 2020-09-15 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US10016465B2 (en) 2004-09-28 2018-07-10 Atrium Medical Corporation Cured gel and method of making
US20060188542A1 (en) * 2005-02-22 2006-08-24 Bobyn John D Implant improving local bone formation
US8071574B2 (en) 2005-02-22 2011-12-06 John Dennis Bobyn Implant improving local bone formation
US8309536B2 (en) 2005-02-22 2012-11-13 John Dennis Bobyn Implant improving local bone formation
US9278161B2 (en) 2005-09-28 2016-03-08 Atrium Medical Corporation Tissue-separating fatty acid adhesion barrier
US11083823B2 (en) 2005-09-28 2021-08-10 Atrium Medical Corporation Tissue-separating fatty acid adhesion barrier
US8124127B2 (en) * 2005-10-15 2012-02-28 Atrium Medical Corporation Hydrophobic cross-linked gels for bioabsorbable drug carrier coatings
US8501229B2 (en) 2005-10-15 2013-08-06 Atrium Medical Corporation Hydrophobic cross-linked gels for bioabsorbable drug carrier coatings
US20080032989A1 (en) * 2006-05-31 2008-02-07 Robinson William H Method of treating inflammatory diseases using tyroskine kinase inhibitors
US20140227254A1 (en) * 2006-11-02 2014-08-14 Acceleron Pharma, Inc. Antagonists of bmp9, bmp10, alk1 and other alk1 ligands, and uses thereof
US9452197B2 (en) * 2006-11-02 2016-09-27 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
US9492596B2 (en) 2006-11-06 2016-11-15 Atrium Medical Corporation Barrier layer with underlying medical device and one or more reinforcing support structures
US9592324B2 (en) 2006-11-06 2017-03-14 Atrium Medical Corporation Tissue separating device with reinforced support for anchoring mechanisms
US8574627B2 (en) 2006-11-06 2013-11-05 Atrium Medical Corporation Coated surgical mesh
US8496967B2 (en) 2006-11-14 2013-07-30 Ariad Pharmaceuticals, Inc. Oral formulations
US11166929B2 (en) 2009-03-10 2021-11-09 Atrium Medical Corporation Fatty-acid based particles
US10285964B2 (en) 2009-03-10 2019-05-14 Atrium Medical Corporation Fatty-acid based particles
US9427423B2 (en) 2009-03-10 2016-08-30 Atrium Medical Corporation Fatty-acid based particles
US10864304B2 (en) 2009-08-11 2020-12-15 Atrium Medical Corporation Anti-infective antimicrobial-containing biomaterials
US11097035B2 (en) 2010-07-16 2021-08-24 Atrium Medical Corporation Compositions and methods for altering the rate of hydrolysis of cured oil-based materials
US10322213B2 (en) 2010-07-16 2019-06-18 Atrium Medical Corporation Compositions and methods for altering the rate of hydrolysis of cured oil-based materials
US11793819B2 (en) 2011-11-23 2023-10-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8987237B2 (en) 2011-11-23 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8633178B2 (en) 2011-11-23 2014-01-21 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11103516B2 (en) 2011-11-23 2021-08-31 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9248136B2 (en) 2011-11-23 2016-02-02 Therapeuticsmd, Inc. Transdermal hormone replacement therapies
US8846649B2 (en) 2011-11-23 2014-09-30 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10675288B2 (en) 2011-11-23 2020-06-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8846648B2 (en) 2011-11-23 2014-09-30 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9867880B2 (en) 2012-06-13 2018-01-16 Atrium Medical Corporation Cured oil-hydrogel biomaterial compositions for controlled drug delivery
US10888617B2 (en) 2012-06-13 2021-01-12 Atrium Medical Corporation Cured oil-hydrogel biomaterial compositions for controlled drug delivery
US11110099B2 (en) 2012-06-18 2021-09-07 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10052386B2 (en) 2012-06-18 2018-08-21 Therapeuticsmd, Inc. Progesterone formulations
US10471148B2 (en) 2012-06-18 2019-11-12 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US11865179B2 (en) 2012-06-18 2024-01-09 Therapeuticsmd, Inc. Progesterone formulations having a desirable PK profile
US9289382B2 (en) 2012-06-18 2016-03-22 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US9301920B2 (en) 2012-06-18 2016-04-05 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10639375B2 (en) 2012-06-18 2020-05-05 Therapeuticsmd, Inc. Progesterone formulations
US11529360B2 (en) 2012-06-18 2022-12-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11166963B2 (en) 2012-06-18 2021-11-09 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8933059B2 (en) 2012-06-18 2015-01-13 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9012434B2 (en) 2012-06-18 2015-04-21 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US10806740B2 (en) 2012-06-18 2020-10-20 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US9006222B2 (en) 2012-06-18 2015-04-14 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US8987238B2 (en) 2012-06-18 2015-03-24 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11033626B2 (en) 2012-06-18 2021-06-15 Therapeuticsmd, Inc. Progesterone formulations having a desirable pk profile
US11065197B2 (en) 2012-12-21 2021-07-20 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11304959B2 (en) 2012-12-21 2022-04-19 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10888516B2 (en) 2012-12-21 2021-01-12 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US10537581B2 (en) 2012-12-21 2020-01-21 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10568891B2 (en) 2012-12-21 2020-02-25 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10835487B2 (en) 2012-12-21 2020-11-17 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10471072B2 (en) 2012-12-21 2019-11-12 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11622933B2 (en) 2012-12-21 2023-04-11 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11497709B2 (en) 2012-12-21 2022-11-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11351182B2 (en) 2012-12-21 2022-06-07 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10806697B2 (en) 2012-12-21 2020-10-20 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11266661B2 (en) 2012-12-21 2022-03-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11116717B2 (en) 2012-12-21 2021-09-14 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11123283B2 (en) 2012-12-21 2021-09-21 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US9180091B2 (en) 2012-12-21 2015-11-10 Therapeuticsmd, Inc. Soluble estradiol capsule for vaginal insertion
US11246875B2 (en) 2012-12-21 2022-02-15 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US11241445B2 (en) 2012-12-21 2022-02-08 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10206932B2 (en) 2014-05-22 2019-02-19 Therapeuticsmd, Inc. Natural combination hormone replacement formulations and therapies
US11103513B2 (en) 2014-05-22 2021-08-31 TherapeuticsMD Natural combination hormone replacement formulations and therapies
US10398708B2 (en) 2014-10-22 2019-09-03 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10668082B2 (en) 2014-10-22 2020-06-02 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10258630B2 (en) 2014-10-22 2019-04-16 Therapeuticsmd, Inc. Vaginal inserted estradiol pharmaceutical compositions and methods
US10328087B2 (en) 2015-07-23 2019-06-25 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US10912783B2 (en) 2015-07-23 2021-02-09 Therapeuticsmd, Inc. Formulations for solubilizing hormones
US9931349B2 (en) 2016-04-01 2018-04-03 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US10286077B2 (en) 2016-04-01 2019-05-14 Therapeuticsmd, Inc. Steroid hormone compositions in medium chain oils
US10532059B2 (en) 2016-04-01 2020-01-14 Therapeuticsmd, Inc. Steroid hormone pharmaceutical composition
US11633405B2 (en) 2020-02-07 2023-04-25 Therapeuticsmd, Inc. Steroid hormone pharmaceutical formulations

Also Published As

Publication number Publication date
CN100522967C (zh) 2009-08-05
EA011488B1 (ru) 2009-04-28
KR20090118118A (ko) 2009-11-17
US7091213B2 (en) 2006-08-15
JP2005516065A (ja) 2005-06-02
EP1478648A4 (en) 2006-06-14
JP4547911B2 (ja) 2010-09-22
US20100197907A1 (en) 2010-08-05
CN101717410A (zh) 2010-06-02
WO2003064383A3 (en) 2004-01-22
US20060264456A1 (en) 2006-11-23
IL199153A (en) 2010-11-30
MXPA04007402A (es) 2005-06-17
US20070190106A1 (en) 2007-08-16
AU2009202913A1 (en) 2009-08-06
PL216224B1 (pl) 2014-03-31
DK1478648T3 (da) 2014-07-28
EA200700193A1 (ru) 2007-12-28
KR100956195B1 (ko) 2010-05-06
AU2009202913B2 (en) 2012-09-13
US9024014B2 (en) 2015-05-05
PT1478648E (pt) 2014-07-15
US8058426B2 (en) 2011-11-15
EP1478648B1 (en) 2014-04-30
AU2003210787B2 (en) 2009-04-23
KR101004224B1 (ko) 2010-12-27
EP2407473A3 (en) 2012-03-21
EP2407473A2 (en) 2012-01-18
EP1478648A2 (en) 2004-11-24
CA2472341A1 (en) 2003-08-07
IL162734A (en) 2009-11-18
US20120178714A1 (en) 2012-07-12
EA200401008A1 (ru) 2005-02-24
ES2485841T3 (es) 2014-08-14
PL371526A1 (en) 2005-06-27
US7709020B2 (en) 2010-05-04
WO2003064383A2 (en) 2003-08-07
CN1646537A (zh) 2005-07-27
BR0307544A (pt) 2004-12-07
CN101717410B (zh) 2015-04-29
US20050032825A1 (en) 2005-02-10
CA2472341C (en) 2011-06-21
EA008379B1 (ru) 2007-04-27
US20040073024A1 (en) 2004-04-15
KR20040083104A (ko) 2004-09-30
US7186826B2 (en) 2007-03-06
IL162734A0 (en) 2005-11-20

Similar Documents

Publication Publication Date Title
US8058426B2 (en) Phosphorus-containing compounds and uses thereof
AU2003210787A1 (en) Phosphorus-containing compounds & uses thereof
JP2005516065A6 (ja) リン含有化合物およびその用途
US20050026868A1 (en) Phosphorus-containing macrocycles
AU2013296627C1 (en) Deuterated ibrutinib
KR20090035716A (ko) 라파마이신 유사체의 결정형 형태
KR20230154191A (ko) 신규 방법
US7432277B2 (en) Phosphorus-containing macrocycles

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARIAD GENE THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:METCALF, III, CHESTER A.;ROZAMUS, LEONARD W.;WANG, YIHAN;AND OTHERS;REEL/FRAME:014100/0777;SIGNING DATES FROM 20030509 TO 20030521

STCB Information on status: application discontinuation

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