WO2002055064A2 - Traitement des mucopolysaccharidoses - Google Patents

Traitement des mucopolysaccharidoses Download PDF

Info

Publication number
WO2002055064A2
WO2002055064A2 PCT/US2002/000813 US0200813W WO02055064A2 WO 2002055064 A2 WO2002055064 A2 WO 2002055064A2 US 0200813 W US0200813 W US 0200813W WO 02055064 A2 WO02055064 A2 WO 02055064A2
Authority
WO
WIPO (PCT)
Prior art keywords
mps
inhibitor
glucosylceramide synthesis
synthesis
use according
Prior art date
Application number
PCT/US2002/000813
Other languages
English (en)
Other versions
WO2002055064A3 (fr
Inventor
Steven Walkley
Original Assignee
Oxford Glycosciences (Uk) Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0100889.5A external-priority patent/GB0100889D0/en
Application filed by Oxford Glycosciences (Uk) Ltd. filed Critical Oxford Glycosciences (Uk) Ltd.
Priority to AU2002241853A priority Critical patent/AU2002241853A1/en
Publication of WO2002055064A2 publication Critical patent/WO2002055064A2/fr
Publication of WO2002055064A3 publication Critical patent/WO2002055064A3/fr
Priority to US10/619,378 priority patent/US20050032841A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic 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
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system

Definitions

  • the present invention relates to the use of inhibitors of glucosylceramide synthesis or agents capable of increasing the rate of neuronal glycolipid degradation in the treatment of mucopolysaccharidosis diseases.
  • Mucopolysaccharidosis (MPS) diseases are autosomal recessive disorders of proteoglycan metabolism.
  • a list of the major examples of the currently known mucopolysaccharidosis diseases and the enzyme defects that underlie them is provided in Table 1.
  • the failure to break down proteoglycans may result in the storage of fragments of these materials (glycosaminoglycans (GAGs)) in cells and tissues such as the joints, skeleton, bone marrow, arteries, eyes, skin, lungs, liver, spleen, blood cells and the central nervous system.
  • GAGs glycosaminoglycans
  • neurons in these MPS animal models conspicuously store the glycolipids, ganglioside GM2 and GM3 (J. Neurochem., 1994, 62(5), 1852; Neuroscience, 1995, 68(4), 1027 and "Multiple substrate accumulation in MPS disorders and the efficacy of substrate deprivation as therapy" S. Walkley et al, abstract presented at Strategies for Therapy of MPS and Related Diseases, 16 th Annual MPS Conference, University of California at Los Angeles, June 21-24, 2001).
  • NB-DNJ The imino sugar N-butyldeoxynojirimycin
  • NB-DNJ is a potent inhibitor of ceramide glucosyltransferase.
  • NB-DNJ is currently undergoing clinical trials as a treatment for Gaucher and Fabry diseases, glycolipid storage disorders resulting from mutations in glucocerebrosidase and alpha-galactosidase A, respectively.
  • NPC Niemann-Pick disease type C
  • Glucosylceramide synthesis inhibitors have been shown to slow the onset of NPC symptoms of neuronal degeneration and to have an effect on the long-term health of NPC animal models, increasing the longevity of treated animals by more than 20% (Current Biology, 2001, 11:1283-1287).
  • neuronal glycolipid storage seen in MPS patients may be reduced by treatment with inhibitors of glucosylceramide synthesis or agents capable of increasing the rate of neuronal glycolipid degradation.
  • the present invention provides the use of an inhibitor of glucosylceramide synthesis in the manufacture of a medicament for the treatment of MPS disease.
  • the invention also provides a method of treating MPS disease, in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of an inhibitor of glucosylceramide synthesis.
  • the invention also provides an inhibitor of glucosylceramide synthesis for use in the treatment of MPS disease.
  • the MPS disease to be treated according to the invention may be selected from MPS I (MPS ffl, IS or 1H/S), MPS II, MPS IIIA, IHB , mC or DID, MPS IVA or IVB, MPS VI or MPS v ⁇ . 12.
  • the invention also provides the use of an inhibitor of glucosylceramide synthesis in the manufacture of a medicament for reducing neuronal glycolipid storage in MPS disease.
  • the invention also provides a method reducing neuronal glycolipid storage in MPS disease, in a mammal in need thereof, comprising administering to said mammal an inhibitor of glucosylceramide synthesis.
  • the invention also provides an inhibitor of glucosylceramide synthesis for use in the reduction of neuronal glycolipid storage in MPS disease. More specifically, inhibitors of glucosylceramide synthesis may be used to reduce neuronal glycolipid storage in the brain in MPS disease.
  • the inhibitor of glucosylceramide synthesis is an inhibitor of ceramide glucosyltransferase.
  • Inhibitors of glucosylceramide synthesis include small molecules and peptides.
  • Small molecules which may be used in the method of the invention include N- butyldeoxynojirimycin, N-butyldeoxygalactonojirimycin, and other imino sugar-structured inhibitors of glucosylceramide synthesis.
  • Other small molecule inhibitors of glycosylceramide synthesis include agents such as l- ⁇ henyl-2-decanoylamino-3-morpholino-l- ⁇ ropanol (PDMP), D- threo-l-phenyl-2-decanoylamino-3-mor ⁇ holino-l-propanol and structurally related analogues thereof.
  • inhibitors of glucosylceramide synthesis is also intended to encompass genetic approaches, based on the introduction of nucleic acid coding for proteins or peptides capable of inhibiting glucosylceramide synthesis or antisense sequences or catalytic RNA capable of interfering with the expression of enzymes responsible for glucosylceramide synthesis (e.g. glucosylceramide synthase).
  • the inhibitor of glucosylceramide synthesis is an antibody capable of binding to and interfering with an enzyme required for glucosylceramide synthesis, for example, glucosylceramide synthase.
  • the invention extends to the preparation of appropriate constructs and compositions containing antibodies to an enzyme required for glucosylceramide synthesis.
  • the antibodies are polyclonal, monoclonal, or chimeric, including humanized antibodies.
  • compounds for use in the present invention include N- butyldeoxynojirimycin and N-butyldeoxygalactonojirimycin.
  • the inhibitor compound is N-butyldeoxynojirimycin.
  • N-butyldeoxynojirimycin and N- butyldeoxygalactojirimycin can be found for example in US-A-4182767, EP-B-0012278, EP-A- 0624652, US-A-4266025, US-A-4405714 and US-A-5151519.
  • the invention also provides the use of an agent capable of increasing the rate of neuronal glycolipid degradation in the manufacture of a medicament for the treatment of MPS disease.
  • the invention also provides a method for the treatment of MPS disease which comprises administering to a subject in need thereof a therapeutically effective amount of an agent capable of increasing the rate of degradation of neuronal glycolipids.
  • the invention also provides an agent capable of increasing the rate of degradation of neuronal glycolipids for use in the treatment of MPS disease.
  • Agents capable of increasing the rate of neuronal glycolipid degradation may be used, in particular, to reduce neuronal glycolipid storage in the brain in MPS disease.
  • agents capable of increasing the rate of neuronal glycolipid degradation include a nucleic acid sequence (DNA or RNA) which codes for the enzymes which degrade neuronal glycolipids, e.g. glucocerebrosidase, lysosomal hexoseaminidases, galactosidases, sialidases and glucosylceramide glucosidase, i.e. such sequences could be introduced to increase natural production of such enzymes.
  • DNA or RNA nucleic acid sequence which codes for the enzymes which degrade neuronal glycolipids, e.g. glucocerebrosidase, lysosomal hexoseaminidases, galactosidases, sialidases and glucosylceramide glucosidase, i.e. such sequences could be introduced to increase natural production of such enzymes.
  • inhibitors of glucosylceramide synthesis or agent capable of increasing the rate of neuronal glycolipid degradation may be co-administered with enzyme replacement therapy, the enzyme replacement therapy being replacement of enzymes known to be defective in MPS diseases, these are listed in Table 1.
  • Specific enzymes which may be mentioned for use in combination therapy according to the invention include alpha-L-iduronidase and iduronate sulfatase.
  • FIG. 1 shows photomicrographs of brain tissue from A untreated controls (i: upper cortex; ⁇ : lower cortex) and B NB-DNJ treated mice (i: upper cortex; ii: lower cortex).
  • FIG. 2 shows photomicrographs of the hippocampus from A untreated control or B NB-DNJ treated mice.
  • FIG. 3 shows photomicrographs of the cerebral cortex from A untreated control or B NB-DNJ treated mice.
  • FIG. 4 shows photomicrographs of the cerebral cortex from A untreated control or B NB-DNJ treated mice.
  • references to “an inhibitor of glucosylceramide synthesis” includes mixtures of such inhibitors
  • reference to “the formulation” or “the method” includes one or more formulations, methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure and so forth.
  • inhibitor of glucosylceramide synthesis includes molecules such as N-butyldeoxynojirimycin or N-butyldeoxygalactonojirimycin, and other imino sugar-structured inhibitors of glucosylceramide synthesis. Also included are agents such as l-phenyl-2-decanoylamino-3-morpholino-l-propanol (PDMP), D-threo-l- ⁇ henyl-2- decanoylamino-3-morpholino-l-propanol and structurally related analogues thereof which exhibit the ability to inhibit glucosylceramide synthesis.
  • PDMP l-phenyl-2-decanoylamino-3-morpholino-l-propanol
  • D-threo-l- ⁇ henyl-2- decanoylamino-3-morpholino-l-propanol and structurally related analogues thereof which exhibit the ability to inhibit glucosylceramide synthesis.
  • inhibition can also be achieved by the use of genetic approaches, based on the introduction of nucleic acid coding for proteins or peptides capable of inhibiting glucosylceramide synthesis or antisense sequences or catalytic RNA capable of interfering with the expression of enzymes responsible for glucosylceramide synthesis (e.g. glucosylceramide synthase).
  • Genetic approaches based on the introduction of nucleic acid coding for proteins or peptides capable of inhibiting glucosylceramide synthesis or antisense sequences or catalytic RNA capable of interfering with the expression of enzymes responsible for glucosylceramide synthesis (e.g. glucosylceramide synthase).
  • a combination of any of the above approaches can be used.
  • substantially pure when referring to a polypeptide, means a polypeptide that is at least 60%, by weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated.
  • a substantially pure glucosylceramide synthesis inhibitor is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, glucosylceramide synthesis inhibitor.
  • a substantially pure glucosylceramide synthesis inhibitor such as N-butyldeoxynojirimycin (NB-DNJ), can be obtained, for example, by chemical synthesis. Purity can be measured by any appropriate method, e.g., column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
  • Treatment refers to the administration of medicine or the performance of medical procedures with respect to a patient, for either prophylaxis (prevention) or to cure the infirmity or malady in the instance where the patient is afflicted.
  • a “therapeutically effective amount” is an amount of a reagent sufficient to achieve the desired treatment effect.
  • the medicaments described herein and which are also for use in the methods provided herein may include one or more of the following: preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifiers, sweeteners, colorants, odorants, salts, buffers, coating agents or antioxidants. They may also contain therapeutically active agents in addition to the compounds and/or agents described herein.
  • the medicaments may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.
  • a composition may be prepared by any method known in the art of pharmacy, for example by admixing the active ingredient with a carrier under sterile conditions.
  • Small molecule inhibitors of glycosylceramide synthesis are preferably formulated for oral administration. Oral Administration
  • Medicaments adapted for oral administration may be provided as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids); as edible foams or whips; or as emulsions.
  • Tablets or hard gelatine capsules may comprise lactose, maize starch or derivatives thereof, stearic acid or salts thereof.
  • Soft gelatine capsules may comprise vegetable oils, waxes, fats, semi-solid, or liquid polyols etc.
  • Solutions and syrups may comprise water, polyols and sugars.
  • suspensions oils e.g. vegetable oils
  • oil-in-water or water-in-oil suspensions may be used to provide oil-in-water or water-in-oil suspensions.
  • Medicaments adapted for transdermal administration may be provided as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the active ingredient may be delivered from the patch by iontophoresis (Iontophoresis is described in Pharmaceutical Research, 3(6):318 (1986)).
  • Medicaments adapted for topical administration may be provided as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • a topical ointment or cream is preferably used.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water base or a water-in-oil base.
  • Medicaments adapted for topical administration to the eye include eye drops.
  • the active ingredient can be dissolved or suspended in a suitable carrier, e.g. in an aqueous solvent.
  • Medicaments adapted for topical administration in the mouth include lozenges, pastilles and mouthwashes.
  • Medicaments adapted for rectal administration may be provided as suppositories or enemas.
  • Medicaments adapted for nasal administration which use solid carriers include a coarse powder (e.g. having a particle size in the range of 20 to 500 microns). This can be administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nose from a container of powder held close to the nose.
  • a coarse powder e.g. having a particle size in the range of 20 to 500 microns. This can be administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nose from a container of powder held close to the nose.
  • compositions adopted for nasal administration which use liquid carriers include nasal sprays or nasal drops. These may comprise aqueous or oil solutions of the active ingredient.
  • Medicaments adapted for administration by inhalation include fine particle dusts or mists, which may be generated by means of various types of apparatus, e.g. pressurized aerosols, nebulizers or insufflators. Such apparatus can be constructed so as to provide predetermined dosages of the active ingredient.
  • Medicaments adapted for vaginal admimstration may be provided as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • Medicaments adapted for parenteral administration include aqueous and non-aqueous sterile injectable solutions or suspensions. These may contain antioxidants, buffers, bacteriostats and solutes which render the compositions substantially isotonic with the blood of an intended recipient. Other components which may be present in such compositions include water, alcohols, polyols, glycerine and vegetable oils, for example.
  • Compositions adapted for parenteral administration may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, e.g. sterile water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • compositions may contain from 0.1% by weight, preferably from 10-60% by weight, of the active material, depending on the method of administration.
  • the amount of the compound of the invention which will be effective in the treatment of mucopolysaccharidosis diseases can be determined by standard clinical techniques based on the present description.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances. However, suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight.
  • Suitable dosage ranges for intranasal admimstration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient.
  • Dosages will be readily determinable by routine trials, and will be under the control of the physician or clinician.
  • the guiding principle for determining a suitable dose will be delivery of a suitably efficacious but non-toxic, or acceptably toxic, amount of material.
  • the optimal quantity and spacing of individual dosages will be determined by the nature and extent of the condition being treated, the form, route and site of administration, and the particular mammal being treated. It will also be appreciated that the optimal course of treatment, i.e., the number of doses of the active material given per day for a defined number of days, can be ascertained by those skilled in the art using conventional course of treatment determination tests.
  • a daily dosage for an adult could be expected to be in the range of from 1 mg to 2 g of active agent, and may be in the range of from 50 to 800 mg, or 150 to 600 mg, or 300 to 600 mg.
  • the dosage may be administered in a single daily dose or alternatively in two, three or more doses during the day.
  • a nucleic acid comprising a sequence encoding a peptide or protein inhibitor of glucosylceramide synthesis is administered.
  • a nucleic acid sequence encoding an agent capable of increasing the rate of neuronal glycolipid degradation e.g., a. glucosylceramide glucosidase, is administered. Any suitable methods for administering a nucleic acid sequence available in the art can be used according to the present invention.
  • the compound comprises a nucleic acid encoding a peptide or protein inhibitor of glucosylceramide synthesis or encoding an enzyme required for neuronal glycolipid degradation, such nucleic acid being part of an expression vector that expresses a the peptide or protein in a suitable host.
  • a nucleic acid has a promoter operably linked to the coding region, said promoter being inducible or constitutive (and, optionally, tissue-specific).
  • a nucleic acid molecule is used in which the coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the nucleic acid (Koller and Smithies (1989) Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al. (1989) Nature 342:435-438).
  • Delivery of the nucleic acid into a subject may be direct, in which case the subject is directly exposed to the nucleic acid or nucleic acid-carrying vector; this approach is known as in vivo gene therapy.
  • delivery of the nucleic acid into the subject may be indirect, in which case cells are first transformed with the nucleic acid in vitro and then transplanted into the subject, known as "ex vivo gene therapy”.
  • the nucleic acid is directly administered in vivo, where it is expressed to produce the encoded product.
  • This can be accomplished by any of numerous methods known in the art, e.g., by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g. , by infection using a defective or attenuated retroviral or other viral vector (see U.S. Patent No.
  • a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180 dated April 16, 1992 (Wu et al.); WO 92/22635 dated December 23, 1992 (Wilson et al); WO92/20316 dated November 26, 1992 (Findeis etal); W093/14188 dated July 22, 1993 (Clarke et al.), WO 93/20221 dated October 14, 1993 (Young)).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935; Zijlstra et al. (1989) Nature 342:435-438).
  • a viral vector that contains a nucleic acid encoding a glycolipid degrading enzyme is used, for example, a retroviral vector can be used (see Miller et al. (1993) Meth. Enzymol. 217:581-599). These retroviral vectors have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
  • the nucleic acid encoding the enzyme to be used in gene therapy is cloned into the vector, which facilitates delivery of the gene into a subject. More detail about retroviral vectors can be found in Boesen et al.
  • Adenoviruses are other viral vectors that can be used in gene therapy. Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothehal cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson (1993) Current Opinion in Genetics and Development 3:499-503 present a review of adenovirus-based gene therapy. Bout et al.
  • Another suitable approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a subject.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, and the like.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr (1993) Mefh. Enzymol. 217:599-618; Cohen et al. (1993) Meth. Enzymol.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a subject by various methods known in the art.
  • epithelial cells are injected, e.g., subcutaneously.
  • recombinant skin cells may be applied as a skin graft onto the subject; recombinant blood cells (e.g., hematopoietic stem or progenitor cells) are preferably administered intravenously.
  • recombinant blood cells e.g., hematopoietic stem or progenitor cells
  • the amount of cells envisioned for use depends on the desired effect, the condition of the subject, etc., and can be determined by one skilled in the art.
  • Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to neuronal cells, glial cells (e.g., oligodendrocytes or astrocytes), epithelial cells, endothehal cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood or fetal liver.
  • the cell used for gene therapy is autologous to the subject that is treated.
  • a nucleic acid encoding a peptide or protein inhibitor of glucosylceramide synthesis, or an agent capable of increasing the rate of neuronal glycolipid degradation is introduced into the cells such that it is expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem or progenitor cells which can be isolated and maintained in vitro can be used in accordance with this embodiment of the present invention (see e.g.
  • the nucleic acid to be introduced for purposes of gene therapy may comprise an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • Direct injection of a DNA coding for a peptide or protein inhibitor of glucosylceramide synthesis or an agent capable of increasing the rate of neuronal glycolipid degradation may also be performed according to, for example, the techniques described in United States Patent No. 5,589,466. These techniques involve the injection of "naked DNA", i.e., isolated DNA molecules in the absence of liposomes, cells, or any other material besides a suitable carrier.
  • the injection of DNA encoding a protein and operably linked to a suitable promoter results in the production of the protein in cells near the site of injection and the elicitation of an immune response in the subject to the protein encoded by the injected DNA.
  • a mucopolysaccharidosis disease is treated or prevented by administration of a compound that inhibits the expression of one or more enzymes responsible for glucosylceramide synthesis.
  • Compounds useful for this purpose may include antibodies directed to glucosylceramide synthesis enzymes (and fragments and derivatives containing the binding region thereof), and antisense or ribozyme nucleic acids.
  • an enzyme involved in neuronal glucosylceramide synthesis is inhibited by use of antisense nucleic acids.
  • the present invention provides the therapeutic or prophylactic use of nucleic acids comprising at least six nucleotides that are antisense to a gene or cDNA encoding an enzyme involved in glucosylceramide synthesis or a portion thereof.
  • an "antisense" nucleic acid refers to a nucleic acid capable of hybridizing by virtue of some sequence complementarity to a portion of an RNA (preferably mRNA) encoding an enzyme involved in glucosylceramide synthesis.
  • the antisense nucleic acid may be complementary to a coding and/or noncoding region of an mRNA encoding an enzyme involved in glucosylceramide synthesis.
  • Such antisense nucleic acids have utility as compounds that inhibit expression of an enzyme involved in glucosylceramide synthesis, and can be used in the treatment or prevention of neurological disorder.
  • the antisense nucleic acids of the invention are double-stranded or single-stranded oligonucleotides, RNA or DNA or a modification or derivative thereof, and can be directly administered to a cell or produced intracellularly by transcription of exogenous, introduced sequences.
  • the invention further provides pharmaceutical compositions comprising a therapeutically effective amount of an antisense nucleic acid that inhibits the expression of an enzyme involved in glucosylceramide synthesis, and a pharmaceutically-acceptable carrier, vehicle or diluent.
  • the antisense nucleic acids are of at least six nucleotides and are preferably oligonucleotides ranging from 6 to about 50 oligonucleotides. In specific aspects, the oligonucleotide is at least 10 nucleotides, at least 15 nucleotides, at least 100 nucleotides, or at least 200 nucleotides.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof and can be single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide may include other appended groups such as peptides; agents that facilitate transport across the cell membrane (see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Acad. Sci. 84:648-652; PCT Publication No.
  • an antisense oligonucleotide is provided, preferably of single-stranded DNA.
  • the oligonucleotide may be modified at any position on its structure with substituents generally known in the art.
  • the antisense oligonucleotide may comprise any suitable of the following modified base moieties, e.g., 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thio ⁇ ridine, 5-carboxymemylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosyl
  • the oligonucleotide comprises at least one modified sugar moiety, e.g., one of the following sugar moieties: arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the oligonucleotide comprises at least one of the following modified phosphate backbones: a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiarnidate, a methylphosphonate, an alkyl phosphotriester, a formacetal, or an analog of formacetal.
  • the oligonucleotide is an ⁇ -anomeric oligonucleotide.
  • An ⁇ -anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual, ⁇ -units, the strands run parallel to each other (Gautier et al. (1987) Nucl. Acids Res. 15:6625-6641).
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent.
  • Oligonucleotides of the invention may he synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, and the like).
  • an automated DNA synthesizer such as are commercially available from Biosearch, Applied Biosystems, and the like.
  • phosphorothioate oligonucleotides may be synthesized by the method of Stein et al. (1988) Nucl. Acids Res. 16:3209, and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al. (1988) Proc. Natl. Acad. Sci. USA 85:7448-7451).
  • the antisense nucleic acid of the invention is produced intracellularly by transcription from an exogenous sequence.
  • a vector can be introduced in vivo such that it is taken up by a cell, within which cell the vector or a portion thereof is transcribed, producing an antisense nucleic acid (RNA) of the invention.
  • RNA antisense nucleic acid
  • Such a vector would contain a sequence encoding the antisense nucleic acid.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA technology standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used for replication and expression in mammalian cells.
  • Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human, cells. Such promoters can be inducible or constitutive. Examples of such promoters are outlined above.
  • the antisense nucleic acids of the invention comprise a sequence complementary to at least a portion of an RNA transcript of a gene encoding an enzyme involved in glucosylceramide synthesis, preferably a human gene encoding an enzyme involved in glucosylceramide synthesis, however, absolute complementarity, although preferred, is not required.
  • a sequence "complementary to at least a portion of an RNA,” as referred to herein, means a sequence having sufficient complementarity to be able to hybridize under stringent conditions (e.g., highly stringent conditions comprising hybridization in 7% sodium dodecyl sulfate (SDS), 1 M EDTA at 65 °C and washing in O.lxSSC/0.1% SDS at 68°C, or moderately stringent conditions comprising washing in 0.2xSSC/0.1% SDS at 42°C with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • stringent conditions e.g., highly stringent conditions comprising hybridization in 7% sodium dodecyl sulfate (SDS), 1 M EDTA at 65 °C and washing in O.lxSSC/0.1% SDS at 68°C, or
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA encoding an enzyme involved in glucosylceramide synthesis it may contain and still form a stable duplex (or triplex, as the case may be).
  • One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
  • compositions of the invention comprising an effective amount of an antisense nucleic acid of the invention in a pharmaceutically acceptable carrier, vehicle or diluent can be administered to a subject having neurological disorder.
  • the amount of antisense nucleic acid which will be effective in the treatment of a neurological disorder can be determined by standard clinical techniques.
  • compositions comprising one or more antisense nucleic acids to an enzyme involved in glucosylceramide synthesis are administered via liposomes, microparticles, or microcapsules.
  • such compositions may be used to achieve sustained release of the antisense nucleic acids.
  • symptoms of MPS disease may be ameliorated by decreasing the level of an enzyme involved in glucosylceramide synthesis by using gene sequences encoding the an enzyme involved in glucosylceramide synthesis in conjunction with well-known gene "knock-out,” ribozyme or triple helix methods to decrease gene expression of an enzyme involved in glucosylceramide synthesis.
  • ribozyme or triple helix molecules are used to modulate the activity, expression or synthesis of the gene encoding the enzyme involved in glucosylceramide synthesis, and thus to ameliorate the symptoms of the disorder.
  • Such molecules may be designed to reduce or inhibit expression of a mutant or non-mutant target gene. Techniques for the production and use of such molecules are well known to those of skill in the art.
  • Ribozyme molecules designed to catalytically cleave gene mRNA transcripts , encoding an enzyme involved in glucosylceramide synthesis can be used to prevent translation of target gene mRNA and, therefore, expression of the gene product.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage event.
  • the composition of ribozyme molecules must include one or more sequences complementary to the target gene mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see, e.g., U.S. Patent No. 5,093,246, which is incorporated herein by reference in its entirety.
  • ribozymes that cleave mRNA at site specific recognition sequences can be used to destroy mRNAs encoding an enzyme involved in glucosylceramide synthesis, the use of hammerhead ribozymes is preferred.
  • Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA have the following sequence of two bases: 5'-UG-3'.
  • the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the mRNA encoding the enzyme involved in glucosylceramide synthesis, i.e., to increase efficiency and minimize the intracellular accumulation of non-functional mRNA transcripts.
  • the ribozymes of the present invention also include RNA endoribonucleases (hereinafter "Cech-type ribozymes”) such as the one that occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and that has been extensively described by Thomas Cech and collaborators (Zaug, et al. (1984) Science, 224, 574-578; Zaug and Cech (1986) Science, 231, 470-475; Zaug, et al. (1986) Nature, 324, 429-433; published International patent application No. WO 88/04300 by University Patents Inc.; Been and Cech
  • the Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA sequence whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in the gene encoding the enzyme involved in glucosylceramide synthesis.
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.) and should be delivered to cells that express the enzyme involved in glucosylceramide synthesis in vivo.
  • a preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol HI or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous mRNA encoding the enzyme involved in glucosylceramide synthesis and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficacy.
  • Endogenous expression of an enzyme involved in glucosylceramide synthesis can also be reduced by inactivating or "knocking out" the gene encoding an enzyme involved in glucosylceramide synthesis, or the promoter of such a gene, using targeted homologous recombination (e.g., see Smithies et al. 1985) Nature 317:230-234; Thomas and Capecchi
  • a mutant gene encoding a non-functional an enzyme involved in glucosylceramide synthesis (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous gene (either the coding regions or regulatory regions of the gene encoding an enzyme involved in glucosylceramide synthesis) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express the target gene in vivo.
  • Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive target gene.
  • ES embryonic stem
  • this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors.
  • the endogenous expression of a gene encoding an enzyme involved in glucosylceramide synthesis can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the gene (i.e., the gene promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene encoding an enzyme involved in glucosylceramide synthesis in target cells in the body.
  • deoxyribonucleotide sequences complementary to the regulatory region of the gene i.e., the gene promoter and/or enhancers
  • triple helical structures that prevent transcription of the gene encoding an enzyme involved in glucosylceramide synthesis in target cells in the body.
  • Nucleic acid molecules to be used in triplex helix formation for the inhibition of transcription in the present invention should be single stranded and composed of deoxynucleotides.
  • the base composition of these oligonucleotides must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC+ triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand, i addition, nucleic acid molecules may be chosen that are purine-rich, for example, contain a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with frst one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • the technique may so efficiently reduce or inhibit the transcription (triple helix) or translation (antisense, ribozyme) of mRNA produced by normal gene alleles of an enzyme involved in glucosylceramide synthesis that the situation may arise wherein the concentration of such an enzyme involved in glucosylceramide synthesis present may be lower than is necessary for a normal phenotype.
  • gene therapy may be used to introduce into cells nucleic acid molecules that encode and express an enzyme involved in glucosylceramide synthesis that exhibit normal gene activity and that do not contain sequences susceptible to whatever antisense, ribozyme, or triple helix treatments are being utilized.
  • a normal enzyme can be co-administered in order to maintain the requisite level of activity.
  • Antisense RNA and DNA, ribozyme, and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules, as discussed above. These include techniques for chemically synthesizing oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
  • a mucopolysaccharidosis disease is treated or prevented by administration of one or more antibodies that inhibit the expression of one or more enzymes responsible for glucosylceramide synthesis.
  • Any of the enzymes required for glucosylceramide synthesis, or a fragment or derivative thereof, may be used as an immunogen to generate antibodies which immunospecifically bind such an immunogen.
  • Such immunogens can be isolated by any convenient means, including the methods described above.
  • Antibodies of the invention include, but are not limited to polyclonal, monoclonal, bispecific, humanized or chimeric antibodies, single chain antibodies, Fab fragments and F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen.
  • the immunoglobulin molecules of the invention can be of any class (e.g., IgG, IgE, IgM, IgD and IgA ) or subclass of immunoglobulin molecule.
  • antibodies that recognize an enzyme required for glucosylceramide synthesis may be prepared.
  • antibodies may be prepared that recognize glucosylceramide glucosyltransferase, glucosylceramide synthase, etc., Alternatively, such antibodies may be purchased from commercial sources where available.
  • methods known to those skilled in the art are used to produce antibodies that recognize an enzyme required for glucosylceramide synthesis.
  • ELISA enzyme-linked immunosorbent assay
  • Polyclonal antibodies which may be used in the methods of the invention are heterogeneous populations of antibody molecules derived from the sera of immunized animals. Unfractionated immune serum can also be used.
  • Various procedures known in the art may be used for the production of polyclonal antibodies to a protein enzyme required for glucosylceramide synthesis, or a peptide or fragment thereof.
  • rabbit polyclonal antibodies to an epitope of such an immunogen polypeptide can be obtained.
  • various host animals can be immunized by injection with the native or a synthetic (e.g., recombinant) version of an enzyme required for glucosylceramide synthesis, or peptide or fragment thereof, including but not limited to rabbits, mice, rats, etc.
  • Isolated enzymes suitable for such immunization may be obtained by the use of discovery techniques, such as the preferred technology described herein. If the enzyme is purified by gel electrophoresis, the enzyme can be used for immunization with or without prior extraction from the polyacrylamide gel.
  • adjuvants may be used to enhance the immunological response, depending on the host species, including, but not limited to, complete or incomplete Freund's adjuvant, a mineral gel such as aluminum hydroxide, surface active substance such as lysolecithin, pluronic polyol, a polyanion, a peptide, an oil emulsion, keyhole limpet hemocyanin, dinitrophenol, and an adjuvant such as BCG (bacille Calmette-Guerin) or corynebacterium parvum. Additional adjuvants are also well known in the art.
  • any technique which provides for the production of antibody molecules by continuous cell lines in culture may be used.
  • the hybridoma technique originally developed by Kohler and Milstein (1975) Nature 256:495-497) as well as the trioma technique, the human B-cell hybridoma technique (Kozbor et al. (1983) Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al. (1985) in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof.
  • the hybridoma producing the mAbs of the invention may be cultivated in vitro or in vivo.
  • monoclonal antibodies can be produced in germ-free animals utilizing known technology (PCT/US90/02545, incorporated herein by reference).
  • the monoclonal antibodies include but are not limited to human monoclonal antibodies and chimeric monoclonal antibodies (e.g., human-mouse chimeras).
  • Humanized antibodies are antibody molecules from non-human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule. (See, e.g., Queen, U.S. Patent No. 5,585,089, which is incorporated herein by reference in its entirety.)
  • Chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671 ; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No. WO 86/01533; U.S. Patent No. 4,816,567; European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043; Liu et al. (1987) Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al. (1987) J. Immunol.
  • Completely human antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Such antibodies can be produced using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes.
  • the transgenic mice are immunized in the normal fashion with selected antigens, e.g., all or a portion of glucosylceramide synthase.
  • Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies.
  • Completely human antibodies that recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a. mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et ⁇ l. (1994) Biotechnology 12:899-903).
  • the antibodies of the present invention can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene HI or gene VHI protein.
  • Phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et ⁇ l. (1995) J. Immunol. Methods 182:41-50; Ames et al. (1995) J. Immunol. Methods 184:177-186; Kettleborough et al. (1994) Eur. J. Immunol. 24:952-958; Persic et ⁇ l.
  • the invention further provides for the use of bispecific antibodies, which can be made by methods known in the art.
  • Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Milstein et al. (1983) Nature 305:537-539). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al. (1991) EMBO J. 10:3655-3659 .
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation.
  • This approach is disclosed in WO 94/04690 published March 3,1994.
  • For further details for generating bispecific antibodies see, for example, Suresh et al. (1986) Methods in Enzymology 121:210.
  • the invention provides functionally active fragments, derivatives or analogs of the specific immunoglobulin molecules.
  • Functionally active means that the fragment, derivative or analog is able to elicit anti-anti-idiotype antibodies (i.e., tertiary antibodies) that recognize the same antigen that is recognized by the antibody from wliich the fragment, derivative or analog is derived.
  • antigenicity of the idiotype of the immunoglobulin molecule may be enhanced by deletion of framework and CDR sequences that are C-terminal to the CDR sequence that specifically recognizes the antigen.
  • synthetic peptides containing the CDR sequences can be used in binding assays with the antigen by any suitable binding assay known in the art.
  • the present invention provides antibody fragments such as, but not limited to, F(ab')2 fragments and Fab fragments. Antibody fragments wliich recognize specific epitopes may be generated by known techniques. F(ab')2 fragments consist of the variable region, the light chain constant region and the CHI domain of the heavy chain and are generated by pepsin digestion of the antibody molecule. Fab fragments are generated by reducing the disulfide bridges of the F(ab')2 fragments. The invention also provides heavy chain and light chain dimers of the antibodies of the invention, or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs) (e.g., as described in U.S. Patent No.
  • SCAs single chain antibodies
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may be used (Skerra et al. (1988) Science 242:1038-1041).
  • the invention provides fusion proteins of the immunoglobulins of the invention (or functionally active fragments thereof), for example in which the immunoglobulin is fused via a covalent bond (e.g., a peptide bond), at either the N-terminus or the C-terminus to an amino acid sequence of another protein (or portion thereof, preferably at least 10, 20 or 50 amino acid portion of the protein) that is not the immunoglobulin.
  • a covalent bond e.g., a peptide bond
  • the immunoglobulin, or fragment thereof is covalently linked to the other protein at the N-terminus of the constant domain.
  • such fusion proteins may facilitate purification, increase half-life in vivo, and enhance the delivery of an antigen across an epithelial barrier to the immune system.
  • the immunoglobulins of the invention include analogs and derivatives that are either modified, i.e, by the covalent attachment of any type of molecule as long as such covalent attachment that does not impair immunospecific binding.
  • the derivatives and analogs of the immunoglobulins include those that have been further modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, or the like. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, etc. Additionally, the analog or derivative may contain one or more non-classical or unnatural amino acids.
  • a murine model of MPS IDA (Sanfilippo disease) has been described that demonstrates the disorder's characteristic joint and skeletal storage of proteoglycan fragments, and neuronal storage of GM2 and GM3 gangliosides. Colonies of mutant mice expressing the MPS TTTA phenotype have been described, and have been validated by a number of criteria as an authentic model of the disease (Stanley, P et al. (1999) Glycobiology 9: 1389 - 1396).
  • MPS IIIA mice display clinical signs of the disease around 6 months of age with decreased activity, scruffy coat, abdominal distention, hunched posture and waddling gait. By 12 months, the mice exhibit severe ataxia, tremors and weight loss. Death results by 18 months or less.
  • the brains of MPS mice are grossly normal. However, microscopic examination reveals swollen somata, meganeurite formation and enlarged axon hillock regions of cortical pyramidal neurons.
  • White matter and Purkinje cells display axonal spheroids.
  • Anti-heparan sulfate immunostaining indicates accumulation of heparan sulfate fragments in the cerebral cortex of MPS IIIA animals.
  • Anti-ganglioside antibody staining shows increased GM2 and GM3 levels primarily in laminae II III and V pyramidal neurons, and in other neurones of the central nervous system.
  • FIG. 1 shows photomicrographs of: A: Untreated control, i. Upper cortex, ii. Lower cortex; B: NB-DNJ Treated, i. Upper cortex, ii. Lower cortex.
  • FIG. 1 shows photomicrographs of: A: Untreated control; B: NB-DNJ Treated.
  • FIG. 4 shows photomicrographs of: A; Untreated control; B: NB-DNJ Treated.
  • glycolipid synthesis inhibitors provide a useful treatment option for the pathologies resulting from glycolipid accumulation in MPS-affected brains.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physical Education & Sports Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne l'utilisation d'inhibiteurs de synthèse de glycosylcéramides dans le traitement des mucopolysaccharidoses.
PCT/US2002/000813 1999-04-20 2002-01-11 Traitement des mucopolysaccharidoses WO2002055064A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2002241853A AU2002241853A1 (en) 2001-01-12 2002-01-11 Mucopolysaccharidosis therapies
US10/619,378 US20050032841A1 (en) 1999-04-20 2003-07-14 Therapeutic compositions and methods of treating glycolipid storage related disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0100889.5 2001-01-12
GBGB0100889.5A GB0100889D0 (en) 2001-01-12 2001-01-12 Compounds
US34723302P 2002-01-10 2002-01-10
US60/347,233 2002-01-10

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/619,378 Continuation US20050032841A1 (en) 1999-04-20 2003-07-14 Therapeutic compositions and methods of treating glycolipid storage related disorders

Publications (2)

Publication Number Publication Date
WO2002055064A2 true WO2002055064A2 (fr) 2002-07-18
WO2002055064A3 WO2002055064A3 (fr) 2002-09-26

Family

ID=26245554

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/000813 WO2002055064A2 (fr) 1999-04-20 2002-01-11 Traitement des mucopolysaccharidoses

Country Status (2)

Country Link
AU (1) AU2002241853A1 (fr)
WO (1) WO2002055064A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT202200007808A1 (it) 2022-04-20 2023-10-20 Luigi Michele Pavone Composizioni terapeutiche per malattie causate da accumulo di eparan solfato

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000062779A1 (fr) * 1999-04-20 2000-10-26 Oxford Glycosciences (Uk) Limited Combinaison d'inhibiteurs de synthese de glucosylceramide et d'enzyme degradant les glycolipides utilisee en therapie
WO2001097829A2 (fr) * 2000-06-19 2001-12-27 Genzyme Corporation Traitement de substitution enzymatique, therapie genique et therapie par petites molecules pour maladies liees au stockage lysosomal

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000062779A1 (fr) * 1999-04-20 2000-10-26 Oxford Glycosciences (Uk) Limited Combinaison d'inhibiteurs de synthese de glucosylceramide et d'enzyme degradant les glycolipides utilisee en therapie
WO2001097829A2 (fr) * 2000-06-19 2001-12-27 Genzyme Corporation Traitement de substitution enzymatique, therapie genique et therapie par petites molecules pour maladies liees au stockage lysosomal

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PLATT F M ET AL: "N-BUTYLDEOXYNOJIRIMYCIN IS A NOVEL INHIBITOR OF GLYCOLIPID BIOSYNTHESIS" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD, US, vol. 269, no. 11, 18 March 1994 (1994-03-18), pages 8362-8365, XP000615445 ISSN: 0021-9258 *
PLATT F M ET AL: "NEW THERAPEUTIC PROSPECTS FOR THE GLYCOSPHINGOLIPID LYSOSOMAL STORAGE DISEASES" BIOCHEMICAL PHARMACOLOGY, PERGAMON, OXFORD, GB, vol. 56, no. 4, 1998, pages 421-430, XP000886851 ISSN: 0006-2952 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT202200007808A1 (it) 2022-04-20 2023-10-20 Luigi Michele Pavone Composizioni terapeutiche per malattie causate da accumulo di eparan solfato
WO2023203004A1 (fr) 2022-04-20 2023-10-26 Pavone Luigi Michele Compositions thérapeutiques avec des iminosucres pour le traitement de maladies d'accumulation du sulfate d'héparane

Also Published As

Publication number Publication date
AU2002241853A1 (en) 2002-07-24
WO2002055064A3 (fr) 2002-09-26

Similar Documents

Publication Publication Date Title
US6683076B2 (en) Methods for therapeutic use of glucosylceramide synthesis inhibitors and composition thereof
US7348000B2 (en) Therapeutic compositions and methods of treating glycolipid storage related disorders
EP3028716B1 (fr) Inhibition du complément pour régénération nerveuse améliorée
RU2566724C9 (ru) Композиции и способы модуляции smn2 сплайсинга у субъекта
SA517390168B1 (ar) تركيبات لتعديل التعبير الوراثي عن c9orf72
US20200101042A1 (en) 4-methylumbelliferone treatment for immune modulation
US20090038022A1 (en) IGF-1 Novel peptides
EP3449001B1 (fr) Inhibition de l'arnmi mir-22 par apt-110
JP2018526368A (ja) Ertナイーブ患者及びert経験患者におけるファブリー病の処置
MX2011010401A (es) Un metodo para regular la proliferacion y deferenciacion de queratinocitos.
JP2021505611A (ja) 免疫疾患の予防または治療のためのripk1阻害剤とikk阻害剤の組み合わせ
EP3656386A1 (fr) Composés et méthodes de modulation de l'expression de l'alipoprotéine c-iii pour améliorer le profil diabétique
Amo-Aparicio et al. Extracellular and nuclear roles of IL-37 after spinal cord injury
Hu et al. A20 inhibits intraocular inflammation in mice by regulating the function of CD4+ T cells and RPE cells
WO2012033518A1 (fr) Procédés et compositions pour le traitement de troubles métaboliques
Martín et al. Administration of a peptide inhibitor of α4-integrin inhibits the development of experimental autoimmune uveitis
US20050032841A1 (en) Therapeutic compositions and methods of treating glycolipid storage related disorders
WO2002055064A2 (fr) Traitement des mucopolysaccharidoses
JP2021506794A (ja) 疼痛を治療し、疼痛感受性を増大させるためのペプチド及び他の薬剤
Mich et al. AAV-mediated interneuron-specific gene replacement for Dravet syndrome
WO2020237803A1 (fr) Utilisation de la protéine ou du gène mrg15 comme cible dans le traitement et la prévention de maladies métaboliques
CA2543305A1 (fr) Desoxyoligonucleotides antisens ache utilises comme agents anti-inflammatoires
KR20200122601A (ko) Cd9을 이용한 퇴행성 신경질환의 예방 또는 치료용 조성물과 퇴행성 신경질환 치료제 스크리닝 방법
JP2021534778A (ja) フィードバック使用可能合成遺伝子、標的シードマッチカセット、およびその使用
US20110034381A1 (en) Methods for Therapeutic Treatment of Benign Prostatic Hypertrophy (BPH)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP