US20120088734A1 - Cyclosporin Analogs - Google Patents

Cyclosporin Analogs Download PDF

Info

Publication number
US20120088734A1
US20120088734A1 US13/270,964 US201113270964A US2012088734A1 US 20120088734 A1 US20120088734 A1 US 20120088734A1 US 201113270964 A US201113270964 A US 201113270964A US 2012088734 A1 US2012088734 A1 US 2012088734A1
Authority
US
United States
Prior art keywords
sar
cyclosporin
chr
amide
ethyl
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
US13/270,964
Inventor
Catherine Simone Victoire Frydrych
William Robert Carling
Michael E. Garst
Michael E. Stern
Christopher S. Schaumburg
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.)
Allergan Inc
Original Assignee
Allergan 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 Allergan Inc filed Critical Allergan Inc
Priority to US13/270,964 priority Critical patent/US20120088734A1/en
Assigned to ALLERGAN, INC. reassignment ALLERGAN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GARST, MICHAEL E., SCHAUMBURG, CHRISTOPHER S., STERN, MICHAEL E., CARLING, WILLIAM ROBERT, FRYDRYCH, CATHERINE SIMONE VICTOIRE
Publication of US20120088734A1 publication Critical patent/US20120088734A1/en
Priority to US14/534,677 priority patent/US9493511B2/en
Priority to US15/290,171 priority patent/US10065992B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • C07K7/645Cyclosporins; Related peptides
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/04Artificial tears; Irrigation solutions
    • 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
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/113General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • Cyclosporins are a class of poly-N-methylated cyclic undecapeptides. There are naturally occurring cyclosporins such as cyclosporin A, and non-natural cyclosporin (“Cs”) derivatives.
  • Cyclosporin A for example, has the following structure:
  • the claimed invention relates to novel compounds of the following formula (I) and pharmaceutically acceptable salts thereof:
  • Cs scaffolds refers to different cyclosporins (e.g., Cs A, Cs C, Cs D, etc.) that vary from each other in the identities of one or more of substituents A, B, C, and D.
  • a “Cs scaffold” refers to a novel compound of formula (I) less the moiety at the ⁇ -carbon of the 3-position amino acid.
  • A, B, C, and D are defined as follows.
  • A represents —CH ⁇ CHR, —CH ⁇ CH—CH ⁇ CHR, or —CH 2 CH 2 R, wherein R represents
  • n-propyl i.e., n-propyl
  • D represents —CH 3 or —CH 2 OH.
  • A is —CH ⁇ CHCH 3
  • B is —CH 2 CH 3
  • C is isobutyl
  • D is —CH 3 , as illustrated below in formula (IA):
  • A is —CH ⁇ CHCH 3
  • B is 1-hydroxyethyl
  • C is isobutyl
  • D is —CH 3 , as illustrated below in formula (IB):
  • A is —CH ⁇ CHCH 3
  • B is isopropyl
  • C is isobutyl
  • D is —CH 3 , as illustrated below in formula (IC):
  • A is —CH ⁇ CHCH 3
  • B is n-propyl
  • C is isobutyl
  • D is —CH 3 , as illustrated below in formula (ID):
  • A is —CH ⁇ CHCH 3
  • B is —CH 2 CH 3
  • C is 1-methylpropyl
  • D is —CH 3 , as illustrated below in formula (IE):
  • A is —CH ⁇ CHCH 3
  • B is —CH 2 CH 3
  • C is isobutyl
  • D is —CH 2 OH, as illustrated below in formula (IF):
  • A is —CH ⁇ CHCH 3
  • B is —CH 2 CH 3
  • C is 2-hydroxy isobutyl
  • D is —CH 3 , as illustrated below in formula (IG):
  • the amino acids of various types of cyclosporine scaffold are labeled numerically from 1 to 11.
  • the modifications of the invention occur at the ⁇ -carbon of the position-3 amino acid (sarcosine at the 3-position) of the Cs scaffold.
  • the modification generally comprises replacement of a hydrogen atom at the ⁇ -carbon of the position-3 amino acid with a moiety in formula (I) wherein:
  • R 1 and R 2 which are identical or different, each represents hydrogen or C 1 -C 4 alkyl, or together represent C 3 -C 7 cycloalkyl;
  • X represents sulfur or —S(O) n , wherein n is 1 or 2;
  • R 3 represents hydrogen; straight or branched C 1 -C 6 alkyl; straight or branched C 2 -C 6 alkenyl; straight or branched C 2 -C 6 alkynyl; C 3 -C 7 cycloalkyl; C 4 -C 7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl;
  • n 1, 2, 3, 4, 5, or 6;
  • each R′ is independently H, straight or branched C 1 -C 6 alkyl, straight or branched C 2 -C 6 alkenyl; straight or branched C 2 -C 6 alkynyl; C 3 -C 7 cycloalkyl; C 4 -C 7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl; or is absent (permitting, in such case, —(CHR′) m to represent an unsaturated moiety such as —CH—CH).
  • R 3 may be optionally substituted with one or more groups, identical or different, of C 1 -C 6 alkyl; halogen; hydroxyl; alkoxycarbonyl; carboxyl; cycloalkyl; saturated or partially unsaturated 5-6 member heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur, which heterocyclyl is optionally substituted by one or more groups of C 1 -C 6 alkyl; aryl; heteroaryl; amino; monoalkylamino; dialkylamino; amidino; guanidine; or urea.
  • Embodiments of formula (I) include, but are not limited to, the following compounds of formula (I) (only the moiety at the ⁇ -carbon of the position-3 amino acid is shown; wavy lines represent the rest of the Cs compounds of formula (I)):
  • the present invention also relates to novel intermediates useful for the preparation of compounds of formula (I).
  • the novel intermediates are compounds of the following formula (III):
  • the present invention also relates to a process for the preparation of a compound of formula (III), comprising:
  • Alkyl refers to a monovalent linear or branched hydrocarbon radical having 1 to 6 carbon atoms. Examples include, but are not limited to, methyl, ethyl, propyl (e.g., 1-propyl, isopropyl), butyl (e.g., 1-butyl, isobutyl, sec-butyl, tert-butyl), pentyl (e.g., 1-pentyl, neopentyl), and hexyl (e.g., 3-hexyl).
  • propyl e.g., 1-propyl, isopropyl
  • butyl e.g., 1-butyl, isobutyl, sec-butyl, tert-butyl
  • pentyl e.g., 1-pentyl, neopentyl
  • hexyl e.g., 3-hexyl
  • Alkenyl refers to a monvalent linear or branched hydrocarbon radical having 2 to 6 carbon atoms and one or more double bonds. Examples include, but are not limited to, ethenyl, propenyl, and butenyl.
  • Alkynyl refers to a monovalent linear or branched hydrocarbon radical having 2 to 6 carbon atoms and one or more triple bonds. Examples include, but are not limited to, ethynyl, propynyl and butynyl.
  • Cycloalkyl refers to monovalent saturated or partially unsaturated cyclic hydrocarbon radical having 3 to 6 carbon atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • Heterocyclyl refers to monovalent, saturated or partially unsaturated cyclic hydrocarbon radical having 3 to 6 ring atoms, at least one of which is a heteroatom selected from nitrogen, oxygen and sulfur.
  • the radical may be on a carbon or a heteroatom. Examples include, but are not limited to, morpholinyl, piperidinyl, pyrrolidinyl, pyranyl, and pyrazolinyl.
  • Aryl refers to monovalent 5-7 member aromatic hydrocarbon radical. Examples include, but are not limited to, phenyl.
  • Heteroaryl refers to monovalent 5-7 member aromatic hydrocarbon radical having one or more heteroatoms selected from nitrogen, sulfur and oxygen. Examples include, but are not limited to, imidazolyl, pyridinyl, furyl, pyrimidinyl and pyrazinyl.
  • alkyl, alkenyl, alkynyl, cycloalkyl, heterocycly, aryl, and heteroaryl radicals may be independently substituted with one or more substituents described herein.
  • Amino refers to the —NH 2 or amidogen group.
  • “Monoalkylamino” refers to the —NHR′ group, where R′ represents an alkyl, alkenyl, alkynyl and cycloalkyl as defined herein.
  • Dialkyamino refers to the —NRR′ group where R and R′ independently represents an alkyl, alkenyl, alkynyl and cycloalkyl as defined herein.
  • Halogen refers to bromo, chloro, fluoro, or iodo.
  • Haldroxyl refers to the —OH group.
  • Carboxy refers to the group:
  • Alkoxycarbonyl refers to the group:
  • R represents an alkyl as defined herein.
  • “Pharmaceutically acceptable salt” refers to any salt of compounds claimed in this application that possesses the biological effectiveness to the compounds and are not toxic or otherwise harmful for pharmaceutical use; these salts may be derived from organic and inorganic counter ions which are well known in the art.
  • DIPEA diisopropylethylamine
  • the starting materials for the claimed compounds of formula (I), including the Cs scaffolds, can be prepared using synthesis schemes and reagents available in the art, and may be obtained through commercial suppliers. Reagents used for the synthesis of the novel compounds of the present invention can also be obtained through commercial suppliers.
  • the resulting yellow solid was purified by passing through a 5 g SCX cartridge using methanol as the eluant, followed by MPLC chromatography using a polar gradient of 100% dichloromethane ⁇ >95% dichloromethane/5% methanol containing 10% aqueous ammonia (0.88) to give [(S)-2-(3-carboxypropionylamino)ethyl-1-sulfanylmethyl-Sar] 3 cyclosporinA.
  • the resulting yellow solid was purified by passing through a 2 g SCX cartridge using methanol as the eluant, followed by MPLC chromatography using a gradient of 100% dichloromethane ⁇ 95% dichloromethane/5% methanol containing 10% aqueous ammonia (0.88) to give [(S)-2-(acetylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar] 3 cyclosporinA.
  • the protease-free PPlase assay measures the rate of cis to trans conversion of a peptide substrate catalyzed by the enzyme cyclophilin A. Addition of an inhibitor slows the catalyzed rate and a K i value is obtained.
  • Assay Buffer 35 mM HEPES pH 7.8, filtered through a 0.2 ⁇ m filter. 50 M DTT was added prior to use each day and then the buffer was stored on ice.
  • Enzyme human recombinant Cyp A (Sigma C3805) enzyme was diluted to 1 M with enzyme dilution buffer (20 mM HEPES pH 7.8, 40% glycerol, 50 M DTT and 1 ⁇ M BSA) and stored at ⁇ 20° C.
  • Substrate SUC-AAPF-pNA (from Bachem AG, L-1400), 20 mg/ml prepared 0.5 M LiCl in trifluoroethanol.
  • a first order rate equation was fitted to the absorbance data, for each concentration of inhibitor, to obtain the rate constant (the first 10 to 15 seconds were excluded as mixing causes errors in this portion of curve).
  • the catalytic rate was calculated from the enzymatic rate constant minus the background rate constant.
  • An exponential curve was generated using the catalytic rate constants versus the inhibitor concentration to obtain the K, value for the inhibitor.
  • Calcineurin is a serine-threonine protein phosphatase that on activation dephosphorylates members of the nuclear factor of activated T cells (NFAT), which are important in T lymphocyte activation.
  • Cs A bound to cyclophilin A (“Cyp A”) inhibits calcineurin activity, thus resulting in immunosuppressive effects.
  • Cs A only inhibits calcineurin when bound to Cyp A, some Cs A analogues will also bind calcineurin in the absence of Cyp A.
  • exemplary compounds of Formula (I) which are cyclosporin analogues, their ability to inhibit calcineurin activity was measured in the presence and absence of Cyp A.
  • the CaN assay kit used is based on a colorimetric assay for measuring calcineurin phosphatase activity, and it is commercially available (Enzo Life Sciences and Calbiochem). Calmodulin is also required for calcineurin activity and RII phosphopeptide is used as an efficient peptide substrate for calcineurin.
  • 2 ⁇ assay buffer 100 mM Tris, pH7.5, 200 mM NaCl, 12 mM MgCl 2 , 1 mM DTT, 0.05% NP-40, 1 mM CaCl 2 )
  • Calmodulin Human, recombinant: was thawed on ice, diluted 1:50 with 2 ⁇ assay buffer, and then stored on ice.
  • Calcineurin was thawed quickly, stored on ice immediately, diluted 1:12.5 with 1 ⁇ assay buffer, and then stored on ice.
  • Inhibitors 2.5 mM inhibitor in 100% DMSO.
  • Cyp A recombinant human CypA (Sigma C3805), 1 mg/ml
  • Inhibitor dilutions inhibitor compounds were diluted in UPW in polypropylene low-binding 96 well plates at 5 ⁇ the final assay concentration. For samples ‘without Cyp A’, a 4-point dilution series of the inhibitor was prepared in duplicate to obtain a final assay concentration of 10, 1, 0.1 and 0.01 ⁇ M. For samples ‘with Cyp A’, a 7-point dilution series was prepared to obtain a 1:1 complex of the inhibitor with CypA; the inhibitor and Cyp A final assay concentrations of 10, 3.33, 1.11, 0.37, 0.12, 0.04, 0.014 ⁇ M were prepared. Cs A inhibitor controls were also prepared to obtain a final concentration of 10 ⁇ M Cs A with and without 10 ⁇ M Cyp A.
  • Assay Setup using the half area 96 well plates supplied with the kit, 10 ⁇ l UPW was added to duplicate wells to provide the non-inhibited control. 10 ⁇ l of the inhibitor or the inhibitor/Cyp A complex was added to the appropriate sample wells. 25 ⁇ l of the 2 ⁇ assay buffer with CaM was added to all wells, then 5 ⁇ l of CaN was added to all wells (40 U per well final concentration) except duplicate ‘no calcineurin blank’ wells to which 5 ⁇ L 1 ⁇ assay buffer was added. The assay plate was placed in an oven at 30° C. for 15 minutes to equilibrate to the reaction temperature. The reaction was started by the addition of 10 ⁇ l RII-peptide (0.15 mM final concentration).
  • the reaction was allowed to proceed at 30° C. for a time period in which the reaction is linear for about 60 minutes.
  • the reaction was then terminated by adding 100 ⁇ l of the Malachite Green reagent.
  • the color was allowed to develop for 15-30 minutes at room temperature before the absorbance at 620 nm was measured using a plate reader (Molecular Devices—SpectraMax M5).
  • the data were analyzed by subtracting ‘no Calcineurin blank’ from all the absorbance readings and plotting the background corrected absorbances against Log 10 inhibitor concentration.
  • a sigmoidal-dose response curve was fitted to the data using GraphPad Prism Software.
  • the MLR assay is another means of estimating the immunosuppressive potential of test compounds.
  • Complete medium consisted of RPMI 1640 medium containing 25 mM HEPES buffer (HyClone, Logan, Utah) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Atlanta Biologicals, Lawrenceville, Ga.), 100 ⁇ g/mL streptomycin, 100 U/mL penicillin G, 0.25 ⁇ g/mL amphotericin B (HyClone), 2 mM L-glutamine dipeptide (HyClone), and 2 ⁇ 10 ⁇ 5 M 2-mercaptoethanol (Sigma). Cells were washed twice and resuspended in complete medium. Cell counts were performed using a Beckman Coulter Z-1 particle counter (Fullerton, Calif.). Cell viability was determined by propidium iodide (PI) staining using an Accuri C6 flow cytometer (Ann Arbor, Mich.).
  • PI propidium iodide
  • Spleen cells from C57BL/6 (H-2 b ) and BALB/c (H-2 d ) were used as responder (R) and stimulator (S) cells, respectively.
  • Cells were plated in triplicate in 96-well flat microtiter plates (Costar, Cambridge, Mass.) such that each well contained 2 ⁇ 10 5 R and 8 ⁇ 10 5 S cells.
  • Cultures were incubated in the absence or presence of various concentrations of Cs A, test compounds, or medium at 37° C. in humidified 5% CO 2 for five days, pulsed with 3 H-thymidine ( 3 H-TdR) for the final 16 hours of incubation, and harvested using a Brandel 96-well cell harvester (Gaithersburg, Md.).
  • Proliferation was measured by counting the radioactivity on filter mats in a Wallac 1450 Microbeta TriLux scintillation counter (Turku, Finland). Controls to demonstrate effective inactivation by the x-irradiation were performed by incubating the S cells with 5 ⁇ g/mL of PHA at 2 ⁇ 10 5 cells/well. These control cultures were incubated for 3 days under the same conditions as those described for the MLR; lymphoproliferation was determined in the same manner as described above.
  • compositions of the invention may be used to treat patients suffering from dry eye, to treat blepharitis and meibomian gland disease, to restore corneal sensitivity that has been impaired due to surgery on the cornea or other surface of the eye, to treat allergic conjunctivitis and atopic and vernal keratoconjunctivitis, and to treat ptyregia, ocular symptoms of graft versus host disease, ocular allergy, atopic keratoconjunctivitis, vernal keratoconjunctivitis, uveitis, anterior uveitis, Behcet's disease, Steven Johnson syndrome, ocular cicatricial pemphigoid, chronic ocular surface inflammation caused by viral infection, herpes simplex keratitis, ocular rosacea, and pinguecula, and to prevent corneal transplant rejection.
  • the International Dry Eye Workshop defines dry eye as “a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface, accompanied by increased osmolarity of the tear film and inflammation of the ocular surface.” It includes those conditions, such as keratoconjunctivitis sicca, that are caused by tear deficiency or excessive evaporation of tears.
  • Blepharitis is a chronic disorder producing inflammation of the anterior and posterior lid margin, with involvement of skin and its related structures (hairs and sebaceous glands), the mucocutaneous junction, and the meibomian glands. It can also affect the conjunctiva, tear film, and the corneal surface in advanced stages and may be associated with dry eye. Blepharitis is commonly classified into anterior or posterior blepharitis, with anterior affecting the lash bearing region of the lids, and posterior primarily affecting the meibomian gland orifices.
  • Meibomian gland disease most often occurs as one of three forms: primary meibomitis, secondary meibomitis, and meibomian seborrhea.
  • Meibomian seborrhea is characterized by excessive meibomian secretion in the absence of inflammation (hypersecretory meibomian gland disease).
  • Primary meibomitis by contrast, is distinguished by stagnant and inspissated meibomian secretions (obstructive hypersecretory meibomian gland disease).
  • Secondary meibomitis represents a localized inflammatory response in which the meibomian glands are secondarily inflamed in a spotty fashion from an anterior lid margin blepharitis.
  • Impaired corneal sensitivity often occurs after refractive surgery, such as photorefractive keratectomy, laser assisted sub-epithelium keratomileusis (LASEK), EPI-LASEK, customized transepithelial non-contact ablation, or other procedures in which the corneal nerves are severed. Impaired corneal sensitivity may also occur after viral infection, such as by HSV-1, HSV-2, and VZV viruses. Patients with impaired corneal sensitivity often complain that their eyes feel dry, even though tear production and evaporation may be normal, suggesting that “dryness” in such patients may actually be a form of corneal neuropathy that results when corneal nerves are severed by surgery or inflamed after viral infection.
  • Allergic conjunctivitis is an inflammation of the conjunctiva resulting from hypersensitivity to one or more allergens. It may be acute, intermittent, or chronic. It occurs seasonally, that is, at only certain time of the year, or it occurs perennially, that is, chronically throughout the year. Symptoms of seasonal and perennial allergic conjunctivitis include, in addition to inflammation of the conjunctiva, lacrimation, tearing, conjunctival vascular dilation, itching, papillary hyperlasia, chemosis, eyelid edema, and discharge from the eye. The discharge may form a crust over the eyes after a night's sleep.
  • Atopic keratoconjunctivitis is a chronic, severe form of allergic conjunctivitis that often leads to visual impairment. Symptoms include itching, burning, pain, redness, foreign body sensation, light sensitivity and blurry vision. There is often a discharge, especially on awakening from a night's sleep; the discharge may be stringy, ropy, and mucoid. The lower conjunctiva is often more prominently affected than the upper conjunctiva. The conjunctiva may range from pale, edematous, and featureless to having the characteristics of advanced disease, including papillary hypertrophy, subepithelial fibrosis, formix foreshortening, trichiasis, entropion, and madurosis.
  • the disease progresses to punctate epithelial erosions, corneal neovascularization, and other features of keratopathy which may impair vision.
  • CD25+ T lymphocytes, macrophages, and dendritic cells are significantly elevated in the substantia intestinal.
  • vernal keratoconjunctivitis is a severe form of allergic conjunctivitis, but it tends to affect the upper conjunctiva more prominently than the lower. It occurs in two forms. In the palpebral form, square, hard, flattened, closely packed papillae are present; in the bulbar (limbal) form, the circumcorneal conjunctiva becomes hypertrophied and grayish. Both forms are often accompanied by a mucoid discharge. Corneal epithelium loss may occur, accompanied by pain and photophobia, as may central corneal plaques and Trantas' dots.
  • Uveitis the inflammation of the uvea, is responsible for about 10% of the visual impairment in the United States.
  • Phacoanaphylactic endophthalmitis is a human autoimmune disease.
  • Panuveitis refers to inflammation of the entire uveal (vascular) layer of the eye.
  • Posterior uveitis generally refers to chorioentinitis
  • anterior uveitis refers to iridocyclitis.
  • the inflammatory products i.e. cells, fibrins, excess proteins
  • these inflammations are commonly found in the fluid spaces if the eye, i.e. anterior chamber, posterior chamber and vitreous space as well as infiltrating the tissue intimately involved in the inflammatory response.
  • Uveitis may occur following surgical or traumatic injury to the eye; as a component of an autoimmune disorder, such as rheumatoid arthritis, Behcet's disease, ankylosing spondylitis, and sarcoidosis; as an isolated immune mediated ocular disorder, such as pars planitis, iridocyclitis etc., unassociated with known etiologies; and following certain systemic diseases which cause antibody-antigen complexes to be deposited in the uveal tissues. Together these disorders represent the non-infectious uveitities.
  • an autoimmune disorder such as rheumatoid arthritis, Behcet's disease, ankylosing spondylitis, and sarcoidosis
  • an isolated immune mediated ocular disorder such as pars planitis, iridocyclitis etc., unassociated with known etiologies
  • systemic diseases which cause antibody-antigen complexes to be deposited in the uveal tissues. Together
  • Phacoanaphylaxis is a severe form of uveitis in which the lens in the causative antigen.
  • the lens proteins are normally secluded by the lens capsule since before birth. When these proteins are released into the eye by injury or by surgery or occasionally during cataract development, they can become intensely antigenic and incite an autoimmune response. If the response is moderate it is seen as chronic uveitis. If it is very fast in progression the eye becomes seriously inflamed in all segments. This latter response is named phacoanaphylaxis.
  • Uveitis is a prominent feature of Behcet's disease, a multi-system inflammatory disorder also characterized by oral and genital ulcers, cutaneous, vascular, joint, and neurological manifestations
  • Rosacea is a chronic and common skin disorder with no identified cause or cure.
  • the pathogenesis of rosacea is thought to have multiple factors. Possible factors include exposure to the demodex folliculorum mite, gastrointestinal disease or a vasodilation disorder, and other triggers such as diet or sunlight.
  • Patients may present with a variety of symptoms, including inflammatory papules, edema, telangiectasia, rhinophyma and ocular symptoms.
  • the ocular signs of rosacea include blepharitis, including anterior blepharitis, conjunctivitis, ulceris, iridocyclitis, keratitis, meibomian gland dysfunction, telangiectasia, erythema, chalazion, hordeolum, interpalpebral hyperemia, conjuctival hyperemia, ciliary base injection, bulbar injection, crusts, sleeves, and superficial punctuate keratopathy.
  • blepharitis including anterior blepharitis, conjunctivitis, ulceris, iridocyclitis, keratitis, meibomian gland dysfunction, telangiectasia, erythema, chalazion, hordeolum, interpalpebral hyperemia, conjuctival hyperemia, ciliary base injection, bulbar injection, crusts, sleeves, and superficial punctuate keratopathy.
  • the ocular symptoms are nonspecific and may include burning, tearing, decreased tear secretion, redness, and foreign body or gritty or dry sensation, irritation, Itchiness, Blurred vision, Photosensitivity, Watery eyes, bloodshot eyes, Burning, telangiectasia, irregularity of the lid margins, and meibomian gland dysfunction.
  • Pinguecula is a benign, yellowish brown proliferative growth that forms on the conjunctiva. Pinguecula may cause irritation and scratchiness of the eye, dry eye, inflammation of the conjunctiva and effect appearance of the eye. Inflamed pinguecula, which cause ocular irritation or become unsightly, may require surgical removal. However, the post-operation scar may be as cosmetically objectionable as the pinguecula and pinguecula regrowth may occur following surgical removal.
  • Allogeneic bone marrow transplantation is a well-established treatment for malignant and non-malignant hematological diseases, and is performed in tens of thousands of patients each year.
  • Mature donor T cells within the stem cell graft are the main mediators of the beneficial immune effects, but they are also responsible for the induction of graft-versus-host disease (GVHD), the major cause of morbidity and mortality in BMT patients.
  • GVHD graft-versus-host disease
  • GVHD graft-versus-host disease
  • Acute or chronic GVHD occurs within a 100-day period post-BMT that leads to dermatitis, enteritis, and hepatitis.
  • Ocular symptoms include blurry vision, foreign body sensation, burning sensation, severe light sensitivity, chronic conjunctivitis, dry eye, and eye pain.
  • the present invention also relates to pharmaceutical compositions comprising at least one compound of general formula (I), the compound being present alone or in combination with one or more pharmaceutically acceptable excipients.
  • a “pharmaceutically acceptable excipient” is one that is compatible with the active ingredient of the composition and not harmful to the person being administered the pharmaceutical composition. Mixtures of two or more of such suitable excipients may be used.
  • compositions may be prepared by combining a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, as an active ingredient, with conventional ophthalmically acceptable pharmaceutical excipients, and by preparation of unit dosage forms suitable for topical ocular use.
  • the therapeutically efficient amount typically is between about 0.0001 and about 5% (w/v), preferably about 0.001 to about 1.0% (w/v) in liquid formulations.
  • the actual dose of the active compounds of the present invention depends on the specific compound, and on the condition to be treated; the selection of the appropriate dose is well within the knowledge of one of ordinary skill in the art.
  • a therapeutically effective amount in the claimed pharmaceutical composition is a concentration useful to observe a therapeutic effect as compared to a placebo composition that, except for the absence of a compound of formula (I), is otherwise identical to the pharmaceutical composition.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Virology (AREA)
  • Analytical Chemistry (AREA)
  • Oncology (AREA)
  • Transplantation (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Biotechnology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Disclosed herein are novel analogs of cyclosporin, pharmaceutical compositions containing them, and methods for their use in the treatment of dry eye and other conditions.

Description

    CROSS REFERENCE
  • This application claims the benefit of U.S. Provisional Patent Application Ser. No. 61/392,449, filed on Oct. 12, 2010, the entire disclosure of which is incorporated herein by this specific reference.
  • Disclosed herein are novel analogs of cyclosporin, pharmaceutical compositions containing them, and methods for their use in the treatment of dry eye and other conditions.
  • BACKGROUND
  • Cyclosporins are a class of poly-N-methylated cyclic undecapeptides. There are naturally occurring cyclosporins such as cyclosporin A, and non-natural cyclosporin (“Cs”) derivatives.
  • Cyclosporin A, for example, has the following structure:
  • Figure US20120088734A1-20120412-C00001
  • The following structure shows the 11 amino acid residues of cyclosporin A:
  • Figure US20120088734A1-20120412-C00002
  • DETAILED DESCRIPTION
  • The claimed invention relates to novel compounds of the following formula (I) and pharmaceutically acceptable salts thereof:
  • Figure US20120088734A1-20120412-C00003
  • The claimed novel compounds of formula (I) result from modifications at the α-carbon of the 3-position amino acid (sarcosine) of “Cs scaffolds,” which as used herein refers to different cyclosporins (e.g., Cs A, Cs C, Cs D, etc.) that vary from each other in the identities of one or more of substituents A, B, C, and D. In other words, a “Cs scaffold” refers to a novel compound of formula (I) less the moiety at the α-carbon of the 3-position amino acid.
  • A, B, C, and D are defined as follows. A represents —CH═CHR, —CH═CH—CH═CHR, or —CH2CH2R, wherein R represents
      • (a) —CH3,
      • (b) —CH2SH,
      • (c) —CH2S—Cn wherein n=1-6,
      • (d) —CH2-carboxyl, i.e.,
  • Figure US20120088734A1-20120412-C00004
      • (e) carboxyl, i.e.,
  • Figure US20120088734A1-20120412-C00005
      • (f) alkoxycarbonyl, i.e.,
  • Figure US20120088734A1-20120412-C00006
      • wherein R10=C1-C6 alkyl, or
      • (g) —CH2-alkoxycarbonyl, i.e.,
  • Figure US20120088734A1-20120412-C00007
      • wherein R11=C1-C6 alkyl.
        B represents
  • —CH2CH3, or
  • 1-hydroxyethyl, i.e.,
  • Figure US20120088734A1-20120412-C00008
  • isopropyl, i.e.,
  • Figure US20120088734A1-20120412-C00009
  • or
  • n-propyl, i.e.,
  • Figure US20120088734A1-20120412-C00010
  • C represents
  • isobutyl, i.e.,
  • Figure US20120088734A1-20120412-C00011
  • 2-hydroxyisobutyl, i.e.,
  • Figure US20120088734A1-20120412-C00012
  • or
  • 1-methylpropyl, i.e.,
  • Figure US20120088734A1-20120412-C00013
  • D represents —CH3 or —CH2OH.
  • In one embodiment of the present invention, A is —CH═CHCH3, B is —CH2CH3, C is isobutyl, and D is —CH3, as illustrated below in formula (IA):
  • Figure US20120088734A1-20120412-C00014
  • In another embodiment, A is —CH═CHCH3, B is 1-hydroxyethyl, C is isobutyl, and D is —CH3, as illustrated below in formula (IB):
  • Figure US20120088734A1-20120412-C00015
  • In another embodiment, A is —CH═CHCH3, B is isopropyl, C is isobutyl, and D is —CH3, as illustrated below in formula (IC):
  • Figure US20120088734A1-20120412-C00016
  • In another embodiment, A is —CH═CHCH3, B is n-propyl, C is isobutyl, and D is —CH3, as illustrated below in formula (ID):
  • Figure US20120088734A1-20120412-C00017
  • In another embodiment, A is —CH═CHCH3, B is —CH2CH3, C is 1-methylpropyl, and D is —CH3, as illustrated below in formula (IE):
  • Figure US20120088734A1-20120412-C00018
  • In one embodiment, A is —CH═CHCH3, B is —CH2CH3, C is isobutyl, and D is —CH2OH, as illustrated below in formula (IF):
  • Figure US20120088734A1-20120412-C00019
  • In one embodiment, A is —CH═CHCH3, B is —CH2CH3, C is 2-hydroxy isobutyl, and D is —CH3, as illustrated below in formula (IG):
  • Figure US20120088734A1-20120412-C00020
  • In another embodiment, the variables are as summarized below:
  • Figure US20120088734A1-20120412-C00021
    Figure US20120088734A1-20120412-C00022
    Scaffold R4 R5 R6 R7 R8
    CsA H H H H H
    CsC H OH H H H
    CsD H Me H H H
    CsG H H Me H H
    NIM811 H H H Me H
    DSer8 H H H H OH
    Scy OH H H H H
  • In formula (I), the amino acids of various types of cyclosporine scaffold are labeled numerically from 1 to 11. In one embodiment, the modifications of the invention occur at the α-carbon of the position-3 amino acid (sarcosine at the 3-position) of the Cs scaffold. The modification generally comprises replacement of a hydrogen atom at the α-carbon of the position-3 amino acid with a moiety in formula (I) wherein:
  • R1 and R2, which are identical or different, each represents hydrogen or C1-C4 alkyl, or together represent C3-C7 cycloalkyl;
  • X represents sulfur or —S(O)n, wherein n is 1 or 2;
  • R3 represents hydrogen; straight or branched C1-C6 alkyl; straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl;
  • —(CHR′)m—NH—CNH2NH;
  • —(CHR′)m—COOH;
  • —(CHR′)m—NHR″;
  • —(CHR′)m—NHCOR″;
  • wherein m is 1, 2, 3, 4, 5, or 6;
  • each R′ is independently H, straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl; or is absent (permitting, in such case, —(CHR′)m to represent an unsaturated moiety such as —CH—CH).
  • R″ is —(CHR′)pN(CH3)2, —(CHR′)pNHCOC, —(CHR′)pNHCOOH, wherein p is 0, 1, 2, 3, 4, 5, or 6; and
  • wherein R3 may be optionally substituted with one or more groups, identical or different, of C1-C6 alkyl; halogen; hydroxyl; alkoxycarbonyl; carboxyl; cycloalkyl; saturated or partially unsaturated 5-6 member heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur, which heterocyclyl is optionally substituted by one or more groups of C1-C6 alkyl; aryl; heteroaryl; amino; monoalkylamino; dialkylamino; amidino; guanidine; or urea.
  • Compound Examples
  • Embodiments of formula (I) include, but are not limited to, the following compounds of formula (I) (only the moiety at the α-carbon of the position-3 amino acid is shown; wavy lines represent the rest of the Cs compounds of formula (I)):
  • Figure US20120088734A1-20120412-C00023
    Figure US20120088734A1-20120412-C00024
    Figure US20120088734A1-20120412-C00025
  • The present invention also relates to novel intermediates useful for the preparation of compounds of formula (I). The novel intermediates are compounds of the following formula (III):
  • Figure US20120088734A1-20120412-C00026
  • wherein A, B, C, and D are as defined above.
  • The following is a non-limiting embodiment of formula (III):
  • Formula (III) Compound Example 1 [Methylene-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00027
  • ESMS MH+1214.8, MNa+1236.8
  • All NMR data show selected diagnostic protons.
  • 1H NMR (CDCl3, ppm) δ 4.98 (d, 1H, olefin CH2), 5.25 (d, 1H, olefin CH2), 7.17 (d, 1H, amide NH), 7.52 (d, 1H, amide NH), 7.59 (d, 1H, amide NH), 7.85 (d, 1H, amide NH).
  • 13C NMR (CDCl3, ppm) δ 143.96 (olefin C), 108.09 (olefin CH2).
  • The present invention also relates to a process for the preparation of a compound of formula (III), comprising:
  • reacting a compound of the formula (II)
  • Figure US20120088734A1-20120412-C00028
      • wherein A, B, C, and D are as previously defined, with a strong base (for example, lithium diisopropylamide or other strong base), at a temperature in the range of −78° C. to −70° C., followed by treatment with a flow of carbon dioxide gas between −70° C. and 15° C., followed by treatment with excess chloromethylchloroformate between −50° C. and room temperature, followed by quenching of the reaction mixture with acetic acid at 0° C. to room temperature to generate a compound of formula (III).
  • Figure US20120088734A1-20120412-C00029
  • Definitions
  • “Alkyl” refers to a monovalent linear or branched hydrocarbon radical having 1 to 6 carbon atoms. Examples include, but are not limited to, methyl, ethyl, propyl (e.g., 1-propyl, isopropyl), butyl (e.g., 1-butyl, isobutyl, sec-butyl, tert-butyl), pentyl (e.g., 1-pentyl, neopentyl), and hexyl (e.g., 3-hexyl).
  • “Alkenyl” refers to a monvalent linear or branched hydrocarbon radical having 2 to 6 carbon atoms and one or more double bonds. Examples include, but are not limited to, ethenyl, propenyl, and butenyl.
  • “Alkynyl” refers to a monovalent linear or branched hydrocarbon radical having 2 to 6 carbon atoms and one or more triple bonds. Examples include, but are not limited to, ethynyl, propynyl and butynyl.
  • “Cycloalkyl” refers to monovalent saturated or partially unsaturated cyclic hydrocarbon radical having 3 to 6 carbon atoms. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • “Heterocyclyl” refers to monovalent, saturated or partially unsaturated cyclic hydrocarbon radical having 3 to 6 ring atoms, at least one of which is a heteroatom selected from nitrogen, oxygen and sulfur. The radical may be on a carbon or a heteroatom. Examples include, but are not limited to, morpholinyl, piperidinyl, pyrrolidinyl, pyranyl, and pyrazolinyl.
  • “Aryl” refers to monovalent 5-7 member aromatic hydrocarbon radical. Examples include, but are not limited to, phenyl.
  • “Heteroaryl” refers to monovalent 5-7 member aromatic hydrocarbon radical having one or more heteroatoms selected from nitrogen, sulfur and oxygen. Examples include, but are not limited to, imidazolyl, pyridinyl, furyl, pyrimidinyl and pyrazinyl.
  • The aforementioned alkyl, alkenyl, alkynyl, cycloalkyl, heterocycly, aryl, and heteroaryl radicals may be independently substituted with one or more substituents described herein.
  • “Amino” refers to the —NH2 or amidogen group.
  • “Monoalkylamino” refers to the —NHR′ group, where R′ represents an alkyl, alkenyl, alkynyl and cycloalkyl as defined herein.
  • “Dialkyamino” refers to the —NRR′ group where R and R′ independently represents an alkyl, alkenyl, alkynyl and cycloalkyl as defined herein.
  • “Halogen” refers to bromo, chloro, fluoro, or iodo.
  • “Hydroxyl” refers to the —OH group.
  • “Carboxy” refers to the group:
  • Figure US20120088734A1-20120412-C00030
  • “Alkoxycarbonyl” refers to the group:
  • Figure US20120088734A1-20120412-C00031
  • where R represents an alkyl as defined herein.
  • “Amidino” refers to the groups:
  • Figure US20120088734A1-20120412-C00032
  • “Guanidino” refers to the group:
  • Figure US20120088734A1-20120412-C00033
  • “Urea” refers to the group:
  • Figure US20120088734A1-20120412-C00034
  • “Pharmaceutically acceptable salt” refers to any salt of compounds claimed in this application that possesses the biological effectiveness to the compounds and are not toxic or otherwise harmful for pharmaceutical use; these salts may be derived from organic and inorganic counter ions which are well known in the art.
  • Synthesis Examples
  • The invention is illustrated by the following non-limiting synthesis examples.
  • Unless otherwise indicated, the following chemical abbreviations are used in the synthesis examples:
  • DMF: dimethylformamide
  • DIPEA: diisopropylethylamine
  • Me: methyl
  • THF: tetrahydrofuran
  • i-Pr: isopropyl
  • n-Bu: n-butyl
  • The starting materials for the claimed compounds of formula (I), including the Cs scaffolds, can be prepared using synthesis schemes and reagents available in the art, and may be obtained through commercial suppliers. Reagents used for the synthesis of the novel compounds of the present invention can also be obtained through commercial suppliers.
  • Example 1 [Methylene-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00035
  • To a solution of diisopropylamine (11.2 ml, 80 mmol) in dry THF (240 ml) at −78° C. under an atmosphere of nitrogen was added dropwise n-butyl lithium (2.5M in hexanes, 32 ml, 80 mmol) and the resulting mixture was stirred at −78° C. for 60 minutes.
  • A solution of dry cyclosporine A (dried by azeotroping with 2×40 ml toluene then kept in dessicator o/n in presence of P2O5) (9.6 g, 8.0 mmol) in dry THF (40 ml) was added and the reaction was stirred under the same conditions for 2 h. A flow of carbon dioxide was bubbled through the reaction mixture for 30 minutes with temperature increasing to −50° C. The resulting mixture was allowed to warm to 15° C. over a period of 2 hours then cool back down to −50° C. before the addition of chloromethylchloroformate (7.1 ml, 80 mmol). The reaction mixture was allowed to warm to room temperature overnight then cooled to 0° C. and acetic acid (5 ml, 88 mmol) was added.
  • The mixture was allowed to warm to room temperature, the solvent evaporated and the resultant mixture was partitioned between ethyl acetate and brine. The organic phase was separated, dried (Na2SO4) and concentrated to give a yellow oil.
  • The crude product was purified by MPLC chromatography using a solvent gradient of 100% diethyl ether→96% diethyl ether/4% methanol to give [methylene-Sar]3 cyclosporinA.
  • ESMS MH+1214.8, MNa+1236.8
  • 1H NMR (CDCl3, ppm) δ 4.98 (d, 1H, olefin CH2), 5.25 (d, 1H, olefin CH2), 7.17 (d, 1H, amide NH), 7.52 (d, 1H, amide NH), 7.59 (d, 1H, amide NH), 7.85 (d, 1H, amide NH).
  • 13C NMR (CDCl3, ppm) δ 143.96 (olefin C), 108.09 (olefin CH2).
  • In a similar way the following compounds were prepared:
  • [Dihydro-MeBmt]1 [Methylene-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00036
  • ESMS MH+1216.8, MNa+ 1238.8
  • 1H NMR (CDCl3, ppm) δ 7.18 (d, 1H, amide NH), 7.52 (d, 1H, amide NH), 7.57 (d, 1H, amide NH), 7.75 (d, 1H, amide NH)
  • [Methylene-Sar]3 cyclosporin D
  • Figure US20120088734A1-20120412-C00037
  • ESMS MH+1228.8, MNa+1250.8
  • 1H NMR (CDCl3, ppm) 7.17 (d, 1H, amide NH), 7.59 (d, 1H, amide NH), 7.63 (d, 1H, amide NH), 7.89 (d, 1H, amide NH)
  • Example 2 [(S)-2-Diethylaminoethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00038
  • To a solution of [methylene-Sar]3 cyclosporin A (0.232 g, 0.19 mmol) in acetonitrile (15 ml) was added potassium carbonate (0.523 g, 3.8 mmol) and the white suspension was bubbled through with nitrogen for one hour before adding diethylaminoethylthiol hydrochloride salt (0.322 g, 1.9 mmol). The reaction mixture was bubbled through with nitrogen overnight during which time the solvent evaporated. Acetonitrile (15 ml) was added, the white suspension filtered through a pad of sodium sulphate then concentrated to give 315 mg of a colourless oil. Trituration with hexane under ultrasound conditions, followed by decantation of the solution provided 240 mg of a white solid. Purification by MPLC chromatography using a solvent gradient of 100% diethyl ether→95% diethyl ether/5% methanol then 100% diethyl ether followed by a second solvent gradient 100% diethyl ether→90% diethyl ether/10% methanol containing 10% aqueous ammonia (0.88) gave [(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin A.
  • ESMS MH+1347.9
  • 1H NMR (CDCl3, ppm) δ 0.69 (3H, d), 0.77-1.07 (43H, m) 1.09 (3H, d), 1.18-1.41 (10H, m), 1.41-1.82 (10H, m), 1.93-2.18 (5H, m), 2.33-2.46 (2H, m), 2.53 (4H, q), 2.60-2.72 (6H, m), 2.99 (2H, br d), 3.10 (3H, s), 3.18 (3H, s), 3.27 (6H, s), 3.50 (3H, s, NMe), 3.58 (1H, br d), 3.67 (1H, q), 4.52 (1H, m), 4.60 (1H, t), 4.82 (1H, m), 4.93-5.08 (4H, m), 5.10 (1H, d), 5.26-5.39 (3H, m), 5.51 (1H, dd), 5.69 (1H, dd), 7.16 (1H, d, amide NH), 7.36 (1H, d, amide NH), 7.68 (1H, d, amide NH), 8.13 (1H, d, amide NH).
  • In a similar way the following compounds were prepared:
  • [Dihydro-MeBmt]1 [(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00039
  • ESMS MH+1350.0
  • 1H NMR (CDCl3, ppm) δ7.17 (1H, d, amide NH), 7.37 (1H, d, amide NH), 7.71 (1H, d, amide NH), 8.10 (1H, d, amide NH).
  • [(S)-2-Diethylaminoethylthiomethyl-Sar]3 cyclosporin D
  • Figure US20120088734A1-20120412-C00040
  • ESMS MH+1361.9
  • 1H NMR (CDCl3, ppm) δ7.16 (1H, d, amide NH), 7.49 (1H, d, amide NH), 7.72 (1H, d, amide NH), 8.16 (1H, d, amide NH)
  • Example 3 [(S)-2-(4-pyridyl)ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00041
  • ESMS MH+1354.0
  • 1H NMR (CDCl3, ppm) δ 7.14 (d, 2H, pyridine), 7.18 (d, 1H, amide NH), 7.31 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.17 (d, 1H, amide NH), 8.54 (d, 2H, pyridine).
  • Example 4 [(S)-Methylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00042
  • ESMS MH+1262.8, MNa+1284.8
  • 1H NMR (CDCl3, ppm) δ 2.20 (s, 3H, SMe). 7.18 (d, 1H, amide NH), 7.36 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 5 [(S)-Pentylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00043
  • ESMS MH+1318.6, MNa+1340.6
  • 1H NMR (CDCl3, ppm) δ 7.15 (d, 1H, amide NH), 7.37 (d, 1H, amide NH), 7.67 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 6 [(S)-2-Aminoethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00044
  • ESMS MH+1291.7
  • 1H NMR (CDCl3, ppm) δ 7.18 (d, 1H, amide NH), 7.35 (d, 1H, amide NH), 7.67 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 7 [(S)-{(R)-2-Amino-2-carbomethoxy-ethyl}thiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00045
  • ESMS MH+1349.6
  • 1H NMR (CDCl3, ppm) δ 3.78 (s, 3H, COOMe), 7.15 (d, 1H, amide NH), 7.33 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 8 [(S)-2-Dimethylaminoethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00046
  • ESMS MH+1319.56
  • 1H NMR (CDCl3, ppm) δ 2.25 (s, 6H, NMe2), 7.17 (d, 1H, amide NH), 7.35 (d, 1H, amide NH), 7.68 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 9 [(S)-2-(4-Methylpiperidinyl)thiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00047
  • ESMS MH+1345.6
  • 1H NMR (CDCl3, ppm) δ 2.29 (s, 3H, NMe), 7.17 (d, 1H, amide NH), 7.33 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.17 (d, 1H, amide NH).
  • Example 10 [(S)-2-(Morpholino)ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00048
  • ESMS MH+1361.8
  • 1H NMR (CDCl3, ppm) 7.17 (d, 1H, amide NH), 7.35 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.19 (d, 1H, amide NH)
  • Example 11 [(S)-Carbomethoxymethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00049
  • ESMS MH+1320.4
  • 1H NMR (CDCl3, ppm) δ 3.78 (s, 3H, COOMe), 7.17 (d, 1H, amide NH), 7.32 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 12 [(S)-Carbomethoxyethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00050
  • ESMS MH+1335.05
  • 1H NMR (CDCl3, ppm) δ 3.71 (s, 3H, COOMe), 7.17 (d, 1H, amide NH), 7.34 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.15 (d, 1H, amide NH).
  • Example 13 [(S)-{(S)-2-Amino-2-carbomethoxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00051
  • ESMS MH+1377.4
  • 1H NMR (CDCl3, ppm) δ 3.70 (s, 3H, COOMe), 7.15 (d, 1H, amide NH), 7.33 (d, 1H, amide NH), 7.68 (d, 1H, amide NH), 8.08 (d, 1H, amide NH).
  • [(S)-2-(N-imidazolyl)ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00052
  • ESMS MH+1342.7
  • 1H NMR (CDCl3, ppm) δ6.89 (s, 1H, imidazole), 7.00 (s, 1H, imidazole), 7.10 (d, 1H, amide NH), 7.24 (d, 1H, amide NH), 7.48 (s, 1H, imidazole), 7.60 (d, 1H, amide NH), 8.11 (d, 1H, amide NH)
  • [(S)-2-(N-pyrazolyl)ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00053
  • ESMS MH+1342.9
  • 1H NMR (CDCl3, ppm) δ6.26 (dd, 1H, pyrazole), 7.17 (d, 1H, amide NH), 7.31 (d, 1H, amide NH), 7.46 (d, 1H, pyrazole), 7.51 (d, 1H, pyrazole), 7.70 (d, 1H, amide NH), 8.11 (d, 1H, amide NH).
  • [(S)-2-Diethylamino-1,1-dimethyl-ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00054
  • ESMS MH+1375.9
  • 1H NMR (CDCl3, ppm) δ7.16 (1H, d, amide NH), 7.39 (1H, d, amide NH), 7.68 (1H, d, amide NH), 8.11 (1H, d, amide NH).
  • [(S)-2-Morpholino-1,1-dimethyl-ethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00055
  • ESMS MH+1389.6
  • 1H NMR (CDCl3, ppm) 7.18 (d, 1H, amide NH), 7.38 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.14 (d, 1H, amide NH).
  • [(S)-3-Diethylaminopropylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00056
  • ESMS MH+1362.0
  • 1H NMR (CDCl3, ppm) δ7.19 (1H, d, amide NH), 7.37 (1H, d, amide NH), 7.68 (1H, d, amide NH), 8.13 (1H, d, amide NH).
  • [(S)-3-(Morpholino)-propylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00057
  • ESMS MH+1375.7
  • 1H NMR (CDCl3, ppm) δ7.18 (1H, d, amide NH), 7.33 (1H, d, amide NH), 7.70 (1H, d, amide NH), 8.16 (1H, d, amide NH).
  • [(5)-Mercaptomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00058
  • A solution of [(S)-{(S)-2-amino-2-carbomethoxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporinA (0.028 g, 0.02 mmol) in methanol (3 ml) was heated to 50° C. for nineteen days. The reaction mixture was purified with SCX column/methanol to give [(S)-mercaptomethyl-Sar]3 cyclosporinA.
  • ESMS MH+1247.9
  • 1H NMR (CDCl3, ppm) δ 7.20 (d, 1H, amide NH), 7.26 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.24 (d, 1H, amide NH).
  • [(S)-Allylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00059
  • To a solution of [(S)-mercaptomethyl-Sar]3 cyclosporinA (0.074 g, 0.06 mmol) in dimethylformamide (1.5 ml) was added potassium carbonate (0.021 g, 0.15 mmol) and allyl bromide (0.036 g, 0.3 mmol). The suspension was stirred at room temperature overnight then an additional amount of potassium carbonate (0.021 g, 0.15 mmol) and allyl bromide (0.036 g, 0.3 mmol) were added and the reaction mixture stirred over 2 days. The reaction mixture was concentrated, the residue ultrasonicated in the presence of dichloromethane then filtrated. The dichloromethane was evaporated and the residue purified by MPLC chromatography using a solvent gradient of 100% dichloromethane→95% dichloromethane/5% methanol to give [(S)-Allylthiomethyl-Sar]3 cyclosporinA.
  • ESMS MH+1288.6
  • 1H NMR (CDCl3, ppm) δ 7.18 (d, 1H, amide NH), 7.35 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.13 (d, 1H, amide NH).
  • Example 14 [(S)-{(R)-2-Amino-2-carbohydroxy-ethyl}thiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00060
  • To a solution of [(S)-{(R)-2-amino-2-carbomethoxy-ethyl}thiomethyl-Sar]3 cyclosporinA (0.033 g, 0.024 mmol) in tetrahydrofuran (2 ml) was added a solution of lithium hydroxide monohydrate (0.0043 g, 0.098 mmol) in water (0.3 ml). The solution was stirred at room temperature for 3 hours. Hydrochloric acid (2N, 0.05 ml0 was added then the reaction mixture was concentrated. Purification by SCX column using a gradient 0.035M ammonia in methanol to 0.175M ammonia in methanol provided 20 mg of [(S)-{(R)-2-Amino-2-carbohydroxy-ethyl}thiomethyl-Sar]3 cyclosporinA.
  • ESMS MH+1335.7
  • 1H NMR (CDCl3, ppm) δ 7.19 (d, 1H, amide NH), 7.32 (d, 1H, amide NH), 7.69 (d, 1H, amide NH), 8.2 (d, 1H, amide NH).
  • In a similar way the following compounds were prepared:
  • Example 15 [(S)-Carbohydroxymethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00061
  • ESMS MH+1306.6
  • 1H NMR (CDCl3, ppm) δ 7.18 (d, 1H, amide NH), 7.35 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.17 (d, 1H, amide NH).
  • Example 16 [(S)-{(S)-2-Amino-2-carbohydroxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00062
  • ESMS MH+1363.5
  • 1H NMR (CDCl3, ppm) δ 7.17 (d, 1H, amide NH), 7.30 (d, 1H, amide NH), 7.68 (d, 1H, amide NH), 8.18 (d, 1H, amide NH).
  • Example 17 [(S)-Carbohydroxyethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00063
  • ESMS MH+1320.6
  • 1H NMR (CDCl3, ppm) δ 7.20 (d, 1H, amide NH), 7.36 (d, 1H, amide NH), 7.70 (d, 1H, amide NH), 8.18 (d, 1H, amide NH).
  • Example 18 [(S)-2-Isopropylaminoethylthiomethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00064
  • To a solution of [(S)-2-aminoethylthiomethyl-Sar]3 cyclosporinA (0.129 g, 0.1 mmol) in chloroform (2 ml) and acetone (0.1 ml) was added tetramethylammonium triacetoxy borohydride (0.066 mg, 0.25 mmol) and the reaction mixture was stirred at room temperature for 17 hours. The reaction mixture was partitioned between dichloromethane and water. The organic phase was washed with brine, dried (Na2SO4) and concentrated to give a white solid.
  • Purification by MPLC chromatography using a solvent gradient of 100% diethyl ether→95% diethyl ether/5% methanol then 100% diethyl ether followed by a second solvent gradient 100% diethyl ether→96% diethyl ether/4% methanol containing 10% aqueous ammonia (0.88) gave [(S)-2-isopropylaminoethylthiomethyl-Sar]3 cyclosporinA.
  • ESMS MH+1333.0
  • 1H NMR (CDCl3, ppm) δ 1.05 (d, 6H, 2×CH3), 2.84 (m, 1H, CH), 7.18 (d, 1H, amide NH), 7.38 (d, 1H, amide NH), 7.72 (d, 1H, amide NH), 8.19 (d, 1H, amide NH).
  • Example 19 [(S)-2-Guanidinoethyl-1-sulfanylmethyl-Sar]3 cyclosporin A hydrochloride
  • Figure US20120088734A1-20120412-C00065
  • To a solution of [(S)-2-aminoethylthiomethyl-Sar]3 cyclosporinA (0.055 g, 0.042 mmol) in dimethylformamide (0.6 ml) containing diisopropylethylamine (0.0072 ml) was added 1H-pyrazole-1-carboxamidine hydrochloride (0.0055 mg, 0.042 mmol) and the reaction mixture was stirred at room temperature for 22 hours. The reaction mixture was concentrated, the residue was triturated with diethyl ether to give after decantation of the supernatant, a white solid. Partition between dichloromethane and water followed by separation of the organic phase and concentration provided [(S)-2-guanidinoethyl-1-sulfanylmethyl-Sar]3 cyclosporinA hydrochloride.
  • ESMS MH+1334.5
  • 1H NMR (CDCl3, ppm) δ 7.12 (d, 1H, amide NH), 7.43 (d, 1H, amide NH), 7.67 (d, 1H, amide NH), 8.31 (d, 1H, amide NH).
  • [(S)-2-Diethylaminoethyl-1-sulfinylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00066
  • [(S)-2-Diethylaminoethylthiomethyl-Sar]3 cyclosporinA (0.147 g, 0.11 mmol) was dissolved in dichloromethane (1 ml) containing trifluoroacetic acid (0.0089 ml). After 5 min the solution was concentrated and the resulting white solid redissolved in acetonitrile (6 ml). A solution of sodium periodate (0.058 g) in water (2 ml) was added and the suspension stirred at room temperature overnight. The reaction mixture was filtrated, the solid washed with methanol and the filtrate concentrated. The resulting residue was dissolved in dichloromethane (10 ml) containing triethylamine (0.255 ml) then ultrasonicated for 1 hour. The mixture was filtrated through a phase separation cartridge. The organic phase was concentrated to give 188 mg of a white solid. Purification by MPLC chromatography using a solvent gradient of 100% dichloromethane→96% dichloromethane/4% methanol containing 10% aqueous ammonia (0.88) gave [(S)-2-diethylaminoethyl-1-sulfinylmethyl-Sar]3 cyclosporinA.
  • ESMS MH+1363.8
  • 1H NMR (CDCl3, ppm) δ 7.19 (1H, d, amide NH), 7.38 (1H, d, amide NH), 7.69 (1H, d, amide NH), 8.28 (1H, d, amide NH)
  • In a similar way the following compound was prepared:
  • [Dihydro-MeBmt]1 [(S)-2-diethylaminoethyl-1-sulfinylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00067
  • ESMS MH+1366.8
  • 1H NMR (CDCl3, ppm) δ7.20 (1H, d, amide NH), 7.40 (1H, d, amide NH), 7.71 (1H, d, amide NH), 8.30 (1H, d, amide NH).
  • [(S)-3-Diethylaminopropyl-1-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00068
  • [(S)-3-Diethylaminopropylthiomethyl-Sar]3 cyclosporinA (0.035 g, 0.026 mmol) was dissolved in dichloromethane (2 ml) containing trifluoroacetic acid (0.0021 ml). After 5 min the solution was concentrated and the resulting white solid redissolved in dichloromethane (2 ml). Tetrabutylammonium oxone (0.109 g) was added and the solution stirred at room temperature overnight. The reaction mixture was treated with triethylamine (0.017 ml) then stirred for 90 min before concentrating. The residue was purified by MPLC chromatography using a solvent gradient of 100% dichloromethane→95% dichloromethane/5% methanol containing 10% aqueous ammonia (0.88) to give [(S)-3-diethylaminopropyl-1-sulfonylmethyl-Sar]3 cyclosporinA.
  • ESMS MH+1393.9
  • 1H NMR (CDCl3, ppm) δ 7.15 (1H, d, amide NH), 7.20 (1H, d, amide NH), 7.65 (1H, d, amide NH), 8.29 (1H, d, amide NH).
  • In a similar way the following compound was prepared:
  • [(S)-3-(Morpholino)propyl-1-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00069
  • ESMS MH+1407.5
  • 1H NMR (CDCl3, ppm) δ 7.22 (1H, d, amide NH), 7.27 (1H, d, amide NH), 7.75 (1H, d, amide NH), 8.41 (1H, d, amide NH).
  • [(S)-(4-Methylpiperidinyl)-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00070
  • ESMS MH+1377.43
  • 1H NMR (CDCl3, ppm) δ 7.22 (1H, d, amide NH), 7.29 (1H, d, amide NH), 7.74 (1H, d, amide NH), 8.40 (1H, d, amide NH).
  • [(S)-(2-Diethylamino-1,1-dimethyl)-ethyl-1-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00071
  • ESMS MH+1407.8
  • 1H NMR (CDCl3, ppm) δ7.19 (1H, d, amide NH), 7.37 (1H, d, amide NH), 7.72 (1H, d, amide NH), 8.30 (1H, d, amide NH).
  • [(S)-(2-Morpholino-1,1-dimethyl)-ethyl-1-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00072
  • ESMS MH+1421.6
  • 1H NMR (CDCl3, ppm) δ7.21 (1H, d, amide NH), 7.31 (1H, d, amide NH), 7.75 (1H, d, amide NH), 8.42 (1H, d, amide NH).
  • [(S)-3-(4-oxy-morpholin-4-yl)-propyl-1-sulfonylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00073
  • ESMS MH+1423.7
  • 1H NMR (CDCl3, ppm) δ7.21 (1H, d, amide NH), 7.25 (1H, d, amide NH), 7.73 (1H, d, amide NH), 8.35 (1H, d, amide NH)
  • [(S)-2-(3-Carboxypropionylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00074
  • To a stirred solution of [(S)-2-aminoethylthiomethyl-Sar]3 cyclosporinA (0.30 g, 0.23 mmol) in dichloromethane (25 ml) was added triethylamine (0.05 ml, 0.23 mmol) followed by succinyl chloride (0.05 ml, 0.23 mmol) and the reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with water (20 ml) and the organic layer was separated, dried (MgSO4), filtered and concentrated under vacuum. The resulting yellow solid was purified by passing through a 5 g SCX cartridge using methanol as the eluant, followed by MPLC chromatography using a polar gradient of 100% dichloromethane→>95% dichloromethane/5% methanol containing 10% aqueous ammonia (0.88) to give [(S)-2-(3-carboxypropionylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporinA.
  • ESMS MH+1391.8
  • 1H NMR (CDCl3, ppm) δ7.19 (1H, d, amide NH), 7.35 (1H, d, amide NH), 7.71 (1H, d, amide NH), 8.21 (1H, d, amide NH).
  • In a similar way the following compound was prepared:
  • [(S)-2-(4-Carboxybutyrylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00075
  • ESMS MH+1405.8
  • 1H NMR (CDCl3, ppm) δ7.18 (1H, d, amide NH), 7.35 (1H, d, amide NH), 7.69 (1H, d, amide NH), 8.21 (1H, d, amide NH).
  • [(S)-2-(Acetylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00076
  • To a stirred solution of [(S)-2-aminoethylthiomethyl-Sar]3 cyclosporinA (0.10 g, 0.078 mmol) in dichloromethane (25 ml) was added N-acetylglycine (0.01 g, 0.084 mmol) and triethylamine (0.01 ml, 0.084 mmol) followed by WSC.HCl (0.022 g, 0.084 mmol) and the reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with water (20 ml) and the organic layer was separated, dried (MgSO4), filtered and concentrated under vacuum. The resulting yellow solid was purified by passing through a 2 g SCX cartridge using methanol as the eluant, followed by MPLC chromatography using a gradient of 100% dichloromethane→95% dichloromethane/5% methanol containing 10% aqueous ammonia (0.88) to give [(S)-2-(acetylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporinA.
  • ESMS MH+1390.82
  • 1H NMR (CDCl3, ppm) 7.20 (1H, d, amide NH), 7.35 (1H, d, amide NH), 7.71 (1H, d, amide NH), 8.21 (1H, d, amide NH).
  • In a similar way the following compound was prepared:
  • [(S)-2-(Dimethylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A
  • Figure US20120088734A1-20120412-C00077
  • ESMS MH+1376.89
  • 1H NMR (CDCl3, ppm) δ 7.19 (d, 1H, amide NH), 7.36 (d, 1H, amide NH), 7.71 (d, 1H, amide NH), 8.18 (d, 1H, amide NH)
  • Data Showing Cyp A Inhibitory Activity & Immunosuppressive Potential of Compounds of Formula (I)
  • Compound *Cyp A nM (n = 1) **CaN-Cyp A **CaN + Cyp A
    Tested Protease-free PPlase Assay nM nM ***MLR nM
    Cs A 1.5 >10,000 (n = 2) 210 (n = 3) 56
    Example 1 12 >10,000 (n = 1) 1,000 (n = 1) 540
    [methylene-Sar]3
    Cs A
    Example 2 2.4 3,600 (n = 2) 2,500 (n = 2) 3,500
    [(S)-2
    diethylaminothylthio-
    methyl-Sar]3 Cs A
    Example 3 4.3 11,800 (n = 2) 6,000 (n = 2) 2,000
    [(S)-2-(4-
    pyridyl)ethylthio-
    methyl-Sar]3 Cs A
    Example 4 6.9 2,200 (n = 2) 175 (n = 2)
    [(S)-methylthio-
    methyl-Sar]3 Cs A
    Example 5 2.3 >10,000 (n = 2) 705 (n = 2) 2,700
    [(S)-pentylthio-
    methyl-Sar]3 Cs A
    Example 6 4.7 3,100 (n = 2) 1,400 (n = 2) 10,400
    [(S)-2-
    aminoethylthio-
    methyl-Sar]3 Cs A
    Example 7 2.9 9,800 (n = 2) 1,600 (n = 2)
    [(S)-{(R)-2-amino-
    2-carbomethoxy-
    ethyl} thiomethyl-
    Sar]3 Cs A
    Example 8 4.2 2,900 (n = 2) 4,800 (n = 2) 4,600
    [(S)-2-
    dimethylamino-
    ethylthiomethyl-Sar]3
    Cs A
    Example 9 4.8 3,300 (n = 2) 11,000 (n = 2) 3,800
    [(S)-(4-
    methylpiperidinyl)
    thiomethyl-Sar]3 Cs A
    Example 10 3.9 4,200 (n = 2) 4,800 (n = 2)
    [(S)-2-
    (morpholino)ethyl-
    thiomethyl-Sar]3 Cs A
    Example 11 2.1 3,900 (n = 2) 600 (n = 2) 19,500
    [(S)-
    carbomethoxy-
    methylthiomethyl-
    Sar]3 Cs A
    Example 12 2.7 9,100 (n = 2) 1,200 (n = 2)
    [(S)-
    carbomethoxyethyl-
    thiomethyl-Sar]3 Cs A
    Example 13 9.3 4,100 (n = 2) 4,900 (n = 2)
    [(S)-{(S)-2-amino-
    2-carbomethoxy-1,1-
    dimethylethyl}thio-
    methyl-Sar]3 Cs A
    Example 14 2.9 7,400 (n = 2) 1,200 (n = 2)
    [(S)-{(R)-2-amino-
    2-carbohydroxy-
    ethyl}thiomethyl-
    Sar]3 Cs A
    Example 15 1.9 6,800 (n = 2) 5,300 (n = 2) 18,300
    [(S)-
    carbohydroxy-
    methylthiomethyl-
    Sar]3 Cs A
    Example 16 5.8 2,700 (n = 2) 2,200 (n = 2)
    [(S)-{(S)-2-amino-
    2-carbohydroxy-1,1-
    dimethylethyl}thio-
    methyl-Sar]3 Cs A
    Example 17 2.8 17,000 (n = 2) 3,100 (n = 2)
    [(S)-
    carbohydroxyethyl-
    thiomethyl-Sar]3 Cs A
    Example 18 5.2 4,600 (n = 2) 6,100 (n = 2) 3,300
    [(S)-2-
    isopropylaminoethyl-
    thiomethyl-Sar]3 Cs A
    Example 19 6.8 3,200 (n = 2) 1,900 (n = 2)
    [(S)-2-
    guanidinoethylthio-
    methyl-Sar]3 Cs A
  • General Procedures Followed to Obtain Data *Protease-Free PPlase Assay
  • The protease-free PPlase assay measures the rate of cis to trans conversion of a peptide substrate catalyzed by the enzyme cyclophilin A. Addition of an inhibitor slows the catalyzed rate and a Ki value is obtained.
  • Materials
  • Assay Buffer: 35 mM HEPES pH 7.8, filtered through a 0.2 μm filter. 50 M DTT was added prior to use each day and then the buffer was stored on ice.
  • Enzyme: human recombinant Cyp A (Sigma C3805) enzyme was diluted to 1 M with enzyme dilution buffer (20 mM HEPES pH 7.8, 40% glycerol, 50 M DTT and 1 μM BSA) and stored at −20° C.
  • Substrate: SUC-AAPF-pNA (from Bachem AG, L-1400), 20 mg/ml prepared 0.5 M LiCl in trifluoroethanol.
  • Method
  • All readings were taken with an Agilent 8453 Spectrophotometer which consists of a cuvette holder, stirrer and chiller to maintain a stirred cuvette temperature of 10.0±0.1° C. The temperature is monitored by the use of temperature probe. To prevent UV degradation of test compounds, the light below 290 nm was blocked using a glass slide in the light path. 1.5 ml of assay buffer was put into a 3 ml quartz cuvette and cooled to 10.0±0.1° C. while stirring (vigorous but not so fast as to produce cavitation). The inhibitor was diluted in 100% DMSO, and then added to the assay to a maximum final concentration of 0.5% DMSO in the assay. A blank spectrum was obtained, then 3 μL of enzyme was added (2 nM final concentration) and then 3 μL substrate (60 μM final concentration) added. The absorbance was measured at 330 nm for 300 s or 500 s for blank runs (NOTE: the substrate must be added in one quick injection and the measurements started immediately to minimize mixing errors).
  • A first order rate equation was fitted to the absorbance data, for each concentration of inhibitor, to obtain the rate constant (the first 10 to 15 seconds were excluded as mixing causes errors in this portion of curve). The catalytic rate was calculated from the enzymatic rate constant minus the background rate constant. An exponential curve was generated using the catalytic rate constants versus the inhibitor concentration to obtain the K, value for the inhibitor.
  • **Calcineurin Phosphatase (CaN) Assay
  • Calcineurin is a serine-threonine protein phosphatase that on activation dephosphorylates members of the nuclear factor of activated T cells (NFAT), which are important in T lymphocyte activation. Cs A bound to cyclophilin A (“Cyp A”) inhibits calcineurin activity, thus resulting in immunosuppressive effects. Although Cs A only inhibits calcineurin when bound to Cyp A, some Cs A analogues will also bind calcineurin in the absence of Cyp A. To investigate the immunosuppressive potential of exemplary compounds of Formula (I), which are cyclosporin analogues, their ability to inhibit calcineurin activity was measured in the presence and absence of Cyp A.
  • The CaN assay kit used is based on a colorimetric assay for measuring calcineurin phosphatase activity, and it is commercially available (Enzo Life Sciences and Calbiochem). Calmodulin is also required for calcineurin activity and RII phosphopeptide is used as an efficient peptide substrate for calcineurin. We have modified the method to enable measurement of Cyp A-dependent and Cyp A-independent inhibition of calcineurin through the addition of Cyp A in a 1:1 complex with the inhibitor. The detection of free phosphate released is based on the classic Malachite green assay.
  • Materials Used
  • Enzo Life Sciences CaN Assay Kit: BML-AK804
  • 2× assay buffer: 100 mM Tris, pH7.5, 200 mM NaCl, 12 mM MgCl2, 1 mM DTT, 0.05% NP-40, 1 mM CaCl2)
  • Malachite Green: BIOMOL Green™ reagent
  • Calmodulin (Human, recombinant): was thawed on ice, diluted 1:50 with 2× assay buffer, and then stored on ice.
  • Calcineurin: was thawed quickly, stored on ice immediately, diluted 1:12.5 with 1× assay buffer, and then stored on ice.
  • R-II Substrate: 915 μL ultrapure water (UPW) was added to the 1.5 mg vial substrate to give a final concentration of 0.75 mM.
  • Inhibitors: 2.5 mM inhibitor in 100% DMSO.
  • Cyp A: recombinant human CypA (Sigma C3805), 1 mg/ml
  • Method
  • Inhibitor dilutions: inhibitor compounds were diluted in UPW in polypropylene low-binding 96 well plates at 5× the final assay concentration. For samples ‘without Cyp A’, a 4-point dilution series of the inhibitor was prepared in duplicate to obtain a final assay concentration of 10, 1, 0.1 and 0.01 μM. For samples ‘with Cyp A’, a 7-point dilution series was prepared to obtain a 1:1 complex of the inhibitor with CypA; the inhibitor and Cyp A final assay concentrations of 10, 3.33, 1.11, 0.37, 0.12, 0.04, 0.014 μM were prepared. Cs A inhibitor controls were also prepared to obtain a final concentration of 10 μM Cs A with and without 10 μM Cyp A.
  • Assay Setup: using the half area 96 well plates supplied with the kit, 10 μl UPW was added to duplicate wells to provide the non-inhibited control. 10 μl of the inhibitor or the inhibitor/Cyp A complex was added to the appropriate sample wells. 25 μl of the 2× assay buffer with CaM was added to all wells, then 5 μl of CaN was added to all wells (40 U per well final concentration) except duplicate ‘no calcineurin blank’ wells to which 5 μL 1× assay buffer was added. The assay plate was placed in an oven at 30° C. for 15 minutes to equilibrate to the reaction temperature. The reaction was started by the addition of 10 μl RII-peptide (0.15 mM final concentration). The reaction was allowed to proceed at 30° C. for a time period in which the reaction is linear for about 60 minutes. The reaction was then terminated by adding 100 μl of the Malachite Green reagent. The color was allowed to develop for 15-30 minutes at room temperature before the absorbance at 620 nm was measured using a plate reader (Molecular Devices—SpectraMax M5). The data were analyzed by subtracting ‘no Calcineurin blank’ from all the absorbance readings and plotting the background corrected absorbances against Log10 inhibitor concentration. A sigmoidal-dose response curve was fitted to the data using GraphPad Prism Software.
  • ***Mixed Lymphocyte Reaction (“MLR”) Assay
  • The MLR assay is another means of estimating the immunosuppressive potential of test compounds. Female C57BL/6 and BALB/c mice, 6-8 weeks of age, were obtained from the Frederick Cancer Research and Development Center of the National Cancer Institute (Frederick, Md.). Spleens were harvested aseptically from all mice and single cell suspensions were prepared by disaggregating the cells with frosted glass slides, allowing the debris to settle, and washing the cells twice with complete medium. Complete medium consisted of RPMI 1640 medium containing 25 mM HEPES buffer (HyClone, Logan, Utah) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Atlanta Biologicals, Lawrenceville, Ga.), 100 μg/mL streptomycin, 100 U/mL penicillin G, 0.25 μg/mL amphotericin B (HyClone), 2 mM L-glutamine dipeptide (HyClone), and 2×10−5 M 2-mercaptoethanol (Sigma). Cells were washed twice and resuspended in complete medium. Cell counts were performed using a Beckman Coulter Z-1 particle counter (Fullerton, Calif.). Cell viability was determined by propidium iodide (PI) staining using an Accuri C6 flow cytometer (Ann Arbor, Mich.).
  • Spleen cells from C57BL/6 (H-2b) and BALB/c (H-2d) were used as responder (R) and stimulator (S) cells, respectively. Cells were plated in triplicate in 96-well flat microtiter plates (Costar, Cambridge, Mass.) such that each well contained 2×105 R and 8×105 S cells. Cultures were incubated in the absence or presence of various concentrations of Cs A, test compounds, or medium at 37° C. in humidified 5% CO2 for five days, pulsed with 3H-thymidine (3H-TdR) for the final 16 hours of incubation, and harvested using a Brandel 96-well cell harvester (Gaithersburg, Md.). Proliferation was measured by counting the radioactivity on filter mats in a Wallac 1450 Microbeta TriLux scintillation counter (Turku, Finland). Controls to demonstrate effective inactivation by the x-irradiation were performed by incubating the S cells with 5 μg/mL of PHA at 2×105 cells/well. These control cultures were incubated for 3 days under the same conditions as those described for the MLR; lymphoproliferation was determined in the same manner as described above.
  • Methods of Treatment
  • Compositions of the invention may be used to treat patients suffering from dry eye, to treat blepharitis and meibomian gland disease, to restore corneal sensitivity that has been impaired due to surgery on the cornea or other surface of the eye, to treat allergic conjunctivitis and atopic and vernal keratoconjunctivitis, and to treat ptyregia, ocular symptoms of graft versus host disease, ocular allergy, atopic keratoconjunctivitis, vernal keratoconjunctivitis, uveitis, anterior uveitis, Behcet's disease, Steven Johnson syndrome, ocular cicatricial pemphigoid, chronic ocular surface inflammation caused by viral infection, herpes simplex keratitis, ocular rosacea, and pinguecula, and to prevent corneal transplant rejection.
  • The International Dry Eye Workshop (DEWS) defines dry eye as “a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface, accompanied by increased osmolarity of the tear film and inflammation of the ocular surface.” It includes those conditions, such as keratoconjunctivitis sicca, that are caused by tear deficiency or excessive evaporation of tears.
  • Blepharitis is a chronic disorder producing inflammation of the anterior and posterior lid margin, with involvement of skin and its related structures (hairs and sebaceous glands), the mucocutaneous junction, and the meibomian glands. It can also affect the conjunctiva, tear film, and the corneal surface in advanced stages and may be associated with dry eye. Blepharitis is commonly classified into anterior or posterior blepharitis, with anterior affecting the lash bearing region of the lids, and posterior primarily affecting the meibomian gland orifices.
  • Meibomian gland disease most often occurs as one of three forms: primary meibomitis, secondary meibomitis, and meibomian seborrhea. Meibomian seborrhea is characterized by excessive meibomian secretion in the absence of inflammation (hypersecretory meibomian gland disease). Primary meibomitis, by contrast, is distinguished by stagnant and inspissated meibomian secretions (obstructive hypersecretory meibomian gland disease). Secondary meibomitis represents a localized inflammatory response in which the meibomian glands are secondarily inflamed in a spotty fashion from an anterior lid margin blepharitis.
  • Impaired corneal sensitivity often occurs after refractive surgery, such as photorefractive keratectomy, laser assisted sub-epithelium keratomileusis (LASEK), EPI-LASEK, customized transepithelial non-contact ablation, or other procedures in which the corneal nerves are severed. Impaired corneal sensitivity may also occur after viral infection, such as by HSV-1, HSV-2, and VZV viruses. Patients with impaired corneal sensitivity often complain that their eyes feel dry, even though tear production and evaporation may be normal, suggesting that “dryness” in such patients may actually be a form of corneal neuropathy that results when corneal nerves are severed by surgery or inflamed after viral infection.
  • Allergic conjunctivitis is an inflammation of the conjunctiva resulting from hypersensitivity to one or more allergens. It may be acute, intermittent, or chronic. It occurs seasonally, that is, at only certain time of the year, or it occurs perennially, that is, chronically throughout the year. Symptoms of seasonal and perennial allergic conjunctivitis include, in addition to inflammation of the conjunctiva, lacrimation, tearing, conjunctival vascular dilation, itching, papillary hyperlasia, chemosis, eyelid edema, and discharge from the eye. The discharge may form a crust over the eyes after a night's sleep.
  • Atopic keratoconjunctivitis is a chronic, severe form of allergic conjunctivitis that often leads to visual impairment. Symptoms include itching, burning, pain, redness, foreign body sensation, light sensitivity and blurry vision. There is often a discharge, especially on awakening from a night's sleep; the discharge may be stringy, ropy, and mucoid. The lower conjunctiva is often more prominently affected than the upper conjunctiva. The conjunctiva may range from pale, edematous, and featureless to having the characteristics of advanced disease, including papillary hypertrophy, subepithelial fibrosis, formix foreshortening, trichiasis, entropion, and madurosis. In some patients the disease progresses to punctate epithelial erosions, corneal neovascularization, and other features of keratopathy which may impair vision. There is typically goblet cell proliferation in the conjunctiva, epithelial pseudotubular formation, and an increased number of degranulating eosinophils and mast cells in the epithelium. CD25+ T lymphocytes, macrophages, and dendritic cells (HLA-DR.sup.+, HLA-CD1+) are significantly elevated in the substantia propria.
  • Like atopic keratoconjunctivitis, vernal keratoconjunctivitis is a severe form of allergic conjunctivitis, but it tends to affect the upper conjunctiva more prominently than the lower. It occurs in two forms. In the palpebral form, square, hard, flattened, closely packed papillae are present; in the bulbar (limbal) form, the circumcorneal conjunctiva becomes hypertrophied and grayish. Both forms are often accompanied by a mucoid discharge. Corneal epithelium loss may occur, accompanied by pain and photophobia, as may central corneal plaques and Trantas' dots.
  • Uveitis, the inflammation of the uvea, is responsible for about 10% of the visual impairment in the United States. Phacoanaphylactic endophthalmitis is a human autoimmune disease. Panuveitis refers to inflammation of the entire uveal (vascular) layer of the eye. Posterior uveitis generally refers to chorioentinitis, and anterior uveitis refers to iridocyclitis. The inflammatory products (i.e. cells, fibrins, excess proteins) of these inflammations are commonly found in the fluid spaces if the eye, i.e. anterior chamber, posterior chamber and vitreous space as well as infiltrating the tissue intimately involved in the inflammatory response. Uveitis may occur following surgical or traumatic injury to the eye; as a component of an autoimmune disorder, such as rheumatoid arthritis, Behcet's disease, ankylosing spondylitis, and sarcoidosis; as an isolated immune mediated ocular disorder, such as pars planitis, iridocyclitis etc., unassociated with known etiologies; and following certain systemic diseases which cause antibody-antigen complexes to be deposited in the uveal tissues. Together these disorders represent the non-infectious uveitities.
  • Phacoanaphylaxis is a severe form of uveitis in which the lens in the causative antigen. The lens proteins are normally secluded by the lens capsule since before birth. When these proteins are released into the eye by injury or by surgery or occasionally during cataract development, they can become intensely antigenic and incite an autoimmune response. If the response is moderate it is seen as chronic uveitis. If it is very fast in progression the eye becomes seriously inflamed in all segments. This latter response is named phacoanaphylaxis.
  • Uveitis is a prominent feature of Behcet's disease, a multi-system inflammatory disorder also characterized by oral and genital ulcers, cutaneous, vascular, joint, and neurological manifestations
  • Rosacea is a chronic and common skin disorder with no identified cause or cure. The pathogenesis of rosacea is thought to have multiple factors. Possible factors include exposure to the demodex folliculorum mite, gastrointestinal disease or a vasodilation disorder, and other triggers such as diet or sunlight. Patients may present with a variety of symptoms, including inflammatory papules, edema, telangiectasia, rhinophyma and ocular symptoms. The ocular signs of rosacea include blepharitis, including anterior blepharitis, conjunctivitis, iritis, iridocyclitis, keratitis, meibomian gland dysfunction, telangiectasia, erythema, chalazion, hordeolum, interpalpebral hyperemia, conjuctival hyperemia, ciliary base injection, bulbar injection, crusts, sleeves, and superficial punctuate keratopathy. The ocular symptoms are nonspecific and may include burning, tearing, decreased tear secretion, redness, and foreign body or gritty or dry sensation, irritation, Itchiness, Blurred vision, Photosensitivity, Watery eyes, bloodshot eyes, Burning, telangiectasia, irregularity of the lid margins, and meibomian gland dysfunction.
  • Pinguecula is a benign, yellowish brown proliferative growth that forms on the conjunctiva. Pinguecula may cause irritation and scratchiness of the eye, dry eye, inflammation of the conjunctiva and effect appearance of the eye. Inflamed pinguecula, which cause ocular irritation or become unsightly, may require surgical removal. However, the post-operation scar may be as cosmetically objectionable as the pinguecula and pinguecula regrowth may occur following surgical removal.
  • Allogeneic bone marrow transplantation (BMT) is a well-established treatment for malignant and non-malignant hematological diseases, and is performed in tens of thousands of patients each year. Mature donor T cells within the stem cell graft are the main mediators of the beneficial immune effects, but they are also responsible for the induction of graft-versus-host disease (GVHD), the major cause of morbidity and mortality in BMT patients. GVHD occurs when transplanted donor-derived T cells recognize proteins expressed by recipient antigen-presenting cells. Consequently, this recognition induces donor T-cell activation, proliferation, and differentiation, leading to a cellular and inflammatory attack on recipient target tissues. Acute or chronic GVHD occurs within a 100-day period post-BMT that leads to dermatitis, enteritis, and hepatitis. Ocular symptoms include blurry vision, foreign body sensation, burning sensation, severe light sensitivity, chronic conjunctivitis, dry eye, and eye pain.
  • Pharmaceutical Compositions
  • The present invention also relates to pharmaceutical compositions comprising at least one compound of general formula (I), the compound being present alone or in combination with one or more pharmaceutically acceptable excipients. A “pharmaceutically acceptable excipient” is one that is compatible with the active ingredient of the composition and not harmful to the person being administered the pharmaceutical composition. Mixtures of two or more of such suitable excipients may be used.
  • For topical ocular applications, pharmaceutical compositions may be prepared by combining a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, as an active ingredient, with conventional ophthalmically acceptable pharmaceutical excipients, and by preparation of unit dosage forms suitable for topical ocular use. The therapeutically efficient amount typically is between about 0.0001 and about 5% (w/v), preferably about 0.001 to about 1.0% (w/v) in liquid formulations. The actual dose of the active compounds of the present invention depends on the specific compound, and on the condition to be treated; the selection of the appropriate dose is well within the knowledge of one of ordinary skill in the art.
  • U.S. Pat. No. 5,474,979, the entire contents of which are incorporated herein by reference, provides examples of ophthalmically acceptable pharmaceutical excipients. The patent discloses the vehicle used in Restasis®, cyclosporin A 0.05%, manufactured by Allergan, Inc.
  • A therapeutically effective amount in the claimed pharmaceutical composition is a concentration useful to observe a therapeutic effect as compared to a placebo composition that, except for the absence of a compound of formula (I), is otherwise identical to the pharmaceutical composition.

Claims (21)

1. A compound of formula (I)
Figure US20120088734A1-20120412-C00078
wherein:
A is —CH═CHR, —CH═CH—CH═CHR, or —CH2CH2R,
wherein R is —CH3; —CH2SH; —CH2S—Cn wherein n is 1, 2, 3, 4, 5, or 6; —CH2-carboxyl; carboxyl;
Figure US20120088734A1-20120412-C00079
wherein R10 is C1-C6 alkyl;
or
Figure US20120088734A1-20120412-C00080
wherein R11=C1-C6 alkyl;
B is —CH2CH3, 1-hydroxyethyl, isopropyl, or n-propyl;
C is isobutyl, 2-hydroxyisobutyl, or 1-methylpropyl;
D is —CH3 or —CH2OH;
R1 and R2, which are identical or different, each represents hydrogen or C1-C4 alkyl, or together represent C3-C7 cycloalkyl;
X is sulfur or —S(O)n, wherein n is 1 or 2;
R3 is hydrogen; straight or branched C1-C6 alkyl; straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl;
—(CHR′)m—NH—CNH2NH;
—(CHR′)m—COOH;
—(CHR′)m—NHR″;
—(CHR′)m—NHCOR″;
wherein m is 1, 2, 3, 4, 5, or 6;
each R′ is independently H, straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl; or is absent;
R″ is —(CHR)pN(CH3)2, —(CHR′)pNHCOC, —(CHR′)pNHCOOH, wherein p is 0, 1, 2, 3, 4, 5, or 6; and
wherein R3 may be optionally substituted with one or more groups, identical or different, of C1-C6 alkyl; halogen; hydroxyl; alkoxycarbonyl; carboxyl; cycloalkyl; saturated or partially unsaturated 5-6 member heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur, which heterocyclyl is optionally substituted by one or more groups of C1-C6 alkyl; aryl; heteroaryl; amino; monoalkylamino; dialkylamino; amidino; guanidine; or urea.
or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 selected from the group consisting of:
[(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-(4-pyridyl)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-methylthiomethyl-Sar]3 cyclosporin A;
[(S)-pentylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-aminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(R)-2-amino-2-carbomethoxy-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-2-dimethylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-(4-methylpiperidinyl)thiomethyl-Sar]3 cyclosporin A;
[(S)-2-(morpholino)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-carbomethoxymethylthiomethyl-Sar]3 cyclosporin A;
[(S)-carbomethoxyethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(S)-2-amino-2-carbomethoxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-{(R)-2-amino-2-carbohydroxy-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-carbohydroxymethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(S)-2-amino-2-carbohydroxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-carbohydroxyethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-isopropylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-guanidinoethylthiomethyl-Sar]3 cyclosporin A;
[methylene-Sar]3 cyclosporin A;
[dihydro-MeBmt]1 [Methylene-Sar]3 cyclosporin A;
[methylene-Sar]3 cyclosporin D;
[(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin A;
[dihydro-MeBmt]1 [(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-diethylaminoethylthiomethyl-Sar]3 cyclosporin D;
[(S)-2-(4-pyridyl)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-methylthiomethyl-Sar]3 cyclosporin A;
[(S)-pentylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-aminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(R)-2-amino-2-carbomethoxy-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-2-dimethylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-(4-methylpiperidinyl)thiomethyl-Sar]3 cyclosporin A;
[(S)-2-(morpholino)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-carbomethoxymethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(S)-2-Amino-2-carbomethoxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-2-(N-imidazolyl)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-(N-pyrazolyl)ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-diethylamino-1,1-dimethyl-ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-morpholino-1,1-dimethyl-ethylthiomethyl-Sar]3 cyclosporin A;
[(S)-3-diethylaminopropylthiomethyl-Sar]3 cyclosporin A;
[(S)-3-(morpholino)-propylthiomethyl-Sar]3 cyclosporin A;
[(S)-mercaptomethyl-Sar]3 cyclosporin A;
[(S)-allylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(R)-2-amino-2-carbohydroxy-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-carbohydroxymethylthiomethyl-Sar]3 cyclosporin A;
[(S)-{(S)-2-amino-2-carbohydroxy-1,1-dimethyl-ethyl}thiomethyl-Sar]3 cyclosporin A;
[(S)-carbohydroxyethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-isopropylaminoethylthiomethyl-Sar]3 cyclosporin A;
[(S)-2-guanidinoethyl-1-sulfanylmethyl-Sar]3 cyclosporin A hydrochloride;
[(S)-2-diethylaminoethyl-1-sulfinylmethyl-Sar]3 cyclosporin A;
[dihydro-MeBmt]1 [(S)-2-diethylaminoethyl-1-sulfinylmethyl-Sar]3 cyclosporin A;
[(S)-3-diethylaminopropyl-1-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-3-(morpholino)propyl-1-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-(4-methylpiperidinyl)-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-(2-diethylamino-1,1-dimethyl)-ethyl-1-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-(2-morpholino-1,1-dimethyl)-ethyl-1-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-3-(4-oxy-morpholin-4-yl)-propyl-1-sulfonylmethyl-Sar]3 cyclosporin A;
[(S)-2-(3-carboxypropionylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A;
[(S)-2-(4-carboxybutyrylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A;
[(S)-2-(acetylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A;
[(S)-2-(dimethylamino-acetylamino)ethyl-1-sulfanylmethyl-Sar]3 cyclosporin A;
and pharmaceutically acceptable salts of any of the foregoing compounds.
3. A compound according to claim 1, wherein A is —CH═CHCH3, B is —CH2CH3, C is isobutyl, and D is —CH3.
4. A compound according to claim 1, wherein A is —CH═CHCH3, B is 1-hydroxyethyl, C is isobutyl, and D is —CH3.
5. A compound according to claim 1, wherein A is —CH═CHCH3, B is isopropyl, C is isobutyl, and D is —CH3.
6. A compound according to claim 1, wherein A is —CH═CHCH3, B is n-propyl, C is isobutyl, and D is —CH3.
7. A compound according to claim 1, wherein A is —CH═CHCH3, B is —CH2CH3, C is 1-methylpropyl, and D is —CH3.
8. A compound according to claim 1, wherein A is —CH═CHCH3, B is —CH2CH3, C is isobutyl, and D is —CH2OH.
9. A compound according to claim 1, wherein A is —CH═CHCH3, B is —CH2CH3, C is 2-hydroxy isobutyl, and D is —CH3.
10. A compound according to claim 1, wherein R3 is selected from the group consisting of
—(CHR′)m—NH—CNH2NH;
—(CHR′)m—COOH;
—(CHR′)m—NHR″;
—(CHR′)m—NHCOR″;
wherein m is 1, 2, 3, 4, 5, or 6;
each R′ is independently H, straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl or is absent; and
R″ is —(CHR)pN(CH3)2, —(CHR′)pNHCOC, or —(CHR′)pNHCOOH, wherein p is 0, 1, 2, 3, 4, 5, or 6.
11. A compound according to claim 10, wherein m is 1, 2, or 3; each R′ is independently H or is absent; and R″ is —(CHR)pN(CH3)2, —(CHR′)pNHCOC, or —(CHR′)pNHCOOH, wherein p is 0, 1, or 2.
12. A pharmaceutical composition comprising at least one compound of claim 1, the compound being present alone or in combination with one or more pharmaceutically acceptable excipients.
13. A pharmaceutical composition comprising at least one compound of claim 1, the compound being present alone or in combination with one or more pharmaceutically acceptable excipients, wherein the concentration of the compound in the composition is about 0.01 to about 0.05 weight percent.
14. A method for treating a condition selected from dry eye, blepharitis, meibomian gland disease, allergic conjunctivitis, atopic and vernal keratoconjunctivitis, ptyregia, ocular symptoms of graft versus host disease, ocular allergy, atopic keratoconjunctivitis, vernal keratoconjunctivitis, uveitis, anterior uveitis, Behcet's disease, Steven Johnson syndrome, ocular cicatricial pemphigoid, chronic ocular surface inflammation caused by viral infection, herpes simplex keratitis, ocular rosacea, pinguecula, to prevent corneal transplant rejection, and to restore corneal sensitivity that has been impaired due to surgery on the cornea or other surface of the eye, the method comprising administering to a mammal a compound of claim 1.
15. The method according to claim 14, wherein the mammal is a human.
16. The method of claim 15, comprising administering the compound of claim 1 wherein R3 is selected from the group consisting of
—(CHR′)m—NH—CNH2NH;
—(CHR′)m—COOH;
—(CHR′)m—NHR″;
—(CHR′)m—NHCOR″;
wherein m is 1, 2, 3, 4, 5, or 6;
each R′ is independently H, straight or branched C1-C6 alkyl, straight or branched C2-C6 alkenyl; straight or branched C2-C6 alkynyl; C3-C7 cycloalkyl; C4-C7 heterocyclyl having 1-3 heteroatoms selected from nitrogen, oxygen, and sulfur; aryl; heteroaryl or is absent; and
R″ is —(CHR)pN(CH3)2, —(CHR′)pNHCOC, or —(CHR′)pNHCOOH, wherein p is 0, 1, 2, 3, 4, 5, or 6.
17. The method of claim 16, wherein m is 1, 2, or 3; each R′ is independently H or is absent; and R″ is —(CHR)pN(CH3)2, —(CHR′)pNHCOC, or —(CHR′)pNHCOOH, wherein p is 0, 1, or 2.
18. A compound of formula (III):
Figure US20120088734A1-20120412-C00081
wherein:
A represents —CH═CHR or —CH2CH2R, wherein R represents —CH3, —CH2SH; —CH2S—Cn, wherein n=1-6; —CH2-carboxyl, carboxyl, alkoxycarbonyl, or —CH2-alkoxycarbonyl;
B represents —CH2CH3, 1-hydroxyethyl, isopropyl, or n-propyl;
C represents isobutyl, 2-hydroxyisobutyl, or 1-methylpropyl;
D represents —CH3 or —CH2OH;
or a pharmaceutically acceptable salt thereof.
19. A compound according to claim 18, wherein A is —CH═CHCH3, B is CH2CH3, C is isobutyl, and D is —CH3.
20. A process for the preparation of a compound of formula (III)
Figure US20120088734A1-20120412-C00082
wherein:
A represents —CH═CHR or —CH2CH2R, wherein R represents —CH3, —CH2SH; —CH2S—Cn, wherein n=1-6; —CH2-carboxyl, carboxyl, alkoxycarbonyl, or —CH2-alkoxycarbonyl;
B represents —CH2CH3, 1-hydroxyethyl, isopropyl, or n-propyl;
C represents isobutyl, 2-hydroxyisobutyl, or 1-methylpropyl;
D represents —CH3 or —CH2OH;
the process comprising reacting a compound of the formula (II)
Figure US20120088734A1-20120412-C00083
wherein A, B, C, and D are as previously defined, with a strong base, at a temperature in the range of −78° C. to −70° C., followed by treatment with a flow of carbon dioxide gas between −70° C. and 15° C., followed by treatment with excess chloromethylchloroformate between −50° C. and room temperature, followed by quenching of the reaction mixture with acetic acid at 0° C. to room temperature to generate a compound of formula (III).
21. The method of claim 20, wherein the strong base is lithium diisopropylamide.
US13/270,964 2010-10-12 2011-10-11 Cyclosporin Analogs Abandoned US20120088734A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/270,964 US20120088734A1 (en) 2010-10-12 2011-10-11 Cyclosporin Analogs
US14/534,677 US9493511B2 (en) 2010-10-12 2014-11-06 Cyclosporin analogs
US15/290,171 US10065992B2 (en) 2010-10-12 2016-10-11 Cyclosporin analogs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US39244910P 2010-10-12 2010-10-12
US13/270,964 US20120088734A1 (en) 2010-10-12 2011-10-11 Cyclosporin Analogs

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/534,677 Continuation US9493511B2 (en) 2010-10-12 2014-11-06 Cyclosporin analogs

Publications (1)

Publication Number Publication Date
US20120088734A1 true US20120088734A1 (en) 2012-04-12

Family

ID=44883784

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/270,964 Abandoned US20120088734A1 (en) 2010-10-12 2011-10-11 Cyclosporin Analogs
US14/534,677 Active US9493511B2 (en) 2010-10-12 2014-11-06 Cyclosporin analogs
US15/290,171 Active US10065992B2 (en) 2010-10-12 2016-10-11 Cyclosporin analogs

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/534,677 Active US9493511B2 (en) 2010-10-12 2014-11-06 Cyclosporin analogs
US15/290,171 Active US10065992B2 (en) 2010-10-12 2016-10-11 Cyclosporin analogs

Country Status (13)

Country Link
US (3) US20120088734A1 (en)
EP (1) EP2627664B1 (en)
JP (2) JP5805774B2 (en)
KR (1) KR20140026329A (en)
CN (1) CN103328497A (en)
AU (1) AU2011316690C1 (en)
BR (1) BR112013008864A2 (en)
CA (1) CA2814192A1 (en)
IL (1) IL225617A0 (en)
MX (1) MX2013004061A (en)
RU (1) RU2013119869A (en)
SG (1) SG189880A1 (en)
WO (1) WO2012051194A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130210704A1 (en) * 2010-12-03 2013-08-15 S&T Global Inc. Novel cyclosporin derivatives and uses thereof
WO2013181339A3 (en) * 2012-06-01 2014-03-13 Allergan, Inc. Cyclosporin a analogs
US9175042B2 (en) 2010-10-12 2015-11-03 Allergan, Inc. Cyclosporin analogs
US9402913B2 (en) 2013-03-08 2016-08-02 Allergan, Inc. Cyclosporine A steroid conjugates
US9914755B2 (en) 2015-01-08 2018-03-13 Allergan, Inc. Cyclosporin derivatives wherein the MeBmt sidechain has been cyclized
WO2018112255A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with an immunosuppressant
WO2019246312A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012021796A2 (en) 2010-08-12 2012-02-16 S&T Global, Inc. Novel cyclosporin derivatives for the treatment and prevention of a viral infection
JP5805774B2 (en) * 2010-10-12 2015-11-10 アラーガン、インコーポレイテッドAllergan,Incorporated Cyclosporine analog
HUE042490T2 (en) * 2010-12-15 2019-07-29 Contravir Pharmaceuticals Inc Cyclosporine analogue molecules modified at amino acid 1 and 3
GB201222455D0 (en) * 2012-12-13 2013-01-30 Perioc Ltd Novel pharmaceutical formulations and their use in the treatment of periodontaldisease
EP3277704A1 (en) 2015-03-31 2018-02-07 Allergan, Inc. Cyclosporins modified on the mebmt sidechain by heterocyclic rings
EP3458470A4 (en) * 2016-05-17 2020-04-22 S&T Global Inc. Novel cyclosporin derivatives and uses thereof
WO2021173474A1 (en) * 2020-02-26 2021-09-02 Bacainn Biotherapeutics, Ltd. Cyclosporine compositions and methods of use
CN113173974A (en) * 2021-03-18 2021-07-27 北京大学深圳研究生院 Synthesis method of cyclosporine derivative

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5474979A (en) * 1994-05-17 1995-12-12 Allergan, Inc. Nonirritating emulsions for sensitive tissue
WO2013028615A2 (en) * 2011-08-19 2013-02-28 S&T Golbal Inc. Novel cyclosporin derivatives for the treatment and prevention of viral infections

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0194972B1 (en) 1985-03-11 1992-07-29 Sandoz Ag Novel cyclosporins
US4649047A (en) * 1985-03-19 1987-03-10 University Of Georgia Research Foundation, Inc. Ophthalmic treatment by topical administration of cyclosporin
US4839342A (en) 1987-09-03 1989-06-13 University Of Georgia Research Foundation, Inc. Method of increasing tear production by topical administration of cyclosporin
FR2757521B1 (en) 1996-12-24 1999-01-29 Rhone Poulenc Rorer Sa NOVEL CYCLOSPORIN DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2757522B1 (en) * 1996-12-24 1999-01-29 Rhone Poulenc Rorer Sa CYCLOSPORIN DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2762843B1 (en) 1997-04-30 1999-12-10 Rhone Poulenc Rorer Sa NOVEL CYCLOSPORIN DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
PT1086124E (en) * 1998-06-12 2004-04-30 Chem Ag C NEW CYCLOSPORINS
US6254860B1 (en) * 1999-04-13 2001-07-03 Allergan Sales, Inc. Ocular treatment using cyclosporin-A derivatives
US20050059583A1 (en) * 2003-09-15 2005-03-17 Allergan, Inc. Methods of providing therapeutic effects using cyclosporin components
US7676680B2 (en) 2005-07-08 2010-03-09 Morgan Stanley Systems and methods for distributing private placement documents
US7288520B2 (en) 2005-07-13 2007-10-30 Allergan, Inc. Cyclosporin compositions
US8188052B2 (en) 2006-05-19 2012-05-29 Scynexis, Inc. Method for the treatment and prevention of ocular disorders
CN101130058B (en) * 2006-08-23 2011-10-05 华北制药集团新药研究开发有限责任公司 Emulsion of cyclosporin A for eyes and method of producing the same
EP2152291B1 (en) * 2007-05-04 2012-10-24 Allergan, Inc. Use of cyclosporines in the treatment of patients with intraocular lenses
CN102149402B (en) * 2008-07-10 2014-07-09 阿勒根公司 Cyclosporin derivatives for treating ocular and dermal diseases and conditions
BR112012031153A2 (en) * 2010-07-02 2016-11-08 Exxonmobil Upstream Res Co low emission triple-cycle power generation systems and methods
WO2012005494A2 (en) 2010-07-06 2012-01-12 엘지전자 주식회사 Method and device for allocating wireless resources for a machine type communication device in a wireless communication system
JP5805774B2 (en) * 2010-10-12 2015-11-10 アラーガン、インコーポレイテッドAllergan,Incorporated Cyclosporine analog
US9890198B2 (en) 2010-12-03 2018-02-13 S&T Global Inc. Cyclosporin derivatives and uses thereof
WO2012075494A1 (en) * 2010-12-03 2012-06-07 S&T Global Inc. Novel cyclosporin derivatives for the treatment and prevention of a viral infection

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5474979A (en) * 1994-05-17 1995-12-12 Allergan, Inc. Nonirritating emulsions for sensitive tissue
WO2013028615A2 (en) * 2011-08-19 2013-02-28 S&T Golbal Inc. Novel cyclosporin derivatives for the treatment and prevention of viral infections

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9175042B2 (en) 2010-10-12 2015-11-03 Allergan, Inc. Cyclosporin analogs
US9561259B2 (en) 2010-10-12 2017-02-07 Allergan, Inc. Cyclosporin analogs
US9890198B2 (en) * 2010-12-03 2018-02-13 S&T Global Inc. Cyclosporin derivatives and uses thereof
US10647747B2 (en) * 2010-12-03 2020-05-12 S&T Global Inc. Cyclosporin derivatives and uses thereof
US20130210704A1 (en) * 2010-12-03 2013-08-15 S&T Global Inc. Novel cyclosporin derivatives and uses thereof
US20180105558A1 (en) * 2010-12-03 2018-04-19 S&T Global Inc. Novel cyclosporin derivatives and uses thereof
US9266927B2 (en) 2012-06-01 2016-02-23 Allergan, Inc. Cyclosporin A analogs
EP3263587A1 (en) * 2012-06-01 2018-01-03 Allergan, Inc. Cyclosporin a analogs
WO2013181339A3 (en) * 2012-06-01 2014-03-13 Allergan, Inc. Cyclosporin a analogs
US9402913B2 (en) 2013-03-08 2016-08-02 Allergan, Inc. Cyclosporine A steroid conjugates
US9914755B2 (en) 2015-01-08 2018-03-13 Allergan, Inc. Cyclosporin derivatives wherein the MeBmt sidechain has been cyclized
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems
WO2018112255A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with an immunosuppressant
US11523772B2 (en) 2016-12-14 2022-12-13 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with an immunosuppressant
WO2019246312A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator

Also Published As

Publication number Publication date
MX2013004061A (en) 2013-07-22
JP2016029050A (en) 2016-03-03
AU2011316690B2 (en) 2016-07-07
EP2627664A1 (en) 2013-08-21
CA2814192A1 (en) 2012-04-19
JP5805774B2 (en) 2015-11-10
AU2011316690C1 (en) 2017-01-19
CN103328497A (en) 2013-09-25
JP2013540776A (en) 2013-11-07
JP6250608B2 (en) 2017-12-20
IL225617A0 (en) 2013-06-27
US20170029469A1 (en) 2017-02-02
US9493511B2 (en) 2016-11-15
SG189880A1 (en) 2013-06-28
WO2012051194A1 (en) 2012-04-19
US10065992B2 (en) 2018-09-04
EP2627664B1 (en) 2018-04-04
AU2011316690A1 (en) 2013-05-02
US20150065433A1 (en) 2015-03-05
RU2013119869A (en) 2014-11-20
KR20140026329A (en) 2014-03-05
BR112013008864A2 (en) 2016-06-28

Similar Documents

Publication Publication Date Title
US10065992B2 (en) Cyclosporin analogs
US9561259B2 (en) Cyclosporin analogs
US10077289B2 (en) Cyclosporins modified on the MeBmt sidechain by heterocyclic rings
US9266927B2 (en) Cyclosporin A analogs

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALLERGAN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRYDRYCH, CATHERINE SIMONE VICTOIRE;CARLING, WILLIAM ROBERT;GARST, MICHAEL E.;AND OTHERS;SIGNING DATES FROM 20111103 TO 20111116;REEL/FRAME:027236/0813

STCB Information on status: application discontinuation

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