WO2011131926A1 - Novel iminosugar combinations - Google Patents

Novel iminosugar combinations Download PDF

Info

Publication number
WO2011131926A1
WO2011131926A1 PCT/GB2011/000594 GB2011000594W WO2011131926A1 WO 2011131926 A1 WO2011131926 A1 WO 2011131926A1 GB 2011000594 W GB2011000594 W GB 2011000594W WO 2011131926 A1 WO2011131926 A1 WO 2011131926A1
Authority
WO
WIPO (PCT)
Prior art keywords
combination
compound
formula
alkyl
dnj
Prior art date
Application number
PCT/GB2011/000594
Other languages
French (fr)
Inventor
Jonathon Mark Tinsley
Original Assignee
Summit Corporation Plc
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 Summit Corporation Plc filed Critical Summit Corporation Plc
Publication of WO2011131926A1 publication Critical patent/WO2011131926A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/008Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy comprising drug dissolved or suspended in liquid propellant for inhalation via a pressurized metered dose inhaler [MDI]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system

Definitions

  • the present invention relates to combinations of N-butyldeoxynojirimycin (a.k.a. Miglustat, ⁇ / ⁇ -DNJ and Zavesca®) with certain carbon-branched iminosugars, to compositions (including pharmaceutical compositions) containing them and to methods of treatment and prophylaxis using the compounds and compositions.
  • N-butyldeoxynojirimycin a.k.a. Miglustat, ⁇ / ⁇ -DNJ and Zavesca®
  • Cystic fibrosis (referred to herein as "CF”, and also known as mucoviscidosis) is a common hereditary disease which affects the entire body, causing progressive disability and often early death. Respiratory dysfunction is the most serious symptom and results from frequent lung infections. Most individuals die in their 20s and 30s from lung failure and lung transplantation is often necessary as CF worsens. A multitude of other symptoms, including sinus infections, poor growth, diarrhoea and infertility result from the effects of CF on other parts of the body. CF is an autosomal recessive disease caused by mutation of the cystic fibrosis
  • CFTR transmembrane conductance regulator
  • the most common mutation comprising over 60% of all mutant alleles, results in the deletion of phenylalanine in the ATP binding cassette at position 508 of the CFTR protein.
  • the resultant AF508 CFTR polypeptide is expressed as a large, misfolded nascent polypeptide which is prematurely destroyed via the ubiquitin pathway but which aggregates following defective processing in the translocation machinery.
  • WO2005/046672 describes the use of various DNJ derivatives having glucosidase inhibitory activity in the treatment of CF.
  • WO2007/123403 describes the use of various iminosugars (and in particular DNJ derivatives) having non-lysosomal glucosylceramidase (glucosidase beta (bile acid) 2) inhibitory activity in the treatment of CF.
  • DNJ derivatives non-lysosomal glucosylceramidase (glucosidase beta (bile acid) 2) inhibitory activity in the treatment of CF.
  • Actelion Pharmaceuticals Limited initiated a Phase lla proof-of-concept clinical trial with ⁇ / ⁇ -DNJ for CF involving 25 patients affected by the AF508 CFTR mutation.
  • R is selected from H; linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl and aralkyl and wherein the optional substitution may be with one or more groups independently selected from: -OH; -F; -CI; -Br; -I; -NH 2 ; alkylamino;
  • dialkylamino linear or branched alkyl, alkenyl, alkynyl and aralkyl; aryl; heteroaryl; linear or branched alkoxy; aryloxy; aralkoxy; -(alkylene)oxy(alkyl); -CN; -N0 2 ; - COOH; -COO(alkyl); -COO(aryl); -C(0)NH(alkyl); -C(0)NH(aryl); sulfonyl; alkylsulfonyl; arylsulfonyl; sulfamoyl; alkylsulfamoyl; alkylthio; alkylsulfonamide; arylsulfonamide; -NHNH 2 ; and -NHOH; or a bioisostere, pharmaceutically acceptable salt or derivative thereof.
  • R 1 is selected from H, C,. 18 alkyl (for example, C 1-9 alkyl, e.g. C,. 6 alkyl), C 2 .i 8 alkenyl (for example, C 2 . 9 alkenyl, e.g. C 2-6 alkenyl) and C 2 .i 8 alkynyl (for example, C 2-9 alkynyl, e.g. C 2 . 6 alkynyl).
  • R 1 may be -H and R 1 selected from C1.18 alkyl (for example, alkyl, e.g. Ci -6 alkyl), C 2 . 18 alkenyl (for example, C 2 . 9 alkenyl, e.g. C 2 -6 alkenyl) and C 2 . 18 alkynyl (for example, C 2 . 9 alkynyl, e.g. C 2 . 6 alkynyl).
  • R 1 represents H; C1-15 alkyl, C1-15 alkenyl or C1 -15 alkynyl, optionally substituted with one or more R 2 ; oxygen or an oxygen containing group such that the compound is an N-oxide; C(0)OR 3 ; C(0)NR 3 R 4 ; S0 2 NR 3 ; OH, OR 3 , or formyl.
  • R 1 may represent C1-9 alkyl, optionally substituted with up to 6 OH, NR 3 R 4 , aryl, 0-C1-3 alkyl, 0-C1-3 alkenyl, C0 2 H, NH(NH)NH 2 , CONR 3 R 4 ; C(0)OR 3 ;
  • the compound of formula I is (3S, 4S)-3-(hydroxymethyl)pyrrolidine-3,4-diol (isoLAB).
  • “comprising,” are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, element, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers.
  • a recited integer e.g. a feature, element, characteristic, property, method/process step or limitation
  • group of integers e.g. features, element, characteristics, properties, method/process steps or limitations
  • treatment refers to an intervention (e.g. the administration of an agent to a subject) which cures, ameliorates or lessens the symptoms of a disease or removes (or lessens the impact of) its cause(s).
  • intervention e.g. the administration of an agent to a subject
  • cures e.g. the administration of an agent to a subject
  • the term is used synonymously with the term "therapy”.
  • treatment refers to an intervention (e.g. the administration of an agent to a subject) which prevents or delays the onset or progression of a disease or reduces (or eradicates) its incidence within a treated population.
  • intervention e.g. the administration of an agent to a subject
  • treatment is used synonymously with the term “prophylaxis”.
  • subject (which is to be read to include “individual”, “animal”, “patient” or “mammal” where context permits) defines any subject, particularly a mammalian subject, for whom treatment is indicated.
  • Mammalian subjects include, but are not limited to, humans, primates, domestic animals, farm animals, pet animals and rodents such as mice, rats, hamsters and guinea pigs. In preferred embodiments, the subject is a human.
  • pharmacoperone is a term of art (from “pharmacological chaperone") used to define a class of biologically active small molecules (sometimes also referred to in the art as “chemical chaperones”) that serve as molecular scaffolds, causing otherwise misfolded mutant proteins to fold and route correctly within the cell.
  • an effective amount of a compound or composition defines an amount that can be administered to a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, but one that is sufficient to provide the desired effect, e.g. the treatment or prophylaxis manifested by a permanent or temporary improvement in the subject's condition.
  • the amount will vary from subject to subject, depending on the age and general condition of the individual, mode of administration and other factors. Thus, while it is not possible to specify an exact effective amount, those skilled in the art will be able to determine an appropriate "effective" amount in any individual case using routine experimentation and background general knowledge.
  • a therapeutic result in this context includes eradication or lessening of symptoms, reduced pain or discomfort, prolonged survival, improved mobility and other markers of clinical improvement. A therapeutic result need not be a complete cure.
  • the term /VB-DNJ is used sensu stricto to cover N-butyldeoxynojirimycin (a.k.a. iglustat, /VB-DNJ and Zavesca®), as well as its pharmaceutically acceptable salts and derivatives.
  • the term is also used sensu lato to cover various derivatives and analogues of deoxynojirimycin (DNJ) which can restore CFTR function, including for example those described in WO2005/046672 and WO2007/123403 (the disclosure of which relating to the structure of the DNJ derivatives is hereby incorporated by reference).
  • DNJ derivatives may have glucosidase inhibitory activity.
  • association of the two or more compounds/agents in a combination may be physical or non-physical.
  • Examples of physically associated combined compounds/agents include:
  • compositions e.g. unitary formulations comprising the two or more
  • compositions comprising material in which the two or more compounds/agents are chemically/physicochemically linked (for example by crosslinking, molecular agglomeration or binding to a common vehicle moiety);
  • compositions comprising material in which the two or more compounds/agents are chemically/physicochemically co-packaged (for example, disposed on or within lipid vesicles, particles (e.g. micro- or nanoparticles) or emulsion droplets); • pharmaceutical kits, pharmaceutical packs or patient packs in which the two or more compounds/agents are co-packaged or co-presented (e.g. as part of an array of unit doses); Examples of non-physically associated combined compounds/agents include:
  • material e.g. a non-unitary formulation
  • material comprising at least one of the two or more compounds/agents together with instructions for the extemporaneous association of the at least one compound/agent to form a physical association of the two or more compounds/agents
  • material e.g. a non-unitary formulation
  • material comprising at least one of the two or more compounds/agents together with instructions for combination therapy with the two or more compounds/agents
  • material comprising at least one of the two or more compounds/agents together with instructions for administration to a patient population in which the other(s) of the two or more compounds/agents have been (or are being) administered;
  • combination therapy is intended to define therapies which comprise the use of a combination of two or more compounds/agents (as defined above).
  • compounds/agents "in combination" in this application may refer to compounds/agents that are administered as part of the same overall treatment regimen. As such, the posology of each of the two or more compounds/agents may differ: each may be administered at the same time or at different times. It will therefore be appreciated that the compounds/agents of the combination may be administered sequentially (e.g. before or after) or
  • the term "pharmaceutical kit” defines an array of one or more unit doses of a pharmaceutical composition together with dosing means (e.g. measuring device) and/or delivery means (e.g. inhaler or syringe), optionally all contained within common outer packaging.
  • dosing means e.g. measuring device
  • delivery means e.g. inhaler or syringe
  • the individual compounds/agents may unitary or non-unitary formulations.
  • the unit dose(s) may be contained within a blister pack.
  • the pharmaceutical kit may optionally further comprise instructions for use.
  • the term "pharmaceutical pack” defines an array of one or more unit doses of a pharmaceutical composition, optionally contained within common outer packaging.
  • pharmaceutical packs comprising a combination of two or more compounds/agents
  • the individual compounds/agents may unitary or non-unitary formulations.
  • the unit dose(s) may be contained within a blister pack.
  • the pharmaceutical pack may optionally further comprise instructions for use.
  • the present invention contemplates all optical isomers, racemic forms and diastereoisomers of the compounds described herein.
  • the compounds may be produced in optically active and racemic forms. If a chiral centre or another form of isomeric centre is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereoisomers, are intended to be covered herein.
  • references to the compounds (e.g. iminosugars) of the present invention encompass the products as a mixture of diastereoisomers, as individual diastereoisomers, as a mixture of enantiomers as well as in the form of individual enantiomers. Therefore, the present invention contemplates all optical isomers and racemic forms thereof of the compounds of the invention, and unless indicated otherwise (e.g. by use of dash-wedge structural formulae) the compounds shown herein are intended to encompass all possible optical isomers of the compounds so depicted. In cases where the
  • bioisostere or simply isostere
  • replacement atoms or groups of atoms having similar steric and/or electronic features to those atoms which they replace.
  • substitution of a hydrogen atom or a hydroxyl group with a fluorine atom is a commonly employed bioisosteric replacement.
  • Sila-substitution C/Si-exchange
  • sila-substituted isosteres may exhibit improved pharmacological properties, and may for example be better tolerated, have a longer half-life or exhibit increased potency (see for example Englebienne (2005) Med. Chem., 1 (3): 215-226).
  • replacement of an atom by one of its isotopes, for example hydrogen by deuterium may also lead to improved pharmacological properties, for example leading to longer half-life (see for example Kushner et al (1999) Can J Physiol Pharmacol. 77(2):79-88).
  • the present invention contemplates all bioisosteres (and specifically, all silicon bioisosteres) of the compounds of the invention.
  • the pharmaceutically acceptable derivatives are therefore suitable for administration to or use in contact with mammalian tissues without undue toxicity, irritation or allergic response (i.e. commensurate with a reasonable benefit/risk ratio).
  • Preferred derivatives are those obtained (or obtainable) by alkylation, esterification or acylation of the parent compounds of the invention.
  • the derivatives may be active perse, or may be inactive until processed in vivo. In the latter case, the derivatives of the invention act as prodrugs.
  • Particularly preferred prodrugs are ester derivatives which are esterified at one or more of the free hydroxyls and which are activated by hydrolysis in vivo.
  • Other preferred prodrugs are covalently bonded
  • pharmaceutically acceptable salt as applied to the inhibitors of the invention defines any non-toxic organic or inorganic acid addition salt of the free base which are suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and which are commensurate with a reasonable benefit/risk ratio. Suitable pharmaceutically acceptable salts are well known in the art.
  • Examples are the salts with inorganic acids (for example hydrochloric, hydrobromic, sulphuric and phosphoric acids), organic carboxylic acids (for example acetic, propionic, glycolic, lactic, pyruvic, malonic, succinic, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, dihydroxymaleic, benzoic, phenylacetic, 4-aminobenzoic, 4- hydroxybenzoic, anthranilic, cinnamic, salicylic, 2-phenoxybenzoic, 2-acetoxybenzoic and mandelic acid) and organic sulfonic acids (for example methanesulfonic acid and p- toluenesulfonic acid).
  • organic carboxylic acids for example acetic, propionic, glycolic, lactic, pyruvic, malonic, succinic, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic
  • compositions containing the compounds of the invention defines compositions in which the compound of the invention is at least 90% pure, preferably at least 95% pure and most preferably at least 99% pure.
  • alkyl defines a straight or branched saturated hydrocarbon chain.
  • the term “CrC 6 alkyl” refers to a straight or branched saturated hydrocarbon chain having one to six carbon atoms. Examples include methyl, ethyl, n- propyl, isopropyl, t-butyl, n-hexyl.
  • C Cg alkyl refers to a straight or branched saturated hydrocarbon chain having one to nine carbon atoms.
  • C Ci 5 alkyl refers to a straight or branched saturated hydrocarbon chain having one to fifteen carbon atoms.
  • the alkyl groups of the invention may be optionally substituted by one or more halogen atoms.
  • alkyl may define the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloaikyl groups, alkyl substituted cycloaikyl groups and cycloaikyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C C 3 o for straight chain, C 3 -C 30 for branched chain), and more preferably up to 20, 15, 12, 10, 8 or 6.
  • preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure.
  • aralkyl defines an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
  • CrC alkyl has a similar meaning except that it contains from one to four carbon atoms.
  • C 2 -C 6 alkenyl refers to a straight or branched hydrocarbon chain having from two to six carbon atoms and containing at least one carbon-carbon double bond. Examples include ethenyl, 2-propenyl, and 3-hexenyl.
  • C C 6 haloalkyl refers to a C ⁇ e alkyl group as defined above substituted by one or more halogen atoms.
  • alkenyl defines a straight or branched hydrocarbon chain having containing at least one carbon-carbon double bond.
  • d-Ce alkenyl refers to a straight or branched unsaturated hydrocarbon chain having one to six carbon atoms.
  • Ci-C 9 alkenyl refers to a straight or branched unsaturated hydrocarbon chain having one to nine carbon atoms.
  • Ci-C 15 alkenyl refers to a straight or branched unsaturated hydrocarbon chain having one to fifteen carbon atoms.
  • Preferred is ( Ce alkenyl. Examples include ethenyl, 2-propenyl, and 3-hexenyl.
  • the alkenyl groups of the invention may be optionally substituted by one or more halogen atoms.
  • alkynyl defines a straight or branched hydrocarbon chain having containing at least one carbon-carbon triple bond.
  • C C 6 alkynyl refers to a straight or branched unsaturated hydrocarbon chain having one to six carbon atoms.
  • C1-C9 alkynyl refers to a straight or branched unsaturated hydrocarbon chain having one to nine carbon atoms.
  • C C 5 alkynyl refers to a straight or branched unsaturated hydrocarbon chain having one to fifteen carbon atoms.
  • Preferred is C C 6 alkynyl. Examples include ethynyl, 2-propynyl, and 3-hexynyl.
  • the alkynyl groups of the invention may be optionally substituted by one or more halogen atoms.
  • heterocyclyl defines a saturated or partially saturated 3 to 14 membered ring system (except when alternative numbers of ring atoms are specified) similar to cycloalkyl but in which at least one of the carbon atoms has been replaced by N, O, S, SO or S0 2 .
  • Examples include piperidine, piperazine, morpholine, tetrahydrofuran and pyrrolidine.
  • carbocyclyl means a mono- or poiycyclic residue containing 3 or more (e.g. 3-14, 3-10 or 3-8) carbon atoms.
  • the carbocyclyl residues of the invention may be optionally substituted by one or more halogen atoms.
  • Mono- and bicyclic carbocyclyl residues are preferred.
  • the carbocyclyl residues can be saturated or partially unsaturated and include fused bicyclic or tricyclic systems. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl and also bridged systems such as norbornyl and adamantyl.
  • cycloalkyls Saturated carbocyclyl residues are preferred and are referred to herein as "cycloalkyls" and the term “cycloalkyi” is used herein to define a saturated 3 to 14 membered carbocyclic ring including fused bicyclic or tricyclic systems. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and also bridged systems such as norbornyl and adamantyl.
  • the cycloalkyi residues of the invention may be optionally substituted by one or more halogen atoms.
  • aryl defines a 5-14 (e.g. 5-10) membered aromatic mono-, bi- or tricyclic group at least one ring of which is aromatic.
  • bicyclic aryl groups may contain only one aromatic ring.
  • aromatic moieties are benzene, naphthalene, imidazole and pyridine.
  • the term also includes bicyclic or tricyclic systems in which one or more of the rings has aromatic character. Indane is an example of this type of system.
  • heteroaryl are aryl moieties as defined above which contain heteroatoms (e.g. nitrogen, sulphur and/or oxygen).
  • heteroatoms e.g. nitrogen, sulphur and/or oxygen.
  • the term also includes systems in which a ring having aromatic character is fused to a saturated or partially saturated ring. Examples include pyridine, pyrimidine, furan, thiophene, indole, isoindole, indoline, benzofuran, benzimidazole, benzimidazoline quinoline, isoquinoline,
  • aryl is to be interpreted to include heteroaryl groups as defined above.
  • aryl and heteroaryl groups of the invention may optionally be substituted by one or more halogen atoms.
  • halo refers to fluoro, chloro, bromo or iodo.
  • N-alkyl derivates of the compounds of formula la and lb may be prepared by techniques known to those skilled in the art. Typical approaches involve: (a) reductive amination by hydrogenantion of the amine in the presence of palladium with an aldehyde; (b) reductive amination by sodium cyanoborohydride of the amine with an aldehyde; (c) reductive amination by sodium triacetoxyborohydride of the amine with an aldehyde; and (d) N- alkylation of the amine with an akyl halide or other leaving group such as a tosylate, etc. (all of the preceding in water, alcohols or other suitable solvents).
  • Preferred compounds of the invention are able to rescue mutant CFTR activity.
  • the ability of the compounds of the invention to rescue mutant CFTR activity may be determined by routine assays known to those skilled in the art (an example of which is described in the Exemplification section (Example 3), below).
  • Preferred are compounds which can rescue the activity of the mutant AF508 CFTR polypeptide. Without wishing to be bound by any theory, it is thought that the compounds of the invention act as CFTR pharmacoperones, restoring mutant CFTR function via a
  • preferred compounds of the invention are CFTR pharmacoperones. Such compounds find application in the treatment of CF.
  • the compounds may act as an indirect chaperone of CFTR via a chaperone effect attendant on binding to a protein (e.g. enzyme) which itself acts as a chaperone or co- chaperone of CFTR.
  • the compounds of the invention may bind to (or otherwise inhibit) chaperone proteins such as calnexin and so influence protein trafficking through the Golgi apparatus.
  • chaperone proteins such as calnexin and so influence protein trafficking through the Golgi apparatus.
  • compounds of the invention may prevent the interaction of mutant CFTR polypeptides (e.g. the AF508 CFTR polypeptide) to the chaperone calnexin.
  • preferred compounds of the invention inhibit the interaction of calnexin with CFTR polypeptide.
  • glycosidase inhibitors i.e. compounds which inhibit one or more glycosidase enzymes.
  • specific inhibitors of a- glucosidases i.e. compounds which inhibit one or more a-glucosidase enzymes but which are inactive against members of other classes of glycosidases.
  • the compounds of the present invention can be administered by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), nasal, rectal, vaginal and topical (including buccal and sublingual) administration. Preferred is oral administration.
  • the amount of the compound administered can vary widely according to the particular dosage unit employed, the period of treatment, the age and sex of the patient treated, the nature and extent of the disorder treated, and the particular compound selected.
  • the desired dose is preferably presented as a single dose for daily administration.
  • sub-doses administered at appropriate intervals throughout the day may also be employed. These sub-doses may be employed.
  • unit dosage forms for example, containing 0.001 to 100 mg, preferably 0.01 to 10 mg, and most preferably 0.5 to 1.0 mg of active ingredient per unit dosage form.
  • a number of factors are considered by the attending physician, including, but not limited to, the potency and duration of action of the inhibitors used, the nature and severity of the illness to be treated, as well as the sex, age, weight, general health and individual responsiveness of the patient to be treated, and other relevant circumstances.
  • dosages can also be determined with guidance from Goodman & Goldman's The Pharmacological Basis of Therapeutics, Ninth Edition (1996), Appendix II, pp. 1707-1711.
  • the effectiveness of a particular dosage of the compound of the invention can be determined by monitoring the effect of a given dosage on the progression of the disease or its prevention.
  • compositions of the invention may be delivered to the respiratory tract and lungs by inhalation. They may be delivered systemically by oral administration.
  • Illustrative pharmaceutically acceptable salts are prepared from formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, stearic, salicylic, p- hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic,
  • ethanesulfonic benzenesulfonic, pantothenic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, cyclohexylaminosulfonic, algenic, b-hydroxybutyric, galactaric and galacturonic acids.
  • Suitable pharmaceutically-acceptable base addition salts include metallic ion salts and organic ion salts.
  • Metallic ion salts include, but are not limited to, appropriate alkali metal (group la) salts, alkaline earth metal (group lla) salts and other physiologically acceptable metal ions.
  • Such salts can be made from the ions of aluminium, calcium, lithium, magnesium, potassium, sodium and zinc.
  • Organic salts can be made from tertiary amines and quaternary ammonium salts, including in part, trimethylamine, diethylamine, N, N - dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. All of the above salts can be prepared by those skilled in the art by conventional means from the corresponding compound.
  • compositions can include stabilizers, antioxidants, colorants and diluents.
  • Pharmaceutically acceptable carriers and additives are chosen such that side effects from the pharmaceutical compound are minimized and the performance of the compound is not compromised to such an extent that treatment is ineffective.
  • the compound of the invention can be administered parenterally, for example
  • sterile injectable aqueous or oleaginous suspensions Such suspensions can be formulated according to known art using suitable dispersing or wetting agents and suspending agents such as those mentioned above or other acceptable agents.
  • a sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example a solution in 1 ,3- butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono-or diglycerides.
  • omega-3 polyunsaturated fatty acids can find use in preparation of injectables. Administration can also be by inhalation, in the form of aerosols or solutions for nebulizers, or rectally, in the form of suppositories prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary
  • buccal and sub-lingual administration including administration in the form of lozenges, pastilles or a chewable gum comprising the inhibitors set forth herein.
  • the inhibitors can be deposited in a flavoured base, usually sucrose, and acacia or tragacanth. Preservatives are optionally employed to prevent microbial growth prior to or during use.
  • Suitable preservatives include polyquaternium-1 , benzalkonium chloride, thimerosal, chlorobutanol, methylparaben, propylparaben, phenylethyl alcohol, edetate disodium, sorbic acid, or other agents known to those skilled in the art. Typically, such preservatives are employed at a level of about 0.001% to about 1.0% by weight of a pharmaceutical composition.
  • Solubility of components of the present compositions can be enhanced by a surfactant or other appropriate cosolvent in the composition.
  • cosolvents include polysorbates 20,60 and 80, polyoxyethylene/polyoxypropylene surfactants (e. g., Pluronic F-68, F-84 and P-103), cyclodextrin, or other agents known to those skilled in the art.
  • cosolvents are employed at a level of about 0.01 % to about 2% by weight of a
  • compositions and carriers encompass all the foregoing and the like.
  • the above considerations concerning effective formulations and administration procedures are well known in the art and are described in standard textbooks. See for example Remington: The Science and Practice of Pharmacy, 20th Edition (Lippincott, Williams and Wilkins), 2000; Lieberman et al., ed. , Pharmaceutical Dosage Forms, Marcel Decker, New York, N. Y. (1980) and Kibbe et al., ed. , Handbook of Pharmaceutical Excipients (3rd Edition), American Pharmaceutical Association, Washington (1999).
  • compositions of the invention are preferably formulated for oral delivery in tablet form.
  • Formulations for delivery to the respiratory tract are preferably formulated for oral delivery in tablet form.
  • the compounds of the invention can be formulated into a solution and/or a suspension of particles in a carrier appropriate for inhalation into the respiratory tract and the lungs.
  • a carrier appropriate for inhalation into the respiratory tract and the lungs.
  • suitable carriers are known to those skilled in the art.
  • powders, mists or aerosols with particle sizes of 0.5 to 1 micron may be delivered to the respiratory tract.
  • particle size ranges are commonly achieved by micronisation or spray drying and such delivery methods are described for example in Remington: The Science and Practice of Pharmacy, 20th Edition (Lippincott, Williams and Wilkins), 2000; Lieberman et al., ed. , Pharmaceutical Dosage Forms, Marcel Decker, New York, N. Y. (1980) and Kibbe er a/., ed. , Handbook of Pharmaceutical Excipients (3rd Edition), American Pharmaceutical Association, Washington (1999).
  • the compound of the invention forms part of a powdered composition within a gelatin capsule, blister pack and a multi-dose metering device.
  • the capsule or blister is ruptured within the device enabling the powder to be inhaled.
  • Powdered compositions typically comprise the compounds of the invention blended or mixed with an inert carrier.
  • the inert carrier has a mean particle size substantially larger than that of the drug. This provides, among other advantages, an improvement in the flow properties and dispensing accuracy of the composition.
  • Suitable carriers include calcium carbonate and sugars.
  • the compound of the invention is formulated as an aerosol, for example by preparing a suspension of the compound as a finely divided powder in a liquefied propellant gas. Alternatively, a solution can be prepared which may contain solubilizers and co-solvents. Pressurized metered dose inhalers (pMDI) are normally used to dispense such formulations to a patient.
  • Suitable propellants include
  • chlorofluorocarbons fluorocarbons and hydrofluoroalkanes.
  • Inhalation devices such as inhalers (including dry powder inhaler and metered dose inhalers (MDIs)) and nebulizers (also known as atomizers) may be used to deliver the compounds of the invention to the respiratory tract and/or lungs.
  • Metered dose inhalers are designed to deliver a fixed unit dosage of medicament per actuation.
  • Exemplary nebulizers for delivering an aerosolized solution include the AERxTM (Aradigm), the Ultravent® (Mallinkrodt), the Pari LC PlusTM or the Pari LC StarTM (Pari GmbH, Germany), the DeVilbiss Pulmo-Aide, and the Acorn II® (Marquest Medical Products).
  • IsoLAB was synthesized from D-tagatose [Scheme 1].
  • the C2 hydroxymethyl group in the diacetonide 17 was introduced in D-tagatose by a Kiliani synthesis, followed by
  • Trifluoromethanesulfonic anhydride (1.9 mL, 1 1.3 mmol) was added dropwise to a stirred solution of 2,3:5,6-di-0-isopropylidene-2-C-hydroxymethyl-D-talono-1 ,4-lactone (2.07g, 7.19 mmol) and pyridine (1.75 mL, 21.7 mmol) in DCM (40 mL) at -30°C. After 2.5 h TLC analysis (1 :1 EtOAc/cyclohexane) indicated the conversion of starting material (R f 0.41 ) into one major product (R f 0.77).
  • isoLAB can also be prepared by routes from other sugars.
  • suitable starting monosaccharides include (but are not restricted to) L-ribose, D- lyxose, L-psicose, D-mannose, D-fructose and L-sorbose, D-mannose, L-ribonolactone, L- gulose, D-lyxonolactone, L-gulonolactone, D-mannonolactone, L-gulonolactone and suitably protected derivatives of any of the foregoing.
  • protection may be of cis-1 ,2-diols by acetone to acetonides, or any suitable ketone (such as cyclohexanone, pentan-3-one or other ketones) to its corresponding ketal.
  • the human tracheal gland serous epithelial cell line CF-KM4 is derived from a CF patient homozygous for the AF508 mutation. The details of the generation, characterization, and routine propagation have been described elsewhere (Kammouni et al. (1999) Resp. Cell Mol. Biol. 20(4): 684-91 ).
  • CFTR ion channel functions were assessed by single-cell fluorescence imaging, using the potential-sensitive probe bis-(1 ,3-diethylthiobarbituric acid)trimethine oxonol (DiSBAC 2 (3); Molecular Probes, Eugene, OR), as previously reported (see Norez et al. (2009) Am. J. Respir. Cell Mol. Biol. 41 (2): 217-225). Fluorescence intensity was recorded by confocal laser scanning microscopy using Bio-Rad MRC 1024 equipped with 15 mW Ar/Kr gas laser (Hemel Hempstead, UK). Maximal resolution was obtained with Olympus plan apo X60 oil, 1.4 NA, objective lens.
  • Fluorescence signal collection was performed through the control software Lasersharp 3.2 (Hemel Hempstead, UK). The resolution time was 30 s.
  • Bis- oxonol slowly distributes across biological membrane according to the membrane potential and binds to hydrophobic cell components; since the quantum yield of the dye increases impressively upon the binding, the fluorescence of cells incubated in a medium containing bis-oxonol increases upon depolarization and, conversely, decreases with
  • the CF-KM4 cells were treated 2 hours with 10 ⁇ of test compound and then CFTR proteins were stimulated by a cocktail of forskolin (Fsk) + genistein (Gst).
  • Fsk forskolin + genistein
  • Fig. 1 shows the functional evaluation of AF508 CFTR by DiSBAC2(3) assay in CF-K 4 cells treated (100 ⁇ for 2h) or not (negative control) with isoLAB.
  • ⁇ / ⁇ -DNJ 100 ⁇ for 2h served as a positive control.
  • a mixture of forskolin (Fsk, 10 ⁇ ) + genistein (Gst, 30 ⁇ ) is used to activate CFTR.
  • CFTRinh-172 (10 ⁇ ) is used to inhibit CFTR.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Otolaryngology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dispersion Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A combination comprising NΒ-DNJ and a compound of formula (I) wherein R1 is selected from H; linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl and araikyl and wherein the optional substitution may be with one or more groups independently selected from: -OH; -F; -CI; -Br; -I; -NH2; alkylamino; dialkylamino; linear or branched alkyl, alkenyl, alkynyl and araikyl; aryl; heteroaryl; linear or branched alkoxy; aryloxy; aralkoxy; -(alkylene)oxy(alkyl); -CN; -NO2; -COOH; -COO(alkyl); -COO(aryl); -C(0)NH(alkyl); -C(0)NH(aryl); sulfonyl; alkylsulfonyl; arylsulfonyl; sulfamoyl; alkylsulfamoyl; alkylthio; alkylsulfonamide; arylsulfonamide; -NHNH2; and -NHOH; or a bioisostere, pharmaceutically acceptable salt or derivative thereof, finds application in the treatment of cystic fibrosis.

Description

NOVEL IMINOSUGAR COMBINATIONS
Field of the Invention The present invention relates to combinations of N-butyldeoxynojirimycin (a.k.a. Miglustat, Λ/Β-DNJ and Zavesca®) with certain carbon-branched iminosugars, to compositions (including pharmaceutical compositions) containing them and to methods of treatment and prophylaxis using the compounds and compositions. Background to the Invention
Cystic fibrosis (referred to herein as "CF", and also known as mucoviscidosis) is a common hereditary disease which affects the entire body, causing progressive disability and often early death. Respiratory dysfunction is the most serious symptom and results from frequent lung infections. Most individuals die in their 20s and 30s from lung failure and lung transplantation is often necessary as CF worsens. A multitude of other symptoms, including sinus infections, poor growth, diarrhoea and infertility result from the effects of CF on other parts of the body. CF is an autosomal recessive disease caused by mutation of the cystic fibrosis
transmembrane conductance regulator (CFTR) gene. CFTR is a chloride ion channel important in creating sweat, digestive juices and mucus and cystic fibrosis occurs when the mutation leads to reduced ion channel activity (via increased clearance of the misfolded CFTR proteins). This leads to sodium hyperabsorption by the airways, profound lung inflammation and dysregulation of calcium homeostasis.
The most common mutation, comprising over 60% of all mutant alleles, results in the deletion of phenylalanine in the ATP binding cassette at position 508 of the CFTR protein. The resultant AF508 CFTR polypeptide is expressed as a large, misfolded nascent polypeptide which is prematurely destroyed via the ubiquitin pathway but which aggregates following defective processing in the translocation machinery.
Recently, it has been reported that several iminosugars, including N-butyldeoxynojirimycin (a.k.a. Miglustat, Λ/Β-DNJ and Zavesca®), bind to nascent mutant CFTR protein during cotranslational processing. This appears to restore CFTR function via a postulated pharmacological chaperone mechanism and/or glucosidase inhibition (see Zhang et al. (2003) J. Biol. Chem. 278: 51232-51242; Norez et al. (2006) FEBS Lett. 580: 2081-2086); Dechecchi et al. (2008) J. Cystic Fibrosis 7(6): 555-565; Antigny et al. (2008) Cell Calcium 43(2): 175-193; Norez er al. (2008) J. Pharm. Exp. Ther. 325(1 ): 89-99; Cox et al. (2007) Medicinal use of iminosugars, in Iminosugars From Synthesis to Therapeutic Applications: Compain, Philippe / Martin, Olivier R. (eds.) ISBN-13: 978-0-470-03391-3 - John Wiley & Sons, pages 3 8-319); Asano, N. (2009) Cell. Mol. Life Sci. 66: 1479-1492.
More recently, Lubamba er al. (2009) Am. J. Resp. Crit. Care 179 (11): 1022-1028 have shown that nasal delivery of Λ Β-DNJ, at picomolar doses, normalizes sodium and CFTR- dependent chloride transport in AF508 transgenic mice, while Norez et al. (2009) Am. J. Resp. Cell Mol. Biol. 41 (2): 217-225 have shown that a human respiratory epithelial cell line of the AF508 CFTR phenotype can acquire a non-CF-like phenotype when chronically treated with low concentrations of Λ/Β-DNJ.
WO2005/046672 describes the use of various DNJ derivatives having glucosidase inhibitory activity in the treatment of CF.
WO2007/123403 describes the use of various iminosugars (and in particular DNJ derivatives) having non-lysosomal glucosylceramidase (glucosidase beta (bile acid) 2) inhibitory activity in the treatment of CF.
In October 2007, Actelion Pharmaceuticals Limited initiated a Phase lla proof-of-concept clinical trial with Λ/Β-DNJ for CF involving 25 patients affected by the AF508 CFTR mutation.
Notwithstanding the promising findings and clinical research summarized above, there is at present no cure for CF. Current treatments are merely palliative: they include antibiotic treatment (for lung infections), chest physiotherapy/mechanical expecoration (for mucus accumulation), surgery and mechanical ventilation. There is therefore a need for improved treatments for CF which at least partially reverse the underlying proteostatic dysfunction.
Summary of the Invention The present inventors have now unexpectedly discovered that the carbon-branched pyrrolidine iminosugar (3S, 4S)-3-(hydroxymethyl)pyrrolidine-3,4-diol (1 ,4-dideoxy-2- hydroxymethyl-1 ,4-imino-L-threitol, referred to herein as isoLAB) can be used in comination with Λ/Β-DNJ (and various derivatives thereof) in the treatment of cystic fibrosis.
Without wishing to be bound by any theory, it is thought that the strikingly different glycosidase inhibitory profile of isoLAB as compared with M3-DNJ (no inhibitory activity of the former has been detected against a range of different glycosidases (see infra), whereas /VB-DNJ exhibits potent inhibition of a wide range of a-glucosidases, including those involved in digestive and ER processing functions) may produce an improved
protective/therapeutic index when used in combination with Λ/Β-DNJ compared with Λ/Β- DNJ or isoLAB alone.
Thus, combinations of Λ/Β-DNJ and isoLAB (and its bioisosteres, pharmaceutically acceptable salts and pharmaceutically acceptable derivatives) therefore find application in improved treatments for CF, as described herein.
Therefore, in a first aspect of the present invention, there is provided a combination comprising Λ/Β-DNJ and a compound of formula (I):
Figure imgf000004_0001
wherein:
R is selected from H; linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl and aralkyl and wherein the optional substitution may be with one or more groups independently selected from: -OH; -F; -CI; -Br; -I; -NH2; alkylamino;
dialkylamino; linear or branched alkyl, alkenyl, alkynyl and aralkyl; aryl; heteroaryl; linear or branched alkoxy; aryloxy; aralkoxy; -(alkylene)oxy(alkyl); -CN; -N02; - COOH; -COO(alkyl); -COO(aryl); -C(0)NH(alkyl); -C(0)NH(aryl); sulfonyl; alkylsulfonyl; arylsulfonyl; sulfamoyl; alkylsulfamoyl; alkylthio; alkylsulfonamide; arylsulfonamide; -NHNH2; and -NHOH; or a bioisostere, pharmaceutically acceptable salt or derivative thereof.
In preferred embodiments, R1 is selected from H, C,.18 alkyl (for example, C1-9 alkyl, e.g. C,. 6 alkyl), C2.i8 alkenyl (for example, C2.9 alkenyl, e.g. C2-6 alkenyl) and C2.i8 alkynyl (for example, C2-9 alkynyl, e.g. C2.6 alkynyl). For example, R1 may be -H and R1 selected from C1.18 alkyl (for example,
Figure imgf000005_0001
alkyl, e.g. Ci-6 alkyl), C2.18 alkenyl (for example, C2.9 alkenyl, e.g. C2-6 alkenyl) and C2.18 alkynyl (for example, C2.9 alkynyl, e.g. C2.6 alkynyl).
In preferred embodiments, R1 represents H; C1-15 alkyl, C1-15 alkenyl or C1 -15 alkynyl, optionally substituted with one or more R2; oxygen or an oxygen containing group such that the compound is an N-oxide; C(0)OR3; C(0)NR3R4; S02NR3; OH, OR3, or formyl.
In other embodiments, R1 may represent C1-9 alkyl, optionally substituted with up to 6 OH, NR3R4, aryl, 0-C1-3 alkyl, 0-C1-3 alkenyl, C02H, NH(NH)NH2, CONR3R4; C(0)OR3;
C(0)NR3R4; or S02NR3. Preferably, the compound of formula I is (3S, 4S)-3-(hydroxymethyl)pyrrolidine-3,4-diol (isoLAB).
Other aspects of the invention are as defined in the claims attached hereto. Detailed Description of the Invention
Definitions and general preferences
Where used herein and unless specifically indicated otherwise, the following terms are intended to have the following meanings in addition to any broader (or narrower) meanings the terms might enjoy in the art:
Unless otherwise required by context, the use herein of the singular is to be read to include the plural and vice versa. The term "a" or "an" used in relation to an entity is to be read to refer to one or more of that entity. As such, the terms "a" (or "an"), "one or more," and "at least one" are used interchangeably herein.
As used herein, the term "comprise," or variations thereof such as "comprises" or
"comprising," are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, element, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers. Thus, as used herein the term "comprising" is inclusive or open-ended and does not exclude additional, unrecited integers or method/process steps.
As used herein, the term "treatment" or "treating" refers to an intervention (e.g. the administration of an agent to a subject) which cures, ameliorates or lessens the symptoms of a disease or removes (or lessens the impact of) its cause(s). In this case, the term is used synonymously with the term "therapy".
Additionally, the terms "treatment" or "treating" refers to an intervention (e.g. the administration of an agent to a subject) which prevents or delays the onset or progression of a disease or reduces (or eradicates) its incidence within a treated population. In this case, the term treatment is used synonymously with the term "prophylaxis".
The term "subject" (which is to be read to include "individual", "animal", "patient" or "mammal" where context permits) defines any subject, particularly a mammalian subject, for whom treatment is indicated. Mammalian subjects include, but are not limited to, humans, primates, domestic animals, farm animals, pet animals and rodents such as mice, rats, hamsters and guinea pigs. In preferred embodiments, the subject is a human.
The term pharmacoperone is a term of art (from "pharmacological chaperone") used to define a class of biologically active small molecules (sometimes also referred to in the art as "chemical chaperones") that serve as molecular scaffolds, causing otherwise misfolded mutant proteins to fold and route correctly within the cell.
As used herein, an effective amount of a compound or composition defines an amount that can be administered to a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, but one that is sufficient to provide the desired effect, e.g. the treatment or prophylaxis manifested by a permanent or temporary improvement in the subject's condition. The amount will vary from subject to subject, depending on the age and general condition of the individual, mode of administration and other factors. Thus, while it is not possible to specify an exact effective amount, those skilled in the art will be able to determine an appropriate "effective" amount in any individual case using routine experimentation and background general knowledge. A therapeutic result in this context includes eradication or lessening of symptoms, reduced pain or discomfort, prolonged survival, improved mobility and other markers of clinical improvement. A therapeutic result need not be a complete cure.
As used herein, the term /VB-DNJ is used sensu stricto to cover N-butyldeoxynojirimycin (a.k.a. iglustat, /VB-DNJ and Zavesca®), as well as its pharmaceutically acceptable salts and derivatives. However, the term is also used sensu lato to cover various derivatives and analogues of deoxynojirimycin (DNJ) which can restore CFTR function, including for example those described in WO2005/046672 and WO2007/123403 (the disclosure of which relating to the structure of the DNJ derivatives is hereby incorporated by reference). Such DNJ derivatives may have glucosidase inhibitory activity.
As used herein, the term "combination", as applied to two or more compounds and/or agents (also referred to herein as the components), is intended to define material in which the two or more compounds/agents are associated. The terms "combined" and
"combining" in this context are to be interpreted accordingly.
The association of the two or more compounds/agents in a combination may be physical or non-physical. Examples of physically associated combined compounds/agents include:
• compositions (e.g. unitary formulations) comprising the two or more
compounds/agents in admixture (for example within the same unit dose);
• compositions comprising material in which the two or more compounds/agents are chemically/physicochemically linked (for example by crosslinking, molecular agglomeration or binding to a common vehicle moiety);
• compositions comprising material in which the two or more compounds/agents are chemically/physicochemically co-packaged (for example, disposed on or within lipid vesicles, particles (e.g. micro- or nanoparticles) or emulsion droplets); • pharmaceutical kits, pharmaceutical packs or patient packs in which the two or more compounds/agents are co-packaged or co-presented (e.g. as part of an array of unit doses); Examples of non-physically associated combined compounds/agents include:
• material (e.g. a non-unitary formulation) comprising at least one of the two or more compounds/agents together with instructions for the extemporaneous association of the at least one compound/agent to form a physical association of the two or more compounds/agents;
• material (e.g. a non-unitary formulation) comprising at least one of the two or more compounds/agents together with instructions for combination therapy with the two or more compounds/agents;
• material comprising at least one of the two or more compounds/agents together with instructions for administration to a patient population in which the other(s) of the two or more compounds/agents have been (or are being) administered;
• material comprising at least one of the two or more compounds/agents in an
amount or in a form which is specifically adapted for use in combination with the other(s) of the two or more compounds/agents.
As used herein, the term "combination therapy" is intended to define therapies which comprise the use of a combination of two or more compounds/agents (as defined above). Thus, references to "combination therapy", "combinations" and the use of
compounds/agents "in combination" in this application may refer to compounds/agents that are administered as part of the same overall treatment regimen. As such, the posology of each of the two or more compounds/agents may differ: each may be administered at the same time or at different times. It will therefore be appreciated that the compounds/agents of the combination may be administered sequentially (e.g. before or after) or
simultaneously, either in the same pharmaceutical formulation (i.e. together), or in different pharmaceutical formulations (i.e. separately). Simultaneously in the same formulation is as a unitary formulation whereas simultaneously in different pharmaceutical formulations is non-unitary. The posologies of each of the two or more compounds/agents in a combination therapy may also differ with respect to the route of administration. As used herein, the term "pharmaceutical kit" defines an array of one or more unit doses of a pharmaceutical composition together with dosing means (e.g. measuring device) and/or delivery means (e.g. inhaler or syringe), optionally all contained within common outer packaging. In pharmaceutical kits comprising a combination of two or more
compounds/agents, the individual compounds/agents may unitary or non-unitary formulations. The unit dose(s) may be contained within a blister pack. The pharmaceutical kit may optionally further comprise instructions for use.
As used herein, the term "pharmaceutical pack" defines an array of one or more unit doses of a pharmaceutical composition, optionally contained within common outer packaging. In pharmaceutical packs comprising a combination of two or more compounds/agents, the individual compounds/agents may unitary or non-unitary formulations. The unit dose(s) may be contained within a blister pack. The pharmaceutical pack may optionally further comprise instructions for use.
In its broadest aspect, the present invention contemplates all optical isomers, racemic forms and diastereoisomers of the compounds described herein. Those skilled in the art will appreciate that, owing to the asymmetrically substituted carbon atoms present in the compounds of the invention, the compounds may be produced in optically active and racemic forms. If a chiral centre or another form of isomeric centre is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereoisomers, are intended to be covered herein. Compounds of the invention containing a chiral centre (or multiple chiral centres) may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone. Thus, references to the compounds (e.g. iminosugars) of the present invention encompass the products as a mixture of diastereoisomers, as individual diastereoisomers, as a mixture of enantiomers as well as in the form of individual enantiomers. Therefore, the present invention contemplates all optical isomers and racemic forms thereof of the compounds of the invention, and unless indicated otherwise (e.g. by use of dash-wedge structural formulae) the compounds shown herein are intended to encompass all possible optical isomers of the compounds so depicted. In cases where the
stereochemical form of the compound is important for pharmaceutical utility, the invention contemplates use of an isolated eutomer. The term bioisostere (or simply isostere) is a term of art used to define drug analogues in which one or more atoms (or groups of atoms) have been substituted with replacement atoms (or groups of atoms) having similar steric and/or electronic features to those atoms which they replace. The substitution of a hydrogen atom or a hydroxyl group with a fluorine atom is a commonly employed bioisosteric replacement. Sila-substitution (C/Si-exchange) is a relatively recent technique for producing isosteres. This approach involves the replacement of one or more specific carbon atoms in a compound with silicon (for a review, see Tacke and Zilch (1986) Endeavour, New Series 10: 191-197). The sila-substituted isosteres (silicon isosteres) may exhibit improved pharmacological properties, and may for example be better tolerated, have a longer half-life or exhibit increased potency (see for example Englebienne (2005) Med. Chem., 1 (3): 215-226). Similarly, replacement of an atom by one of its isotopes, for example hydrogen by deuterium, may also lead to improved pharmacological properties, for example leading to longer half-life (see for example Kushner et al (1999) Can J Physiol Pharmacol. 77(2):79-88). In its broadest aspect, the present invention contemplates all bioisosteres (and specifically, all silicon bioisosteres) of the compounds of the invention.
The term pharmaceutically acceptable derivative as applied to the compounds of the invention define compounds which are obtained (or obtainable) by chemical derivatization of the parent compounds of the invention. The pharmaceutically acceptable derivatives are therefore suitable for administration to or use in contact with mammalian tissues without undue toxicity, irritation or allergic response (i.e. commensurate with a reasonable benefit/risk ratio). Preferred derivatives are those obtained (or obtainable) by alkylation, esterification or acylation of the parent compounds of the invention. The derivatives may be active perse, or may be inactive until processed in vivo. In the latter case, the derivatives of the invention act as prodrugs. Particularly preferred prodrugs are ester derivatives which are esterified at one or more of the free hydroxyls and which are activated by hydrolysis in vivo. Other preferred prodrugs are covalently bonded
compounds which release the active parent drug according to general formula (I) after cleavage of the covalent bond(s) in vivo.
The term pharmaceutically acceptable salt as applied to the inhibitors of the invention defines any non-toxic organic or inorganic acid addition salt of the free base which are suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and which are commensurate with a reasonable benefit/risk ratio. Suitable pharmaceutically acceptable salts are well known in the art. Examples are the salts with inorganic acids (for example hydrochloric, hydrobromic, sulphuric and phosphoric acids), organic carboxylic acids (for example acetic, propionic, glycolic, lactic, pyruvic, malonic, succinic, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, dihydroxymaleic, benzoic, phenylacetic, 4-aminobenzoic, 4- hydroxybenzoic, anthranilic, cinnamic, salicylic, 2-phenoxybenzoic, 2-acetoxybenzoic and mandelic acid) and organic sulfonic acids (for example methanesulfonic acid and p- toluenesulfonic acid).
The term substantially pure as applied to compositions containing the compounds of the invention defines compositions in which the compound of the invention is at least 90% pure, preferably at least 95% pure and most preferably at least 99% pure. In the present specification the term "alkyl" defines a straight or branched saturated hydrocarbon chain. The term "CrC6 alkyl" refers to a straight or branched saturated hydrocarbon chain having one to six carbon atoms. Examples include methyl, ethyl, n- propyl, isopropyl, t-butyl, n-hexyl. The term "C Cg alkyl" refers to a straight or branched saturated hydrocarbon chain having one to nine carbon atoms. The term "C Ci5 alkyl" refers to a straight or branched saturated hydrocarbon chain having one to fifteen carbon atoms. The alkyl groups of the invention may be optionally substituted by one or more halogen atoms. Thus, the term alkyl may define the radical of saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloaikyl groups, alkyl substituted cycloaikyl groups and cycloaikyl substituted alkyl groups. In preferred embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C C3o for straight chain, C3-C30 for branched chain), and more preferably up to 20, 15, 12, 10, 8 or 6. Likewise, preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 5, 6 or 7 carbons in the ring structure. The term aralkyl defines an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
"CrC alkyl" has a similar meaning except that it contains from one to four carbon atoms. "C2-C6 alkenyl" refers to a straight or branched hydrocarbon chain having from two to six carbon atoms and containing at least one carbon-carbon double bond. Examples include ethenyl, 2-propenyl, and 3-hexenyl.. The term "C C6 haloalkyl" refers to a C^e alkyl group as defined above substituted by one or more halogen atoms.
In the present specification the term "alkenyl" defines a straight or branched hydrocarbon chain having containing at least one carbon-carbon double bond. The term "d-Ce alkenyl" refers to a straight or branched unsaturated hydrocarbon chain having one to six carbon atoms. The term "Ci-C9 alkenyl" refers to a straight or branched unsaturated hydrocarbon chain having one to nine carbon atoms. The term "Ci-C15 alkenyl" refers to a straight or branched unsaturated hydrocarbon chain having one to fifteen carbon atoms. Preferred is ( Ce alkenyl. Examples include ethenyl, 2-propenyl, and 3-hexenyl. The alkenyl groups of the invention may be optionally substituted by one or more halogen atoms.
In the present specification the term "alkynyl" defines a straight or branched hydrocarbon chain having containing at least one carbon-carbon triple bond. The term "C C6 alkynyl" refers to a straight or branched unsaturated hydrocarbon chain having one to six carbon atoms. The term "C1-C9 alkynyl" refers to a straight or branched unsaturated hydrocarbon chain having one to nine carbon atoms. The term "C C 5 alkynyl" refers to a straight or branched unsaturated hydrocarbon chain having one to fifteen carbon atoms. Preferred is C C6 alkynyl. Examples include ethynyl, 2-propynyl, and 3-hexynyl. The alkynyl groups of the invention may be optionally substituted by one or more halogen atoms.
The term "heterocyclyl" defines a saturated or partially saturated 3 to 14 membered ring system (except when alternative numbers of ring atoms are specified) similar to cycloalkyl but in which at least one of the carbon atoms has been replaced by N, O, S, SO or S02. Examples include piperidine, piperazine, morpholine, tetrahydrofuran and pyrrolidine.
As used herein, the term "carbocyclyl" means a mono- or poiycyclic residue containing 3 or more (e.g. 3-14, 3-10 or 3-8) carbon atoms. The carbocyclyl residues of the invention may be optionally substituted by one or more halogen atoms. Mono- and bicyclic carbocyclyl residues are preferred. The carbocyclyl residues can be saturated or partially unsaturated and include fused bicyclic or tricyclic systems. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl and also bridged systems such as norbornyl and adamantyl.
Saturated carbocyclyl residues are preferred and are referred to herein as "cycloalkyls" and the term "cycloalkyi" is used herein to define a saturated 3 to 14 membered carbocyclic ring including fused bicyclic or tricyclic systems. Examples of such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and also bridged systems such as norbornyl and adamantyl. The cycloalkyi residues of the invention may be optionally substituted by one or more halogen atoms.
In the present specification the term "aryl" defines a 5-14 (e.g. 5-10) membered aromatic mono-, bi- or tricyclic group at least one ring of which is aromatic. Thus, bicyclic aryl groups may contain only one aromatic ring. Examples of aromatic moieties are benzene, naphthalene, imidazole and pyridine. The term also includes bicyclic or tricyclic systems in which one or more of the rings has aromatic character. Indane is an example of this type of system.
As used herein, the term "heteroaryl" are aryl moieties as defined above which contain heteroatoms (e.g. nitrogen, sulphur and/or oxygen). The term also includes systems in which a ring having aromatic character is fused to a saturated or partially saturated ring. Examples include pyridine, pyrimidine, furan, thiophene, indole, isoindole, indoline, benzofuran, benzimidazole, benzimidazoline quinoline, isoquinoline,
tetrahydroisoquinoline, quinazoline, thiazole, benzthiazole, benzoxazole, indazole and imidazole ring systems. Unless otherwise indicated, the term "aryl" is to be interpreted to include heteroaryl groups as defined above.
The aryl and heteroaryl groups of the invention may optionally be substituted by one or more halogen atoms. In the present specification, "halo" refers to fluoro, chloro, bromo or iodo.
N-alkylation
N-alkyl derivates of the compounds of formula la and lb may be prepared by techniques known to those skilled in the art. Typical approaches involve: (a) reductive amination by hydrogenantion of the amine in the presence of palladium with an aldehyde; (b) reductive amination by sodium cyanoborohydride of the amine with an aldehyde; (c) reductive amination by sodium triacetoxyborohydride of the amine with an aldehyde; and (d) N- alkylation of the amine with an akyl halide or other leaving group such as a tosylate, etc. (all of the preceding in water, alcohols or other suitable solvents).
Exemplary techniques and reaction schemes which may be adapted for use in the synthesis of compounds of formula (I) are described for example in: (a) Rawlings et al. (2009) Chembiochem 10: 1 101 -1 105; (b) Mellor et al. (2002) Biochem. J. 366: 225-233; (c) WO2001/010429; and (d) Butters et al. (2000) Tetrahedron Asymm. 1 1 : 1 13-125.
CFTR protein rescue
Preferred compounds of the invention are able to rescue mutant CFTR activity. The ability of the compounds of the invention to rescue mutant CFTR activity may be determined by routine assays known to those skilled in the art (an example of which is described in the Exemplification section (Example 3), below). Preferred are compounds which can rescue the activity of the mutant AF508 CFTR polypeptide. Without wishing to be bound by any theory, it is thought that the compounds of the invention act as CFTR pharmacoperones, restoring mutant CFTR function via a
pharmacological chaperone mechanism.
Thus, preferred compounds of the invention are CFTR pharmacoperones. Such compounds find application in the treatment of CF.
Some or all of the observed CFTR rescue activity may arise from other mechanisms. For example, the compounds may act as an indirect chaperone of CFTR via a chaperone effect attendant on binding to a protein (e.g. enzyme) which itself acts as a chaperone or co- chaperone of CFTR. For example, the compounds of the invention may bind to (or otherwise inhibit) chaperone proteins such as calnexin and so influence protein trafficking through the Golgi apparatus. Thus, compounds of the invention may prevent the interaction of mutant CFTR polypeptides (e.g. the AF508 CFTR polypeptide) to the chaperone calnexin. Thus, preferred compounds of the invention inhibit the interaction of calnexin with CFTR polypeptide.
Glvcosidase inhibition
The ability to inhibit various glycosidases underpins the therapeutic utility of a number of known iminosugar therapeutics (see for example Cox ef a/. (2007) Medicinal use of iminosugars, in Iminosugars From Synthesis to Therapeutic Applications: Co pain, Philippe / Martin, Olivier R. (eds.) ISBN-13: 978-0-470-03391-3 - John Wiley & Sons, pages 318-319); Asano, N. (2009) Cell. Mol. Life Sci. 66: 1479-1492).
Thus, preferred compounds of the invention are glycosidase inhibitors (i.e. compounds which inhibit one or more glycosidase enzymes). Preferred are specific inhibitors of a- glucosidases (i.e. compounds which inhibit one or more a-glucosidase enzymes but which are inactive against members of other classes of glycosidases).
Posology
The compounds of the present invention can be administered by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), nasal, rectal, vaginal and topical (including buccal and sublingual) administration. Preferred is oral administration.
The amount of the compound administered can vary widely according to the particular dosage unit employed, the period of treatment, the age and sex of the patient treated, the nature and extent of the disorder treated, and the particular compound selected.
The desired dose is preferably presented as a single dose for daily administration.
However, two, three, four, five or six or more sub-doses administered at appropriate intervals throughout the day may also be employed. These sub-doses may be
administered in unit dosage forms, for example, containing 0.001 to 100 mg, preferably 0.01 to 10 mg, and most preferably 0.5 to 1.0 mg of active ingredient per unit dosage form.
In determining an effective amount or dose, a number of factors are considered by the attending physician, including, but not limited to, the potency and duration of action of the inhibitors used, the nature and severity of the illness to be treated, as well as the sex, age, weight, general health and individual responsiveness of the patient to be treated, and other relevant circumstances. Those skilled in the art will appreciate that dosages can also be determined with guidance from Goodman & Goldman's The Pharmacological Basis of Therapeutics, Ninth Edition (1996), Appendix II, pp. 1707-1711.
The effectiveness of a particular dosage of the compound of the invention can be determined by monitoring the effect of a given dosage on the progression of the disease or its prevention.
Compositions of the invention may be delivered to the respiratory tract and lungs by inhalation. They may be delivered systemically by oral administration.
Formulation
Illustrative pharmaceutically acceptable salts are prepared from formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, stearic, salicylic, p- hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic,
ethanesulfonic, benzenesulfonic, pantothenic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, cyclohexylaminosulfonic, algenic, b-hydroxybutyric, galactaric and galacturonic acids.
Suitable pharmaceutically-acceptable base addition salts include metallic ion salts and organic ion salts. Metallic ion salts include, but are not limited to, appropriate alkali metal (group la) salts, alkaline earth metal (group lla) salts and other physiologically acceptable metal ions. Such salts can be made from the ions of aluminium, calcium, lithium, magnesium, potassium, sodium and zinc. Organic salts can be made from tertiary amines and quaternary ammonium salts, including in part, trimethylamine, diethylamine, N, N - dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. All of the above salts can be prepared by those skilled in the art by conventional means from the corresponding compound.
Pharmaceutical compositions can include stabilizers, antioxidants, colorants and diluents. Pharmaceutically acceptable carriers and additives are chosen such that side effects from the pharmaceutical compound are minimized and the performance of the compound is not compromised to such an extent that treatment is ineffective.
The compound of the invention can be administered parenterally, for example
subcutaneously, intravenously, or intramuscularly, or by infusion techniques, in the form of sterile injectable aqueous or oleaginous suspensions. Such suspensions can be formulated according to known art using suitable dispersing or wetting agents and suspending agents such as those mentioned above or other acceptable agents. A sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example a solution in 1 ,3- butanediol. Among acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed, including synthetic mono-or diglycerides. In addition, omega-3 polyunsaturated fatty acids can find use in preparation of injectables. Administration can also be by inhalation, in the form of aerosols or solutions for nebulizers, or rectally, in the form of suppositories prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary
temperature, but liquid at rectal temperature and will therefore, melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols. Also encompassed by the present invention is buccal and sub-lingual administration, including administration in the form of lozenges, pastilles or a chewable gum comprising the inhibitors set forth herein. The inhibitors can be deposited in a flavoured base, usually sucrose, and acacia or tragacanth. Preservatives are optionally employed to prevent microbial growth prior to or during use. Suitable preservatives include polyquaternium-1 , benzalkonium chloride, thimerosal, chlorobutanol, methylparaben, propylparaben, phenylethyl alcohol, edetate disodium, sorbic acid, or other agents known to those skilled in the art. Typically, such preservatives are employed at a level of about 0.001% to about 1.0% by weight of a pharmaceutical composition.
Solubility of components of the present compositions can be enhanced by a surfactant or other appropriate cosolvent in the composition. Such cosolvents include polysorbates 20,60 and 80, polyoxyethylene/polyoxypropylene surfactants (e. g., Pluronic F-68, F-84 and P-103), cyclodextrin, or other agents known to those skilled in the art. Typically, such cosolvents are employed at a level of about 0.01 % to about 2% by weight of a
pharmaceutical composition.
Pharmaceutically acceptable excipients and carriers encompass all the foregoing and the like. The above considerations concerning effective formulations and administration procedures are well known in the art and are described in standard textbooks. See for example Remington: The Science and Practice of Pharmacy, 20th Edition (Lippincott, Williams and Wilkins), 2000; Lieberman et al., ed. , Pharmaceutical Dosage Forms, Marcel Decker, New York, N. Y. (1980) and Kibbe et al., ed. , Handbook of Pharmaceutical Excipients (3rd Edition), American Pharmaceutical Association, Washington (1999).
The compounds and compositions of the invention are preferably formulated for oral delivery in tablet form. Formulations for delivery to the respiratory tract
The compounds of the invention can be formulated into a solution and/or a suspension of particles in a carrier appropriate for inhalation into the respiratory tract and the lungs. A wide range of suitable carriers are known to those skilled in the art. In general, powders, mists or aerosols with particle sizes of 0.5 to 1 micron may be delivered to the respiratory tract. Such particle size ranges are commonly achieved by micronisation or spray drying and such delivery methods are described for example in Remington: The Science and Practice of Pharmacy, 20th Edition (Lippincott, Williams and Wilkins), 2000; Lieberman et al., ed. , Pharmaceutical Dosage Forms, Marcel Decker, New York, N. Y. (1980) and Kibbe er a/., ed. , Handbook of Pharmaceutical Excipients (3rd Edition), American Pharmaceutical Association, Washington (1999).
In a preferred formulation for inhalation, the compound of the invention forms part of a powdered composition within a gelatin capsule, blister pack and a multi-dose metering device. The capsule or blister is ruptured within the device enabling the powder to be inhaled.
Powdered compositions typically comprise the compounds of the invention blended or mixed with an inert carrier. Usually the inert carrier has a mean particle size substantially larger than that of the drug. This provides, among other advantages, an improvement in the flow properties and dispensing accuracy of the composition. Suitable carriers include calcium carbonate and sugars. In other embodiments, the compound of the invention is formulated as an aerosol, for example by preparing a suspension of the compound as a finely divided powder in a liquefied propellant gas. Alternatively, a solution can be prepared which may contain solubilizers and co-solvents. Pressurized metered dose inhalers (pMDI) are normally used to dispense such formulations to a patient. Suitable propellants include
chlorofluorocarbons, fluorocarbons and hydrofluoroalkanes.
Inhalation devices, such as inhalers (including dry powder inhaler and metered dose inhalers (MDIs)) and nebulizers (also known as atomizers) may be used to deliver the compounds of the invention to the respiratory tract and/or lungs. Metered dose inhalers are designed to deliver a fixed unit dosage of medicament per actuation. Exemplary nebulizers for delivering an aerosolized solution include the AERx™ (Aradigm), the Ultravent® (Mallinkrodt), the Pari LC Plus™ or the Pari LC Star™ (Pari GmbH, Germany), the DeVilbiss Pulmo-Aide, and the Acorn II® (Marquest Medical Products). Exemplification
The invention will now be described with reference to specific Examples. These are merely exemplary and for illustrative purposes only: they are not intended to be limiting in any way to the scope of the monopoly claimed or to the invention described. These examples constitute the best mode currently contemplated for practicing the invention.
(a) Example 1 : Preparation of isoLAB
Reaction scheme summary
Figure imgf000019_0001
Scheme 1
IsoLAB was synthesized from D-tagatose [Scheme 1]. The C2 hydroxymethyl group in the diacetonide 17 was introduced in D-tagatose by a Kiliani synthesis, followed by
acetonation. The diacetonide 17 was converted into the corresponding triflate on reaction with triflic anhydride in dichloromethane in the presence of pyridine; subsequent reaction of the triflate with sodium azide in DMF gave the D-fa/ono-azidolactone 18 [mp. 81-83 °C;
[cc]D 25 -82.9 (c 1.0)] in 94% yield. Two-step reduction of the lactone 18 by DIBALH in dichloromethane, followed by sodium borohydride in methanol, afforded the diol 19 [oil;
[oc]D 25 -52.2 (c 1.0)] in 90% yield. Selective hydrolysis of the terminal acetonide in 19 formed the tetraol 20; [a]D 25 -98.1 (c 1.0, CHCI3)] (83% yield) which on oxidation with sodium periodate cleaved the C4-C5 and C5-C6 bonds to give the isopropylidene 3-C-azidomethyl- D-erythrose 15D [oil, [a]D 25 -102.1 (c 1.0)] in 91 % yield. Hydrolysis of the acetonide 15D with acid ion exchange resin afforded the deprotected azidolactol 16D [oil; [a]D 25 +1.7 (c 1.3, MeOH)] which on hydrogenation gave isoLAB 1 L [oil, [a]D 25 +35.6 (c 0.27, H20)] in 71 % yield. The overall yield of isoLAB 1 L from the ia/ono-diacetonide 17 was 45%.
2-C-Azidomethyl-2,3:5,6-di-0-isopropylidene-D-talono-1 ,4-lactone
Trifluoromethanesulfonic anhydride (1.9 mL, 1 1.3 mmol) was added dropwise to a stirred solution of 2,3:5,6-di-0-isopropylidene-2-C-hydroxymethyl-D-talono-1 ,4-lactone (2.07g, 7.19 mmol) and pyridine (1.75 mL, 21.7 mmol) in DCM (40 mL) at -30°C. After 2.5 h TLC analysis (1 :1 EtOAc/cyclohexane) indicated the conversion of starting material (Rf 0.41 ) into one major product (Rf 0.77). The reaction mixture was diluted with DCM (70 mL) and washed with aqueous hydrochloric acid (1 M, 60 mL). The organic residue was dried (MgS04), filtered and concentrated in vacuo to afford the triflate, which was used without further purification. Sodium azide (701 mg, 10.78 mmol) was added to a stirred solution of the crude triflate in dry DMF (35 mL) at RT. The reaction mixture was stirred for 3 h after which TLC analysis (1 : 1 EtOAc/cyclohexane) showed the conversion of starting material to one major product (Rf 0.73). The reaction mixture was concentrated in vacuo and the residue was partitioned between EtOAc (90 mL) and brine (50 mL). The aqueous layer was extracted with EtOAc (40 mL) and the combined organic layers were dried (MgS04), filtered and concentrated in vacuo. The resulting residue was purified by flash column chromatography (1 :6 - 1 :2 EtOAc/cyclohexane) to afford the azide (2.12 g, 94% over 2 steps). HRMS (ESI +ve): found 336.1 162 ( +Na)+; Ci3Hi9N3Na06 requires 336.1166; [a]D 25 -82.9 (c 1.0, CHCI3); m.p. 81-83°C; vmax (thin film): 2107 (s, N3), 1786 (s, C=0); δΗ (CDCI3, 400 MHz): 1.32, 1.36, 1.47, 1.54 (4 x 3H, s, Me), 3.57 (1 H, d, H2'a, Jgem 13.3), 3.86 (1 H, d, H2'b, Jgem 13.3), 3.97 (1 H, a-t, H6a, 7.9), 4.14 (1 H, dd, H6b, gem 8.3, J6b,5 6.9), 4.29-4.34 (1 H, m, H5), 4.53 (1 H, s, H4), 4.76 (1 H, s, H3); 5C (CHCI3, 100 MHz): 25.5, 25.5, 26.4, 27.2 (C(CH3)2), 49.9 (C21), 65.3 (C6), 75.2 (C5), 79.8 (C3), 81.4 (C4), 85.2 (C2), 1 10.7, 1 14.3 (C(CH3)2), 173.8 (C1); m/z (ESI +ve): 336 (75% M+Na+), 649 (100% 2M+Na+);
2-C-Azidomethyl-2,3:5,6-di-0-isopropylidene-D-talitol
Diisobutylaluminium hydride solution (1.5M in toluene, 6.2 mL, 9.3 mmol) was added dropwise to a stirred solution of the lactone (2.09 g, 6.68 mmol) in DCM (25 mL) at -78°C. The reaction mixture was stirred for 2 h at -78°C after which time TLC analysis (1 :2 EtOAc/cyclohexane) revealed the conversion of starting material (Rf 0.54) into one major product (Rf 0.42). The reaction was quenched with methanol (5.7 mL) and allowed to warm to RT. Saturated aqueous potassium sodium tartrate solution (48 mL) was added and the reaction was stirred for 16 h at RT. The aqueous phase was extracted with DCM (3 * 30 mL), dried (MgS04), filtered and concentrated in vacuo. The crude lactol was dissolved in methanol (25 mL) and stirred with sodium borohydride (244 mg, 6.68 mmol) at 0°C for 30 min and allowed to warm to RT for a further 2 h, after which time TLC analysis (1 :2 EtOAc/cyclohexane) revealed the formation of one major product (Rf 0.21 ). The reaction was neutralised with Dowex® (50W-X8, H+), filtered and concentrated in vacuo. The resulting residue was purified by flash column chromatography (1 :5 - 1 :1 EtOAc/cyclohexane) to afford the diol as a colourless oil (1.91 g, 90% over 2 steps).
HRMS (ESI -ve): found 340.1484 (M+Na)+; C13H23N3Na06 requires 340.1479; [a]D 25 -52.2 (c 1.0, CHCI3); vmax (thin film): 3417 (OH), 2110 (N3); rjH (400 MHz, CDCI3): 1.39 (3 H, s, CH3), 1.43 (3 H, s, CH3), 1.46 (3 H, s, CH3), 1.47 (3 H, s, CH3), 2.89 (1 H, s, OH1 ), 2.97 (1 H, s, OH4), 3.38 (1 H, d, H2'a, Jgem 13.2), 3.64-3.70 (2H, m, H1 a, H4), 3.74 (1 H, m, H1 b), 3.77 (1 H, d, H2'b, gem 13.2), 3.92 (1 H, dd, H6a, Jgem 8.6, J6a,5 6.4), 4.03 (1 H, d, H3, J3,4 9.5), 4.08 (1 H, dd, H6b, gem 8.6, J6b,5 6.6), 4.24 (1 H, dt, H5, J5,6a /5,6b 6.4, JS 4.7); Dc (100 MHz, CDCI3): 25.1 (CH3), 26.1 (CH3), 26.5 (CH3), 28.5 (CH3), 54.2 (C2'), 62.6 (C1 ), 66.2 (C6), 69.7 (C4), 76.7 (C5), 77.7 (C3), 84.7 (C2), 109.3 (CMe2), 109.5 (CMe2); m/z (ESI +ve): 340 (100% M+Na+), 657 (100% 2M+Na+);
2-C-Azidomethyl-2,3-0-isopropylidene-D-talitol A solution of 2-C-azidomethyl-2,3:5,6-di-0-isopropylidene-D-talitol (337 mg, 1.06 mmol) in AcOH:H20 (1 :1 , 7 mL) was stirred for 16 h at RT. After this time TLC analysis (EtOAc) showed complete conversion of starting material (Rf 0.78) to a single product (Rf 0.22). The reaction mixture was concentrated in vacuo, coevaporated with toluene (3 x 15 mL) and purified by flash column chromatography (EtOAc) to afford the tetraol as white crystals (244 mg, 83%).
HRMS (ESI -ve): found 300.1168 (M+Na)+; C10H19 3NaO6 requires 300.1166; [oc]D 25 -98.1 (c 1.0, CHCI3); vmax (thin film): 3424 (w, br, OH), 2107 (s, N3); DH (100 MHz, (CD3)2CO): 3.40 (1 H,d, H2'a, Jgem 12.9), 3.57-3.68 (3H, m, H1 a, H6a, H1 b or H6b), 3.70-3.80 (5H, m, H5, H2'b, H1 b or H6b, OH1 or OH6, OH5), 3.91 (1 H, ddd, H4, J4I3 9.6, ΟΗ4 6.2, J4,5 1.5, H4), 4.18 (1 H, d, OH4, J0H4,4 6.3), 4.27 (1 H, d, H3, J3 9.6), 4.40 (1 H, t, OH1 or OH6, J 5.4); rjc (100 MHz, (CD3)2CO): 26.8, 29.2 (2 x 3H, s, C(CH3)2), 55.6 (C2') , 63.2, 64.7 (C1 , C6), 70.2 (C4), 72.3 (C5), 78.0 (C3), 85.8 (C2), 109.0 (C(CH3)2); m/z (ESI +ve): 300 (98% M+Na+), 577 (100% 2M+Na+).
3-C-Azidomethyl-2,3-0-isopropylidene-D-erythrose
Sodium periodate (1.83 g, 8.57 mmol) was added to a solution of 2-C-azidomethyl-2,3-0- isopropylidene-D-talitol (1.19 g, 0.74 mmol) in water (30 mL) at RT and stirred for 3 h after which time TLC analysis (EtOAc) showed the complete conversion of the starting material (Rf 0.22) to one major product (Rf 0.88). The reaction mixture was extracted with EtOAc (3 x 15 mL), the organic layers were combined, dried (MgS04) and concentrated in vacuo. The resulting residue was purified by flash column chromatography (1 :4 - 1 :2 EtOAc/cyclohexane) to afford 3-C-azidomethyl-2,3-0-isopropylidene-D-erythrose (829 mg, 90%).
HRMS (ESI -ve): found 238.0798 (M+Na)+; C8H13N3Na04 requires 238.0798; [<x]D 25 -102.1 (c 1.0, CHCI3); vmax (thin film): 3425 (w, br, OH), 2106 (s, N3); DH (400 MHz, CDCI3) major anomer only: 1.46, 1.49 (2 x 3H, s, CH3), 3.00 (1 H, s, OH), 3.53 (1 H, d, H3'a, Jgem 12.9), 3.61 (1 H, d, H3'b, Jgem 12.9), 3.97 (1 H, d, H4a, J9em 10.1 ), 4.03 (1 H, d, H4b, Jgem 10.1), 4.34 (1 H, s, H2), 5.43 (1H, s, H1 ); Dc (100 MHz, CDCI3) major anomer only: 27.2, 27.3 (C(CH3)2), 54.7 (C3'), 74.6 (C4), 86.7 (C2), 97.3 (C3), 101.4 (C1 ), 113.8 (C(CH3)2); m/z (ESI +ve): 238 (70% M+Na+), (100% M+CH3CN+H+).
3-C-Azidomethyl-D-erythrose
Dowex® (50W-X8, H+) (1.73 g) was added to a solution of 3-C-azidomethyl-2,3-0- isopropylidene-D-erythrose (770 mg, 3.58 mmol) in 1 : 1 water: 1 ,4-dioxane (18 mL) and stirred at 60°C. After 18 h TLC analysis (1 :1 EtOAc/cyclohexane) showed conversion of starting material (Rf 0.64) to a single product (Rf 0.12). The crude mixture was filtered and concentrated under reduced pressure to afford 3-C-azidomethyl-D-erythrose (572 mg, 91%) as a pale brown oil, in a 3:2 ratio of anomers.
HRMS (ESI -ve): found 174.0518 [M-H]"; C5H8N304 requires 174.0520; [a]D 25 +1.7 (c 1.3, MeOH); vmax (thin film, Ge): 3356 (m, br, OH), 2110 (s, N3); δΗ ((CD3)2CO, 400 MHz): 3.36 (1 H, d, H3'aB, Jgem 10.9), 3.41 (1H, d, H3'bB, Jgem 10.9), 3.45 (2H, s, H3,A), 3.72 (1 H, d, H4aA, Jgem 9.6), 3.73-3.78 (1 H, m, H2A), 3.79 (1 H, d, H4aB, Jgem 8.6), 3.80-3.84 (1 H, m, H2B), 3.85 (1 H, d, H4bB, Jaem 9.6), 3.94 (1 H, d, H4bA, Jgem 9.6), 4.31 (1 H, s, OH3B), 4.32 (1 H, s, OH3A), 4.55 (1 H, d, OH2B, J0H2,2 .3), 4.90 (1 H, d, OH2A, JOH2,2 5.6), 5.18 (1 H, d, H1A, J1 i0Hi 2.3), 5.23 (1 H, d, H1 B, J1 I0H1 4.6), 5.40 (1 H, d, OH1B, J0HI,I 3.5), 5.53 (1 H, d, OH1A, J0H1.1 4.5); 5c ((CD3)2CO, 100 MHz); 56.9 (C3,B), 57.0 (C3,A), 73.6 (C2B), 74.0 (C4B), 74.6 (C4A), 78.6 (C3B), 79.1 (C2A), 79.8 (C3A), 97.4 (C1B), 104.1 (C1A); m/z (ESI -ve): 174 (100% [M-H]').
1 ,4-Dideoxy-2-C-hydroxymethyl-1 ,4-imino-L-threitol (iso-LAB)
Palladium on activated carbon (10%, 112 mg, 3.08 mmol) was added to a solution of 3-C- azidomethyl-D-erythrose (545 mg, 3.11 mmol) in 9:1 water/acetic acid (70 mL) and the reaction was purged with argon then hydrogen. The reaction mixture was stirred under hydrogen at RT for 24 h. The reaction was monitored by LRMS until no starting material was detected. The reaction mixture was filtered through Celite® and concentrated under reduced pressure to approximately 5 mL and absorbed onto a column of Dowex® (50W-X8, H+). The resin was washed with water before liberation of the amine with 2M aqueous ammonia. The ammoniacal fractions were concentrated under reduced pressure to afford 1 ,4-dideoxy-2-C-hydroxymethyl-1 ,4-imino-L-threitol (333 mg, 80%) as a brown oil.
HRMS (ESI +ve): found 134.0806 (M+H)+; C5H12N03 requires 134.0812; [a]D 25 +35.6 (c 0.27, H20); (thin film, Ge): 3326 (w, br, OH); δΗ (D20, 400 MHz): 2.89 (1 H, d, H4a, Jgem 12.9), 2.89 (1 H, d, H1 a, Jgem 12.6), 3.00 (1 H, d, H1b, Jgem 12.6), 3.41 (1 H, dd, H4b, Jgem 12.7, 34.5), 3.70 (1 H, d, H2'a, Jgem 12.0), 3.82 (1 H, d, H2'b, Jgem 12.0), 4.10 (1H, d, H3, J3,44.3); δο (D20, 100 MHz): 53.0 (C4), 53.3 (C1), 63.1 (C2'), 76.6 (C3), 83.7 (C2); m/z (ESI +ve): 134 (100% M+H+).
The above exemplified synthesis starts from D-tagatose, but those skilled in the art will recognize that isoLAB can also be prepared by routes from other sugars. Examples of the many suitable starting monosaccharides include (but are not restricted to) L-ribose, D- lyxose, L-psicose, D-mannose, D-fructose and L-sorbose, D-mannose, L-ribonolactone, L- gulose, D-lyxonolactone, L-gulonolactone, D-mannonolactone, L-gulonolactone and suitably protected derivatives of any of the foregoing. In the latter case, protection may be of cis-1 ,2-diols by acetone to acetonides, or any suitable ketone (such as cyclohexanone, pentan-3-one or other ketones) to its corresponding ketal.
(b) Example 2: CFTR rescue activity
Materials and methods
Cell culture
The human tracheal gland serous epithelial cell line CF-KM4 is derived from a CF patient homozygous for the AF508 mutation. The details of the generation, characterization, and routine propagation have been described elsewhere (Kammouni et al. (1999) Resp. Cell Mol. Biol. 20(4): 684-91 ).
Functional analysis of CFTR activity
CFTR ion channel functions were assessed by single-cell fluorescence imaging, using the potential-sensitive probe bis-(1 ,3-diethylthiobarbituric acid)trimethine oxonol (DiSBAC2(3); Molecular Probes, Eugene, OR), as previously reported (see Norez et al. (2009) Am. J. Respir. Cell Mol. Biol. 41 (2): 217-225). Fluorescence intensity was recorded by confocal laser scanning microscopy using Bio-Rad MRC 1024 equipped with 15 mW Ar/Kr gas laser (Hemel Hempstead, UK). Maximal resolution was obtained with Olympus plan apo X60 oil, 1.4 NA, objective lens. Fluorescence signal collection was performed through the control software Lasersharp 3.2 (Hemel Hempstead, UK). The resolution time was 30 s. Bis- oxonol slowly distributes across biological membrane according to the membrane potential and binds to hydrophobic cell components; since the quantum yield of the dye increases impressively upon the binding, the fluorescence of cells incubated in a medium containing bis-oxonol increases upon depolarization and, conversely, decreases with
hyperpolarization (Dall'Asta et al. (1997) Exp. Cell Res. 231 : 260-268). CFTR-dependent current was stimulated by application of Forskolin + Genistein (Fsk+Gst), inducing a depolarization characterized by an increase of the fluorescence, while CFTR-dependent current was inhibited by application of CFTRinh-172 characterized by a decrease of the fluorescence. Results are presented as transformed data to obtain the percentage signal variation (Fx) relative to the time of addition of the stimulus, according to the equation : Fx = ([Ft-F0]/F0]X100 where Ft and FO are the fluorescent values at the time t and at the time of addition of the stimulus, respectively. For histogram representation, the values correspond to the level of stable variation of fluorescence induced by each drug.
The CF-KM4 cells were treated 2 hours with 10ΌμΜ of test compound and then CFTR proteins were stimulated by a cocktail of forskolin (Fsk) + genistein (Gst).
Results
Fig. 1 shows the functional evaluation of AF508 CFTR by DiSBAC2(3) assay in CF-K 4 cells treated (100μΜ for 2h) or not (negative control) with isoLAB. Λ/Β-DNJ (100μΜ for 2h) served as a positive control. Fig. 1A shows examples of typical time courses obtained with untreated (NT) or treated with isoLAB. Data represent the mean (± SEM) of the relative fluorescence collected from all the cells of a field (n=12). A mixture of forskolin (Fsk, 10 μΜ) + genistein (Gst, 30 μΜ) is used to activate CFTR. CFTRinh-172 (10 μΜ) is used to inhibit CFTR. Fig. 1 B shows histograms report the mean of the relative fluorescence collected from separate experiments (N=2) with a total of 24 cells with the test compound (isoLAB) and positive (NB-DNJ) and negative (NT) controls.
As expected, no variation of the fluorescence was observed after the cocktail stimulation in untreated CF-KM4 bearing the defective AF508 CFTR protein (Fig. 1A). In contrast, treatment of CF-KM4 cells with isoLAB produced a strong increase in the recorded fluorescence signal (greater than that produced by the positive control M3-DNJ) after Fsk+Gst stimulation (Fig. 1A and B). The signal was fully inhibited by the CFTR inhibitor CFTRinh-172 (Fig. 1A). In a separate study, the cellular toxicity of isoLAB was evaluated on CF-K 4 cells using the MTT test (Noel et al. (2006) Pharmacol. Expt. Therapeut. 319: 349-359). No apparent cell toxicity (a range of concentration was tested up to 1 mM) was detected (data not shown). (c) Example 3: Glycosidase inhibitory profile of isoLAB
The CFTR-resuing activity of Λ/Β-DNJ has been ascribed (at least in part) to its a- glucosidase inhibitory activity (Norez et al. (2006) FEBS Lett. 580: 2081 -2086). Assays of isoLAB with various glycosidases were conducted to evaluate its glycosidase inhibitory profile (see Table 1 , below).
Table 1 : Glycosidase inhibitory profile of isoLAB
Enzyme isoLAB
a-Glucosidase
Rice Nl
Yeast Nl
Rat intestinal maltase Nl
Rat intestinal isomaltase l
Rat intestinal sucrase Nl
β-Glucosidase
Almond Nl
Bovine liver Nl
Rat intestinal cellobiase Nl a-Galactosidase
Coffee beans Nl
Human lysosome Nl
β-Galactosidase
Bovine liver Nl
Rat intestinal lactase Nl
a-Mannosidase
Jack beans l
β-Mannosidase
Snail Nl
oc-L-Rhamnosidase
P. decumbens Nl
cc-L-Fucosidase
Bovine epididymis Nl
Trehalase
Rat intestinal trehalase Nl
Nl : No inhibition (less than 50% inhibition at 1000 μΜ) The results show that isoLAB has no inhibitory activity on any of the glycosidase enzymes listed. In contrast, Λ/Β-DNJ exhibits potent inhibition of a wide range of a-glucosidases (including those involved in digestive and ER processing functions). Since such nonspecific inhibitory activity is implicated in a variety of undesirable side effects (including gastric toxicity), the absence of such inhibitory activity is an unexpected and significant technical advantage of isoLAB versus Λ/Β-DNJ.
Equivalents The foregoing description details presently preferred embodiments of the present invention. Numerous modifications and variations in practice thereof are expected to occur to those skilled in the art upon consideration of these descriptions. Those modifications and variations are intended to be encompassed within the claims appended hereto.

Claims

CLAIMS:
1. A combination comprising Λ/Β-DNJ and a compound of formula (I):
Figure imgf000028_0001
wherein:
R1 is selected from H; linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl and aralkyl and wherein the optional substitution may be with one or more groups independently selected from: -OH; -F; -CI; -Br; -I; -NH2; alkylamino;
dialkylamino; linear or branched alkyl, alkenyl, alkynyl and aralkyl; aryl; heteroaryl; linear or branched alkoxy; aryloxy; aralkoxy; -(alkylene)oxy(alkyl); -CN; -N02; -
COOH; -COO(alkyl); -COO(aryl); -C(0)NH(alkyl); -C(0)NH(aryl); sulfonyl;
alkylsulfonyl; arylsulfonyl; sulfamoyl; alkylsulfamoyl; alkylthio; alkylsulfonamide; arylsulfonamide; -NHNH2; and -NHOH; or a bioisostere, pharmaceutically acceptable salt or derivative thereof.
2. The combination of claim 1 wherein R1 is selected from H, C,.^ alkyl (for example, C -9 alkyl, e.g. C1.6 alkyl), C2.18 alkenyl (for example, C2.9 alkenyl, e.g. C2.6 alkenyl) and C2-18 alkynyl (for example, C2.9 alkynyl, e.g. C2-6 alkynyl). For example, R1 may be -H and Rr selected from C1-18 alkyl (for example, Ci-9 alkyl, e.g. Ci-6 alkyl), C2. 8 alkenyl (for example, C2-9 alkenyl, e.g. C2.6 alkenyl) and C2.i8 alkynyl (for example, C2-9 alkynyl, e.g. C2^ alkynyl).
3. The combination of claim 1 wherein R1 is selected from H; C1-15 alkyl, C1-15 alkenyl or C1-15 alkynyl, optionally substituted with one or more R2; oxygen or an oxygen containing group such that the compound of formula (I) is an N-oxide; C(0)OR3; C(0)NR3R4; S02NR3; OH, OR3, or formyl.
4. The combination of claim 1 wherein R1 is selected from C1 -9 alkyl, optionally substituted with up to 6 OH, NR3R4, aryl, 0-C1-3 alkyl, 0-C1-3 alkenyl, C02H, NH(NH)NH2, CONR3R4; C(0)OR3; C(0)NR3R4; or S02NR3.
5. The combination of claim 1 wherein R1 is H, the compound of formula (I) being (3S,4S)- 3-(hydroxymethyl)pyrrolidine-3,4-diol (isoLAB).
6. The combination of any one of the preceding claims wherein the compound of formula (I) rescues mutant CFTR activity.
7. The combination of claim 6 wherein the compound of formula (I) rescues AF508 CFTR activity.
8. The combination of any one of the preceding claims wherein the compound of formula (I) is a CFTR pharmacoperone.
9. The combination of any one of the preceding claims wherein the compound of formula (I) and Λ/Β-DNJ are physically associated.
10. The combination of claim 9 wherein the compound of formula (I) and Λ/Β-DNJ are: (a) in admixture (for example within the same unit dose); (b) chemically/physicochemically linked (for example by crosslinking, molecular agglomeration or binding to a common vehicle moiety); (c) chemically/physicochemically co-packaged (for example, disposed on or within lipid vesicles, particles (e.g. micro- or nanoparticles) or emulsion droplets); or (d) unmixed but co-packaged or co-presented (e.g. as part of an array of unit doses).
1 1. The combination ff any one of claims 1 to 8 wherein the compound of formula (I) and Λ/Β-DNJ are non-physically associated.
12. The combination of claim 11 wherein the combination comprises: (a) at least one of the compound and Λ/Β-DNJ together with instructions for their extemporaneous association to form a physical association; or (b) at least one of the compound and Λ/Β-DNJ together with instructions for combination therapy with the compound and Λ/Β-DNJ; or (c) at least one of the compound and Λ/Β-DNJ together with instructions for administration to a patient population in which either the compound and Λ/Β-DNJ have been (or are being) administered; or (d) at least one of the compound and Λ/Β-DNJ in an amount or in a form which is specifically adapted for use in combination.
13. The combination as defined in any one of claims 9 to 12: (a) in the form of a pharmaceutical pack, kit or patient pack; (b) in a pharmaceutical excipient; or (c) in unit dosage form.
14. The combination of any one of the preceding claims wherein the compound of formula (I): (a) is an an a-glucosidase inhibitor; or (b) is not an a-glucosidase inhibitor.
15. The combination of any one of the preceding claims wherein the compound of formula (I) is not an α-glucosidase inhibitor.
16. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit cellular ER processing glucosidases.
17. The combination of any one of the preceding claims wherein the compound of formula (I) exhibits less than 50% inhibition of cellular ER processing glucosidases at 1000 μΜ.
18. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit glucosylceramide synthase.
19. The combination of any one of the preceding claims wherein the compound of formula (I) exhibits less than 50% inhibition of glucosylceramide synthase at 1000 μΜ.
20. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit disaccharidases, for example sucrase and/or maltase.
21. The combination of any one of the preceding claims wherein the compound of formula (I) exhibits less than 50% inhibition of disaccharidases, for example sucrase and/or maltase, at 1000 μΜ.
22. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit lysosomal and/or non-lysosomal glucocerebrosidase.
23. The combination of any one of the preceding claims wherein the compound of formula (I) exhibits less than 50% inhibition of lysosomal and/or non-lysosomal glucocerebrosidase at 1000 μΜ.
24. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit a-glucosidase I and/or II.
25. The combination of any one of the preceding claims wherein the compound of formula (I) exhibits less than 50% inhibition of a-glucosidase I and/or II at 1000 μΜ.
26. The combination of any one of the preceding claims wherein the compound of formula (I) does not exhibit inhibition of glycosidase activity sufficient to elicit gastric toxicity in humans.
27. The combination of any one of the preceding claims wherein the compound of formula (I) does not inhibit the glycosidase enzymes listed in Table 1 herein.
28. The combination of any one of the preceding claims which exhibits less than 50% inhibition at 1000 μΜ of each of the glycosidase enzymes listed in Table 1 herein.
29. The combination of any one of the preceding claims which further comprises a pharmaceutically acceptable excipient.
30. The combination of any one of the preceding claims wherein the Λ/Β-DNJ is an Λ/Β- DNJ derivative which can restore CFTR function.
31. The combination of any one of the preceding claims which is formulated for oral delivery.
32. The combination of any one of claims 1 to 30 which is formulated for delivery to the lung.
33. An inhalation device comprising the combination of claim 32.
34. The inhalation device of claim 33 wherein the compound or composition is present in the form of a solution, a suspension or a powder.
35. The inhalation device of claim 33 or 34 which is an MDI or nebulizer.
36. The combination of any one of claims 1 to 30 for use in therapy or prophylaxis.
37. The combination of any one of claims 1 to 30 for use in a method of treating of cystic fibrosis.
38. A method of treating cystic fibrosis comprising administering an effective amount of the combination as defined in any one of claims 1 to 30 to a subject in need thereof.
PCT/GB2011/000594 2010-04-19 2011-04-19 Novel iminosugar combinations WO2011131926A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1006434.3A GB201006434D0 (en) 2010-04-19 2010-04-19 Novel iminosugar combinations
GB1006434.3 2010-04-19

Publications (1)

Publication Number Publication Date
WO2011131926A1 true WO2011131926A1 (en) 2011-10-27

Family

ID=42245370

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/000594 WO2011131926A1 (en) 2010-04-19 2011-04-19 Novel iminosugar combinations

Country Status (2)

Country Link
GB (1) GB201006434D0 (en)
WO (1) WO2011131926A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014110270A1 (en) * 2013-01-09 2014-07-17 Amicus Therapeutics, Inc. Stable parenteral dnj compositions

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001010429A2 (en) 1999-08-10 2001-02-15 The Chancellor, Masters, And Scholars Of The University Of Oxford Long chain n-alkyl compounds and oxa-derivatives thereof and use as antiviral compositions
WO2005046672A2 (en) 2003-11-07 2005-05-26 Centre National De La Recherche Scientifique Use of glucosidase inhibitors for therapy of mucovisidosis
WO2007123403A1 (en) 2006-04-24 2007-11-01 Academisch Medisch Centrum Improved treatment of cystic fibrosis

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001010429A2 (en) 1999-08-10 2001-02-15 The Chancellor, Masters, And Scholars Of The University Of Oxford Long chain n-alkyl compounds and oxa-derivatives thereof and use as antiviral compositions
WO2005046672A2 (en) 2003-11-07 2005-05-26 Centre National De La Recherche Scientifique Use of glucosidase inhibitors for therapy of mucovisidosis
US20070213357A1 (en) * 2003-11-07 2007-09-13 Becq Frederic Use of Glucosidase Inhibitors for Therapy of Mucovisidosis
WO2007123403A1 (en) 2006-04-24 2007-11-01 Academisch Medisch Centrum Improved treatment of cystic fibrosis

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
"Handbook of Pharmaceutical Excipients", 1999, AMERICAN PHARMACEUTICAL ASSOCIATION
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS AND WILKINS
ANTIGNY ET AL., CELL CALCIUM, vol. 43, no. 2, 2008, pages 175 - 193
ASANO, N., CELL. MOL. LIFE SCI., vol. 66, 2009, pages 1479 - 1492
BEST D ET AL: "Cystic fibrosis and diabetes: isoLAB and isoDAB, enantiomeric carbon-branched pyrrolidine iminosugars", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 51, no. 32, 11 August 2010 (2010-08-11), pages 4170 - 4174, XP027129775, ISSN: 0040-4039, [retrieved on 20100608] *
BOLS M ET AL: "Synthesis of a Ribofuranosyl Cation Mimic", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 37, no. 12, 18 March 1996 (1996-03-18), pages 2097 - 2100, XP004029871, ISSN: 0040-4039, DOI: DOI:10.1016/0040-4039(96)00202-X *
BUTTERS ET AL., TETRAHEDRON ASYMM, vol. 11, 2000, pages 113 - 125
COX ET AL.: "Iminosugars From Synthesis to Therapeutic Applications", 2007, JOHN WILEY & SONS, article "Medicinal use of iminosugars", pages: 318 - 319
CRUZ F P D ET AL: "4-C-Me-DAB and 4-C-Me-LAB-enantiomeric alkyl-branched pyrrolidine iminosugars-are specific and potent alpha-glucosidase inhibitors; acetone as the sole protecting group", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 52, no. 2, 12 January 2011 (2011-01-12), pages 219 - 223, XP027549745, ISSN: 0040-4039, [retrieved on 20101207] *
DALL'ASTA ET AL., EXP. CELL RES, vol. 231, 1997, pages 260 - 268
DECHECCHI ET AL., J. CYSTIC FIBROSIS, vol. 7, no. 6, 2008, pages 555 - 565
ENGLEBIENNE, MED. CHEM., vol. 1, no. 3, 2005, pages 215 - 226
GOODMAN, GOLDMAN'S: "The Pharmacological Basis of Therapeutics", 1996, pages: 1707 - 1711
KAMMOUNI ET AL., RESP. CELL MOL. BIOL., vol. 20, no. 4, 1999, pages 684 - 91
KUSHNER ET AL., CAN J PHYSIOL PHARMACOL., vol. 77, no. 2, 1999, pages 79 - 88
LUBAMBA BOB ET AL: "Airway delivery of low-dose miglustat normalizes nasal potential difference in F508del cystic fibrosis mice.", AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE 1 JUN 2009 LNKD- PUBMED:19299496, vol. 179, no. 11, 1 June 2009 (2009-06-01), pages 1022 - 1028, XP002655391, ISSN: 1535-4970 *
LUBAMBA ET AL., AM. J. RESP. CRIT. CARE, vol. 179, no. 11, 2009, pages 1022 - 1028
MELLOR ET AL., BIOCHEM. J., vol. 366, 2002, pages 225 - 233
NOEL ET AL., PHARMACOL. EXPT. THERAPEUT., vol. 319, 2006, pages 349 - 359
NOREZ ET AL., AM. J. RESP. CELL MOL. BIOL., vol. 41, no. 2, 2009, pages 217 - 225
NOREZ ET AL., AM. J. RESPIR. CELL MOL. BIOL., vol. 41, no. 2, 2009, pages 217 - 225
NOREZ ET AL., FEBS LETT., vol. 580, 2006, pages 2081 - 2086
NOREZ ET AL., J. PHARM. EXP. THER., vol. 325, no. 1, 2008, pages 89 - 99
RAWLINGS ET AL., CHEMBIOCHEM, vol. 10, 2009, pages 1101 - 1105
TACKE, ZILCH, ENDEAVOUR, vol. 10, 1986, pages 191 - 197
ZHANG ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 51232 - 51242

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014110270A1 (en) * 2013-01-09 2014-07-17 Amicus Therapeutics, Inc. Stable parenteral dnj compositions
CN104918619A (en) * 2013-01-09 2015-09-16 阿米库斯治疗学公司 Stable parenteral dnj compositions
US9827189B2 (en) 2013-01-09 2017-11-28 Amicus Therapeautics, Inc. Stable parenteral DNJ compositions
AU2014205378B2 (en) * 2013-01-09 2018-08-30 Amicus Therapeutics, Inc. Stable parenteral DNJ compositions
US10117830B2 (en) 2013-01-09 2018-11-06 Amicus Therapeutics, Inc. Stable parenteral DNJ compositions
CN104918619B (en) * 2013-01-09 2019-04-02 阿米库斯治疗学公司 Stable parenteral DNJ composition

Also Published As

Publication number Publication date
GB201006434D0 (en) 2010-06-02

Similar Documents

Publication Publication Date Title
EP3440057B1 (en) Silicone atoms containing ivacaftor analogues
EP3615528B1 (en) 4-sulfonylaminocarbonylquinoline derivatives for increasing cftr activity
EP3472156B1 (en) Compounds, compositions, and methods for increasing cftr activity
JP4767172B2 (en) Hydroxypiperidine derivatives for the treatment of Gaucher disease
JP5292605B2 (en) Isofagomine tartrate and methods of use
TWI587859B (en) Novel compositions for preventing and/or treating lysosomal storage disorders
US20110160223A1 (en) NMDA Receptor Antagonists for the Treatment of Neuropsychiatric Disorders
EA021755B1 (en) PYRAZINE DERIVATIVES AS ENaC BLOCKERS
US9492440B2 (en) Bicyclic amides for enhancing glutamatergic synaptic responses
EP3564253A1 (en) Antidepressant compound and preparation method and application thereof
US11498896B2 (en) Dopamine D2 receptor ligands
KR20110017452A (en) Novel compounds active as muscarinic receptor antagonists
US7541466B2 (en) Tetrahydroisoquinoline derivatives for treating protein trafficking diseases
Pandey et al. Convergent approach toward the synthesis of the stereoisomers of C-6 homologues of 1-deoxynojirimycin and their analogues: evaluation as specific glycosidase inhibitors
CN113166051A (en) Pegylated prodrugs of phenolic TRPV1 agonists
US8680279B2 (en) Compounds for the treatment of neurological disorders
WO2011131926A1 (en) Novel iminosugar combinations
EP3233799B1 (en) Dopamine d2 receptor ligands
US11512052B2 (en) Dihydropyridines for the treatment of cognitive impairment or traumatic brain injury
WO2011086347A1 (en) Pyrrolidine iminosugars used in the treatment of cystic fibrosis
WO2011095772A2 (en) Novel iminosugar therapeutics
JP2008546744A (en) Novel compounds of the imino sugar family, their use for the treatment of lysosomal diseases and methods for their production
WO2013062680A1 (en) Novel compounds and compositions thereof for treating nervous system disorders
US20240025852A1 (en) T-type calcium channel modulators and methods of use thereof
TW202235079A (en) Progranulin modulators and methods of using the same

Legal Events

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

Ref document number: 11731448

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11731448

Country of ref document: EP

Kind code of ref document: A1