AU2016202625A1 - Methods and compositions for increasing alpha-iduronidase activity in the CNS - Google Patents

Methods and compositions for increasing alpha-iduronidase activity in the CNS Download PDF

Info

Publication number
AU2016202625A1
AU2016202625A1 AU2016202625A AU2016202625A AU2016202625A1 AU 2016202625 A1 AU2016202625 A1 AU 2016202625A1 AU 2016202625 A AU2016202625 A AU 2016202625A AU 2016202625 A AU2016202625 A AU 2016202625A AU 2016202625 A1 AU2016202625 A1 AU 2016202625A1
Authority
AU
Australia
Prior art keywords
amino acid
acid sequence
seq
idua
iduronidase
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2016202625A
Inventor
Ruben J. Boado
William M. Pardridge
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.)
Armagen Inc
Original Assignee
Armagen Technologies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2013206657A external-priority patent/AU2013206657A1/en
Application filed by Armagen Technologies Inc filed Critical Armagen Technologies Inc
Priority to AU2016202625A priority Critical patent/AU2016202625A1/en
Publication of AU2016202625A1 publication Critical patent/AU2016202625A1/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided herein are methods and compositions for treating a subject suffering from a deficiency in a-L-Iduronidase in the CNS. The methods include systemic administration of a bifunctional fusion antibody comprising an antibody to a human insulin receptor and an a-L-Iduronidase. A therapeutically effective systemic dose is based on the specific CNS uptake characteristics of human insulin receptor antibody-at-L Iduronidase fusion antibodies as described herein. 7671816_1 (GHMatters) P83171.AU.2

Description

METHODS AND COMPOSITIONS FOR INCREASING a-L-IDURONIDASE ACTIVITY IN THE CNS CROSS-REFERENCE [0001] The entire disclosure in the complete specification of our Australian Patent Application No. 2013206657 is by this cross-reference incorporated into the present specification. BACKGROUND OF THE INVENTION [0002] Type I mucopolysaccharidosis (MPS), also known as Hurler's syndrome, is an inherited metabolic disease caused by a defect in the enzyme a-L-iduronidase (IDUA), which functions to degrade mucopolysaccharides. An insufficient level of IDUA causes a pathological buildup of heparan sulfate and dermatan sulfate in, e.g., the heart, liver, and central nervous system. Symptoms including neurodegeneration and mental retardation appear during childhood and early death can occur due to organ damage. Typically, treatment includes intravenous enzyme replacement therapy with recombinant IDUA. However, systemically administered recombinant IDUA does not cross the blood brain barrier (BBB), and therefore has little impact on the effects of the disease in the central nervous system (CNS). [0003] It is to be understood that if any prior art publication is referred to herein, such reference does not constitute an admission that the publication forms a part of the common general knowledge in the art in Australia or any other country. SUMMARY OF THE INVENTION [0004] Described herein are methods and compositions for treating a subject suffering from an IDUA deficiency. In particular, the methods allow delivery of IDUA to the CNS by systemically administering a therapeutically effective amount of a bifunctional human insulin receptor antibody IDUA (HIR Ab-IDUA) fusion antibody. The HIR Ab-IDUA fusion antibody binds to the extracellular domain of the insulin receptor and is transported across the blood brain barrier into the CNS, while retaining IDUA activity. A therapeutically effective systemic dose of a HIR Ab-IDUA fusion antibody for systemic administration will be based, in part, on the specific CNS uptake characteristics of the fusion antibody from peripheral blood as described herein. [0005] Accordingly, in one aspect provided herein is a method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the subject a therapeutically effective dose of a fusion antibody having a-L iduronidase activity. The method is characterized by the following: (i) at least about 0.5% of the therapeutically effective dose is delivered to the brain; (ii) the fusion antibody: comprises: (a) a fusion protein containing the amino acid sequence of an immunoglobulin heavy chain and an a-L-iduronidase, and (b) an immunoglobulin light chain; (iii) the fusion antibody binds to an extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate; and (iv) the amino acid sequence of the a L iduronidase is covalently linked to the carboxy terminus of the amino acid sequence of the immunoglobulin heavy chain. -1 7671816_1 (GHMatters) P83171.AU.2 [0006] In some embodiments at least about 50,000 units of a-L-iduronidase activity are delivered to the brain. In some embodiments, the therapeutically effective dose of the fusion antibody comprises at least about 1 x 106 units of a-L-iduronidase activity or at least about 140,000 units/Kg of body weight. In some embodiments the IDUA specific activity of the fusion antibody is at least 200,000 units/mg. In some embodiments, systemic administration is parenteral, intravenous, subcutaneous, intra muscular, trans-nasal, intra-arterial, transdermal, or respiratory. In some embodiments, delivery of at least 0. 5% of the therapeutically effective dose to the brain occurs within two hours or less after the systemic administration. [0007] In some embodiments, the fusion antibody is a chimeric antibody. [0008] In some embodiments, the immunoglobulin heavy chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 4 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions. [0009] In other embodiments, the inmmunoglobulin heavy chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations. [0010] In other embodiments, the inmmunoglobulin heavy chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3. [0011] In further embodiments, the immunoglobulin heavy chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3. [0012] In some embodiments, the immunoglobulin light chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions. [0013] In other embodiments, the inmmunoglobulin light chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations. -2 7671816_1 (GHMatters) P83171.AU.2 [0014] In other embodiments, the inmmunoglobulin light chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6. [0015] In further embodiments, the immunoglobulin light chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6. [0016] In some embodiments, the immunoglobulin heavy chain of the fusion antibody comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3; and the inmmunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6. [0017] In some embodiments, the inmmunoglobulin heavy chain of the fusion antibody is at least 90% identical to SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO:8. [0018] In some embodiments, the inmmunoglobulin heavy chain of the fusion antibody comprises SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin comprises SEQ ID NO: 8 [0019] In yet further embodiments, the a-L-iduronidase comprises an amino acid sequence at least 90% (e.g., 95%, or 100o) identical to SEQ ID NO:9. [0020] In other embodiments, the amino acid sequence of the immunoglobulin heavy chain of the fusion antibody at least 90% identical to SEQ ID NO:7; the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO: 8; and the amino acid sequence of the a-L iduronidase is at least 95% identical to SEQ ID NO:9 or comprises SEQ ID NO:9. [0021] In still other embodiments, the amino acid sequence of the immunoglobulin heavy chain of the fusion antibody comprises SEQ ID NO:8, the amino acid sequence of the immunoglobulin light chain comprises SEQ ID NO:8, and the amino acid sequence of the IDUA comprises SEQ ID NO:9 [0022] In a further aspect provided herein is a method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the subject a therapeutically effective dose of a fusion antibody having a-L-iduronidase activity, where the method is characterized in that (i) at least about 0.5% of the systemically administered therapeutically effective dose is delivered to the brain; (ii) the fusion antibody: comprises: (a) a fusion protein at least 95% identical to SEQ ID NO: 10, and (b) an inmmunoglobulin light chain; and (iii) the fusion antibody binds to an extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate. [0023] In yet another aspect provided herein is a method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the -3 7671816_1 (GHMatters) P83171.AU.2 subject a therapeutically effective dose of a fusion antibody having a-L-iduronidase activity, where the method is characterized in that: (i) at least about 0.5% of the therapeutically effective dose is delivered to the brain; (ii) the fusion antibody: comprises a fusion protein containing the amino acid sequence of an immunoglobulin heavy chain and an a-L-iduronidase; or comprises a fusion protein containing the amino acid sequence of an immunoglobulin light chain and an a-L-iduronidase; binds to the extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate; and (iii) the amino acid sequence of the a-L-iduronidase is covalently linked to the carboxy terminus of the amino acid sequence of the immunoglobulin heavy chain or the immunoglobulin light chain. BRIEF DESCRIPTION OF THE DRAWINGS [0024] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings, as follow: [0025] Figure 1. Amino acid sequence of an immunoglobulin heavy chain variable region from an exemplary human insulin receptor antibody directed against the extracellular domain of the human insulin receptor. The underlined sequences are a signal peptide, CDRI, CDR2, and CDR3, respectively. The heavy chain constant region, taken from human IgGI, is shown in italics. [0026] Figure 2. Amino acid sequence of an immunoglobulin light chain variable region from an exemplary human insulin receptor antibody directed against the extracellular domain of the human insulin receptor. The underlined sequences are a signal peptide, CDRI, CDR2, and CDR3, respectively. The constant region, derived from human kappa light chain, is shown in italics. [0027] Figure 3. A table showing the CDRI, CDR2, and CDR3 amino acid sequences from a heavy and light chain of an exemplary human insulin receptor antibody directed against the extracellular domain of the human insulin receptor. [0028] Figure 4. Amino acid sequence of human a-L-iduronidase (IDUA) (GenBank NP_000194), not including the initial 26 amino acid signal peptide (mature IDUA). [0029] Figure 5. Amino acid sequence of a fusion of an exemplary human insulin receptor antibody heavy chain to mature human IDUA. The underlined sequences are, in order, an IgG signal peptide, CDRI, CDR2, CDR3, and a peptide linker linking the carboxy terminus of the heavy chain to the amino terminus of the IDUA. Sequence in italic corresponds to the heavy chain constant region, taken from human IgG 1. The sequence in bold corresponds to human IDUA. [0030] Figure 6. An exemplary HIR Ab-IDUA fusion antibody is formed by fusion of the amino terminus of the mature IDUA to the carboxyl terminus of the CH3 region of the heavy chain of the HIR Ab. The fusion protein is a bi-functional molecule: the fusion protein binds the HIR, at the BBB, to -4 7671816_1 (GHMatters) P83171.AU.2 mediate transport into the brain, and expresses IDUA enzyme activity, which is deficient in MPS Type I (Hurler's syndrome). [0031] Figure 7. Schematic depiction of a "molecular trojan horse" strategy in which the fusion antibody comprises an antibody to the extracellular domain of the human insulin receptor, which acts as a molecular Trojan horse (TH), and IDUA, a lysosomal enzyme (E). By itself, the IDUA normally does not cross the blood-brain barrier (BBB). However, following fusion of the IDUA to the TH, the enzyme is able to cross the BBB, and the brain cell membrane, by trafficking on the IR, which is expressed at both membranes in the brain. [0032] Figure 8. Ethidium bromide stain of agarose gel of human IDUA cDNA (lane 1), which was produced by PCR from human liver cDNA, and IDUA-specific ODN primers (Table I). Lanes 2 and 3: PhiX174 HaeIII digested DNA standard, and Lambda HindlIl digested DNA standard. [0033] Figure 9. Western blot with either anti-human (h) IgG primary antibody (right panel) or rabbit anti-human IDUA primary antiserum (left panel). The immunoreactivity of the HIR Ab-IDUA fusion antibody is compared to the HIR Ab alone. Both the HIR Ab-IDUA fusion antibody and the HIR Ab have identical light chains on the anti-hIgG Western. The HIR Ab-IDUA fusion heavy chain reacts with both the anti-hIgG and the anti-human IDUA antibody, whereas the HIR Ab heavy chain only reacts with the anti-hIgG antibody. The size of the HIR Ab-IDUA fusion heavy chain, 130 kDa, is about 80 kDa larger than the size of the heavy chain of the HIR Ab, owing to the fusion of the 80 kDa IDUA to the 50 kDa HIR Ab heavy chain. [0034] Figure 10. Binding of either the chimeric HIR Ab or the HIR Ab-IDUA fusion protein to the HIR extracellular domain (ECD) is saturable. The ED 50 of HIR Ab-IDUA binding to the HIR ECD is comparable to the ED 50 of the binding of the chimeric HIR Ab. [0035] Figure 11. (A) Intracellular IDUA enzyme activity is increased in Hurler fibroblasts in proportion to the concentration of medium HIR Ab-IDUA fusion protein. Data are mean ± SE (n=3 dishes/point). The horizontal bar is the IDUA enzyme activity in healthy human fibroblasts (284 ± 5 units/mg protein). (B) Reversal of glycosaminoglycan (GAG) accumulation in Hurler fibroblasts with a single treatment of 0.3 pg/mL of HIR Ab-IDUA fusion protein in the medium. There is a 70% reduction in GAG accumulation, as compared to the 35S incorporation in healthy human fibroblasts (p<0.0005). Data are mean SE (n=5 dishes/point). [0036] Figure 12. (A, B, C, D) Hurler fibroblasts were incubated with HIR Ab-IDUA fusion protein for 24 hours and then fixed and immune stained for confocal microscopy. The fixed cells were stained with a rabbit polyclonal antibody to human IDUA (panel A: red channel signal, shown here in black and white), and a mouse monoclonal antibody to human lysosomal associated membrane protein (LAMP)- 1 (panel B: green channel signal, shown here in black and white). The overlap image in panel C shows sequestration of the HIR Ab-IDUA fusion protein within lysosomes. Panel D is an overlap image of negative control primary antibodies: rabbit serum and mouse IgG. (E) Film autoradiography of Rhesus monkey brain removed 2 hours after an intravenous administration of [1251] -HIR Ab-IDUA fusion protein. Coronal sections through the forebrain (top panel), midbrain (middle panel), and hindbrain/cerebellum (bottom panel) are shown. -5 7671816_1 (GHMatters) P83171.AU.2 [0037] Figure 13. Pharmacokinetics and brain uptake of fusion protein in the adult Rhesus monkey. (A) The serum concentration, expressed as a percent of injected dose (ID)/mL, of the [..1]-HIR Ab IDUA fusion protein is plotted vs. time after a single intravenous injection of the protein in the anesthetized adult Rhesus monkey; the serum concentration is expressed as either 1251 radioactivity (closed symbol) or IDUA enzyme activity (open symbol). (B) The volume of distribution (VD) at 120 min after injection of the [1251] -HIR Ab-IDUA fusion protein is shown for the total brain homogenate and the post-vascular supernatant. The equivalence of the VD in both compartments is evidence for transport of the fusion protein through the BBB in vivo (Methods). The data for the [ 3 H] -mouse IgG2a is from Pardridge et al (1995). [0038] Figure 14. Genetic engineering of tandem vector (TV-HIRMAb-IDUA) encoding 4 separate and tandem expression cassettes encoding the heavy chain (HC) fusion gene, the light chain (LC) gene, the DHFR gene, and the neo gene. [0039] Figure 15. The 3-column purification of CHO derived HIRMAb-IDUA fusion protein uses protein A affinity chromatography (A), SP Sepharose cation exchange (CATEX) chromatography (B), and Q Sepharose anion exchange (ANEX) chromatography (C). The peak of fusion protein elution for each column is bracketed in the figure. [0040] Figure 16. The HIRMAb-IDUA fusion protein, derived from CHO cells, is purified to homogeneity on reducing SD S-PAGE, as shown in lane 3. Lane 2 is the chimeric HIRMAb without the fused IDUA. The MW of the HC of the HIRMAb-IDUA fusion protein is about 85 kDa larger than the HC of the HIRMAb, owing to the fusion of the IDUA enzyme. Lanes 1 and 4 are MW standards. [0041] Figure 17. Western blot of the HIRMAb-IDUA fusion protein, derived from CHO cells, using primary antibodies to either the human IgG heavy chain (lane 1) or to human IDUA (lane 2). Both antibodies react equally to the 130 kDa HIRMAb-IDUA fusion protein heavy chain. [0042] Figure 18. Binding of either the chimeric HIRMAb or the CHO cell derived HIRMAb-IDUA fusion protein to the HIR extracellular domain (ECD) is saturable. The ED 50 of HIRMAb-IDUA binding to the HIR ECD is comparable to the ED 50 of the binding of the chimeric HIRMAb, which indicates the affinity for the HIR is not impaired by fusion of the IDUA to the HIRMAb heavy chain. [0043] Figure 19. The IDUA enzyme activity of the CHO derived HIRMAb-IDUA fusion protein is 291+ 9 units/pg protein, where 1 unit=1 nmol/hr, based on a fluorometric enzymatic assay that uses 4-methylumbelliferyl L-a-iduronide (MUBI) as a substrate, and 4-methylumbelliferone (4-MU) as an assay standard. The IDUA enzyme activity is linear with respect to time and mass of HIRMAb-IDUA fusion protein. The IDUA enzyme specific activity of the HIRMAb-IDUA fusion protein is comparable to recombinant IDUA. [0044] Figure 20. Size exclusion chromatography (SEC) HPLC using 2 TosoHaas G3000SWXL columns in series. The CHO derived HIRMAb-IDUA fusion protein elutes as a single species without aggregates. -6 7671816_1 (GHMatters) P83171.AU.2 DETAILED DESCRIPTION OF THE INVENTION Introduction [0045] The blood brain barrier is a severe impediment to the delivery of systemically administered IDUA (e.g., recombinant IDUA) to the central nervous system. The methods and compositions described herein address three factors that are important in delivering a therapeutically significant level of IDUA activity across the BBB to the CNS: 1) Modification of an IDUA to allow it to cross the BBB; 2) the amount and rate of uptake of systemically administered modified IDUA into the CNS, and 3) Retention of IDUA activity once across the BBB. Various aspects of the methods and compositions described herein address these factors, by (1) providing human insulin receptor (HIR) antibody (Ab) IDUA fusion antibodies comprising an IDUA (i.e., a protein having IDUA activity) fused, with or without intervening sequence, to an immunoglobulin (heavy chain or light chain) directed against the extracellular domain of a human insulin receptor; and (2) establishing therapeutically effective systemic doses of the fusion antibodies based on a characterization of their uptake in the CNS and their specific activity. [0046] Accordingly, the invention provides compositions and methods for treating a a-L-iduronidase deficiency in the central nervous system by systemically administering to a subject in need thereof a therapeutically effective dose of a bifunctional HIR Ab-IDUA fusion antibody having a-L-iduronidase activity and selectively binding to the extracellular domain of a human insulin receptor. Some Definitions [0047] In the claims which follow and in the description of the invention, except where the context requires otherwise due to express language or necessary implication, the word "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention. [0048] "Treatment" or "treating" as used herein includes achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder or condition being treated. For example, in an individual with Hurler's syndrome, therapeutic benefit includes partial or complete halting of the progression of the disorder, or partial or complete reversal of the disorder. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological or psychological symptoms associated with the underlying condition such that an improvement is observed in the patient, notwithstanding the fact that the patient may still be affected by the condition. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition (e.g., slowing the progression of a lysosomal storage disorder), or decreasing the likelihood of occurrence of a condition. As used herein, "treating" or "treatment" includes prophylaxis. [0049] As used herein, the term "effective amount" can be an amount, which when administered systemically, is sufficient to effect beneficial or desired results in the CNS, such as beneficial or -7 7671816_1 (GHMatters) P83171.AU.2 desired clinical results, or enhanced cognition, memory, mood, or other desired CNS results. An effective amount is also an amount that produces a prophylactic effect, e.g., an amount that delays, reduces, or eliminates the appearance of a pathological or undesired condition. Such conditions include, but are not limited to, mental retardation, hearing loss, and neurodegeneration. An effective amount can be administered in one or more administrations. In terms of treatment, an "effective amount" of a composition of the invention is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of a disorder, e.g., a neurological disorder. An "effective amount" may be of any of the compositions of the invention used alone or in conjunction with one or more agents used to treat a disease or disorder. An "effective amount" of a therapeutic agent within the meaning of the present invention will be determined by a patient's attending physician or veterinarian. Such amounts are readily ascertained by one of ordinary skill in the art and will a therapeutic effect when administered in accordance with the present invention. Factors which influence what a therapeutically effective amount will be include, the IDUA specific activity of the HIR Ab-IDUA fusion antibody administered, its absorption profile (e.g., its rate of uptake into the brain), time elapsed since the initiation of the disorder, and the age, physical condition, existence of other disease states, and nutritional status of the individual being treated. Additionally, other medication the patient may be receiving will affect the determination of the therapeutically effective amount of the therapeutic agent to administer. [0050] A "subject" or an "individual," as used herein, is an animal, for example, a mammal. In some embodiments a "subject" or an "individual" is a human. In some embodiments, the subject suffers from Mucopolysaccharidosis Type I H ("Hurler's Syndrome"), Mucopolysaccharidosis Type I S ("Scheie Syndrome"), or Mucopolysaccharidosis Type I H-S ("Hurler-Scheie Syndrome). [0051] In some embodiments, a pharmacological composition comprising an HIR-IDUA fusion antibody is "administered peripherally" or "peripherally administered." As used herein, these terms refer to any form of administration of an agent, e.g., a therapeutic agent, to an individual that is not direct administration to the CNS, i.e., that brings the agent in contact with the non-brain side of the blood-brain barrier. "Peripheral administration," as used herein, includes intravenous, intra-arterial, subcutaneous, intramuscular, intraperitoneal, transdermal, by inhalation, transbuccal, intranasal, rectal, oral, parenteral, sublingual, or trans-nasal. [0052] A "pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" herein refers to any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Such carriers are well known to those of ordinary skill in the art. A thorough discussion of pharmaceutically acceptable carriers/excipients can be found in Remington 's Pharmaceutical Sciences, Gennaro, AR, ed., 20th edition, 2000: Williams and Wilkins PA, USA.. Exemplary pharmaceutically acceptable carriers can include salts, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. For example, compositions of the invention may be provided in liquid form, and formulated in saline based aqueous solution of varying pH (5-8), with or without detergents such polysorbate-80 at 0.0 1-o1%, or carbohydrate additives, such -8 7671816_1 (GHMatters) P83171.AU.2 mannitol, sorbitol, or trehalose. Commonly used buffers include histidine, acetate, phosphate, or citrate. [0053] A "recombinant host cell" or "host cell" refers to a cell that includes an exogenous polynucleotide, regardless of the method used for insertion, for example, direct uptake, transduction, f mating, or other methods known in the art to create recombinant host cells. The exogenous polynucleotide may be maintained as a nonintegrated vector, for example, a plasmid, or alternatively, may be integrated into the host genome. [0054] The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally occurring amino acid, e.g., an amino acid analog. As used herein, the terms encompass amino acid chains of any length, including full length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds. [0055] The term "amino acid" refers to naturally occurring and non-naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally encoded amino acids are the 20 common amino acids (alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, seine, threonine, tryptophan, tyrosine, and valine) and pyrolysine and selenocysteine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. [0056] Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. [0057] The term "nucleic acid" refers to deoxyribonucleotides, deoxyribonucleosides, ribonucleosides, or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers to oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like). Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions -9 7671816_1 (GHMatters) P83171.AU.2 may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic AcidRes. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Cassol et al. (1992); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)). [0058] The terms "isolated" and "purified" refer to a material that is substantially or essentially removed from or concentrated in its natural environment. For example, an isolated nucleic acid may be one that is separated from the nucleic acids that normally flank it or other nucleic acids or components (proteins, lipids, etc...) in a sample. In another example, a polypeptide is purified if it is substantially removed from or concentrated in its natural environment. Methods for purification and isolation of nucleic acids and proteins are well known in the art. The Blood Brain Barrier [0059] In one aspect, the invention provides compositions and methods that utilize an IDUA fused to an HIR Ab capable of crossing the blood brain barrier (BBB). The compositions and methods are useful in transporting IDUA from the peripheral blood and across the blood brain barrier into the CNS. As used herein, the "blood-brain barrier" refers to the barrier between the peripheral circulation and the brain and spinal cord which is formed by tight junctions within the brain capillary endothelial plasma membranes, creates an extremely tight barrier that restricts the transport of molecules into the brain, even molecules as small as urea, molecular weight of 60 Da. The blood-brain barrier within the brain, the blood-spinal cord barrier within the spinal cord, and the blood-retinal barrier within the retina, are contiguous capillary barriers within the central nervous system (CNS), and are collectively referred to as the blood-brain barrier or BBB. [0060] The BBB limits the development of new neurotherapeutics, diagnostics, and research tools for the brain and CNS. Essentially 100% of large molecule therapeutics such as recombinant proteins, antisense drugs, gene medicines, purified antibodies, or RNA interference (RNAi)-based drugs, do not cross the BBB in pharmacologically significant amounts. While it is generally assumed that small molecule drugs can cross the BBB, in fact, <2% of all small molecule drugs are active in the brain owing to the lack transport across the BBB. A molecule must be lipid soluble and have a molecular weight less than 400 Daltons (Da) in order to cross the BBB in pharmacologically significant amounts, and the vast majority of small molecules do not have these dual molecular characteristics. Therefore, most potentially therapeutic, diagnostic, or research molecules do not cross the BBB in pharmacologically active amounts. So as to bypass the BBB, invasive transcranial drug delivery strategies are used, such as intracerebro-ventricular (ICV) infusion, intracerebral (IC) administration, and convection enhanced diffusion (CED). Transcranial drug delivery to the brain is expensive, invasive, and largely ineffective. The ICV route delivers IDUA only to the ependymal surface of the brain, not into brain parenchyma, which is typical for drugs given by the ICV route. The IC administration of an enzyme such as IDUA, only provides local delivery, owing to the very low efficiency of protein diffusion within the brain. The CED results in preferential fluid flow through the white matter tracts of brain, which causes demyelination, and astrogliosis. -10 7671816_1 (GHMatters) P83171.AU.2 [0061] The methods described herein offer an alternative to these highly invasive and generally unsatisfactory methods for bypassing the BBB, allowing a functional IDUA to cross the BBB from the peripheral blood into the CNS following systemic administration of an HIR-IDUA fusion antibody composition described herein. The methods described herein exploit the expression of insulin receptors (e.g., human insulin receptors) on the BBB to shuttle desired a bifunctional HIR-IDUA fusion antibody from peripheral blood into the CNS. Insulin Receptors [0062] The BBB has been shown to have specific receptors, including insulin receptors, that allow the transport from the blood to the brain of several macromolecules. In particular, insulin receptors are suitable as transporters for the HIR Ab-IDUA fusion antibodies described herein. The HIR-IDUA fusion antibodies described herein bind to the extracellular domain (ECD) of the human insulin receptor. [0063] Insulin receptors and their extracellular, insulin binding domain (ECD) have been extensively characterized in the art both structurally and functionally. See, e.g., Yip eta (2003), "1Biol. Chem, 278(30):27329-27332; and Whittaker etal. (2005),JBiol Chem, 280(22):20932-20936. The amino acid and nucleotide sequences of the human insulin receptor can be found under GenBank accession No. NM_000208. Antibodies that bind to an insulin receptor mediated transport system [0064] One noninvasive approach for the delivery of IDUA to the CNS is to fuse the IDUA to an antibody that selectively binds to the ECD of the insulin receptor. Insulin receptors expressed on the BBB can thereby serve as a vector for transport of the IDUA across the BBB. Certain ECD-specific antibodies may mimic the endogenous ligand and thereby traverse a plasma membrane barrier via transport on the specific receptor system. Such insulin receptor antibodies act as molecular "Trojan horses," as depicted schematically in Fig. 7. Thus, despite the fact that antibodies and other macromolecules are normally excluded from the brain, they can be an effective vehicle for the delivery of molecules into the brain parenchyma if they have specificity for the extracellular domain of a receptor expressed on the BBB, e.g., the insulin receptor. In certain embodiments, an HIR Ab-IDUA fusion antibody binds an exofacial epitope on the human BBB HIR and this binding enables the fusion antibody to traverse the BBB via a transport reaction that is mediated by the human BBB insulin receptor. [0065] The term "antibody" describes an immunoglobulin whether natural or partly or wholly synthetically produced. The term also covers any polypeptide or protein having a binding domain which is, or is homologous to, an antigen-binding domain. CDR grafted antibodies are also contemplated by this term. [0066] "Native antibodies" and "native immunoglobulins" are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is typically linked to a heavy chain by one covalent disulfide bond, while the -11 7671816_1 (GHMatters) P83171.AU.2 number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain ("VH") followed by a number of constant domains ("CH"). Each light chain has a variable domain at one end ("VL") and a constant domain ("CL") at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light- and heavy-chain variable domains. [0067] The term "variable domain" refers to protein domains that differ extensively in sequence among family members (i.e. among different isoforms, or in different species). With respect to antibodies, the term "variable domain" refers to the variable domains of antibodies that are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the "framework region" or "FR". The variable domains of unmodified heavy and light chains each comprise four FRs (FR1, FR2, FR3 and FR4, respectively), largely adopting a $-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the D-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991), pages 647-669). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity. [0068] The term "hypervariable region" when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region comprises amino acid residues from three "complementarity determining regions" or "CDRs", which directly bind, in a complementary manner, to an antigen and are known as CDRI, CDR2, and CDR3 respectively. [0069] In the light chain variable domain, the CDRs typically correspond to approximately residues 24-34 (CDRL 1), 50-56 (CDRL2) and 89-97 (CDRL3), and in the heavy chain variable domain the CDRs typically correspond to approximately residues 31-35 (CDRHi), 50-65 (CDRH2) and 95-102 (CDRH3); Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a "hypervariable loop" (i.e. residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) and 96-101 (H3) in the heavy chainvariable domain; Chothia and Lesk J. Mol. Biol. 196:901 917 (1987)). [0070] As used herein, "variable framework region" or "VFR" refers to framework residues that form a part of the antigen binding pocket or groove and/or that may contact antigen. In some embodiments, the framework residues form a loop that is a part of the antigen binding pocket or groove. The amino acids residues in the loop may or may not contact the antigen. In an embodiment, the loop amino acids -12 7671816_1 (GHMatters) P83171.AU.2 of a VFR are determined by inspection of the three-dimensional structure of an antibody, antibody heavy chain, or antibody light chain. The three-dimensional structure can be analyzed for solvent accessible amino acid positions as such positions are likely to form a loop and/or provide antigen contact in an antibody variable domain. Some of the solvent accessible positions can tolerate amino acid sequence diversity and others (e.g. structural positions) can be less diversified. The three dimensional structure of the antibody variable domain can be derived from a crystal structure or protein modeling. In some embodiments, the VFR comprises, consist essentially of, or consists of amino acid positions corresponding to amino acid positions 71 to 78 of the heavy chain variable domain, the positions defined according to Kabat et al., 1991. In some embodiments, VFR forms a portion of Framework Region 3 located between CDRH2 and CDRH3. The VFR can form a loop that is well positioned to make contact with a target antigen or form a part of the antigen binding pocket. [0071] Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these can be further divided into subclasses (isotypes), e.g., IgGI, IgG2, IgG3, IgG4, IgA, and IgA2. The heavy-chain constant domains (Fc) that correspond to the different classes of immunoglobulins are called a, 6, E, y, and p, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. [0072] The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa or ("K") and lambda or ("k"), based on the amino acid sequences of their constant domains. [0073] In referring to an antibody or fusion antibody described herein, the terms "selectively bind," "selectively binding," "specifically binds," or "specifically binding" refer to binding to the antibody or fusion antibody to its target antigen for which the dissociation constant (Kd) is about 10-6 M or lower, i.e., 10-7, 10-8, 10, 10-m, 10-", or 10-2 M. [0074] The term antibody as used herein will also be understood to mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen, (see generally, Holliger et al., Nature Biotech. 23 (9) 1126-1129 (2005)). Non-limiting examples of such antibodies include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH 1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544 546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423 426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879 5883; and Osbourn et al. (1998) Nat. Biotechnol. 16:778). Such single chain antibodies are also intended to be encompassed within the -13 7671816_1 (GHMatters) P83171.AU.2 term antibody. Any VH and VL sequences of specific scFv can be linked to human immunoglobulin constant region cDNA or genomic sequences, in order to generate expression vectors encoding complete IgG molecules or other isotypes. VH and VL can also be used in the generation of Fab, Fv or other fragments of inmmunoglobulins using either protein chemistry or recombinant DNA technology. Other forms of single chain antibodies, such as diabodies are also encompassed. [0075] "F(ab')2" and "Fab"' moieties can be produced by treating immunoglobulin (monoclonal antibody) with a protease such as pepsin and papain, and includes an antibody fragment generated by digesting immunoglobulin near the disulfide bonds existing between the hinge regions in each of the two H chains. For example, papain cleaves IgG upstream of the disulfide bonds existing between the hinge regions in each of the two H chains to generate two homologous antibody fragments in which an L chain composed of VL (L chain variable region) and CL (L chain constant region), and an H chain fragment composed of VH (H chain variable region) and CHyl (yl region in the constant region of H chain) are connected at their C terminal regions through a disulfide bond. Each of these two homologous antibody fragments is called Fab'. Pepsin also cleaves IgG downstream of the disulfide bonds existing between the hinge regions in each of the two H chains to generate an antibody fragment slightly larger than the fragment in which the two above-mentioned Fab' are connected at the hinge region. This antibody fragment is called F(ab')2. [0076] The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH 1) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH 1 domain including one or more cysteine(s) from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known. [0077] "Fv" is the minimum antibody fragment which contains a complete antigen-recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. [0078] "Single-chain Fv" or "sFv" antibody fragments comprise a VH, a VL, or both a VH and VL domain of an antibody, wherein both domains are present in a single polypeptide chain. In some embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding. For a review of sFv see, e.g., Pluckthun in The Pharmacology of Monoclonal Antibodies, Vol. 113, Rosenburg and Moore eds. Springer-Verlag, New York, pp. 269 315 (1994). -14 7671816_1 (GHMatters) P83171.AU.2 [0079] A "chimeric" antibody includes an antibody derived from a combination of different mammals. The mammal may be, for example, a rabbit, a mouse, a rat, a goat, or a human. The combination of different mammals includes combinations of fragments from human and mouse sources. [0080] In some embodiments, an antibody of the present invention is a monoclonal antibody (MAb), typically a chimeric human-mouse antibody derived by humanization of a mouse monoclonal antibody. Such antibodies are obtained from, e.g., transgenic mice that have been "engineered" to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic mice can synthesis human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas. [0081] For use in humans, a chimeric HIR Ab is preferred that contains enough human sequence that it is not significantly immunogenic when administered to humans, e.g., about 80% human and about 20% mouse, or about 85% human and about 15% mouse, or about 90% human and about 10% mouse, or about 95% human and 5% mouse, or greater than about 95% human and less than about 5% mouse. Chimeric antibodies to the human BBB insulin receptor with sufficient human sequences for use in the invention are described in, e.g., Boado et al. (2007), Biotechnol Bioeng, 96(2):381-391. A more highly humanized form of the HIR MAb can also be engineered, and the humanized HIR Ab has activity comparable to the murine HIR Ab and can be used in embodiments of the invention. See, e.g., U.S. Patent Application Publication Nos. 20040101904, filed Nov. 27, 2002 and 20050142141, filed Feb. 17, 2005. [0082] In exemplary embodiments, the HIR antibodies or HIR-IDUA fusion antibodies derived therefrom contain an immunoglobulin heavy chain comprising CDRs corresponding to the sequence of at least one of the HC CDRs listed in Fig. 3 (SEQ ID NOs 1-3) or a variant thereof. For example, a HC CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 1, 2, 3, 4, 5, or 6 single amino acid mutations, a HC CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 single amino acid mutations, or a HC CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 1, or 2 single amino acid mutations, where the single amino acid mutations are substitutions, deletions, or insertions. [0083] In other embodiments, the HIR Abs or HIR Ab-IDUA fusion Abs contain an immunoglobulin HC the amino acid sequence of which is at least 50% identical ( i.e., at least, 55, 60, 65, 70, 75, 80, 85, 90, 95, or any other percent up to 100% identical) to SEQ ID NO:7 (shown in Fig. 1). [0084] In some embodiments, the HIR Abs or HIR AB-IDUA fusion Abs include an immunoglobulin light chain comprising CDRs corresponding to the sequence of at least one of the LC CDRs listed in Fig. 3 (SEQ ID NOs: 4-6) or a variant thereof. For example, a LC CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 1, 2, 3, 4, or 5 single amino acid mutations, a LC CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 1, 2, 3, or 4 single amino acid -15 7671816_1 (GHMatters) P83171.AU.2 mutations, or a LC CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 1, 2, 3, 4, or 5 single amino acid mutations. [0085] In other embodiments, the HIR Abs or HIR AB-IDUA fusion Abs contain an immunoglobulin LC the amino acid sequence of which is at least 50% identical (i.e., at least, 55, 60, 65, 70, 75, 80, 85, 90, 95, or any other percent up to 100% identical) to SEQ ID NO:8 (shown in Fig. 2). [0086] In yet other embodiments, the HIR Abs or HIR Ab-IDUA fusion Abs contain both a heavy chain and a light chain corresponding to any of the above-mentioned HIR heavy chains and HIR light chains. [0087] HIR antibodies used in the invention may be glycosylated or non-glycosylated. If the antibody is glycosylated, any pattern of glycosylation that does not significantly affect the function of the antibody may be used. Glycosylation can occur in the pattern typical of the cell in which the antibody is made, and may vary from cell type to cell type. For example, the glycosylation pattern of a monoclonal antibody produced by a mouse myeloma cell can be different than the glycosylation pattern of a monoclonal antibody produced by a transfected Chinese hamster ovary (CHO) cell. In some embodiments, the antibody is glycosylated in the pattern produced by a transfected Chinese hamster ovary (CHO) cell. [0088] One of ordinary skill in the art will appreciate that current technologies permit a vast number of sequence variants of candidate HIR Abs or known HIR Abs to be readily generated be (e.g., in vitro) and screened for binding to a target antigen such as the ECD of the human insulin receptor or an isolated epitope thereof. See, e.g., Fukuda et al. (2006) "In vitro evolution of single-chain antibodies using mRNA display," Nuc. AcidRes., 34(19) (published online) for an example of pltra high throughput screening of antibody sequence variants. See also, Chen et al. (1999), "In vitro scanning saturation mutagenesis of all the specificity determining residues in an antibody binding site," Prot Eng, 12(4): 349-356. An insulin receptor ECD can be purified as described in, e.g., Coloma et al. (2000) Pharm Res, 17:266-274, and used to screen for HIR Abs and HIR Ab sequence variants of known HIR Abs. [0089] Accordingly, in some embodiments, a genetically engineered HIR Ab, with the desired level of human sequences, is fused to an IDUA, to produce a recombinant fusion antibody that is a bi functional molecule. The HIR Ab-IDUA fusion antibody: (i) binds to an extracellular domain of the human insulin receptor; (ii) catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate; and (iii) is able to cross the BBB, via transport on the BBB HIR, and retain IDUA activity once inside the brain, following peripheral administration. -L-Iduronidase (IDUA) [0090] Systemic administration (e.g., by intravenous injection) of recombinant IDUA (e.g., Aldurazyme*) fails to rescue a deficiency of IDUA in the CNS of patients suffering from Hurler's syndrome. IDUA does not cross the BBB, and the lack of transport of the enzyme across the BBB prevents it from having a significant therapeutic effect in the CNS following peripheral administration. However, when the IDUA is fused to an HIR Ab (e.g., by a linker), this enzyme is now able to enter the CNS from blood following a non-invasive peripheral route of administration such as intravenous, -16 7671816_1 (GHMatters) P83171.AU.2 intra-arterial, intramuscular, subcutaneous, intraperitoneal, or even oral administration. Administration of a HIR Ab-IDUA fusion antibody enables delivery of IDUA activity into the brain from peripheral blood. Described herein is the determination of a systemic dose of the HIR Ab-IDUA fusion antibody that is therapeutically effective for treating an IDUA deficiency in the CNS. As described herein, appropriate systemic doses of an HIR Ab-IDUA fusion antibody are established based on a quantitative determination of CNS uptake characteristics and enzymatic activity of an HIR Ab-IDUA fusion antibody. [0091] As used herein, IDUA refers to any naturally occurring or artificial enzyme that can catalyze the hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate, e.g., the human IDUA sequence listed under GenBank Accession No. NP_000194. [0092] In some embodiments, IDUA has an amino acid sequence that is a at least 50% identical (i.e., at least, 55, 60, 65, 70, 75, 80, 85, 90, 95, or any other percent up to 100% identical) to the amino acid sequence of human IDUA, a 653 amino acid protein listed under GenBank Accession No. NP 000194, or a 627 amino acid subsequence thereof, which lacks a 26 amino acid signal peptide, and corresponds to SEQ ID NO:9 (Fig. 4). The structure-function relationship of human IDUA is well established, as described in, e.g., Rempel et al. (2005), "A homology model for human a-L Iduronidase: Insights into human disease," Mol. Genetics andMet., 85:28-37. In particular, residues that are critical to the function of IDUA include, e.g., Gly 51, Ala 75, Ala 160, Glu 182, Gly 208, Leu 218, Asp 315, Ala 327, Asp 349, Thr 366, Thr 388, Arg 489, Arg 628, Ala 79, His 82, Glu 178, Ser 260, Leu 346, Asn 350, Thr 364, Leu 490, Pro 496, Pro 533, Arg 619, Arg 89, Cys 205, His 240, Ala 319, Gln 380, Arg 383, and Arg 492. [0093] In some embodiments, IDUA has an amino acid sequence at least 50% identical (i.e., at least, 55, 60, 65, 70, 75, 80, 85, 90, 95, or any other percent up to 100% identical) to SEQ ID NO:9 (shown in Fig. 4). Sequence variants of a canonical IDUA sequence such as SEQ ID NO:9 can be generated, e.g., by random mutagenesis of the entire sequence or specific subsequences corresponding to particular domains. Alternatively, site directed mutagenesis can be performed reiteratively while avoiding mutations to residues known to be critical to IDUA function such as those given above. Further, in generating multiple variants of an IDUA sequence, mutation tolerance prediction programs can be used to greatly reduce the number of non-functional sequence variants that would be generated by strictly random mutagenesis. Various programs) for predicting the effects of amino acid substitutions in a protein sequence on protein function (e.g., SIFT, PolyPhen, PANTHER PSEC, PMUT, and TopoSNP) are described in, e.g., Henikoff et al. (2006), "Predicting the Effects of Amino Acid Substitutions on Protein Function," Annu. Rev. Genomics Hum. Genet., 7:61-80. IDUA sequence variants can be screened for of IDUA activity/retention of IDUA activity by, e.g., 4-methylumbelliferyl a-L-iduronide (MUBI) flurometric IDUA assays known in the art. See, e.g., Kakkis et al. (1994), Prot Expr Purif 5:225-232. One unit of IDUA activity is defined as the hydrolysis of 1 nmole substrate/hour. Accordingly, one of ordinary skill in the art will appreciate that a very large number of operable IDUA sequence variants can be obtained by generating and screening -17 7671816_1 (GHMatters) P83171.AU.2 extremely diverse "libraries" of IDUA sequence variants by methods that are routine in the art, as described above. [0094] Percent sequence identity is determined by conventional methods. See, for example, Altschul et al., Bull. Math. Bio. 48:603 (1986), and Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1, and the "BLOSUM62" scoring matrix of Henikoff and Henikoff (ibid.). The percent identity is then calculated as: ([Total number of identical matches]/[length of the longer sequence plus the number of gaps introduced into the longer sequence in order to align the two sequences])(100). [0095] Those skilled in the art appreciate that there are many established algorithms available to align two amino acid sequences. The "FASTA" similarity search algorithm of Pearson and Lipman is a suitable protein alignment method for examining the level of identity shared by an amino acid sequence disclosed herein and the amino acid sequence of another peptide. The FASTA algorithm is described by Pearson and Lipman, Proc. Nat'l Acad. Sci. USA 85:2444 (1988), and by Pearson, Meth. Enzymol. 183:63 (1990). Briefly, FASTA first characterizes sequence similarity by identifying regions shared by the query sequence (e.g., SEQ ID NO:24 or SEQ ID NO: 39) and a test sequence that have either the highest density of identities (if the ktup variable is 1) or pairs of identities (if ktup=2), without considering conservative amino acid substitutions, insertions, or deletions. The ten regions with the highest density of identities are then rescored by comparing the similarity of all paired amino acids using an amino acid substitution matrix, and the ends of the regions are "trimmed" to include only those residues that contribute to the highest score. If there are several regions with scores greater than the "cutoff' value (calculated by a predetermined formula based upon the length of the sequence and the ktup value), then the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps. Finally, the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman-Wunsch-Sellers algorithm (Needleman and Wunsch, J. Mol. Biol. 48:444 (1970); Sellers, SIAM J. Appl. Math. 26:787 (1974)), which allows for amino acid insertions and deletions. Illustrative parameters for FASTA analysis are: ktup=1, gap opening penalty= 10, gap extension penalty=1, and substitution matrix=BLOSUM62. These parameters can be introduced into a FASTA program by modifying the scoring matrix file ("SMATRIX"), as explained in Appendix 2 of Pearson, Meth. Enzymol. 183:63 (1990). [0096] The present invention also includes proteins having a conservative amino acid change, compared with an amino acid sequence disclosed herein. Among the common amino acids, for example, a "conservative amino acid substitution" is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) seine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine. The BLOSUM62 table is an amino acid substitution matrix derived from about 2,000 local multiple alignments of protein sequence segments, representing highly conserved regions of more than 500 groups of related proteins (Henikoff -18 7671816_1 (GHMatters) P83171.AU.2 and Henikoff, Proc. Nat'l Acad. Sci. USA 89:10915 (1992)). Accordingly, the BLOSUM62 substitution frequencies can be used to define conservative amino acid substitutions that may be introduced into the amino acid sequences of the present invention. Although it is possible to design amino acid substitutions based solely upon chemical properties (as discussed above), the language "conservative amino acid substitution" preferably refers to a substitution represented by a BLOSUM62 value of greater than -1. For example, an amino acid substitution is conservative if the substitution is characterized by a BLOSUM62 value of 0, 1, 2, or 3. According to this system, preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 1 (e.g., 1, 2 or 3), while more preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 2 (e.g., 2 or 3). [0097] It also will be understood that amino acid sequences may include additional residues, such as additional N- or C-terminal amino acids, and yet still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence retains sufficient biological protein activity to be functional in the compositions and methods of the invention. Compositions [0098] Strikingly, it has been found that the bifunctional HIR Ab-IDUA fusion antibodies described herein, retain a high proportion of the activity of their separate constituent proteins, i.e., binding of the HIR Ab to the IR ECD and transport across the BBB, and the enzymatic activity of IDUA. Construction of cDNAs and expression vectors encoding any of the proteins described herein, as well as their expression and purification are well within those of ordinary skill in the art, and are described in detail herein in, e.g., Examples 1-3, and , in Boado et al (2007), Biotechnol Bioeng 96:381-391, U.S Patent Application Serial No. 11/061,956, and U.S. Patent Application Serial No. 11/245,710. [0099] Described herein are bifunctional HIR Ab-IDUA fusion antibodies containing a HIR Ab, as described herein, capable of crossing the BBB fused to IDUA, where the HIR Ab is capable of crossing the blood brain barrier and the IDUA each retain an average of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, 99, or 100% of their activities, compared to their activities as separate entities. In some embodiments, the invention provides a HIR Ab-IDUA fusion antibody where the HIR Ab and IDUA each retain an average of at least about 50% of their activities, compared to their activities as separate entities. In some embodiments, the invention provides a HIR Ab-IDUA fusion antibody where the HIR Ab and IDUA each retain an average of at least about 60% of their activities, compared to their activities as separate entities. In some embodiments, the invention provides a HIR Ab-IDUA fusion antibody where the HIR Ab and IDUA each retain an average of at least about 70% of their activities, compared to their activities as separate entities. In some embodiments, the invention provides a HIR Ab-IDUA fusion antibody where the HIR Ab and IDUA each retain an average of at least about 80% of their activities, compared to their activities as separate entities. In some embodiments, the invention provides a fusion HIR Ab-IDUA fusion antibody where the HIR Ab and IDUA each retain an average of at least about 90% of their activities, compared to their activities as separate entities. In some embodiments, the HIR Ab retains at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, 99, or 100% of its -19 7671816_1 (GHMatters) P83171.AU.2 activity, compared to its activity as a separate entity, and the IDUA retains at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, 99, or 100% of its activity, compared to its activity as a separate entity. Accordingly, described herein are compositions containing a bifunctional HIR Ab-IDUA fusion antibody capable of crossing the BBB, where the constituent HIR Ab and IDUA each retain, as part of the fusion antibody, an average of at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 95, 99, or 100% of their activities, i.e., HIR binding and IDUA activity, respectively, compared to their activities as separate proteins. An HIR Ab IDUA fusion antibody refers to a fusion protein comprising any of the HIR antibodies and IDUAs described herein. [00100] In the HIR Ab-IDUA fusion antibodies described herein, the covalent linkage between the antibody and the IDUA may be to the carboxy or amino terminal of the HIR antibody and the amino or carboxy terminal of the IDUA as long as the linkage allows the HIR Ab-IDUA fusion antibody to bind to the ECD of the IR and cross the blood brain barrier, and allows the IDUA to retain a therapeutically useful portion of its activity. In certain embodiments, the covalent link is between an HC of the antibody and the IDUA or a LC of the antibody and the IDUA. Any suitable linkage may be used, e.g., carboxy terminus of light chain to amino terminus of IDUA, carboxy terminus of heavy chain to amino terminus of IDUA, amino terminus of light chain to amino terminus of IDUA, amino terminus of heavy chain to amino terminus of IDUA, carboxy terminus of light chain to carboxy terminus of IDUA, carboxy terminus of heavy chain to carboxy terminus of IDUA, amino terminus of light chain to carboxy terminus of IDUA, or amino terminus of heavy chain to carboxy terminus of IDUA. In some embodiments, the linkage is from the carboxy terminus of the HC to the amino terminus of the IDUA. [00101] It will be appreciated that a linkage between terminal amino acids can be accomplished by an intervening peptide linker sequence that forms part of the fused amino acid sequence. The peptide sequence linker may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 amino acids in length. In some embodiments, a two amino acid linker is used. In some embodiments, the linker has the sequence ser ser. The peptide linker sequence may include a protease cleavage site, however this is not a requirement for activity of the IDUA; indeed, an advantage of these embodiments of the present invention is that the bifunctional HIR Ab-IDUA fusion antibody, without cleavage, is partially or fully active both for transport and for activity once across the BBB. Fig. 5 shows an exemplary embodiment of the amino acid sequence of a HIR Ab-IDUA fusion antibody (SEQ ID NO:10) in which the HC is fused through its carboxy terminus via a two amino acid "ser-ser" linker to the amino terminus of the IDUA. In some embodiments, the fused IDUA sequence is devoid of its 26 amino acid signal peptide, as shown in Fig. 4. [00102] In some embodiments, a HIR Ab-IDUA fusion antibody comprises both a HC and a LC. In some embodiments, the HIR Ab-IDUA fusion antibody is a monovalent antibody. In other embodiments, the HIR Ab-IDUA fusion antibody is a divalent antibody, as described herein in the Example section. -20 7671816_1 (GHMatters) P83171.AU.2 [00103] The HIR Ab used as part of the HIR Ab-IDUA fusion antibody can be glycosylated or nonglycosylated; in some embodiments, the antibody is glycosylated, e.g., in a glycosylation pattern produced by its synthesis in a CHO cell. [00104] As used herein, "activity" includes physiological activity (e.g., ability to cross the BBB and/or therapeutic activity), binding affinity of the HIR Ab for the IR ECD, or the enzymatic activity of IDUA. [00105] Transport of a HIR Ab-IDUA fusion antibody across the BBB may be compared to transport across the BBB of the HIR Ab alone by standard methods. For example, pharmacokinetics and brain uptake of the HIR Ab-IDUA fusion antibody by a model animal, e.g., a mammal such as a primate, may be used. Such techniques are illustrated in Example 5, which demonstrates pharmacokinetics and brain uptake of a fusion protein of the invention by the adult Rhesus monkey. Similarly, standard models for determining IDUA activity may also be used to compare the function of the IDUA alone and as part of a HIR Ab-IDUA fusion antibody. See, e.g., Example 3, which demonstrates the enzymatic activity of IDUA versus HIR Ab-IDUA fusion antibody. Binding affinity for the IR ECD can be compared for the HIR Ab-IDUA fusion antibody versus the HIR Ab alone. See, e.g., Example 4 herein. [00106] Also included herein are pharmaceutical compositions that contain one or more HIR Ab IDUA fusion antibodies described herein and a pharmaceutically acceptable excipient. A thorough discussion of pharmaceutically acceptable carriers/excipients can be found in Remington's Pharmaceutical Sciences, Gennaro, AR, ed., 20th edition, 2000: Williams and Wilkins PA, USA. Pharmaceutical compositions of the invention include compositions suitable for administration via any peripheral route, including intravenous, subcutaneous, intramuscular, intraperitoneal injection; oral, rectal, transbuccal, pulmonary, transdermal, intranasal, or any other suitable route of peripheral administration. [00107] The compositions of the invention are particular suited for injection, e.g., as a pharmaceutical composition for intravenous, subcutaneous, intramuscular, or intraperitoneal administration. Aqueous compositions of the present invention comprise an effective amount of a composition of the present invention, which may be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. The phrases "pharmaceutically or pharmacologically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, e.g., a human, as appropriate. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. [00108] Exemplary pharmaceutically acceptable carriers for injectable compositions can include salts, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. -21 7671816_1 (GHMatters) P83171.AU.2 For example, compositions of the invention may be provided in liquid form, and formulated in saline based aqueous solution of varying pH (5-8), with or without detergents such polysorbate-80 at 0.01 1%, or carbohydrate additives, such mannitol, sorbitol, or trehalose. Commonly used buffers include histidine, acetate, phosphate, or citrate. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol; phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate, and gelatin. [00109] For human administration, preparations meet sterility, pyrogenicity, general safety, and purity standards as required by FDA and other regulatory agency standards. The active compounds will generally be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, subcutaneous, intralesional, or intraperitoneal routes. The preparation of an aqueous composition that contains an active component or ingredient will be known to those of skill in the art in light of the present disclosure. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use in preparing solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. [00110] Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation include vacuum-drying and freeze drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. [00111] Upon formulation, solutions will be systemically administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective based on the criteria described herein. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed [00112] The appropriate quantity of a pharmaceutical composition to be administered, the number of treatments, and unit dose will vary according to the CNS uptake characteristics of a HIR Ab-IDUA fusion antibody as described herein, and according to the subject to be treated, the state of the subject and the effect desired. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. [00113] In addition to the compounds formulated for parenteral administration, such as intravenous or intramuscular injection, other alternative methods of administration of the present invention may also -22 7671816_1 (GHMatters) P83171.AU.2 be used, including but not limited to intradermal administration (See U.S. Pat. Nos. 5,997,501; 5,848,991; and 5,527,288), pulmonary administration (See U.S. Pat.. Nos. 6,361,760; 6,060,069; and 6,041,775), buccal administration (See U.S. Pat. Nos. 6,375,975; and 6,284,262), transdermal administration (See U.S. Pat. Nos. 6,348,210; and 6,322,808) and transmucosal administration (See U.S. Pat. No. 5,656,284). Such methods of administration are well known in the art. One may also use intranasal administration of the present invention, such as with nasal solutions or sprays, aerosols or inhalants. Nasal solutions are usually aqueous solutions designed to be administered to the nasal passages in drops or sprays. Nasal solutions are prepared so that they are similar in many respects to nasal secretions. Thus, the aqueous nasal solutions usually are isotonic and slightly buffered to maintain a pH of 5.5 to 6.5. In addition, antimicrobial preservatives, similar to those used in ophthalmic preparations and appropriate drug stabilizers, if required, may be included in the formulation. Various commercial nasal preparations are known and include, for example, antibiotics and antihistamines and are used for asthma prophylaxis. [00114] Additional formulations, which are suitable for other modes of administration, include suppositories and pessaries. A rectal pessary or suppository may also be used. Suppositories are solid dosage forms of various weights and shapes, usually medicated, for insertion into the rectum or the urethra. After insertion, suppositories soften, melt or dissolve in the cavity fluids. For suppositories, traditional binders and carriers generally include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in any suitable range, e.g., in the range of 0.5% to 10%, preferably 1%-2%. [00115] Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations, or powders. In certain defined embodiments, oral pharmaceutical compositions will comprise an inert diluent or assimilable edible carrier, or they may be enclosed in a hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations can contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied, and may conveniently be between about 2 to about 75% of the weight of the unit, or between about 25-60%. The amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained. [00116] The tablets, troches, pills, capsules and the like may also contain the following: a binder, such as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various -23 7671816_1 (GHMatters) P83171.AU.2 other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup of elixir may contain the active compounds sucrose as a sweetening agent, methylene and propyl parabens as preservatives, a dye and flavoring, such as cherry or orange flavor. In some embodiments, an oral pharmaceutical composition may be enterically coated to protect the active ingredients from the environment of the stomach; enteric coating methods and formulations are well-known in the art. Methods [00117] Described herein are methods for delivering an effective dose of IDUA to the CNS across the BBB by systemically administering a therapeutically effective amount of a HIR Ab-IDUA fusion antibody, as described herein. Suitable systemic doses for delivery of a HIR Ab-IDUA fusion antibody is based on its CNS uptake characteristics and IDUA specific activity as described herein. Systemic administration of a HIR Ab-IDUA fusion antibody to a subject suffering from an IDUA deficiency is an effective approach to the non-invasive delivery of IDUA to the CNS. [00118] The amount of a HIR-IDUA fusion antibody that is a therapeutically effective systemic dose of a HIR Ab-IDUA fusion antibody depends, in part, on the CNS uptake characteristics of the HIR IDUA fusion antibody to be administered, as described herein., e.g., the percentage of the systemically administered dose to be taken up in the CNS, [00119] In some embodiments, 0.3% (i.e., about 0.32%, 0.4%, 0.48%, 0.6%, 0.74%, 0.8%, 0.
9 %, 1.05, 1.1, 1.2, 1.3%, 1.5%, 2%, 2.5%, 5%, or any %from about 0.3%to about 12%N) of the systemically administered HIR Ab-IDUA fusion antibody is delivered to the brain as a result of its uptake from peripheral blood across the BBB. In some embodiments, at least 0.5%, (i.e., about 0.32%, 0.4%, 0.48%, 0.6%, 0.74%, 0.8%, 0.9%, 1.05, 1.1, 1.2, 1.3%, 1.5%, 2%, 2.5%, 5%, or any % from about 0.3% to about 12%) of the systemically administered dose of the HIR Ab-IDUA fusion antibody is delivered to the brain within two hours or less, i.e., 1.8, 1.7, 1.5, 1.4, 1.3, 1.2, 1.1, 0.9, 0.8, 0.6, 0.5 or any other period from about 0.5 to about two hours after systemic administration. [00120] Accordingly, in some embodiments the invention provides methods of administering a therapeutically effective amount of a HIR Ab-IDUA fusion antibody systemically, such that the amount of the HIR Ab-IDUA fusion antibody to cross the BBB provides at least 0.2 units of IDUA activity/mg protein in the subject's brain, i.e., 0.21, 0.22, 0.25, 0.4, 0.5, 0.6, 0.7, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 2, 2.2, 2.4, 2.5, 2.7, 2.8, 3, 4, or any other value from 0.2 to 4 of units of IDUA activity/mg protein in the subject's brain. [00121] In some embodiments, the total number of units of IDUA activity delivered to a subject's brain is at least, 25,000 units, i.e., at least 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 60,000, 70,000, 80,000, 90,000, 110,000, 120,000, 130,000, 140,000, 200,000, 210,000, 220,000, 230,000, 240,000, 250,000, 260,000, 270,000, 280,000, 300,000 or any other total number of IDUA units from about 25,000 to 300,000 units of IDUA activity. [00122] In some embodiments, a therapeutically effective systemic dose comprises at least 5 x 10 5 , 1 x 106,2 x 106, 3 x 106, 4, 106, 5 x 106,6 x 106,7 x 106, 8 x 106, 9 x 106, 1 X 10 7 , 1.1 X 10 7 , 1.2 x 107, 1.5 x -24 7671816_1 (GHMatters) P83171.AU.2 10', 1.6 x 10', 1.7 x 10', 1.8 x 10, 1.9 x 107, 2 x 107, 2.1 x 107, 3 x 107, or any other systemic dose from about 5 x 10 5 to 3 x 107 units of IDUA activity. [00123] In other embodiments, a therapeutically effective systemic dose is at least about 100,000 units of IDUA activity/kg body weight, i.e., at least about 110,000, 120,000, 130,000, 140,000, 200,000, 210,000, 220,000, 230,000, 240,000, 250,000, 260,000, 270,000, 280,000, 300,000 or any other number of IDUA units from about 110,000 to 300,000 units of IDUA activity/kg of body weight. [00124] One of ordinary skill in the art will appreciate that the mass amount of a therapeutically effective systemic dose of a HIR Ab-IDUA fusion antibody will depend, in part, on its IDUA specific activity. In some embodiments, the IDUA specific activity of a HIR Ab-IDUA fusion antibody is at least 100,000 U/mg of protein, i.e., at least about 110,000, 120,000, 130,000, 140,000, 200,000, 210,000, 220,000, 230,000, 240,000, 250,000, 260,000, 270,000, 280,000, 300,000, 320,000, 340,000, 350,000, 360,000, 370,000, 373,000, 400,000, 500,000, or any other specific activity value from about 100,000 units/mg to about 500,000 units/mg. [00125] Thus, with due consideration of the specific activity of a HIR Ab-IDUA fusion antibody and the body weight of a subject to be treated, a systemic dose of the HIR Ab-IDUA fusion antibody can be at least 2 mg, i.e., 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 100, or any other value from about 2 mg to about 100 mg of HIR Ab-IDUA fusion antibody. [00126] The term "systemic administration" or "peripheral administration," as used herein, includes any method of administration that is not direct administration into the CNS, i.e., that does not involve physical penetration or disruption of the BBB. "Systemic administration" includes, but is not limited to, intravenous , intra-arterial intramuscular, subcutaneous, intraperitoneal, intranasal, transbuccal, transdermal, rectal, transalveolar (inhalation), or oral administration. Any suitable HIR Ab-IDUA fusion antibody, as described herein, may be used. [00127] An IDUA deficiency as referred to herein includes, one or more conditions known as Hurler's syndrome, Hurler's disease, mucopolysaccharidosis type I, Scheie sydrome (MPS I S), and Hurler Scheie (MPS I H-S). The IDUA deficiency is characterized by the buildup of heparan sulfate and dermatan sulfate occurs in the body (the heart, liver, brain etc.). [00128] The compositions of the invention, i.e., an HIR Ab-IDUA fusion antibody may be administered as part of a combination therapy. The combination therapy involves the administration of a composition of the invention in combination with another therapy for treatment or relief of symptoms typically found in a patient suffering from an IDUA deficiency. If the composition of the invention is used in combination with another CNS disorder method or composition, any combination of the composition of the invention and the additional method or composition may be used. Thus, for example, if use of a composition of the invention is in combination with another CNS disorder treatment agent, the two may be administered simultaneously, consecutively, in overlapping durations, in similar, the same, or different frequencies, etc. In some cases a composition will be used that contains a composition of the invention in combination with one or more other CNS disorder treatment agents. -25 7671816_1 (GHMatters) P83171.AU.2 [00129] In some embodiments, the composition, e.g., an HIR Ab-IDUA fusion antibody is co administered to the patient with another medication, either within the same formulation or as a separate composition. For example, the HIR Ab-IDUA fusion antibody could be formulated with another fusion protein that is also designed to deliver across the human blood-brain barrier a recombinant protein other than IDUA. Further, the fusion HIR Ab-IDUA fusion antibody may be formulated in combination with other large or small molecules. EXAMPLES [00130] The following specific examples are to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. Without further elaboration, it is believed that one skilled in the art can, based on the description herein, utilize the present invention to its fullest extent. All publications cited herein are hereby incorporated by reference in their entirety. Where reference is made to a URL or other such identifier or address, it is understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information. Example 1 Construction of human HIR Ab heavy chain-IDUA fusion protein expression vector [00131] Human IDUA cDNA corresponding to amino acids Metl-Pro653 of the mature human IDUA protein, including the 26 amino acid signal peptide (NP 00194), was cloned by reverse transcription (RT) polymerase chain reaction (PCR) using the oligodeoxynucleotides (ODNs) listed in Table 1, designated 'IDUA forward primer' and 'IDUA reverse primer', and human liver polyA+ RNA (Clontech). Human liver cDNA was prepared using the Super Script first-strand synthesis kit (Invitrogen, San Diego, CA) and oligodeoxythymidine priming as per the manufacturer's instructions. The IDUAcDNA was cloned by PCR using 2 pl liver cDNA reverse transcription reaction, 0.2 pM IDUA forward and reverse ODN primers (Table 1), 0.2 mM dNTPs and 2.5 U PfuUltraDNA polymerase (Stratagene, San Diego, CA) in a 50 pl Pfu buffer (Stratagene). The amplification was performed in a Master cycler temperature cycler (Eppendorf, Hamburg, Germany) with an initial denaturing step of 95 0 C for 2 min followed by 30 cycles of denaturing at 95 0 C for 30 sec, annealing at 55 0 C for 30 sec and amplification at 72 0 C for 1 min. PCR products were resolved in 1% agarose gel electrophoresis, and the expected major single band of ~1.9 kb corresponding to the human IDUA cDNA was isolated (Fig. 8). The cloned human IDUA was inserted into the pcDNA eukaryotic expression plasmid at the EcoRV site, and this IDUA expression plasmid was designated pCD-IDUA. The entire expression cassette of the plasmid was confirmed by sequencing both strands. -26 7671816_1 (GHMatters) P83171.AU.2 Table 1 Nucleotide Oligodeoxynucleotide primers used in the RT-PCR cloning of human IDUA IDUA FORWARD PRIMER: 5'-phosphate-GCGTGGCCATGCGTCCCCTGCGCCCCCGCGCCGCGCTGCTGGCGCTCCTG (SEQ IDNO:11) IDUA-signal peptide FORWARD PRIMER: 5'-phosphate-CAGAGGCCCCGCACCTGGTGCAGGTGGACGCGGCCCGCGCGCTGTG (SEQ ID NO:12) IDUA REVERSE PRIMER: 5'-phosphate-TCATGGATTGCCCGGGGATGGGGGCCCTCTTGGCACAGGGACC (SEQ ID NO:13) [00132] DNA sequencing of the expression cassette of the pCD-IDUA encompassed 3,085 nucleotides (nt), including a 715 nt CMV promoter, a 1,962 nt IDUA open reading frame, and a 401 nt BGH sequence, which predicted a 653 amino acid human IDUA protein, including a 26 amino acid signal peptide with 100% identity with the known sequence for human IDUA (GenBank Accession No.: NP_000194). [00133] The pHIR Ab-HC plasmid encodes the heavy chain of a human-mouse chimeric HIR Ab, and pHIR Ab-LC encodes the LC of the chimeric HIR Ab. The HC and LC expression vectors are comprised of intronless cDNA open reading frames (orf), and these cDNAs were obtained by RT-PCR of NS0/1 myeloma cell lines transfected with chromosomal derived HIR Ab HC and LC intron-bearing vectors as described in detail inBoado et al (2007), BiotechnolBioeng 96:381-391. See also U.S Patent Application Serial No. 11/061,956. The sequence of the HIR Ab HC (SEQ ID NO:7) HIR Ab LC (SEQ ID NO:8) are shown in Figs 1 and 2, respectively. The sequences of the CDRs of the HIR Ab HC (SEQ ID NOs: 1-3) and HIR Ab LC (SEQ ID NOs:4-6) are shown in Fig. 3. [00134] The HIR Ab HC and LC intronless cDNA expression vectors are driven by the cytomegalovirus (CMV) promoter and contain the bovine growth hormone (BGH) polyadenylation (pA) sequence. The engineering of a universal pHIR Ab-HC vector was performed by insertion of a single HpaI site at the end of the HIR Ab HC CH3 open reading frame (ORF) by site directed mutagenesis (SDM), as described previously in Boado et al. supra. All constructs were validated by bi-directional DNA sequencing. [00135] For the engineering of the expression plasmid encoding the fusion protein of the heavy chain (HC) of the HIR Ab and IDUA, designated pCD-HC-IDUA, the human IDUA, minus the 26 amino acid signal peptide, was cloned again by PCR, using the ODNs designated 'IDUA-signal peptide forward primer' and 'IDUA reverse primer.' in Table 1. The ODNs used for PCR are 5' phosphorylated for direct insertion of the PCR product into the HpaI site of the pHIR Ab-HC -27 7671816_1 (GHMatters) P83171.AU.2 expression plasmid. The IDUA-signal peptide forward PCR primer (Table 1) introduces "CA" nucleotides to maintain the open reading frame and to introduce a Ser-Ser linker between the carboxyl terminus of the CH3 region of the HIR Ab HC and the amino terminus of the IDUA minus the 26 amino acid signal peptide of the enzyme. The IDUA reverse PCR primer introduces a stop codon, "TGA," immediately after the terminal Pro of the mature human IDUA protein. The fused IDUA amino acid sequence (SEQ ID NO:9) and the sequence of HIR Ab HC-IDUA fusion protein (SEQ ID NO: 10) are shown in Figs. 4 and 5, respectively. A schematic depiction of the bivalent HIR Ab-IDUA fusion antibody is shown in Fig. 6. [00136] DNA sequencing of the expression cassette of the pCD-HC-IDUA encompassed 4,369 nt, including a 714 nt CMV promoter, a 9 nt full Kozak site (GCCGCCACC), a 3,276 nt HIR Ab HC IDUA fusion protein open reading frame, and a 370 nt BGH sequence. The plasmid encoded for a 1,091 amino acid protein, comprised of a 19 amino acid IgG signal peptide, the 443 amino acid HIR Ab HC, a 2 amino acid linker (Ser-Ser), and the 627 amino acid human IDUA minus the enzyme signal peptide. The predicted molecular weight of the heavy chain fusion protein, minus glycosylation, is 118,836 Da, with a predicted isoelectric point (pI) of 8.89. Example 2 Expression analysis of IDUA and HIR Ab-IDUA fusion protein in COS cells [00137] COS cells were plated in 6-well cluster dishes, and were either transfected with the pCD IDUA, or dual transfected with pHIR Ab-LC and pCD-HC-IDUA using Lipofectamine 2000, with a ratio of 1:2.5, pg DNA:uL Lipofectamine, and conditioned serum free medium was collected at 3 and 7 days. IDUA enzyme activity was measured in both the medium and in the intracellular compartment. The washed monolayer was lysed in 0.4 M sodium formate, pH=3.5, 0.2% Triton X-100; the lysate was sonicated 7 sec 3 times on ice, centrifuged, and the supernatant was taken for IDUA enzyme assay (He et al, (1999), Mol GenetMetab, 67:106-112). Transfection of COS cells with pCD-IDUA resulted in high levels of IDUA enzyme activity in both the intracellular compartment and in the medium at 3 and 7 days following transfection, as shown in Table 2. -28 7671816_1 (GHMatters) P83171.AU.2 Table 2 IDUA enzyme activity in COS cells following transfection with pCD-IDUA or cotransfection with pCD-HC-IDUA and pHIR Ab-LC Treatment kntrac&eiuar actdy Medium actd y (nrJmd~hig: (nmcdlmO) 3 days Upofectaminfe 2000 75 i7 5.2 i 1-4 pCD-IDUA 4070 ±108 1S74 156 pCD-HG-DUA, plus 530 i 34 240 ± 25 pCD-LC-1 7 days Upofectamine 2000 203 ± 80 27 ± 8 pCD-IDUA 79619 858 286 25 pCD-HCADUA plus 146 ±D 136 11 4 ± 83 pCD-LC-1 Meain SE (n=3 dishes per point). [00138] Dual transfection of COS cells with the pCD-HC-IDUA and the pHIR Ab-LC resulted in medium levels of IDUA enzyme activity of 240 ± 25 and 1,194 ± 83 nmol/hr/mL at 3 and 7 days after transfection, respectively (Table 2). The COS intracellular IDUA enzyme activity at 3 and 7 days is 530 ± 34 and 1,460 ± 136 nmol/hr/mg protein, respectively (Table 2). For production of larger amounts of fusion protein, COS cells were transfected in 10 x T500 flasks. The 3 day and 7 day medium was pooled, and the 2 L of serum free conditioned medium was concentrated to 400 mL with tangential flow filtration (Millipore) followed by purification with protein A affinity chromatography. [00139] The purity of protein A purified fusion protein produced by COS cells was evaluated with 12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDSPAGE) with 5% D mercaptoethanol. Immunoreactivity was tested with a primary rabbit antibody to human IDUA, provided by Prof. E. Neufeld, UCLA, or a primary goat antiserum against human IgG heavy and light chains (Vector Labs, Burlingame, CA). [00140] On Western blotting of the purified HIR Ab-IDUA fusion protein, the anti-human IgG antibody reacts with a 130 kDa HC for the fusion protein, and a 50 kDa HC for the chimeric HIR Ab, and the difference in size, 80 kDa, is due to the fusion of IDUA (Fig. 9, right panel). The anti-human IgG antibody reacts equally with the light chain of either the HIR Ab-IDUA fusion protein or the HIR Ab, since both proteins are comprised of the same light chain. The anti-IDUA antibody reacts with the 130 kDa HC of the fusion protein, but not with the HC of the chimeric HIR Ab (Fig. 9, left panel). -29 7671816_1 (GHMatters) P83171.AU.2 Example 3 Analysis of HIR binding and IDUA activity [00141] The affinity of the fusion protein for the HIR extracellular domain (ECD) was determined with an ELISA. CHO cells permanently transfected with the HIR ECD were grown in serum free media (SFM), and the HIR ECD was purified with a wheat germ agglutinin affinity column, as previously described in Coloma et al. (2000) Pharm Res, 17:266-274. The HIR ECD was plated on Nunc-Maxisorb 96 well dishes and the binding of the HIR Ab, or the HIR Ab-IDUA fusion protein, to the HIR ECD was detected with a biotinylated goat anti-human IgG (H+L) secondary antibody, followed by avidin and biotinylated peroxidase (Vector Labs, Burlingame, CA). The concentration of either HIR Ab or HIR Ab-IDUA fusion protein that gave 50% maximal binding was determined with a non-linear regression analysis. [00142] As shown in Fig. 10 there was comparable binding of either the chimeric HIR Ab or the HIR Ab-IDUA fusion protein for the HIR ECD with ED50 of 0.61+0.15 nM and 0.93+0.07 nM, respectively. [00143] The IDUA enzyme activity was determined with a fluorometric assay using 4-methylumbelliferyl a-L-iduronide (MUBI), which was purchased from Glycosynth, Ltd. (Cheshire, England). This substrate is hydolyzed to 4-methylumbelliferone (4-MU) by IDUA, and the 4-MU is detected fluorometrically with a Farrand filter fluorometer using an emission wavelength of 450 nm and an excitation wavelength of 365 nm. A standard curve was constructed with known amounts of 4-MU (Sigma-Aldrich, St. Louis, MO). The assay was performed at 37C at pH=3.5, and was terminated by the addition of 1 nmL of 0.5 M glycine (pH=10.3). One unit = 1 nmole substrate/hr (see Kakkis et al., (1994), Prot Expr Purif, 5:225-232). The IDUA enzyme activity of the purified HIR Ab-IDUA fusion protein was 363 + 37 nmol/hr/ug protein; the assay was linear with respect to both time and mass of fusion protein. Based on these results, we concluded that the HIR Ab-HC-IDUA fusion/HIR Ab-LC antibody is a bifunctional antibody that binds selectively to the IR ECD and retains a high level of IDUA activity. Example 4 HIR Ab-IDUA fusion protein uptake and biological activity in MPS Type I fibroblasts [00144] Type I MPS Hurler fibroblasts and healthy human fibroblasts were grown in 6-well cluster dishes to confluency. The medium was aspirated, wells washed with phosphate buffered saline (PBS), and incubated with 1 mL of Dulbecco's modified Eagle medium (DMEM) without serum, along with a range of concentrations of the HIR Ab-IDUA fusion protein, for 60 min at 37C. The medium was aspirated, and the wells were washed extensively (1 mL/well, 5 washes) with PBS, and the monolayer was taken up in 0.4 mL/well of lysis buffer (0.4 M sodium formate, 0.2% Triton X-100,pH=3.5), sonicated on ice 7 seconds 3 times, and microfuged 10 min 4C. The supernatant was removed for IDUA enzyme activity and bicinchoninic acid (BCA) protein assay. The uptake of the fusion protein was expressed as nmol/hr of IDUA enzyme activity per mg protein. [00145] The HIR Ab-IDUA fusion protein was taken up by MPS Type I fibroblasts (Fig. 11A). The basal IDUA activity in these cells without treatment is very low (less than 5 nmol/hr/mg of protein). -30 7671816_1 (GHMatters) P83171.AU.2 The intracellular IDUA enzyme activity increased in proportion to the concentration of medium HIR Ab-IDUA. The uptake of the HIR Ab-IDUA by the cells was inhibited 55% by the addition of 10 pg/ml murine HIR Ab (p<0.00 1), but was not inhibited by the addition of 4 mM mannose-6-phosphate (p>0.05). The IDUA enzyme activity in the Hurler fibroblasts approximates 250 mnol/hr/mgp at a medium HIR Ab-IDUA concentration of 2000 ng/mL (Fig. 11A). The horizontal line in Fig. 11A denotes the IDUA activity level in healthy human fibroblasts. [00146] The effect of the HIR Ab-IDUA fusion protein on cell glycosoaminoglycan (GAG) accumulation was assessed with a 35S incorporation assay (Unger et al, 1994). Type I MPS or healthy human fibroblasts were plated to 6-well cluster dishes at 250,000 cells/well and grown for 4 days in DMEM with 10% fetal bovine serum (FBS). The medium was discarded, the wells were washed with PBS, and 1 mL/well of low sulfate F12 medium with 10% dialyzed FBS was added, along with 5 mM CaCl2, the HIR Ab-IDUA fusion protein (0.3 g/mL), and 10 pCi/mL of 35S-sodium sulfate (Amersham, Chicago, IL). Following a 48 hr incubation at 37C, the medium was aspirated, the wells were washed with cold PBS (1 mL, 5 washes), and the cells were lysed with 0.4 mL/well of 1 N NaOH. The lysate was heated 60C 60 min to solubilize protein, an aliquot was removed for BCA protein assay, and the sample was counted for radioactivity with a Perkin Elmer Tri-Carb 2100 liquid scintillation counter. The data were expressed as 35S CPM per pg protein. [00147] The Hurler fibroblasts, with or without treatment by the HIR Ab-IDUA fusion protein, and the healthy human fibroblasts, were incubated for 48 hours in the presence of 35S-sodium sulfate, which is incorporated into intracellular GAGs. Treatment with the HIR Ab-IDUA fusion protein reduced GAG accumulation in Hurler fibroblasts by 70% as compared to healthy fibroblasts (p<0.0005) (Fig. 1IB). [00148] The prevention of GAG accumulation in Hurler fibroblasts (Fig. 1 IB) indicated that the HIR Ab-IDUA fusion antibody was directed to the lysosomal compartment of the cell, where GAG accumulates. [00149] To confirm targeting of the HIR Ab-IDUA fusion antibody to the lysosome, confocal microscopy was performed. Type I MPS Hurler fibroblasts were grown overnight in DMEM with 10% FBS to 50% confluency. The medium was aspirated, the wells washed well with PBS, and the cells were treated with fresh DMEM with no serum and containing 20 ptg/mL of the HIR Ab-IDUA fusion protein. Following a 24 hr incubation at 37C, the medium was aspirated, the wells washed extensively with cold PBS, and the cells were fixed with either 100% cold acetone for 20 min at -20C, or 4% paraformaldehyde at 4C for 20 miin. Following a PBS wash, the plates were blocked with 10% donkey serum, and then co-labeled with a 1:2000 dilution of the rabbit anti-IDUA antiserum, and 10 ptg/ml of a mouse MAb to human lysosomal associated membrane protein (LAMP)-1 (BD Pharmingen). Negative control antibodies were the same dilutions of either rabbit serum or mouse IgG. The secondary antibodies (Molecular Probes/Invitrogen) were 5 pg/mL each of 488 conjugated donkey anti-mouse IgG (green channel) and 594 conjugated donkey anti-rabbit IgG (red channel). The slides were imaged with a Zeiss LSM 5 PASCAL confocal microscope with dual argon and helium/neon lasers equipped with Zeiss LSM software, as described previously (Zhang et al, (2004), Hum Gene Ther, 15:339-350). -31 7671816_1 (GHMatters) P83171.AU.2 [00150] The cell IDUA and LAMP-I immunoreactivity is shown in Figs. 12A and 12B, respectively. The overlap image in Fig. 12C shows that the fusion protein was immunoreactive with an anti-IDUA antiserum, and the lysosomal marker, LAMP-1. No immune staining was detected with the control antibodies (Fig. 12D). Based on these results, we concluded that the HIR Ab-IDUA fusion antibody was targeted to lysosomes, as would be expected for IDUA. Example 5 Analysis of brain delivery and pharmacokinetics of HIR Ab-IDUA fusion protein in the Rhesus monkey [00151] The HIR Ab-IDUA fusion protein was iodinated with [1251]-iodine to a specific activity of 24 paCi/pg, and a trichloroacetic acid (TCA) precipitability of 99%. The fusion protein was iodinated on the same day as injection into the primate. A 7-year old female Rhesus monkey, weighing 7.2 kg, was obtained from Covance, Inc. (Alice, TX), and anesthetized with intramuscular ketamine, and isoflurane by inhalation. The anesthetized primate was administered by a single intravenous injection a dose of 957 pCi of [1251] -HIR Ab-IDUA fusion protein mixed with 400 pg (0.06 mg/kg) of unlabeled HIR Ab IDUA fusion protein in a final volume of 3 mL. Serum was collected at multiple time points over a 120 min period and analyzed for (a) serum 1251 radioactivity, and (b) serum IDUA enzyme activity. The serum glucose of the anesthetized, overnight-fasted primate was constant throughout the 120 min study period, and averaged 88 ± 1 mg%, which indicates that the administration of the HIR Ab fusion protein caused no interference of the endogenous insulin receptor, and had no effect on glycemic control. At 120 minutes after drug injection, the animal was euthanized, and brain and organ radioactivity was analyzed with a gamma counter; brain was also analyzed with the capillary depletion method, as described previously (Triguero et al., (1990), JNeurochem, 54:1882-1888), similar to prior work on the brain delivery of [1251] -labeled murine HIR Ab in the Rhesus monkey (Pardridge et al, (1995), Pharm Res, 12:807-816). The capillary depletion technique demonstrates the transcytosis of the fusion protein through the BBB in vivo, and into brain. [00152] The delivery of the fusion protein by brain, and other organs, at 120 minutes after intravenous injection is expressed as a % of injected dose (ID)/gram organ, and these data are given in Table 3. The fusion protein is delivered to all parts of brain, as shown by the film autoradiogram of the primate brain at 2 hours after intravenous injection of the [1251] -HIR Ab-IDUA fusion protein (Figure 12E). -32 7671816_1 (GHMatters) P83171.AU.2 Table 3. Brain and organ delivery of an HIR Ab-IDUA Fusion Antibody Tissue %ID/100 g VD (ul/g) Cerebrum gray matter 1.05 ± 0.07 134 8 Cerebrum white matter 0.32 ± 0.02 41±2 Cerebellum gray/white 0.74 ± 0.17 95 22 Liver 11.7 ±0.15 1485±18 Spleen 12.1 ±0.16 1541±20 Lung 5.2 ±0.4 655+48 Kidney 4.0 ±0.1 505±14 Heart 1.4 ±0.08 183±9 Skeletal muscle (triceps) 0.48 ± 0.002 61 1 Fat (omental) 1.7 ±0.4 221±58 Mean ± SD (n=3 replicates) [00153] The serum 1251 radioactivity concentration profile, expressed as % injected dose (ID)/mL (Fig. 13A), was fit to a bi-exponential equation, as described previously (Pardridge et al, (1995), Pharm Res, 12:807-816), to yield the pharmacokinetic (PK) parameters in Table 4. The parameters for the [125I] fusion protein are compared to the PK parameters reported previously (Coloma et al, (2000), Pharma Res, 17:266-274) for the ["..In]-HIR Ab (Table 4). [00154] The decline in serum radioactivity with time after injection was paralleled by the decline in serum IDUA enzyme activity (Fig. 13A). The serum IDUA enzyme activity in the primate before injection of the fusion protein was 1.5 ± 0.4 units/mL, and was 2120 ± 59, 496 ± 5, 194 ± 20, 67 ± 4, 19 ± 1, 12 ± 2, and 14 ± 1 units/mL, at 1, 2.5, 5, 15, 30, 60, and 120 min after injection, respectively. [00155] The brain delivery of the fusion protein at 2 hours after injection in the primate is expressed as a volume of distribution (VD), which is the ratio of DPM/gram brain divided by the DPM/uL serum (Fig. 13B). The brain VD for the fusion protein exceeds 140 pl/gram in the brain homogenate, and is much larger than the VD for [ 3 H] -mouse IgG2a, which has no receptor binding, and does not cross the BBB (Pardridge et al, supra). The brain VD for the [ 3 H]-mouse IgG2a, 18 pl/gram, equals the arterial blood volume of the brain (Ito et al, 2005), which is indicative of lack of transport across the BBB of an antibody that has no BBB receptor specificity. The brain VD for the [ 12 5 1]-fusion protein is also ~140 pl/gram in the post-vascular supernatant (Fig. 13B), as determined with the capillary depletion method. -33 7671816_1 (GHMatters) P83171.AU.2 Table 4. Pharmacokinetic parameters for [12sI]-HIR Ab-IDUA fusion protein and [m.In]-HIR Ab Parameter [..I]-HIR Ab-IDUA [m'In]-HIR Ab
A
1 (%ID/ml) 1.00 0.22 0.15 0.01
A
2 (%ID/ml) 0.077 0.013 0.10 0.01 ki (min- 0.65 +0.11 0.12 +0.01 k 2 (hr-') 0.42 + 0.26 0.11 + 0.06 t 1 2 (min) 1.1 0.2 5.8 0.6 t1/2 (hr) 1.7 1.0 6.3 0.6 CL (ml/min/kg) 1.11 0.47 0.22 0.08 Vss (ml/kg) 139 37 116 11 [00156] In table 4A 1 , A 2 , k 1 , and k 2 are the intercepts and slopes of the bi-exponential function describing the decay in plasma concentration with time. The parameters for the HIR Ab-IDUA fusion protein were determined for the Rhesus monkey in this study, and the parameters for HIR Ab were determined previously in the adult Rhesus monkey (Coloma et al, 2000). All data are normalized for differences in body weight. The ti 2 and ti 2 2 are computed from ki and k 2 , respectively, and are the half-times of the decay curves for each exponent. Clearance (CL) and steady state volume of distribution (Vss) are computed from A 1
A
2 k 1 and k 2 using previously described pharmacokinetic formulations (Pardridge et al, 1995). [00157] The brain VD of the post-vascular supernatant of the [ 125 1]-fusion protein is equal to the VD of the brain homogenate (Fig. 13B), which indicated that the fusion protein was transcytosed through the BBB and into brain parenchyma. The brain VD for the vascular pellet was low, 1.1 + 0.1 pl/g. [00158] Based on these data, we concluded that the HIR Ab-IDUA fusion antibody was taken up at a high rate into the primate protein, as shown in Table 3. This high rate of delivery into the brain was due to the targeting of the insulin receptor on the BBB. The fusion protein underwent transcytosis across the primate BBB in vivo, as demonstrated by the capillary deletion technique (Fig. 13B). [00159] Importantly, brain delivery of the HIR Ab-IDUA fusion antibody was 1.05 + 0.05% of injected dose per 100 gram brain (Table 3). The size of the Rhesus monkey brain is approximately 100 grams; therefore, about 1% of the injected dose is distributed to the primate brain. Owing to this high rate of delivery of the fusion antibody into the brain, it will be possible to produce normal levels of IDUA enzyme activity in the brain of patients with Hurler's syndrome. The delivery of the fusion protein by brain, expressed as a % of ID/gram, in the human will be reduced, as compared to the primate, in proportion to body weight. Therefore, the expected brain delivery of the fusion protein in the human brain is about 0.l1% of the injected dose per 100 gram brain, or about 1% of the ID per 1000 g human brain. A normal level of IDUA enzyme activity for the human brain ranges from 0.5-1.5 -34 7671816_1 (GHMatters) P83171.AU.2 units/mg protein (Crow et al, (1983), J C/in Pathol, 36:415-430) and there is a total of about 100,000 mg of protein in an average size human brain. Thus, it would be expected that delivery to the brain of between 50,000 units to about 150,000 units of IDUA activity should suffice to rescue a deficit in brain IDUA activity as observed in, e.g., Hurler's syndrome. As recombinant IDUA itself does not cross the BBB, this has not been feasible. In contrast, given the observed delivery of the HIR Ab-IDUA fusion antibody into the brain, and its high IDUA specific activity, we conclude that achieving delivery of a normalizing level of IDUA activity in the brain of patients suffering from an IDUA deficiency (e.g., as in Hurler's syndrome) will be achieved by systemic administration of an HIR Ab-IDUA fusion antibody. Further, due to the broad distribution of the fusion antibody in all organs examined (Table 3), systemic administration of the HIR Ab-IDUA fusion antibody may also normalize IDUA enzyme activity outside the CNS in Hurler's patients. Example 6. Expression vectors for permanent transfection of host cell [00160] The genetic engineering of TV-HIRMAb-IDUA was accomplished in several linear steps, comprised of the following: (1) A "double gene" expression plasmid, designated pCD-HC-IDUA-LC (Figure 14) was engineered from 2 precursor plasmids, pCD-HC-IDUA, and pCD-LC, following linearization of pCD-HC-IDUA by Afel, and release of the LC expression cassette with NruI and Afel, and closure of the new plasmid with T4 ligase, as shown in Figure 14. (2) A "triple gene" tandem vector (TV) expression plasmid, designated TV-HIRMAb-IDUA (Figure 14) was engineered from 2 precursor plasmids, pCD-HC-IDUA-LC and pwtDHFR, where pwtDHFR encodes for the wild type (wt) murine dihydrofolate reductase (DHFR). The DHFR expression cassette was released from pwtDHFR with Smal and SalI. The end of SalI was filled with T4 DNA polymerase and deoxynucleotide triphosphates. In parallel, the pCD-HC-IDUA-LC was opened with Afel. The new TV was closed with T4 ligase. [00161] The engineering of the TV was validated by (a) agarose gel electrophoresis, (b) IDUA expression in COS cells, and (c) by bi-directional DNA sequencing. The entire 7,822 nucleotides (nt) of the TV-HIRMAb-IDUA was subjected to bi-directional DNA sequencing using custom oligodeoxynucleotides (ODNs), and the nt sequence is given in SEQ ID NO. 14. The DNA sequence was comprised of 7,822 nt, which included the following domains: * 714 nt cytomegalovirus (CMV) promoter * 9 nt Kozak sequence (GCCGCCACC) * 3,276 nt open reading frame (orf) encoding the fusion gene of the HIRMAb HC and IDUA * 297 nt bovine growth hormone (BGH) polyA (pA) sequence * 23 nt linker * 731 nt CMV promoter * 9 nt Kozak sequence * 705 orf encoding the HIRMAb LC * 291 nt BGH pA -35 7671816_1 (GHMatters) P83171.AU.2 * 254 SV40 promoter * 9 nt Kozak sequence * 564 murine DHFR orf * 940 hepatitis B virus (HBV) pA [00162] The TV-HIRMAb-IDUA also included the expression cassette encoding neo, the neomycin resistance gene, to enable selection with G418 (Figure 14). It was necessary to include the HC fusion gene, the LC gene, and the DHFR gene on a single piece of DNA, or tandem vector (Figure 14) to allow for equally high expression of all 3 genes in the transfected host cell. [00163] The TV-HIRMAb-IDUA sequence, from nt 724-3,999 (SEQ ID NO. 14), encoded for a 1,091 amino acid (AA) HC fusion protein, which was comprised of a 19 AA IgG signal peptide, the 442 AA HIRMAb HC, a 3 AA linker, and the 627 AA human IDUA enzyme, and is given in SEQ ID. NO. 15. The predicted molecular weight (MW) of the non-glycosylated HC was 118,795 Daltons (Da) and the predicted isolectric point (pI) of the fusion HC protein was 8.85. The TV-HIRMAb-IDUA sequence, from nt 5,060-5,764 (SEQ ID NO. 14), encoded for a 234 AA LC protein (SEQ ID NO. 16), which was comprised of a 20 AA IgG signal peptide, and the 214 AA HIRMAb LC. The predicted MW of the LC was 23,398 Da and the predicted pI of the LC protein was 5.45. The TV-HIRMAb IDUA sequence, from nt 6,319-6,882 (SEQ ID NO. 14), encoded for a DHFR protein (SEQ ID NO. 17) that was comprised of 187 AA. Example 7. Permanent transfection of Chinese hamster ovary cells with TV-HIRMAb-IDUA [00164] DG44 Chinese hamster ovary (CHO) cells were grown in serum free HyQ SFM4CHO utility medium (HyClone), containing 1 x HT supplement (hypoxanthine and thymidine). DG44 CHO cells (5 x 106 viable cells) were electroporated with 5 pg PvuI-linearized TV-HIRMAb-IDUA plasmid DNA. The cell-DNA suspension is then incubated for 10 min on ice. Cells are electroporated with BioRad pre-set protocol for CHO cells, i.e. square wave with pulse of 15 msec and 160 volts. After electroporation, cells are incubated for 10 min on ice. The cell suspension is transferred to 50 nil culture medium and plated at 125 pl per well in 4 x 96-well plates (10,000 cells per well). A total of 10 electroporations and 4,000 wells are performed per study. [00165] Following electroporation (EP), the CHO cells are placed in the incubator at 37 C and 8% C02. Owing to the presence of the neo gene in the TV, transfected cell lines are initially selected with G418. The TV-HIRMAb-IDUA also contains the gene for DHFR (Figure 14), so the transfected cells are also selected with 20 nM methotrexate (MTX) and HT deficient medium. Once visible colonies are detected at about 21 days after EP, the conditioned medium is sampled for human IgG by ELISA. Wells with high human IgG signals in the ELISA are transferred from the 96-well plate to a 24-well plate with 1mL of HyQ SFM4CHO-Utility. The 24-well plates are returned to the incubator at 37 C and 8% C02. The following week IgG ELISA is performed on the clones in the 24-well plates. This is repeated through the 6-well plates to T75 flasks and finally to 60 mL and 125 mL square plastic bottles on an orbital shaker. At this stage, the final MTX concentration is 80 nM, and the medium IgG -36 7671816_1 (GHMatters) P83171.AU.2 concentration, which is a measure of HIRMAb-IDUA fusion protein in the medium is >10 mg/L at a cell density of 106/mL. [00166] Clones selected for dilutional cloning (DC) are removed from the orbital shaker in the incubator and transferred to the sterile hood. The cells are diluted to 500 mL in F- 12K medium with 5% dialyzed fetal bovine serum (d-FBS) and Penicillin/Streptomycin, and the final dilution is 8 cells per niL, so that 4,000 wells in 40 x 96-well plates can be plated at a cell density of 1 cell per well (CPW). Once the cell suspension is prepared, within the sterile hood, a 125uL aliquot is dispensed into each well of a 96-well plate using an 8-channel pipettor or a precision pipettor system. The plates are returned to the incubator at 37 C and 8% C02. The cells diluted to 1 cell/well cannot survive without serum. On day 6 or 7, DC plates are removed from the incubator and transferred to the sterile hood where 125 pl of F-12K medium with 5% dialyzed fetal bovine serum (d-FBS) is added to each well. This selection media now contains 5% d-FB S, 30 nM MTX and 0.25 mg/mL Geneticin. On day 21 after the initial 1 CPW plating, aliquots from each of the 4,000 wells are removed for human IgG ELISA, using robotics equipment. DC plates are removed from the incubator and transferred to the sterile hood, where 100 pl of media is removed per well of the 96-well plate and transferred into a new, sterile sample 96-well plate using an 8-channel pipettor or the precision pipettor system. [00167] On day 20 after the initial 1 CPW plating, 40 x 96-well Inmmunoassay plates are plated with l00uL of 1tg/mL solution of Primary antibody, a mouse anti-human IgG in 0.1M NaH CO3. Plates are incubated overnight in the 4C refrigerator. The following day, the ELISA plates are washed with 1x TBST 5 times, and 100uL of lug/mL solution of secondary antibody and blocking buffer are added. Plates are washed with 1x TBST 5 times. 100uL of 1mg/mL of 4-nitrophenyl phosphate di(2-amino-2 ethyl- 1,3-propanediol) salt in 0. IM glycine buffer are added to the 96-well immunoassay plates. Plates are read on a microplate reader. The assay produces IgG output data for 4,000 wells/experiment. The highest producing 24-48 wells are selected for further propagation. [00168] The highest producing 24-well plates from the 1 CPW DC are transferred to the sterile hood are gradually subcloned through 6-well dishes, T75 flasks, and 125 mL square plastic bottles on an orbital shaker. During this process the serum is reduced to zero, at the final stage of centrifugation of the cells and resuspension in SFM. [00169] The above procedures are repeated with a second round of dilutional cloning, at 0.5 cells/well (CPW). At this stage, approximately 40% of the wells show any cell growth, and all wells showing growth also secrete human IgG. These results confirm that on average only 1 cell is plated per well with these procedures, and that the CHO cell line originates from a single cell. Example 8. Manufacturing of CHO-derived HIRMAb-IDUA fusion protein [00170] Following the second round of dilutional cloning, the highest producing cell line secreting the HIRMAb-IDUA fusion protein was propagated in serum free medium to a total volume of 2,000 mL in several IL square plastic bottles on an orbital shaker. The HIRMAb-IDUA fusion protein was purified from the CHO cell conditioned medium using the following down-stream processing: * Depth filtration with a 0.2 m 2 0.65 ptm GF filter in series with an 0.05 m 2 0.2 ptm Sartopore-2 -37 7671816_1 (GHMatters) P83171.AU.2 pltrafilter * Volume reduction to 400 mL using tangential flow filtration (TFF) system * Ultra-filtration with a 0.2 mm pltra-filter and application to a column of protein A Sepharose 4 Fast Flow. Following application to the column, the column was eluted with 1 M NaCl, which elutes DNA non-specifically absorbed to the column, and the product is eluted as a single peak with 0.1 M sodium acetate/pH=3.7 (Figure 15A). The acid eluate was neutralized with 1 M Tris base and concentrated to 5 mL with a Centriprep-30 * Cation exchange (CATEX) chromatography in bind-elute mode was performed with a column of SP Sepharose FF equilibrated with 0.02 M MES and 0.05 M NaCl. The conductivity of the sample was reduced to <5 mS/cm prior to application to the column. The column was successively eluted with step gradients of 0.02 M MES/pH=5.5 containing 0.25 M NaCl, 0.35 M NaCl, 0.5 M NaCl, and IM NaCl. The HIRMAb-IDUA fusion protein eluted in 0.5 M NaCl, as shown in Figure 15B. * Anion exchange (ANEX) chromatography in flow-through mode was performed with a column of Q Sepharose FF equilibrated with 0.025 M MES/pH=5.5 and 0.05 M NaCl. The conductivity of the sample was reduced to <7 mS/cm. The HIRMAb-IDUA fusion protein eluted in the flow-through as shown in Figure 15C. [00171] The purity and potency of the CHO derived HIRMAb-IDUA fusion protein was assessed with the following procedures: [00172] (a) SDS-PAGE. The CHO-derived HIRMAb-IDUA fusion protein was purified to homogeneity based on reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS PAGE), as demonstrated in Figure 16 (lane 3). The chimeric HIRMAb is applied to lane 2 of Figure 16, and HIRMAb-IDUA fusion protein is applied to lane 3 of Figure 16. The size of the light chain (LC) of both proteins is the same as both proteins are comprised of the same LC. The size of the heavy chain (HC) of HIRMAb-IDUA fusion protein is 130 kDa (lane 3, Figure 16), whereas the size of the HC of the chimeric HIRMAb is 50 kDa (lane 2, Figure 16), and the difference in size is due to fusion of the 80 kDa IDUA to the HC of the chimeric HIRMAb. [00173] (b) IDUA and human IgG Western blot. The CHO derived HIRMAb-IDUA fusion protein was electrophoresed on a 7.5% SD S-PAGE gel and blotted to nitrocellulose for Western blotting with primary antibodies to either human IgG (lane 1, Figure 17), or to human IDUA (lane 2, Figure 17). Both the anti-human IgG antibody and the anti-human IDUA antibody reacted specifically with the heavy chain of HIRMAb-IDUA fusion protein, which migrated with a molecular weight of 130 kDa in this reducing gel (Figure 17). [00174] (c) Human insulin receptor (HIR) binding assay. The extracellular domain (ECD) of the HIR was purified by lectin affinity chromatography from serum free medium conditioned by CHO cells that were permanently transfected with the HIR ECD. The HIR ECD was plated in ELISA wells to bind the chimeric HIRMAb without IDUA fused, and the CHO-derived HIRMAb-IDUA fusion protein. As shown in Figure 18, the ED50 of the chimeric HIRMAb or the HIRMAb-IDUA fusion protein binding to the HIR is not significantly different, and is 0.75 nM. These data indicate the affinity of the HIRMAb for the HIR is not affected by the fusion of IDUA to the carboxyl terminus of -38 7671816_1 (GHMatters) P83171.AU.2 the IgG. The binding constants shown in Figure 18 were determined by non-linear regression analysis of the binding isotherms. [00175] (d) IDUA enzyme activity of the HIRMAb-IDUA fusion protein. The IDUA enzyme activity of the CHO-derived HIRMAb-IDUA fusion protein was determined with a fluorometric assay using 4-methylumbelliferyl a-L-iduronide (MUBI) as the assay substrate. This substrate is hydolyzed to 4-methylumbelliferone (4-MU) by IDUA, and the 4-MU is detected fluorometrically with a filter fluorometer using an emission wavelength of 450 nm and an excitation wavelength of 365 nm. A standard curve was constructed with known amounts of 4-MU. The assay was performed at 37C at pH=3.5, and was terminated by the addition of 1 mL of 0.1 M glycine (pH=10.3). One unit-1 nmol/hr. IDUA enzyme activity of the HIRMAb-IDUA fusion protein was linear with respect to time and concentration (Figure 19). The IDUA enzyme specific activity of the CHO derived the HIRMAb IDUA fusion protein is 291 ± 9 units per pg protein (Figure 19). The IDUA enzyme specific activity of recombinant IDUA is 240 units per pg protein; therefore, the IDUA enzyme activity of the HIRMAb-IDUA fusion protein is comparable to that of recombinant IDUA. [00176] (e) Size exclusion high performance liquid chromatography. The absence of aggregates in the purified HIRMAb-IDUA fusion protein was demonstrated with size exclusion chromatography (SEC) high performance liquid chromatography (HPLC) using 2 G3000 SWXL columns, 0.78x30 cm, in series, and an HPLC pump at 0.5 mL/min with detection at 280 nm. As shown in Figure 20, the CHO-derived HIRMAb-IDUA fusion protein elutes as a single peak, removed from the void volume, with no detectable aggregates. [00177] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby. -39 7671816_1 (GHMatters) P83171.AU.2

Claims (51)

1. A method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the subject a therapeutically effective dose of a fusion antibody having a-L-iduronidase activity, wherein: (i) at least about 0.5% of the therapeutically effective dose is delivered to the brain; (ii) the fusion antibody: comprises: (a) a fusion protein containing the amino acid sequence of an immunoglobulin heavy chain and an a-L-iduronidase, and (b) an imnmunoglobulin light chain; (iii) the fusion antibody binds to an extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate; and (iv) the amino acid sequence of the a-L-iduronidase is covalently linked to the carboxy terminus of the amino acid sequence of the immunoglobulin heavy chain.
2. The method of claim 1, wherein at least about 25,000 units of a-L-iduronidase activity are delivered to the brain.
3. The method of claim 1, wherein the therapeutically effective dose comprises at least about 1 x 106 units of a-L-iduronidase activity or at least about 140,000 units/Kg of body weight.
4. The method of claim 1, wherein the IDUA specific activity of the fusion antibody is at least 200,000 units/mg.
5. The method of claim 1, wherein the systemic administration is parenteral, intravenous, subcutaneous, intra-muscular, trans-nasal, intra-arterial, transdermal, or respiratoryThe method of claim 1, wherein the delivery occurs within two hours or less after the systemic administration. The method of claim 1, wherein the fusion antibody is a chimeric antibody.
6. The method of claim 1, wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 4 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions.
7. The method of claim 6, wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations. -40 7671816_1 (GHMatters) P83171.AU.2
8. The method of claim 7, wherein the wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3.
9. The method of claim 1, wherein the immunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions.
10. The method of claim 9, wherein the immunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6.
11. The method of claim 10, wherein the amino acid sequence of the heavy chain imnmunoglobulin is at least 90% identical to SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO:8.
12. The method of claim 11, wherein the amino acid sequence of the heavy chain immunoglobulin comprises SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin comprises SEQ ID NO:8.
13. The method of claim 1, wherein the a-L-iduronidase comprises an amino acid sequence at least 90% identical to SEQ ID NO:9.
14. The method of claim 13, wherein the amino acid sequence is at least 95% identical to SEQ ID NO:9.
15. The method of claim 14, wherein the amino acid sequence comprises SEQ ID NO:9.
16. The method of claim 1, wherein the amino acid sequence of the heavy chain immunoglobulin is at least 90% identical to SEQ ID NO:7; the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO:8; and the amino acid sequence of the a-L-iduronidase is at least 95% identical to SEQ ID NO:9.
17. A method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the subject a therapeutically effective dose of a fusion antibody having a-L-iduronidase activity, wherein: (i) at least about 0.5% of the therapeutically effective dose is delivered to the brain; -41 7671816_1 (GHMatters) P83171.AU.2 (ii) the fusion antibody: comprises: (a) a fusion protein at least 95% identical to SEQ ID NO: 10, and (b) an immunoglobulin light chain; (iii) the fusion antibody binds to an extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate.
18. The method of claim 17, wherein at least about 25,000 units of a-L-iduronidase activity are delivered to the brain.
19. The method of claim 17, wherein the therapeutically effective dose comprises at least about 1 x 106 units of a-L-iduronidase activity or at least about 140,000 units of a-L-iduronidase activity/Kg of body weight.
20. The method of claim 17, wherein the IDUA specific activity of the fusion antibody is at least about 200,000 units/mg.
21. The method of claim 17, wherein the systemic administration is parenteral, intravenous, subcutaneous, intra-muscular, trans-nasal, intra-arterial, transdermal, or respiratory.
22. The method of claim 17, wherein the delivery occurs in two hours or less after the systemic administration.
23. The method of claim 17, wherein the fusion antibody is a chimeric antibody.
24. The method of claim 17, wherein the immunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions.
25. The method of claim 24, wherein the immunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO: 5, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6.
26. A method for treating an a-L-iduronidase deficiency in the central nervous system of a subject in need thereof, comprising systemically administering to the subject a therapeutically effective dose of a fusion antibody having a-L-iduronidase activity, wherein: (i) at least about 0.5% of the therapeutically effective dose is delivered to the brain; (ii) the fusion antibody: comprises a fusion protein containing the amino acid sequence of an immunoglobulin heavy chain and an a-L-iduronidase, or a fusion protein containing the amino acid sequence of an immunoglobulin light chain and an a-L-iduronidase; -42 7671816_1 (GHMatters) P83171.AU.2 binds to the extracellular domain of the human insulin receptor; and catalyzes hydrolysis of unsulfated alpha-L-iduronosidic linkages in dermatan sulfate; and (iii) the amino acid sequence of the a-L-iduronidase is covalently linked to the carboxy terminus of the amino acid sequence of the immunoglobulin heavy chain or the immunoglobulin light chain
27. The fusion antibody of claim 26, wherein the fusion antibody comprises an immunoglobulin heavy chain and an immunoglobulin light chain.
28. The method of claim 26, wherein the fusion protein contains the amino acid sequence of an immunoglobulin heavy chain and the a-L-iduronidase.
29. The method of claim 26, wherein at least about 25,000 units of a-L-iduronidase activity are delivered to the brain.
30. The method of claim 26, wherein the therapeutically effective dose comprises at least about 1 x 106 units of a-L-iduronidase activity or at least about 140,000 units of a-L-iduronidase activity/Kg body weight.
31. The method of claim 26, wherein the IDUA specific activity of the fusion antibody is about 200,000 units/mg.
32. The method of claim 26, wherein the systemic administration is parenteral, intravenous, subcutaneous, intra-muscular, trans-nasal, intra-arterial, transdermal, or respiratory.
33. The method of claim 26, wherein the delivery occurs in two hours or less after the systemic administration.
34. The method of claim 26, wherein the fusion antibody is a chimeric antibody.
35. The method of claim 26, wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 4 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions.
36. The method of claim 35, wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2 with up to 6 single amino acid mutations, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3 with up to 3 single amino acid mutations.
37. The method of claim 35, wherein the wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 -43 7671816_1 (GHMatters) P83171.AU.2 corresponding to the amino acid sequence of SEQ ID NO:2, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3.
38. The method of claim 37, wherein the wherein the immunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3.
39. The method of claim 26, wherein the inmmunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations, wherein the single amino acid mutations are substitutions, deletions, or insertions.
40. The method of claim 39, wherein the inmmunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4 with up to 3 single amino acid mutations, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5 with up to 5 single amino acid mutations, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6 with up to 5 single amino acid mutations.
41. The method of claim 39, wherein the inmmunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO: 5, or a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6.
42. The method of claim 27, wherein the inmmunoglobulin heavy chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO: 1, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:2, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:3; and the immunoglobulin light chain comprises a CDR1 corresponding to the amino acid sequence of SEQ ID NO:4, a CDR2 corresponding to the amino acid sequence of SEQ ID NO:5, and a CDR3 corresponding to the amino acid sequence of SEQ ID NO:6.
43. The method of claim 27, wherein the amino acid sequence of the heavy chain imnmunoglobulin is at least 90% identical to SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO:8.
44. The method of claim 43, wherein the amino acid sequence of the heavy chain imnmunoglobulin comprises SEQ ID NO:7 and the amino acid sequence of the light chain immunoglobulin comprises SEQ ID NO:8.
45. The method of claim 26, wherein the a-L-iduronidase comprises an amino acid sequence at least 9 0% identical to SEQ ID NO:9. -44 7671816_1 (GHMatters) P83171.AU.2
46. The method of claim 45, wherein the amino acid sequence is at least 95% identical to SEQ ID NO:9.
47. The method of claim 46, wherein the amino acid sequence comprises SEQ ID NO:9.
48. The method of claim 27, wherein the amino acid sequence of the heavy chain immunoglobulin is at least 90% identical to SEQ ID NO:7; the amino acid sequence of the light chain immunoglobulin is at least 90% identical to SEQ ID NO:8; and the amino acid sequence of the a-L-iduronidase is at least 95% identical to SEQ ID NO:9.
49. The method of claim 48, wherein the amino acid sequence of the heavy chain immunoglobulin comprises SEQ ID NO:7; the amino acid sequence of the light chain immunoglobulin comprises SEQ ID NO: 8; and the amino acid sequence of the a-L-iduronidase comprises SEQ ID NO:9.
50. The method of claim 26, wherein the amino acid sequence of the fusion protein is at least 95% identical to SEQ ID NO: 10.
51. The method of claim 50, wherein the amino acid sequence of the fusion protein comprises SEQ ID NO:10. -45 7671816_1 (GHMatters) P83171.AU.2
AU2016202625A 2007-07-27 2016-04-26 Methods and compositions for increasing alpha-iduronidase activity in the CNS Abandoned AU2016202625A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2016202625A AU2016202625A1 (en) 2007-07-27 2016-04-26 Methods and compositions for increasing alpha-iduronidase activity in the CNS

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/952,547 2007-07-27
AU2013206657A AU2013206657A1 (en) 2007-07-27 2013-07-03 Methods and compositions for increasing alpha-iduronidase activity in the CNS
AU2016202625A AU2016202625A1 (en) 2007-07-27 2016-04-26 Methods and compositions for increasing alpha-iduronidase activity in the CNS

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2013206657A Division AU2013206657A1 (en) 2007-07-27 2013-07-03 Methods and compositions for increasing alpha-iduronidase activity in the CNS

Publications (1)

Publication Number Publication Date
AU2016202625A1 true AU2016202625A1 (en) 2016-05-19

Family

ID=56024487

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2016202625A Abandoned AU2016202625A1 (en) 2007-07-27 2016-04-26 Methods and compositions for increasing alpha-iduronidase activity in the CNS

Country Status (1)

Country Link
AU (1) AU2016202625A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110709090A (en) * 2017-04-27 2020-01-17 益缪索夫特公司 B cells and dosages thereof for in vivo delivery of therapeutic agents

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110709090A (en) * 2017-04-27 2020-01-17 益缪索夫特公司 B cells and dosages thereof for in vivo delivery of therapeutic agents

Similar Documents

Publication Publication Date Title
US20230279156A1 (en) Methods and compositions for increasing alpha-l-iduronidase activity in the cns
US12043661B2 (en) Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US20220127369A1 (en) Methods and compositions for increasing n-acetylglucosaminidase activity in the cns
AU2016202625A1 (en) Methods and compositions for increasing alpha-iduronidase activity in the CNS
AU2013206657A1 (en) Methods and compositions for increasing alpha-iduronidase activity in the CNS

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period