EP1078075A2 - (alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci - Google Patents

(alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci

Info

Publication number
EP1078075A2
EP1078075A2 EP99924155A EP99924155A EP1078075A2 EP 1078075 A2 EP1078075 A2 EP 1078075A2 EP 99924155 A EP99924155 A EP 99924155A EP 99924155 A EP99924155 A EP 99924155A EP 1078075 A2 EP1078075 A2 EP 1078075A2
Authority
EP
European Patent Office
Prior art keywords
iduronidase
cell line
recombinant
enzyme
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP99924155A
Other languages
German (de)
English (en)
Inventor
Emil D. Kakkis
Becky Tanamachi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harbor-Ucla Research and Education Institute
Harbor UCLA Research and Education Institute
Original Assignee
Harbor-Ucla Research and Education Institute
Harbor UCLA Research and Education Institute
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 Harbor-Ucla Research and Education Institute, Harbor UCLA Research and Education Institute filed Critical Harbor-Ucla Research and Education Institute
Publication of EP1078075A2 publication Critical patent/EP1078075A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01076L-Iduronidase (3.2.1.76)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present mvention is in the field of molecular biology, enzymology, biochemistry and clinical medicine.
  • the present invention provides a recombmant ⁇ -L-iduronidase, methods to produce and purify this enzyme as well as methods to treat certain genetic disorders including ⁇ -L-iduronidase deficiency and mucopolysaccha ⁇ dosis I (MPS I).
  • MPS I mucopolysaccha ⁇ dosis I
  • Carbohydrates play a number of important roles in the functioning of living organisms.
  • carbohydrates are structural components of the human body covalently attached to numerous other entities such as proteins and hpids (called glycoconjugates)
  • proteins and hpids called glycoconjugates
  • human connective tissues and cell membranes comprise proteins, carbohydrates and a proteoglycan matrix.
  • the carbohydrate portion of this proteoglycan matrix provides important properties to the body's structure.
  • a genetic deficiency of the carbohydrate-cleaving, lysosomal enzyme ⁇ -L-iduronidase causes a lysosomal storage disorder known as mucopolysaccha ⁇ dosis I (MPS I) (Neufeld, E. F., and Muenzer, J. (1989).
  • MPS I mucopolysaccha ⁇ dosis I
  • MPS I is commonly known as Hurler syndrome and is associated with multiple problems such as mental retardation, clouding of the cornea, coarsened facial features, cardiac disease, respiratory disease, liver and spleen enlargement, hernias, and joint stiffness Patients suffering, from Hurler syndrome usually die before age 10.
  • Hurler-Scheie syndrome mental function is generally not severely affected, but physical problems may lead to death by the teens or twenties.
  • Scheie syndrome is the mildest form of MPS I. It is compatible with a normal life span, but joint stiffness, corneal clouding and heart valve disease cause significant problems.
  • the frequency of MPS I is estimated to be 1:100,000 according to a British Columbia survey of all newborns (Lowry et al , Human Genetics 85:389-390 (1990)) and 1:70,000 according to an Irish study (Nelson, Human Genetics 101:355-358 (1990)). There appears to be no ethnic predilection for this disease It is likely that worldwide the disease is underdiagnosed either because the patient dies of a complication before the diagnosis is made or because the milder forms of the syndrome may be mistaken for arthritis or missed entirely. Effective newborn screening for MPS I would likely find some previously undetected patients.
  • Bone marrow transplants can be effective in treating some of the symptoms of the disorder but have high morbidity and mortality in MPS I and often are not available to patients because of a lack of suitable donors. An alternative therapy available to all affected patients would provide an important breakthrough in treating and managing this disease.
  • Enzyme replacement therapy has long been considered a potential therapy for MPS I following the discovery that ⁇ -L-iduronidase can correct the enzymatic defect in Hurler cells in culture.
  • the enzyme containing a mannose-6-phosphate residue is taken up into cells through receptor-mediated endocytosis and transported to the lysosomes where it clears the stored substrates, heparan sulfate and dermatan sulfate.
  • Application of this therapy to humans has previously not been possible due to inadequate sources of ⁇ -L-iduronidase in tissues.
  • the enzyme replacement concept was first effectively applied to Gaucher patients in a modified placental glucocerebrosidase. The delivery and effective uptake of glucocerebrosidase in Gaucher patients demonstrated that an enzyme could be taken up in vivo in sufficient quantities to provide effective therapy.
  • FIGURE 1 represents the nucleotide and deduced amino acid sequences of cDNA encoding ⁇ -L-iduronidase. Nucleotides 1 through 6200 are provided. Amino acids are provided starting with the first methionine in the open reading frame.
  • FIGURE 2 represents the results from an SDS-PAGE run of eluate obtained according to the procedure set forth in Example 1.
  • Lane 1 is blank.
  • Lane 2 contained high molecular weight standards.
  • Lane 3 is a blank.
  • Lane 4 contained bovine serum albumin in a concentration of 50 ⁇ g.
  • Lanes 5 through 10 represent eluate containing recombinantly produced human ⁇ -L-iduronidase in amounts of 1 ⁇ g, 2 ⁇ g, 5 ⁇ g, 5 ⁇ g, 5 ⁇ g and 5 ⁇ g, respectively.
  • FIGURE 3 reveals the urinary GAG levels in 16 MPS I patients in relation to normal excretion values. There is a wide range of urine GAG values in untreated MPS I patients. A greater than 50% reduction in excretion of undegraded GAGs following therapy with recombinant ⁇ -L-iduronidase is a valid means to measure an individual's response to therapy.
  • FIGURE 4 demonstrates leukocyte iduronidase activity before and after enzyme therapy in MPS I patients.
  • FIGURE 5 demonstrates the buccal iduronidase activity before and after enzyme therapy.
  • FIGURE 6 demonstrates in three patients that a substantial shrinkage of liver and spleen together with significant clinical improvement in joint and soft tissue storage was associated with a greater than 65% reduction in undegraded GAG after only 8 weeks of treatment with recombinant enzyme.
  • FIGURE 7 demonstrates that there is substantial normalization of livers and spleens in patients treated with recombinant enzyme after only 12 weeks of therapy.
  • FIGURE 8 demonstrates a precipitous drop in urinary GAG excretion over 22 weeks of therapy with recombinant enzyme in 6 patients.
  • the present invention features a method to produce ⁇ -L-iduronidase in amounts which enable using the enzyme therapeutically.
  • the method comprises the step of transfecting a cDNA encoding for all or a part of an ⁇ -L-iduronidase into a cell suitable for the expression thereof.
  • a cDNA encoding for a complete ⁇ -L-iduronidase is used, preferably a human ⁇ -L-iduronidase.
  • a cDNA encoding for a biologically active fragment or mutant thereof may be used. Specifically, one or more amino acid substitutions may be made while preserving or enhancing the biological activity of the enzyme.
  • an expression vector is used to transfer the cDNA into a suitable cell or cell line for expression thereof.
  • the cDNA is transfected into a Chinese hamster ovary cell to create cell line 2.131.
  • the production procedure features one or more of the following characteristics which have demonstrated particularly high production levels: (a) the pH of the cell growth culture may be lowered to about 6.5 to 7.0, preferably to about 6.7-6.8 during the production process, (b) about 2/3 to 3/4 of the medium may be changed approximately every 12 hours, (c) oxygen saturation may be optimized at about 80% using intermittent pure oxygen sparging, (d) microcarriers with about 10% serum initially may be used to produce cell mass followed by a rapid washout shift to protein-free medium for production, (e) a protein-free or low protein medium such as a JRH Biosciences PF-CHO product may be optimized to include supplemental amounts of one or more ingredients selected from the group consisting of glutamate, aspartate, glycine
  • the present invention provides a transfected cell line which features the ability to produce ⁇ -L-iduronidase in amounts which enable using the enzyme therapeutically.
  • the present invention features a recombinant Chinese hamster ovary cell line such as the 2.131 cell line that stably and reliably produces amounts of ⁇ -L-iduronidase which enable using the enzyme therapeutically.
  • the cell line may contain at least about 10 copies of a an expression construct.
  • the cell line expresses recombinant ⁇ -L-iduronidase in amounts of at least about 20-40 micrograms per 10 7 cells per day.
  • the present invention provides novel vectors suitable to produce ⁇ -L-iduronidase in amounts which enable using the enzyme therapeutically.
  • the present invention features an expression vector comprising a cytomegalovirus promoter/enhancer element, a 5' intron consisting of a murine Ca intron, a cDNA encoding all or a fragment or mutant of an ⁇ -L-iduronidase, and a 3' bovine growth hormone polyadenylation site.
  • the cDNA encoding all or a fragment or mutant of an ⁇ -L-iduronidase is about 2.2 kb in length.
  • This expression vector may be transfected at, for example, a 50 to 1 ratio with any appropriate common selection vector such as, for example, pSV2NEO, to enhance multiple copy insertions.
  • any appropriate common selection vector such as, for example, pSV2NEO
  • gene amplification may be used to induce multiple copy insertions.
  • the present invention provides novel ⁇ -L-iduronidase produced in accordance with the methods of the present invention and thereby present in amounts which enable using the enzyme therapeutically.
  • the specific activity of the ⁇ -L-iduronidase according to the present invention is in excess of 200,000 units per milligram protein. Preferably, it is in excess of about 240,000 units per milligram protein.
  • the molecular weight of the ⁇ -L-iduronidase of the present invention is about 82,000 daltons, about 70,000 daltons being amino acid and about 12,000 daltons being carbohydrates.
  • the present invention features a novel method to purify ⁇ -L-iduronidase.
  • a cell mass may be grown in about 10% serum containing medium followed by a switch to a modified protein-free production medium without any significant adaptation to produce a high specific activity starting material for purification.
  • a concentration/diafiltration scheme is employed that allows for the removal of exogenous materials that may be required for recombinant production of the same such as, for example, Pluronics F-68, a commonly used surfactant for protecting cells from sparging damage.
  • exogenous materials should normally be separated from the crude bulk to prevent fouling of the columns.
  • a first column load is acidified to minimize the competitive inhibition effect of uronic acids found in protein-free medium formulations.
  • a heparin, phenyl and sizing column purification scheme is used to produce pure enzyme using automatable steps and validatable media.
  • the heparin and phenyl column steps are used to eliminate less desirable ⁇ -L-iduronidase that is nicked or degraded.
  • an acid pH treatment step is used to inactivate potential viruses without harming the enzyme.
  • the present invention features novel methods of treating diseases caused all or in part by a deficiency in ⁇ -L-iduronidase.
  • this method features administering a recombinant ⁇ -L-iduronidase or a biologically active fragment or mutant thereof alone or in combination with a pharmaceutically suitable carrier.
  • this method features transferring a nucleic acid encoding all or a part of an ⁇ -L-iduronidase into one or more host cells in vivo.
  • Preferred embodiments include optimizing the dosage to the needs of the organism to be treated, preferably mammals or humans, to effectively ameliorate the disease symptoms.
  • the disease is mucopolysaccharidosis I (MPS I), Hurler syndrome, Hurler-Scheie syndrome or Scheie syndrome.
  • the present mvention features novel pharmaceutical compositions comprising ⁇ -L-iduronidase useful for treating a disease caused all or in part by a deficiency in ⁇ -L- iduronidase.
  • Such compositions may be suitable for administration in a number of ways such as parenteral, topical, intranasal, inhalation or oral administration.
  • embodiments featuring nucleic acid sequences encoding all or a part of an ⁇ -L-iduronidase which may be administered in vivo into cells affected with an ⁇ -L-iduronidase deficiency.
  • the present invention features a method to produce ⁇ -L-iduronidase in amounts which enable using the enzyme therapeutically.
  • the method features transforming a suitable cell line with the cDNA encoding for all of ⁇ -L-iduronidase or a biologically active fragment or mutant thereof.
  • Those of skill in the art may prepare expression constructs other than those expressly described herein for optimal production of ⁇ -L-iduronidase in suitable cell lines transfected therewith.
  • skilled artisans may easily design fragments of cDNA encoding biologically active fragments and mutants of the naturally occurring ⁇ -L-iduronidase which possess the same or similar biological activity to the naturally occurring full-length enzyme.
  • a large series of expression vectors may be constructed and tested for expression of a ⁇ -L-iduronidase cDNA. Based on transient transfection experiments as well as stable transfections, an expression construct may be identified that provides particularly high level expression.
  • a Chinese hamster cell line 2.131 developed by transfection of the ⁇ -L-iduronidase expression construct and selection for a high expression clone provides particularly high level expression.
  • Such a Chinese hamster cell line according to this embodiment of the present invention may secrete about 5,000 to 7,000 fold more ⁇ -L-iduronidase than normal.
  • the ⁇ -L-iduronidase produced thereby may be properly processed, taken up into cells with high affinity and is corrective for ⁇ -L-iduronidase deficient cells such as those from patients suffering from Hurler's Syndrome.
  • microcarriers are used as a low cost scalable surface on which to grow adherent cells.
  • a culture system is optimized.
  • the culture pH is lowered to about 6.5 to 7.0, preferably to about 6.7-6.8 during the production process.
  • One advantage of such a pH is to enhance accumulation of lysosomal enzymes that are more stable at acidic pH.
  • about 2/3 to 3/4 of the medium is changed approximately every 12 hours.
  • oxygen saturation is optimized at about 80% using intermittent pure oxygen sparging rather than continuous sparging.
  • cytodex 2 microcarriers with about 10% serum initially are used to produce a cell mass followed by a rapid washout shift to a protein-free medium for production.
  • a growth medium such as a JRH Biosciences PF-CHO product may be optimized to include supplemental amounts of one or more ingredients selected from the group consisting of glutamate, aspartate, glycine, ribonucleosides and deoxyribonucleosides.
  • a perfusion wand such as a Bellco perfusion wand may be used in a frequent batch-feed process rather than a standard intended perfusion process.
  • a mild sodium butyrate induction process may be used to induce increased ⁇ -L-iduronidase expression without a substantial effect on the carbohydrate processing and cellular uptake of the enzyme.
  • Such an induction process may provide about a two-fold increase in production without significantly altering post-translational processing.
  • Particularly preferred embodiments of the method for producing ⁇ -L-iduronidase according to the present invention feature one, more than one or all of the optimizations described herein
  • the production method of the present mvention may therefore provide a production culture process having the following features:
  • a microcarrier based culture using Cytodex 2 beads or an equivalent thereof is preferably used in large scale culture flasks with overhead wand stir ⁇ ng using a Bellco perfusion wand or an equivalent thereof. Attachment to these beads may be achieved by culture in a 10% fetal bovme serum in DME F12 1:1 medium modified with ingredients including ribonucleosides, deoxyribonucleosides, pyruvate, non-essential ammo acids, and HEPES and at a pH of about 6.7-6.9.
  • the culture conditions are preferably maintained at a dissolved oxygen of 80% of air saturation at a pH of about 6.7 and at a temperature of about 37° C
  • a dissolved oxygen of 80% of air saturation at a pH of about 6.7 and at a temperature of about 37° C
  • An air saturation of about 80% results in improved ⁇ -L- lduromdase secretion over 40% and 60% air saturation.
  • 90% air saturation does not provide significantly enhanced secretion over 80% air saturation.
  • the dissolved oxygen may be supplied by intermittent pure oxygen sparging using a 5 micron stainless steel sparger or equivalent thereof.
  • a pH of about 6.7 is optimal for the accumulation of the ⁇ -L-iduronidase enzyme.
  • the enzyme is particularly unstable at pH's above about 7.0 Below a pH of about 6.7, the secretion rate may decrease, particularly below a pH of about 6.5.
  • the culture is therefore maintained optimally between a pH of about 6.6 to 6.8 .
  • the production culture medium may be a modified form of the commercially available proprietary medium from JRH Biosciences called Excell PF CHO. This medium supports levels of secretion equivalent to that of serum using a cell line such as the 2.131 cell line. It may be preferably modified to include an acidic pH of about 6.7 (+/- 0. 1), and it may be buffered with HEPES at 7.5 mM.
  • the medium may contain 0.05 to 0.1% of Pluronics F-68 (BASF), a non-ionic surfactant or an equivalent thereof which features the advantage of protecting cells from shear forces associated with sparging.
  • BASF Pluronics F-68
  • the medium may further contain a proprietary supplement that proves to be important in increasing the productivity of the medium over other protem-free mediums that are presently available.
  • BASF Pluronics F-68
  • the medium may further contain a proprietary supplement that proves to be important in increasing the productivity of the medium over other protem-free mediums that are presently available.
  • the production medium may be analyzed using an amino acid analyzer comparing spent medium with starting medium. Such analyses have demonstrated that the 2.131 cell line depletes a standard PF CHO medium of glycine, glutamate and aspartate to a level of around 10% of the starting concentration. Supplementation of these amino acids to higher levels may result in enhanced culture density and productivity that may lead to a 2-3 fold higher production than at baseline. Skilled artisans will appreciate that other cell lines within the scope of the present invention may be equally useful for producing ⁇ -L-iduronidase according to the present method. Hence, more or less supplemental nutrients may be required to optimize the medium. Such optimizations are intended to be within the scope of the present invention and may be practiced without undue experimentation.
  • the medium may be supplemented with ribonucleosides and deoxyribonucleosides to support the dihydrofolate reductase deficient cell line 2.131.
  • Skilled artisans will appreciate that other cell lines within the scope of the present invention may be equally useful for producing ⁇ -L- iduronidase according to the present method. Hence, more or less ribonucleosides and deoxyribonucleosides may be required to optimize the medium. Such optimizations are intended within the scope of the present invention and may be practiced without undue experimentation.
  • approximately 2/3 of the medium may be changed out approximately every 12 hours.
  • the change out of medium may be accomplished using, for instance, a Bellco perfusion wand which is a stirring device with a hollow center and screen filter at its tip. By pumping out medium through the hollow interior of the wand through the 40 micron screen.
  • the microcarriers with the 2.131 cell mass are separated from supernatant containing the enzyme.
  • a 15 liter culture system may produce approximately 25 mg per liter of culture per day, or more at peak culturing density.
  • the present invention provides a transfected cell line which possesses the unique ability to produce ⁇ -L-iduronidase in amounts which enable using the enzyme therapeutically.
  • the present invention features a recombinant Chinese hamster ovary cell line such as the 2.131 cell line that stably and reliably produces amounts of ⁇ -L-iduronidase.
  • the cell line may contain at least about 10 copies of an expression construct comprising a CMV promoter, a Ca intron, a human ⁇ -L-iduronidase cDNA, and a bovine growth hormone polyadenylation sequence.
  • the cell line expresses ⁇ - L-iduronidase at amounts of at least about 20-40 micrograms per 10 7 cells per day in a properly processed, high uptake form appropriate for enzyme replacement therapy.
  • the transfected cell line adapted to produce ⁇ -L- iduronidase in amounts which enable using the enzyme therapeutically possesses one or more of the following features:
  • the cell line of preferred embodiments is derived from a parent cell line wherein the cells are passaged in culture until they have acquired a smaller size and more rapid growth rate and until they readily attach to substrates.
  • the cell line of preferred embodiments is transfected with an expression vector containing the 2 and 3, a human cDNA of about 2.2 kb in length, and a 3' bovine growth hormone cytomegalovirus promoter/enhancer element, a 5' intron consisting of the murine Ca intron between exons polyadenylation site.
  • This expression vector may be transfected at, for example, a 50 to 1 ratio with any appropriate common selection vector such as pSV2NEO.
  • the selection vector pSV2NEO in turn confers G418 resistance on successfully transfected cells.
  • a ratio of about 50 to 1 is used since this ratio enhances the acquisition of multiple copy number inserts.
  • the Chinese hamster ovary cell line 2.131 there are approximately 10 copies of the expression vector for ⁇ -L-iduronidase.
  • a cell line has demonstrated the ability to produce large quantities of human ⁇ -L-iduronidase (minimum 20 micrograms per 10 million cells per day).
  • Particularly preferred embodiments such as the 2.131 cell line possess the ability to produce properly processed enzyme that contains N-linked oligosaccharides containing high mannose chains modified with phosphate at the 6 position in sufficient quantity to produce an enzyme with high affinity (K-uptake of less than 3 nM).
  • the enzyme produced from the cell lines of the present invention such as a Chinese hamster ovary cell line 2.131 is rapidly assimilated into cells, eliminates glycosaminoglycan storage and has a half-life of about 5 days in cells from patients suffering from ⁇ -L-iduronidase deficiency.
  • the cell line of preferred embodiments such as a 2.131 cell line adapts to large scale culture and stably produces human ⁇ -L-iduronidase under these conditions.
  • the cells of preferred embodiments are able to grow and secrete ⁇ -L-iduronidase at the acid pH of about 6.6 to 6.8 at which enhanced accumulation of ⁇ -L-iduronidase can occur.
  • Particularly preferred embodiments of the cell line according to the invention are able to secrete human ⁇ -L-iduronidase at levels exceeding 2,000 units per ml (8 micrograms per ml) twice per day using a specially formulated protein-free medium.
  • the present invention provides novel vectors suitable to produce ⁇ -L- iduronidase in amounts which enable using the enzyme therapeutically.
  • the production of adequate quantities of recombinant ⁇ -L-iduronidase is a critical prerequisite for studies on the structure of the enzyme as well as for enzyme replacement therapy.
  • the cell lines according to the present invention permit the production of significant quantities of recombinant ⁇ -L-iduronidase that is appropriately processed for uptake.
  • Overexpression in Chinese hamster ovary (CHO) cells has been described for three other lysosomal enzymes, ⁇ -galactosidase (Ioannou et al, J Cell. Biol.
  • the present invention features a dihydrofolate reductase- deficient CHO cell line, but according to preferred embodiments of the invention amplification is unnecessary.
  • the present mvention provides a high level of expression of the human ⁇ - L-iduronidase using the CMV immediate early gene promoter/enhancer.
  • the present invention features in preferred embodiments an expression vector comprising a cytomegalovirus promoter/enhancer element, a 5' intron consisting of the murine Ca intron derived from the murine long chain immunoglobulin C ⁇ gene between exons 2 and 3, a human cDNA of about 2.2 kb in length, and a 3' bovine growth hormone polyadenylation site.
  • This expression vector may be transfected at, for example, a 50 to 1 ratio with any appropriate common selection vector such as, for example, pSV2NEO.
  • the selection vector such as pSV2NEO in turn confers G418 resistance on successfully transfected cells.
  • a ratio of about 50 to 1 expression vector to selection vector is used since this ratio enhances the acquisition of multiple copy number inserts.
  • the Chinese hamster ovary cell line 2.131 there are approximately 10 copies of the expression vector for ⁇ -L-iduronidase.
  • Such an expression construct has demonstrated the ability to produce large quantities of human ⁇ -L- iduronidase (minimum 20 micrograms per 10 million cells per day) in a suitable cell line such as, for example, a Chinese hamster ovary cell line 2.131.
  • the present invention provides novel ⁇ -L-iduronidase produced in accordance with the methods of the present invention and thereby present in amounts that enable using the enzyme therapeutically.
  • the methods of the present invention produce a substantially pure ⁇ -L-iduronidase that is properly processed and in high uptake form appropriate for enzyme replacement therapy and that is effective in therapy in vivo.
  • the specific activity of the ⁇ -L-iduronidase according to the present invention is in excess of about 200,000 units per milligram protein. Preferably, it is in excess of about 240,000 units per milligram protein.
  • the molecular weight of the full length ⁇ -L-iduronidase of the present invention is about 82,000 daltons comprising about 70,000 daltons of amino acids and 12,000 daltons of carbohydrates.
  • the recombinant enzyme of the present invention is endocytosed even more efficiently than has been previously reported for a partially purified preparation of urinary enzyme.
  • the recombinant enzyme according to the present invention is effective in reducing the accumulation of radioactive S-labeled GAG in ⁇ -L-iduronidase-deficient fibroblasts, indicating that it is transported to lysosomes, the site of GAG storage.
  • the remarkably low concentration of ⁇ -L-iduronidase needed for such correction (half-maximal correction at 0.7 pM) may be very important for the success of enzyme replacement therapy.
  • the human cDNA of ⁇ -L-iduronidase predicts a protein of 653 amino acids and an expected molecular weight of 70,000 daltons after signal peptide cleavage. Amino acid sequencing reveals alanine 26 at the N-terminus giving an expected protein of 629 amino acids.
  • Human recombinant ⁇ - L-iduronidase has a Histidine at position 8 of the mature protein.
  • the predicted protein sequence comprises six potential N-linked oligosaccharide modification sites. All of these may be modified in the recombinant protein. The third and sixth sites have been demonstrated to contain one or more mannose 6-phosphate residues responsible for high affinity uptake into cells.
  • the following peptide corresponds to Amino Acids 26-45 of Human Recombinant ⁇ -L-iduronidase with an N-terminus alanine and the following sequence: ala-glu-ala-pro-his-leu-val-his-val-asp-ala-ala-arg-ala-leu-t -pro-leu-arg-arg
  • the overexpression of the ⁇ -L-iduronidase of the present invention does not result in generalized secretion of other lysosomal enzymes that are dependent on mannose-6-P targeting.
  • the secreted recombinant ⁇ -L-iduronidase is similar to normal secreted enzyme in many respects. Its molecular size, found in various determinations to be 77, 82, 84, and 89 kDa, is comparable to 87 kDa, found for urinary corrective factor (Barton et al, J. Biol. Chem.
  • the present invention features a novel method to purify ⁇ -L-iduronidase.
  • the present invention features a method to purify recombinant ⁇ -L- iduronidase that has been optimized to produce a rapid and efficient purification with validatible chromatography resins and easy load, wash and elute operation.
  • the method of purifying ⁇ -L- iduronidase of the present invention involves a series of column chromatography steps which allow the high yield purification of enzyme from protein-free production medium.
  • the cell mass is grown in about 10 % serum containing medium followed by a switch to a modified protein-free production medium without any significant adaptation to produce a high specific activity starting material for purification.
  • a concentration/diafiltration scheme is employed that allows for the removal of such exogenous materials as Pluronics F-68 from the crude bulk to prevent fouling of columns.
  • a first column load is acidified to minimize the competitive inhibition effect of such compounds as uronic acids found in protein-free medium formulations.
  • a heparin, phenyl and sizing column purification scheme is used to produce pure enzyme using automatable steps.
  • the heparin and phenyl column steps are used to eliminate less desirable ⁇ -L-iduronidase that is nicked or degraded.
  • an acid pH treatment step is used to inactivate potential viruses without harming the enzyme.
  • Particularly preferred embodiments of the method for purifying ⁇ -L-iduronidase according to the present invention feature more than one or all of the optimizations according to the following particular embodiments.
  • the purification method of the present mvention may therefore provide a purified ⁇ -L-iduronidase having the characteristics described herein.
  • Concentration/diafiltration Crude supernatant is processed with a hollow fiber concentrator (A/G Technologies, 30K cutoff) to reduce fluid volume by about 75% and is then diafiltrated with a heparin load buffer (10 mM NaP0 4 pH 5.3, NaCl 200 mM).
  • the diaf ⁇ ltration is an important step that eliminates undesirable compounds such as Pluronics F-68 from the supernatant, a surfactant needed in many cell cultures of the present invention that can foul columns.
  • the diafilfration may also partly remove competitor inhibitors that may prevent binding to the heparin column. These inhibitors may be found in PF-CHO medium and are believed to be uronic acids derived from a soybean hydrolysate present in this particular medium.
  • Heparin column The load may be adjusted to a pH of about 5.0 before loading on Heparin Sepharose CL-6B.
  • Other types of heparin columns such as a heparin FF (Pharmacia) have different linkages and do not bind ⁇ -L-iduronidase as efficiently. A lower pH neutralizes uronic acids to some extent which lessens their competitive effect. Without the diafiltration and pH adjustment, heparin columns cannot be run using PF-CHO medium without having substantial enzyme flowthrough.
  • the column may be washed with a pH of about 5.3 buffer and then eluted in 0.6 M NaCl. The narrow range of binding and elution salt concentration leads to an efficient purification step and enzyme that is often greater than 90% pure after one step.
  • Phenyl column A Phenyl-Sepharose BP (Pharmacia) may be used in the next step.
  • the heparin eluate may be adjusted to about 1.5 M NaCl and loaded on the column.
  • the choice of resin is important as is the salt concentration in ensuring that the enzyme binds completely (no flow through) and yet elutes easily and completely with about 0.15 M NaCl.
  • the eluate obtained is nearly pure ⁇ -L-iduronidase.
  • a pH inactivation may be performed to provide a robust step for the removal of potential viruses.
  • the phenyl pool is adjusted to a pH of about 3.3 using Citrate pH 3.0 and held at room temperature for about 4 hours.
  • the enzyme may then be neutralized.
  • Embodiments featuring this step have been shown to eliminate viruses at a minimum of about 5 log units. The step does not substantially inactivate or affect the enzyme activity.
  • the enzyme may then be concentrated and injected onto a Sephacryl S-200 column and the peak of enzyme collected. Enzyme purified in this manner has been shown to contain mannose-6-phosphate residues of sufficient quantity at positions 3 and 6 of the N-lmked sugars to give the enzyme uptake affinity of less than 30 units per ml (less than 2 nM) enzyme.
  • the enzyme is substantially corrective for glycosammo glycan storage disorders and has a half-life inside cells of approximately 5 days.
  • the present invention features novel methods of treating diseases caused all or in part by a deficiency in ⁇ -L-iduronidase.
  • Recombinant ⁇ -L-iduronidase provides enzyme replacement therapy in a canme model of MPS 1.
  • This canme model is deficient in ⁇ -L-iduronidase due to a genetic mutation and is similar to human MPS 1.
  • Purified, properly processed ⁇ -L- lduromdase was administered intravenously to 11 dogs. In those dogs treated with weekly doses of 25,000 to 125,000 units per kg for 3, 6 or 13 months, the enzyme was taken up in a variety of tissues and decreased the lysosomal storage m many tissues.
  • the present mvention features novel pharmaceutical compositions comprising human ⁇ -L-iduronidase useful for treating a deficiency in ⁇ -L-iduromdase
  • the recombinant enzyme may be administered in a number of ways such as parenteral, topical, mtranasal, inhalation or oral administration.
  • Another aspect of the invention is to provide for the administration of the enzyme by formulating it with a pharmaceutically-acceptable earner which may be solid, semi- solid or liquid or an mgestable capsule.
  • pharmaceutical compositions include tablets, drops such as nasal drops, compositions for topical application such as ointments, jellies, creams and suspensions, aerosols for inhalation, nasal spray, and liposomes.
  • the recombinant enzyme comprises between 0.05 and 99% or between 0.5 and 99% by weight of the composition, for example, between 0.5 and 20% for compositions intended for injection and between 0.1 and 50% for compositions intended for oral administration.
  • the enzyme may be mixed with a solid, pulverulent carrier, for example lactose, saccharose, sorbitol, mannitol, a starch such as potato starch, corn starch, amylopectin, laminaria powder or citrus pulp powder, a cellulose derivative or gelatine and also may include lubricants such as magnesium or calcium stearate or a Carbowax or other polyethylene glycol waxes and compressed to form tablets or cores for dragees.
  • a solid, pulverulent carrier for example lactose, saccharose, sorbitol, mannitol, a starch such as potato starch, corn starch, amylopectin, laminaria powder or citrus pulp powder, a cellulose derivative or gelatine and also may include lubricants such as magnesium or calcium stearate or a Carbowax or other polyethylene glycol waxes and compressed to form tablets or cores for dragees.
  • the cores may be coated for example with concentrated sugar solutions which may contain gum arabic, talc and/or titanium dioxide, or alternatively with a film forming agent dissolved in easily volatile organic solvents or mixtures of organic solvents.
  • Dyestuffs can be added to these coatings, for example, to distinguish between different contents of active substance.
  • the active substance may be admixed with a Carbowax® or a suitable oil as e.g., sesame oil, olive oil, or arachis oil.
  • Hard gelatine capsules may contain granulates of the active substance with solid, pulverulent carriers such as lactose, saccharose, sorbitol, mannitol, starches such as potato starch, corn starch or amylopectin, cellulose derivatives or gelatine, and may also include magnesium stearate or stearic acid as lubricants.
  • solid, pulverulent carriers such as lactose, saccharose, sorbitol, mannitol, starches such as potato starch, corn starch or amylopectin, cellulose derivatives or gelatine, and may also include magnesium stearate or stearic acid as lubricants.
  • Therapeutic enzymes of the subject invention may also be administered parenterally such as by subcutaneous, intramuscular or intravenous injection or by sustained release subcutaneous implant.
  • the therapeutic enzyme (the active ingredient) may be dissolved or dispersed in a liquid carrier vehicle.
  • the active material may be suitably admixed with an acceptable vehicle, preferably of the vegetable oil variety such as peanut oil, cottonseed oil and the like.
  • an acceptable vehicle preferably of the vegetable oil variety such as peanut oil, cottonseed oil and the like.
  • Other parenteral vehicles such as organic compositions using solketal, glycerol, formal, and aqueous parenteral formulations may also be used.
  • compositions may comprise an aqueous solution of a water soluble pharmaceutically acceptable salt of the active acids according to the invention, desirably in a concentration of 0.5-10%, and optionally also a stabilizing agent and/or buffer substances in aqueous solution. Dosage units of the solution may advantageously be enclosed in ampules.
  • the compound When therapeutic enzymes are administered in the form of a subcutaneous implant, the compound is suspended or dissolved in a slowly dispersed material known to those skilled in the art, or administered in a device which slowly releases the active material through the use of a constant driving force such as an osmotic pump. In such cases, administration over an extended pe ⁇ od of time is possible.
  • the pharmaceutical compositions are suitably in the form of an ointment, gel, suspension, cream or the like.
  • the amount of active substance may vary, for example, between 0.05- 20% by weight of the active substance.
  • Such pharmaceutical compositions for topical application may be prepared in known manner by mixing the active substance with known earner materials such as isopropanol, glycerol, paraffin, stearyl alcohol, polyethylene glycol, etc.
  • the pharmaceutically acceptable carrier may also include a known chemical absorption promoter. Examples of absorption promoters are, e g , dimethylacetamide (U.S. Patent No. 3,472,931), tnchloro ethanol or tnfluoroethanol (U.S. Patent No.
  • the dosage at which the therapeutic enzyme containing pharmaceutical compositions are administered may vary within a wide range and will depend on various factors such as, for example, the severity of the disease, the age of the patient, etc., and may have to be individually adjusted As a possible range for the amount of therapeutic enzyme which may be administered per day be mentioned from about 0.1 mg to about 2000 mg or from about 1 mg to about 2000 mg.
  • compositions containing the therapeutic enzyme may suitably be formulated so that they provide doses within these ranges either as single dosage units or as multiple dosage units
  • the subject formulations may contain one or more substrates or cofactors for the reaction catalyzed by the therapeutic enzyme in the compositions.
  • Therapeutic enzyme containing compositions may also contain more than one therapeutic enzyme.
  • the recombinant enzyme employed in the subject methods and compositions may also be administered by means of transforming patient cells with nucleic acids encoding the recombinant ⁇ - L-iduromdase.
  • the nucleic acid sequence so encoding may be incorporated into a vector for transformation into cells of the subject to be treated. Preferred embodiments of such vectors are described herein.
  • the vector may be designed so as to integrate into the chromosomes of the subject, e.g , retroviral vectors, or to replicate autonomously m the host cells.
  • Vectors containing encoding ⁇ - L-iduromdase nucleotide sequences may be designed so as to provide for continuous or regulated expression of the enzyme.
  • the genetic vector encoding the enzyme may be designed so as to stably integrate into the cell genome or to only be present transiently.
  • the general methodology of conventional genetic therapy may be applied to polynucleotide sequences encoding ⁇ -L- iduronidase. Reviews of conventional genetic therapy techniques can be found in Friedman, Science 244:1275-1281 (1989); Ledley, J. Inherit. Metab. Dis. 13:587-616 (1990); and Tolstoshev et al, Curr Opinions Biotech. L55-61 (1990).
  • a particularly preferred method of administering the recombinant enzyme is intravenously.
  • a particularly preferred composition comprises recombinant ⁇ -L-iduronidase, normal saline, phosphate buffer to maintain the pH at about 5.8 and human albumin. These ingredients may be provided in the following amounts: ⁇ -L-iduronidase 0.05-0.2 mg/mL or 12,500-50,000 units per mL
  • the cassette included 136 bp of the 3' end of exon 2 and 242 bp of the 5' end of exon 3, which would remain in the properly spliced cDNA. No ATG sequences are present in the coding, region of the intron cassette.
  • the intron cassette was cloned into the Hindlll site 5' of the ⁇ - L-iduronidase cDNA. The neo gene was deleted by digestion with Xhol followed by recircularizing the vector to make pCMVhldu.
  • One vial of the master cell bank is thawed and placed in three T150 flasks in DME F12 plus supplements plus 10% FBS and 500 1 lg/ml G418. After 3-4 days, the cells are passaged using trypsin-EDTA to 6 high capacity roller bottles in the same medium.
  • the innoculum of 2 x 10 9 cells is added to a Wheaton microcarrier flask containing 60 grams of Cytodex 2 microcarriers, and DME/F12 plus supplements, 10% FBS and 500 1 lg/ml of G418 at a final volume of 13 liters.
  • the flask is stirred by a Bellco overhead drive with a Perfusion wand stirrer.
  • the culture is monitored by temperature, DO and pH probes and controlled using the Wheaton mini-pilot plant control system with a PC interface (BioPro software). The parameters are controlled at the set points, 37° C, 80% air saturation, and pH 6.7, using a heating- blanket, oxygen sparger and base pump.
  • the culture is incubated for 3-4 days at which time the culture is coming out of log phase growth at 1-3 x 10 6 cells per ml. Thereafter, at 12 hour intervals, the medium is changed with PF-CHO medium (with custom modifications, JRH Biosciences). The first 2 collections are set aside as "washout”. The third collection is the beginning of the production run. Sodium butyrate at final 2 mM is added every 48 hours to induce an increase in iduronidase expression.
  • Production continues with medium changes of 10 liters every 12 hours and the collections filtered through a 1 micron filter to eliminate free cells and debris.
  • the culture is monitored for temperature, pH and DO on a continuous basis. Twice daily, the culture is sampled before the medium change and assayed for cell condition and microorganisms by phase contrast microscopy, glucose content using a portable glucometer, iduronidase activity using a fluorescent substrate assay. Cell mass is assayed several times during the run using a total cellular protein assay. By the middle of the run, cell mass reaches 10 7 cells per ml. Collected production medium containing iduronidase is then concentrated five fold using an A/G Technology hollow fiber molecular filter with a 30,000 molecular weight cutoff.
  • the concentrate is then diafiltrated with a minimum three fold volume of 0.2 M NaCl in 10 mM NaP0 4 , pH 5. 8 over a period of 8 hours. This step removes Pluronics F68 and uronic acids from the concentrate. These molecules can inhibit function of the Heparin column.
  • the concentrate is adjusted to pH 5.0, filtered through 1.0 and 0.2 micron filters and then loaded on a Heparin- Sepharose CL-6B column. The column is washed with 10 column volumes of 0.2 M NaCl, 10 mM NaP0 4 , pH 5.3), and the enzyme eluted with 0.6 MI, 10mMNaPO 4 ,pH 5.8.
  • the eluate is adjusted to 1.5 M NaCl, filtered through a 1 micron filter and loaded on a Phenyl-Sepharose HP column.
  • the column is washed with 10 column volumes of 1.5 M NaCl, 10 mM NaP0 4 , pH 5.8 and the enzyme eluted with 0. 15 M NaCl, 10 mM NaP0 4 , pH 5.8.
  • Viral inactivation is performed by acidifying the enzyme fraction to pH 3.3 using 1 M citric acid pH 2.9 and incubating the enzyme at pH 3.3 at room temperature for 4 hours and readjusting the pH to 5.8 using 1 M phosphate buffer. This step has been demonstrated to remove 5 logs or better of a retrovirus in spiking experiments.
  • the inactivated enzyme is filtered through a 0.2 ⁇ filter, concentrated on an A/G Technologies hollow fiber concentrator apparatus (30,000 molecular weight cutoff) and injected in cycles on a Sephacryl S200 gel filtration column and the peaks collected. The pooled peaks are filtered through a 0.2 ⁇ filter, formulated to 0.1 M NaP0 4 , pH 5.8 and vialed.
  • a set of studies may be performed to assess the quality, purity, potency of the enzyme. Results of an SDS-PAGE analysis of the eluate is provided in Figure 2.
  • One recombinant human ⁇ -L-iduronidase obtained from this procedure demonstrates a potency of 100,000 units per milliliter and has a total protein concentration of 0.313 mg/ml.
  • Short-term intravenous administration of purified human recombinant ⁇ -L-iduronidase to 9 MPS I dogs and 6 MPS I cats has shown significant uptake of enzyme in a variety of tissues with an estimated 50% or more recovery in tissues 24 hours after a single dose.
  • liver and spleen take up the largest amount of enzyme, and have the best pathologic improvement, improvements in pathology and glycosaminoglycan content has been observed in many, but not all tissues.
  • the cartilage, brain and heart valve did not have significant improvement.
  • Clinical improvement was observed in a single dog on long-term treatment for 13 months, but other studies have been limited to 6 months or less. All dogs, and most cats, that received recombinant human enzyme developed antibodies to the human product.
  • the IgG antibodies are of the complement activating type (probable canine IgG equivalent). This phenomena is also observed in at least 13% of alglucerase-treated Gaucher patients. Proteinuria has been observed in one dog which may be related to immune complex disease. No other effects of the antibodies have been observed in the other treated animals. Specific toxicity was not observed and clinical laboratory studies (complete blood counts, electrolytes, BLJN/creatinine, liver enzymes, urinalysis) have been otherwise normal.
  • Enzyme therapy at even small doses of 25,000 units resulted in significant enzyme distribution to some tissues and decreases in GAG storage. If continued for over 1 year, significant clinical effects of the therapy were evident in terms of activity, mobility, growth and overall health. The therapy at this dose did not improve other tissues that are important sites for disease in this entity such as cartilage and brain. Higher doses of 125,000 units (0.5 mg/kg) given 5 times over two weeks demonstrate that improved tissue penetration can be achieved, and a therapeutic effect at the tissue level was accomplished in as little as 2 weeks. Studies at this increased dose are ongoing in two dogs for six months to date. These MPS I dogs showed significant clinical improvement and substantial decreases in urinary GAG excretion into the normal range.
  • the human cDNA of ⁇ -L-iduronidase predicts a protein of 653 amino acids and an expected molecular weight of 70,000 daltons after signal peptide cleavage. Amino acid sequencing reveals alanine 26 at the N-terminus giving an expected protein of 629 amino acids.
  • Human recombinant ⁇ - L-iduronidase has a Histidine at position 8 of the mature protein.
  • the predicted protein sequence comprises six potential N-linked oligosaccharide modification sites. All of these sites are modified in the recombinant protein. The third and sixth sites have been demonstrated to contain one or more mannose 6-phosphate residues responsible for high affinity uptake into cells.
  • This peptide corresponds to Amino Acids 26-45 of Human Recombinant ⁇ -L-iduronidase with an N-terminus alanine and the following sequence: ala-glu-ala-pro-his-leu-val-his-val-asp-ala-ala-arg-ala-leu-trp-pro-leu-arg-arg
  • the recombinant enzyme has an apparent molecular weight of 82,000 daltons on SDS-PAGE due to carbohydrate modifications.
  • Purified human recombinant ⁇ -L-iduronidase has been sequenced by the UCLA Protein Sequencing facility. It is preferred to administer the recombinant enzyme intravenously.
  • Human recombinant ⁇ -L-iduronidase was supplied in 10 mL polypropylene vials at a concentration of 0.05-0.2 mg/ml (12,500-50,000 units per mL).
  • the final dosage form of the enzyme includes human recombinant ⁇ -L-iduronidase, normal saline, phosphate buffer at pH 5.8 and human albumin at 1 mg/ml. These are prepared in a bag of normal saline.
  • Component Composition ⁇ -L-iduronidase 0.05-0.2 mg/mL or 12,500-50,000 units per mL
  • FIGURE 4 demonstrates leukocyte iduronidase activity before and after enzyme therapy in MPS I patients.
  • the buccal iduronidase activity before and after enzyme therapy is depicted in FIGURE 5.
  • FIGURE 6 demonstrates in three patients that a substantial shrinkage of liver and spleen together with significant clinical improvement in joint and soft tissue storage was associated with a greater than 65% reduction in undegraded GAG after only 8 weeks of treatment with recombinant enzyme.
  • FIGURE 7 demonstrates that there is substantial normalization of livers and spleens in patients treated with recombinant enzyme after only 12 weeks of therapy with recombinant enzyme.
  • FIGURE 8 demonstrates a precipitous drop in urinary GAG excretion over 22 weeks of therapy with recombinant enzyme in 11 patients.
  • Clinical assessment of liver and spleen size has been the most widely accepted means for evaluating successful bone marrow transplant treatment in MPS-I patients (Hoogerbrugge et al, Lancet 345:1398 (1995)). Such measurements are highly correlated with a decreased visceral storage of GAGs in MPS-I patients.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

La présente invention concerne une α-L-iduronidase, des fragments et des mutants biologiquement actifs de celle-ci, des procédés de production et de purification de cette enzyme, ainsi que des méthodes de traitement de certaines affections génétiques comprenant un déficit en α-L-iduronidase et la mucopolysaccharidose I (MPS I).
EP99924155A 1998-05-13 1999-05-07 (alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci Withdrawn EP1078075A2 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US7820998A 1998-05-13 1998-05-13
US17097798A 1998-10-13 1998-10-13
US170977 1998-10-13
PCT/US1999/010102 WO1999058691A2 (fr) 1998-05-13 1999-05-07 (alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci
US78209 2008-07-03

Publications (1)

Publication Number Publication Date
EP1078075A2 true EP1078075A2 (fr) 2001-02-28

Family

ID=26760239

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99924155A Withdrawn EP1078075A2 (fr) 1998-05-13 1999-05-07 (alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci

Country Status (8)

Country Link
EP (1) EP1078075A2 (fr)
JP (1) JP2002514429A (fr)
CN (1) CN1300323A (fr)
AU (1) AU4072499A (fr)
BR (1) BR9910323A (fr)
CA (1) CA2328518A1 (fr)
IL (1) IL139616A0 (fr)
WO (1) WO1999058691A2 (fr)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6426208B1 (en) * 1999-11-12 2002-07-30 Harbor-Ucla Research And Education Institute Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating diseases caused by deficiencies thereof
US6585971B1 (en) * 1999-11-12 2003-07-01 Harbor-Ucla Research And Education Institute Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating disease caused by deficiencies thereof
US6569661B1 (en) 1999-11-12 2003-05-27 Biomarin Pharmaceutical Inc. Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating diseases caused by deficiencies thereof
PT1301201E (pt) 2000-07-18 2007-03-30 Univ Duke Tratamento da doença de armazenamento de glicogénio tipo ii
US7442372B2 (en) * 2003-08-29 2008-10-28 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
CN103180445B (zh) * 2010-10-22 2018-02-16 库尔纳公司 通过抑制α‑L‑艾杜糖醛酸酶(IDUA)的天然反义转录物而治疗IDUA相关疾病
CN103487414A (zh) * 2012-06-14 2014-01-01 北京和信非凡生物技术有限公司 一种α-L-艾杜糖醛酸酶活性检测方法、底物和试剂
PT2984166T (pt) * 2013-03-15 2020-07-16 Univ Pennsylvania Composições para tratar mps i
BR112018015751A2 (pt) 2016-02-03 2019-02-05 Univ Pennsylvania terapia gênica para tratamento de mucopolissacaridose tipo i
KR20190109506A (ko) * 2017-01-31 2019-09-25 리젠엑스바이오 인크. 완전히-인간 글리코실화된 인간 알파-l-이두로니다아제(idua)를 이용한 뮤코다당증 1형의 치료
EP3676385A1 (fr) 2017-07-06 2020-07-08 The Trustees of The University of Pennsylvania Thérapie génique médiée par aav9 pour traiter la mucopolysaccharidose de type i
CN107699590A (zh) * 2017-10-13 2018-02-16 成都中医药大学 一种制备重组人α‑L‑艾杜糖醛酸酶的方法
CN115109790A (zh) * 2021-03-23 2022-09-27 北京据德医药科技有限公司 一种重组a-L-艾杜糖醛酸普酶及其制备方法
CN114181318B (zh) * 2021-11-08 2023-08-01 四川大学 一种组织特异性表达穿透血脑屏障的idua融合蛋白的重组腺相关病毒及应用

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2099503C (fr) * 1991-11-14 2007-10-09 Hamish Steele Scott Alpha-l-iduronidase synthetique et sequences de genes la codant
EP0865499B1 (fr) * 1995-09-14 2009-03-18 Virginia Tech Intellectual Properties, Inc. Production d'enzymes lysosomiales par des systemes d'expression vegetaux

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9958691A2 *

Also Published As

Publication number Publication date
BR9910323A (pt) 2001-01-30
AU4072499A (en) 1999-11-29
CA2328518A1 (fr) 1999-11-18
CN1300323A (zh) 2001-06-20
WO1999058691A3 (fr) 2000-02-17
WO1999058691A2 (fr) 1999-11-18
JP2002514429A (ja) 2002-05-21
IL139616A0 (en) 2002-02-10

Similar Documents

Publication Publication Date Title
US7041487B2 (en) Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating diseases caused by deficiencies thereof
US7354576B2 (en) Methods of treating diseases caused by deficiencies of recombinant α-L-iduronidase
EP2292256B1 (fr) Protéines à forte teneur en mannose et procédé pour la fabrication de celles-ci
US8343487B2 (en) Large-scale production of soluble hyaluronidase
WO2002038775A2 (fr) METHODES PERMETTANT DE PRODUIRE ET DE PURIFIER UNE α-L-IDURONIDASE RECOMBINEE
WO1999058691A2 (fr) (alpha)-l-iduronidase recombinee, procedes de production et de purification de celle-ci et methodes de traitement de maladies dues a des deficits en celle-ci
US20060040348A1 (en) Methods for producing and purifying recombinant alpha-L-iduronidase
RAO Methods for treating diseases caused by deficiencies of recombinant alpha-L-iduronidase
AU2014204569B9 (en) High mannose proteins and methods of making high mannose proteins
AU2016277569A1 (en) High mannose proteins and methods of making high mannose proteins
AU2007202256A1 (en) High mannose proteins and methods of making high mannose proteins

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20001106

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20031202