Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INTRATHECAL DELIVERY OF NUCLEIC ACID SEQUENCES ENCODING ABCD1 FOR TREATMENT OF ADRENOMYELONEUROPATHY
Document Type and Number:
WIPO Patent Application WO/2017/079467
Kind Code:
A1
Abstract:
Methods of the invention encompass delivery of nucleic acid sequences encoding ABCD1 for the treatment of X-linked Adrenoleukodystrophy (X-ALD), e.g., for Adrenomyeloneuropathy (AMN).

Inventors:
MAGUIRE CASEY A (US)
EICHLER FLORIAN (US)
Application Number:
PCT/US2016/060375
Publication Date:
May 11, 2017
Filing Date:
November 03, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MASSACHUSETTS GEN HOSPITAL (US)
International Classes:
A61K9/50; A61K48/00; A61P25/28; C12N7/02; C12N15/85; C12N15/86
Domestic Patent References:
WO2015114365A12015-08-06
Foreign References:
US20130004471A12013-01-03
US20130202559A12013-08-08
US6225525B12001-05-01
US7192579B22007-03-20
US4458066A1984-07-03
US5609885A1997-03-11
US5728396A1998-03-17
US5985305A1999-11-16
US5997527A1999-12-07
US6113938A2000-09-05
US6132420A2000-10-17
US6156331A2000-12-05
US6217906B12001-04-17
US6261584B12001-07-17
US6270787B12001-08-07
US6287295B12001-09-11
US6375978B12002-04-23
US6395292B22002-05-28
US6508808B12003-01-21
US6544252B12003-04-08
US6635268B22003-10-21
US6682522B22004-01-27
US6923800B22005-08-02
US6939556B22005-09-06
US6976981B22005-12-20
US6997922B22006-02-14
US7014636B22006-03-21
US7207982B22007-04-24
US7112335B22006-09-26
US7163688B22007-01-16
US20050175701A12005-08-11
US20070281024A12007-12-06
US20080091176A12008-04-17
Other References:
GONG ET AL.: "Adenoassociated Virus Serotype 9-mediated Gene Therapy for X-Linked Adrenoleukodystrophy", MOLECULAR THERAPY, vol. 23, 1 May 2015 (2015-05-01), pages 824 - 834, XP055381024
ENGELEN ET AL., ORPHANET J RARE DIS, vol. 7, 2012, pages 51
PUJOL ET AL., HUM MOL GENET, vol. 11, 2002, pages 499 - 505
AUBOURGCHAUSSAIN, HORM RES, vol. 1, 2003, pages 104 - 5
STEINBERG ET AL., CURR PROTOC HUM GENET, 2008
STEINBERG SJMOSER ABRAYMOND GV, X-LINKED ADRENOLEUKODYSTROPHY, 26 March 1999 (1999-03-26)
"GeneReviews® [Internet]. Seattle (WA", 1993, UNIVERSITY OF WASHINGTON, article "LABORATORY TECHNIQUES IN BIOCHEMISTRY AND MOLECULAR BIOLOGY: HYBRIDIZATION WITH NUCLEIC ACID PROBES, Part I"
BENTONDAVIS, SCIENCE, vol. 196, 1977, pages 180
GRUNSTEINHOGNESS, PROC. NATL. ACAD. SCI., USA, vol. 72, 1975, pages 3961
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 2001, WILEY INTERSCIENCE
BERGERKIMMEL: "Guide to Molecular Cloning Techniques", 1987, ACADEMIC PRESS
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ZHANGMADDEN, GENOME RES., vol. 7, 1997, pages 649 - 656
BRUMMELKAMP ET AL., SCIENCE, vol. 296, 2002, pages 550 - 553
LEE ET AL., NATURE BIOTECHNOL., vol. 20, 2002, pages 500 - 505
MIYAGISHITAIRA, NATURE BIOTECHNOL, vol. 20, 2002, pages 505 - 508
PADDISON ET AL., GENES & DEV., vol. 16, 2002, pages 948 - 958
SUI, PROC. NATL. ACAD. SD. USA, vol. 99, no. 6, 2002, pages 5515 - 5520
YU ET AL., PROC NATLACADSCI USA, vol. 99, 2002, pages 6047 - 6052
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual.", vol. 1-3, 1989, COLD SPRING HARBOR LABORATORY
COFFIN ET AL.: "Retroviruses", 1997, OXFORD UNIVERSITY PRESS, article "RNA Viruses: A Practical Approach"
ADAMS, J. AM. CHEM. SOC., vol. 105, 1983, pages 661
BELOUSOV, NUCLEIC ACIDS RES., vol. 25, 1997, pages 3440 - 3444
FRENKEL, FREE RADIC. BIOL. MED., vol. 19, 1995, pages 373 - 380
BLOMMERS, BIOCHEMISTRY, vol. 33, 1994, pages 7886 - 7896
NARANG, METH. ENZYMOL., vol. 68, 1979, pages 109
BEAUCAGE, TETRA. LETT., vol. 22, 1981, pages 1859
"CURRENT PROTOCOLS IN MOLECULAR BIOLOGY", 1997, JOHN WILEY & SONS, INC.
"Modified-Release Drug Delivery Technology", 2008, INFORMA HEALTHCARE, pages: 143 - 149
Attorney, Agent or Firm:
DEYOUNG, Janice Kugler et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method of increasing adeno-associated virus 9 (AAV9) vector titers in transfected producer cells grown in culture, said method comprising the steps of:

i) incubating a nucleic acid sequence that is complementary to an mR A encoding ATP binding cassette subfamily D member 1 (ABCD l) with the cells, and

ii) transfecting an AAV9 vector comprising a nucleotide sequence encoding ABCD 1 into the cells (AAV9-ABCD 1 vector), wherein the amount of ABCD 1 mRNA expressed from the AAV9 vector is decreased, thereby increasing AAV9-ABCD 1 vector yield in cell lysate and/or media by about 1 fold to about 50 fold compared to a reference standard.

2. The method of claim 1, wherein the nucleic acid sequence that is complementary to an mRNA encoding ABCD l is an interfering RNA.

3. The method of claim 2, wherein the wherein the interfering RNA is an shRNA or siRNA.

4. The method of claim 3, wherein the siRNA comprises SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 7, or a combination thereof.

5. The method of claim 1, wherein the reference standard comprises AAV9-ABCD 1 vector yield in cell lysate and/or media from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCD l .

6. A method of treating X-linked adrenoleukodystrophy (X-ALD) in a subject in need thereof comprising administering to the subject a composition comprising purified AAV9- ABCD 1 vector obtained from the producer cells of claim 1.

7. The method of claim 6, wherein the composition comprising purified AAV9-ABCD 1 vector is administered to the subject by intrathecal administration.

8. A method of treating X-linked adrenoleukodystrophy (X-ALD) in a subject in need thereof comprising administering to the subject an adeno-associated virus (AAV) vector encoding an ATP binding cassette subfamily D member 1 (ABCD l), wherein said vector is administered to the subject by intrathecal administration.

9. The method of claim 8, wherein the intrathecal administration is mediated by an osmotic pump.

10. The method of claim 8, wherein the dose of vector is about lxl013GC to about 10xl013GC.

1 1. The method of claim 8, wherein the AAV is AAV9.

12. A method of providing ATP binding cassette subfamily D member 1 (ABCD l) to a subject having X-linked adrenoleukodystrophy (X-ALD) comprising administering to the subject a vector encoding ABCD l, wherein said vector is administered to the subject by intrathecal administration, and wherein ABCD 1 expression from said vector in the central nervous system is less than ABCD l expression from said vector in peripheral organs.

13. The method of claim 12, wherein ABCD l expression from said vector in the central nervous system is about 3 fold higher than expression of ABCD l in the central nervous system of an untreated subject that does not have X-ALD.

14. The method of claim 13, wherein the ABCD l expression from said vector in peripheral organs is about 90% less than expression of ABCD 1 in the peripheral organs of an untreated subject that does not have X-ALD.

Description:
INTRATHECAL DELIVERY OF NUCLEIC ACID SEQUENCES ENCODING ABCDl FOR TREATMENT OF ADRENOMYELONEUROPATHY

RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. provisional application Ser. No.

62/251,208, filed November 5, 2015 and U.S. provisional application Ser. 62/300,691, filed February 26, 2016. The entire disclosures of the aforementioned applications are incorporated herein by reference.

FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

[0002] This work was supported by Grant Nos. R21 NS081374-01 and R01 NS072446-01 awarded by the National Institutes of Health. The government has certain rights in the invention.

BACKGROUND OF THE INVENTION

[0003] X-linked adrenoleukodystrophy (X-ALD), a progressive genetic disorder, is caused by mutations in the ABCDl gene, which encodes a peroxisomal ATP-binding cassette transporter (ABCD 1) responsible for transport of CoA-activated very long-chain fatty acids (VLCFA) into the peroxisome for degradation leading to the accumulation of high levels of saturated, very long chain fatty acids (VLCFA) in plasma and tissues of the brain and adrenal cortex. Symptoms can begin in childhood or adulthood. Adult ALD patients typically develop adrenomyeloneuropathy (AMN), a debilitating neurological disorder, in their twenties (Engelen et al., Orphanet J Rare Dis. 2012; 7: 51). The Abcdl 7" mouse develops a phenotype similar to AMN, manifesting spinal cord axon degeneration as well as peripheral neuropathy due to affected dorsal root ganglion neurons (DRGs) (Pujol et al., Hum Mol Genet. 2002; 11 :499-505). Transduction of central nervous system cells in vitro and in vivo using recombinant adeno-associated virus serotype 9 (rAAV9) vector for delivery of the human ABCDl gene was previously reported. Unfortunately, intravenous delivery in young mice is associated with cardiac toxicity due to transgene overexpression. Delivery systems that provide non-toxic levels of ABCD 1 in patients suffering from X-ALD or AMN would be highly desirable. SUMMARY OF THE INVENTION

[0004] Other features and advantages of the invention will be apparent from the Detailed Description, and from the claims. Thus, other aspects of the invention are described in the following disclosure and are within the ambit of the invention.

[0005] In one aspect, the invention provides a method of increasing adeno-associated Virus 9 (AAV9) vector titers in transfected producer cells grown in culture, said method comprising the steps of i) incubating a nucleic acid sequence that is complementary to an mRNA encoding ATP binding cassette subfamily D member 1 (ABCD1) with the cells and ii) transfecting an AAV9 vector comprising a nucleotide sequence encoding ABCD 1 into the cells (AAV9-ABCD 1 vector), wherein the amount of ABCD 1 mRNA expressed from the AAV9 vector is decreased, thereby increasing AAV9-ABCD 1 vector yield in cell lysate and/or media by about 1 fold to about 50 fold compared to a reference standard.

[0006] In one embodiment, the nucleic acid sequence that is complementary to an mRNA encoding ABCD1 is an interfering RNA.

[0007] In another embodiment, the interfering RNA is an shRNA or siRNA.

[0008] In yet another embodiment, the siRNA comprises SEQ ID NO. 4, SEQ ID NO. 5, SEQ ID NO. 6, SEQ ID NO. 7 or a combination thereof.

[0009] In yet another embodiment, the reference standard comprises AAV9-ABCD1 vector yield in cell lysate and/or media from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCD 1.

[0010] In yet another aspect, the invention provides a method of treating X-linked adrenoleukodystrophy (X-ALD) in a subject in need thereof comprising administering to the subject a composition comprising purified AAV9-ABCD 1 vector obtained from the producer cells having increased AAV9 vector titers compared to a reference standard.

[0011] In one embodiment, the composition comprising purified AAV9-ABCD1 vector is administered to the subject by intrathecal administration.

[0012] In yet another aspect, the invention provides a method of treating X-linked adrenoleukodystrophy (X-ALD) in a subject in need thereof comprising administering to the subject an adeno-associated Virus (AAV) vector encoding an ATP binding cassette subfamily D member 1 (ABCD1), wherein said vector is administered to the subject by intrathecal administration.

[0013] In one embodiment, the intrathecal administration is mediated by an osmotic pump.

[0014] In another embodiment, the dose of vector is 0.5X10 n GC.

[0015] In yet another embodiment, the AAV is AAV9. [0016] In yet another aspect, the invention provides a method of providing ATP binding cassette subfamily D member 1 (ABCDl) to a subject having X-linked adrenoleukodystrophy (X-ALD) comprising administering to the subject a vector encoding ABCDl, wherein said vector is administered to the subject by intrathecal administration, and wherein ABCDl expression from said vector in the central nervous system is less than ABCDl expression from said vector in peripheral organs.

[0017] In one embodiment, the intrathecal administration is mediated by an osmotic pump.

[0018] In another embodiment, the dose of vector is about lxl0 13 GC to about 10xl0 13 GC.

[0019] In yet another embodiment, the vector is an adeno-associated virus (AAV) vector.

[0020] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

BRIEF DESCRIPTION OF THE DRAWINGS

[0021] The following Detailed Description, given by way of example, but not intended to limit the invention to certain embodiments described, may be understood in conjunction with the accompanying figures, incorporated herein by reference.

Figure 1 depicts improved AAV9-ABCD1 vector titers from transfected 293T cells incubated with siRNA specific for ABCDl mRNA.

Figure 2 depicts reduced ABCD 1 protein in AAV-ABCD 1 transfected cells incubated with an siRNA pool specific for ABCD 1 mRNA.

Figure 3 depicts improved AAV9-ABCD1 vector titers from transfected 293T cells incubated with siRNA specific for ABCDl mRNA in both cell lysates and conditioned media.

Figure 4 depicts distribution of rAAV9-ABCD 1 following intrathecal bolus delivery over 2 minutes.

Figure 5 depicts distribution of rAAV9-ABCD 1 following intrathecal pump infusion of rAAV9-ABCDl over 24 hours.

Figure 6 depicts low dose (0.5xl0 n gc) bolus and pump delivery of AAV9-ABCD1. Figure 7 depicts higher expression of ABCD 1 across peripheral organs (outside the CNS) two weeks after bolus injection of AAV9-ABCD1 compared to pump infusion of AAV9-ABCD1. Figure 8 depicts a vector map of AAV9-ABCD 1.

Figure 9 depicts distribution of endogenous ABCD1 across different organs.

Figure 10 depicts distribution of endogenous ABCD1 across different organs.

Figure 11 depicts expression of ABCD 1 after IT pump in Abcdl-/- mouse.

Figure 12 depicts expression of ABCD 1 after IT pump in Abcdl-/- mouse.

Figure 13 depicts spinal cord C26:0 level 15 days after IT pump and PT bolus injection. Figure 14 depicts ABCD1 expression in different cell types after IT pump delivery of AAV9- hABCDl . SC: spinal cord; DRG: dorsal root ganglion; CD31: endothelial marker; GFAP: astrocyte marker; IBA1 : microglial marker; TOPR03: nuclear counterstain; DRG shows expression speckled pattern in neuron and more prominently around neurons (satellite cells).

DETAILED DESCRIPTION OF THE INVENTION

Definitions

[0022] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In case of conflict, the present application, including definitions will control.

[0023] A "subject" is a vertebrate, including any member of the class mammalia, including humans, domestic and farm animals, and zoo, sports or pet animals, such as mouse, rabbit, pig, sheep, goat, cattle and higher primates.

[0024] As used herein, the terms "treat," "treating," "treatment," and the like refer to reducing or ameliorating X-ALD, e.g., adrenomyeloneuropathy (AMN), and/or symptoms associated therewith. It will be appreciated that, although not precluded, treating X-ALD or AMN does not require that the disorder, condition or symptoms associated therewith be completely eliminated.

[0025] Unless specifically stated or clear from context, as used herein, the term "about" is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. "About" is understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about.

[0026] As used herein "a decrease in expression" refers to an amount of ABCD 1 gene expression or protein expression in peripheral organs of a subject that is at least about 0.05 fold less (for example 0.1, 0.2, 0.3, 0.4, 0.5, 1, 5, 10, 25, 50, 100, 1000, 10,000-fold or more less) than the amount of ABCD1 gene expression or protein expression in the central nervous system of a subject having been administered a vector encoding ABCD1 according to the methods described herein. "Decreased" as it refers to ABCD1 gene expression or protein expression in peripheral organs of a subject also means at least about 5% less (for example 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99 or 100%) than the amount of ABCD 1 gene expression or protein expression in the central nervous system of a subject having been administered a vector encoding ABCD1 according to the methods described herein. Amounts can be measured according to standard methods known in the art for determining amounts of gene expression or protein expression.

[0027] As used herein "an increase in vector titers" refers to an amount of titer from producer cells transfected with a vector encoding ABCD1 that is at least about 0.05 fold more (for example 0.1, 0.2, 0.3, 0.4, 0.5, 1, 5, 10, 25, 50, 100, 1000, 10,000-fold or more) than the amount of titer from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCD1 according to the methods described herein. "Increased" as it refers to an amount of titer (concentration of AAV vector, often described in genome copies per milliliter) from producer cells transfected with a vector encoding ABCD 1 also means at least about 5% more (for example 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 99 or 100%) than the amount of titer from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCD1 according to the methods described herein. Amounts can be measured according to standard methods known in the art for determining amounts of AAV genomes, transgene expression, or protein expression.

[0028] Ranges provided herein are understood to be shorthand for all of the values within the range. For example, a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 (as well as fractions thereof unless the context clearly dictates otherwise).

[0029] As used herein, the term "reference level" refers to the level of titer in a known sample against which another test sample is compared. A reference level can be obtained, for example, from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCD 1 or with a control antisense oligonucleotide or siRNA. A reference level can be obtained, for example, from untreated subjects that do not have X-ALD. "Untreated" refers to the lack of therapy from administration of a vector expressing an ABCD1 transgene.

[0030] In this disclosure, "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. Patent law and can mean "includes," "including," and the like; "consisting essentially of or "consists essentially" likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.

[0031] Other definitions appear in context throughout this disclosure. Compositions and Methods

[0032] Compositions and methods of the invention provide treatments for X-linked adrenoleukodystrophy (X-ALD). X-linked adrenoleukodystrophy is a genetic disorder, caused by mutations in the ABCD1 gene, that occurs primarily in males and mainly affects the nervous system and the adrenal glands. Myelin of the brain and spinal cord deteriorate (demyelination), which reduces the functional ability of the nerves. In addition, damage to the outer layer of the adrenal glands (adrenal cortex) causes a shortage of certain hormones (adrenocortical insufficiency). There are several distinct types of X-linked

adrenoleukodystrophy, including a childhood cerebral form, an adrenomyeloneuropathy (AMN) type, and a form called Addison disease. As used herein, X-ALD does not include "neonatal adrenoleukodystrophy," which belongs to the peroxisomal biogenesis disorders of the Zellweger spectrum and is unrelated to mutations in ABCD 1. Methods for diagnosing or identifying subjects with X-ALD or AMN are known in the art and can include measurement of plasma very long chain fatty acid (VLCFA) levels and/or genetic testing; see, e.g., Engelen et al, Orphanet J Rare Dis. 2012; 7: 51; Aubourg and Chaussain, Horm Res. 2003;59 Suppl 1 : 104-5; Steinberg et al, Curr Protoc Hum Genet. 2008 Chapter 17:Unit 17.6; Steinberg SJ, Moser AB, Raymond GV. X-Linked Adrenoleukodystrophy. 1999 Mar 26 [Updated 2015 Apr 9]. In: Pagon RA, Adam MP, Ardinger HH, et al, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2016. Available from:

ncbi.nlm.nih.gov/books/NBK1315/).

[0033] Mutations in the ABCD1 gene cause X-linked adrenoleukodystrophy. The ABCD1 gene encodes the adrenoleukodystrophy protein (ALDP), which is involved in transporting very long-chain fatty acids (VLCFAs) into peroxisomes. ABCD1 gene mutations result in a deficiency of ALDP. When this protein is lacking, the transport and subsequent breakdown of VLCFAs is disrupted, causing abnormally high levels of these fats in the body. The accumulation of VLCFAs may be toxic to the adrenal cortex and myelin.

[0034] Correction of the genetic defect by gene therapy presents a viable therapy. Targeted, specific delivery of the ABCDl gene to the CNS is essential to avoid toxicity in peripheral organs. This can be achieved, for example, by administering an adeno-associated virus (AAV) vector encoding ABCD l via intrathecal administration.

[0035] Sequences encoding the ABCDl cDNA and its expressed protein are well known, and can be found, for example at Genbank Accession Nos. NG_009022.2 and NP_000024.2.

[0036] "AAV" is adeno-associated virus, and may be used to refer to the recombinant virus vector itself or derivatives thereof. The term covers all subtypes, serotypes and pseudotypes, and both naturally occurring and recombinant forms, except where required otherwise. As used herein, the term "serotype" refers to an AAV which is identified by and distinguished from other AAVs based on its serology, e.g., there are eleven serotypes of AAVs, AAV1- AAV11, and the term encompasses pseudotypes with the same properties. Many of these serotypes have unique biological properties from other AAV serotypes (e.g. cell surface receptor binding, intracellular trafficking). Thus, for example, AAV5 serotypes include AAV with the biological properties of AAV5, e.g., a pseudotyped AAV comprising AAV5 capsid and an AAV genome which is not derived or obtained from AAV5 or which genome is chimeric.

[0037] An "AAV vector" refers to a viral particle composed of at least one AAV capsid protein and an encapsidated polynucleotide. If the particle comprises a heterologous polynucleotide (i.e., a polynucleotide other than a wild-type AAV genome such as a transgene to be delivered to a mammalian cell), it can be referred to as "rAAV (recombinant AAV)." An AAV "capsid protein" includes a capsid protein of a wild-type AAV, as well as modified forms of an AAV capsid protein which are structurally and or functionally capable of packaging an AAV genome and bind to at least one specific cellular receptor which may be different than a receptor employed by wild type AAV. A modified AAV capsid protein includes a chimeric AAV capsid protein such as one having amino acid sequences from two or more serotypes of AAV, e.g., a capsid protein formed from a portion of the capsid protein from AAV5 fused or linked to a portion of the capsid protein from AAV2, and a AAV capsid protein having a tag or other detectable non-AAV capsid peptide or protein fused or linked to the AAV capsid protein, e.g., a portion of an antibody molecule which binds the transferrin receptor may be recombinantly fused to the AAV -2 capsid protein. [0038] Cells capable of producing AAV are known in the art and include, but are not limited to 293 cells, HeLa cells and insect cells.

[0039] In certain embodiments, methods of producing high titers of AAV can be utilized to maximize administration of ABCD 1. Transfected producer cells grown in culture can be incubated with a nucleic acid sequence that is complementary to an mRNA encoding ATP binding cassette subfamily D member 1 (ABCDl) and ii) transfected with an AAV vector comprising a nucleotide sequence encoding ABCDl into the cells (e.g., AAV9-ABCD1 vector). The amount of ABCDl mRNA expressed from the AAV vector is decreased, thereby increasing AAV-ABCD1 vector yield in cell lysate and/or media by about 1 fold to about 50 fold compared to a reference standard. In certain embodiments, the AAV-ABCD 1 vector yield in cell lysate and/or media is increased by about 4 fold. Vector titers can be determined according to methods well known in the art. Typically, this is performed using dot blots or quantitative PCR to measure AAV genomes. In general, AAV vector yields can be about 1X10 10 genome copies/ml (gc/ml) to about 1 xl0 16 gc/ml from cell lysates and from media.

[0040] In specific embodiments, the reference standard comprises AAV-ABCD 1 vector yield in cell lysate and/or media from producer cells that were not incubated with a nucleic acid sequence that is complementary to an mRNA encoding ABCDl .

[0041] This can be achieved, for example, by providing an antisense oligonucleotide that is complementary to ABCDl mRNA. Other nucleic acid sequences for use in practicing the methods of the invention and that are complementary to ABCDl mRNA can be those which inhibit post-transcriptional processing of ABCD l, such as an interfering RNA, including but not limited to an shRNA or siRNA, or an antagomir.

[0042] Sequences encoding the ABCDl mRNA are well known, and can be found, for example at Genbank Accession Nos. NM_000033.3.

[0043] Antisense oligonucleotides are typically designed to block expression of a DNA or RNA target by binding to the target and halting expression at the level of transcription, translation, or splicing. Antisense oligonucleotides of the present invention are

complementary nucleic acid sequences designed to hybridize under stringent conditions to ABCDl mRNA. Thus, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect.

[0044] In the context of this invention, hybridization means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases. For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. Complementary, as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target.

[0045] It is understood in the art that a complementary nucleic acid sequence need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable. A complementary nucleic acid sequence of the invention is specifically hybridizable when binding of the sequence to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of activity, and there is a sufficient degree of complementarity to avoid non-specific binding of the sequence to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed under suitable conditions of stringency. For example, stringent salt concentration will ordinarily be less than about 750 mM NaCl and 75 mM trisodium citrate, preferably less than about 500 mM NaCl and 50 mM trisodium citrate, and more preferably less than about 250 mM NaCl and 25 mM trisodium citrate. Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high stringency hybridization can be obtained in the presence of at least about 35% formamide, and more preferably at least about 50% formamide. Stringent temperature conditions will ordinarily include temperatures of at least about 30° C, more preferably of at least about 37° C, and most preferably of at least about 42° C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed. In a preferred embodiment, hybridization will occur at 30° C in 750 mM NaCl, 75 mM trisodium citrate, and 1% SDS. In a more preferred embodiment, hybridization will occur at 37° C in 500 mM NaCl, 50 mM trisodium citrate, 1% SDS, 35% formamide, and 100 μg/ml denatured salmon sperm DNA (ssDNA). In a most preferred embodiment, hybridization will occur at 42° C in 250 mM NaCl, 25 mM trisodium citrate, 1% SDS, 50% formamide, and 200 μg/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.

[0046] For most applications, washing steps that follow hybridization will also vary in stringency. Wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature. For example, stringent salt concentration for the wash steps will preferably be less than about 30 mM NaCl and 3 mM trisodium citrate, and most preferably less than about 15 mM NaCl and 1.5 mM trisodium citrate. Stringent temperature conditions for the wash steps will ordinarily include a temperature of at least about 25° C, more preferably of at least about 42° C, and even more preferably of at least about 68° C. In a preferred embodiment, wash steps will occur at 25° C in 30 mM NaCl, 3 mM trisodium citrate, and 0.1% SDS. In a more preferred embodiment, wash steps will occur at 42° C. in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS. In a more preferred embodiment, wash steps will occur at 68° C in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art. Hybridization techniques are well known to those skilled in the art and are described, for example, in Benton and Davis (Science 196: 180, 1977); Grunstein and Hogness (Proc. Natl. Acad. Sci., USA 72:3961, 1975); Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, New York, 2001); Berger and Kimmel (Guide to Molecular Cloning Techniques, 1987, Academic Press, New York); and Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York.

[0047] It is preferred that the antisense oligonucleotides of the present invention comprise at least 80% sequence complementarity to a target region within the target nucleic acid, moreover that they comprise 90% sequence complementarity and even more preferable to comprise 95% sequence complementarity to the target region within the target nucleic acid sequence to which they are targeted. For example, an antisense compound in which 18 of 20 nucleobases of the antisense oligonucleotide are complementary, and would therefore specifically hybridize, to a target region would represent 90 percent complementarity.

Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely using basic local alignment search tools (BLAST programs) (Altschul et al., J. Mol. Biol., 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656). Antisense and other compounds of the invention, which hybridize to ABCD1 mRNA, are identified through experimentation, and representative sequences of these compounds are herein below identified as preferred embodiments of the invention.

[0048] In another embodiment, the nucleic acid sequence that is complementary to ABCD 1 mRNA can be an interfering RNA, including but not limited to an shRNA or siRNA.

Interfering RNA includes, but is not limited to small interfering RNA ("siRNA") and small hairpin RNA ("shRNA"). Methods for constructing interfering RNAs are well known in the art. For example, the interfering RNA can be assembled from two separate oligonucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary (i.e., each strand comprises nucleotide sequence that is complementary to nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double stranded structure); the antisense strand comprises nucleotide sequence that is complementary to a nucleotide sequence in a target nucleic acid molecule or a portion thereof (i.e., an undesired gene) and the sense strand comprises nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. Alternatively, interfering RNA is assembled from a single oligonucleotide, where the self-complementary sense and antisense regions are linked by means of nucleic acid based or non-nucleic acid-based linker(s). The interfering RNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self- complementary sense and antisense regions, wherein the antisense region comprises a nucleotide sequence that is complementary to nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. The interfering can be a circular single-stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siRNA molecule capable of mediating RNA interference.

[0049] In certain embodiments of the invention, the interfering RNA coding region encodes a self-complementary RNA molecule having a sense region, an antisense region and a loop region. Such an RNA molecule when expressed desirably forms a "hairpin" structure, and is referred to herein as an "shRNA." The loop region is generally between about 2 and about 10 nucleotides in length. In a preferred embodiment, the loop region is from about 6 to about 9 nucleotides in length. In one such embodiment of the invention, the sense region and the antisense region are between about 15 and about 20 nucleotides in length. Following post- transcriptional processing, the small hairpin RNA is converted into a siRNA by a cleavage event mediated by the enzyme Dicer, which is a member of the RNase III family. The siRNA is then capable of inhibiting the expression of a gene with which it shares homology. For details, see Brummelkamp et al., Science 296:550-553, (2002); Lee et al, Nature Biotechnol., 20, 500-505, (2002); Miyagishi and Taira, Nature Biotechnol 20:497-500, (2002); Paddison et al. Genes & Dev. 16:948-958, (2002); Paul, Nature Biotechnol, 20, 505- 508, (2002); Sui, Proc. Natl. Acad. Sd. USA, 99(6), 5515-5520, (2002); Yu et al. Proc NatlAcadSci USA 99:6047-6052, (2002).

[0050] The target RNA cleavage reaction guided by siRNAs is highly sequence specific. In general, siRNA containing a nucleotide sequences identical to a portion of the target gene (i.e., ABCD1) are preferred for inhibition. However, 100% sequence identity between the siRNA and the target gene is not required to practice the present invention. Thus the invention has the advantage of being able to tolerate sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence. For example, siRNA sequences with insertions, deletions, and single point mutations relative to the target sequence have also been found to be effective for inhibition. Alternatively, siRNA sequences with nucleotide analog substitutions or insertions can be effective for inhibition.

[0051] In yet another embodiment, the nucleic acid sequence that is complementary to

ABCD 1 mRNA is an antagomir. Antagomirs are single stranded, double stranded, partially double stranded and hairpin structured chemically modified oligonucleotides that target a microRNA. Preferably, an antagomir featured in the invention includes a nucleotide sequence sufficiently complementary to hybridize to a miRNA target sequence of about 10 to 25 nucleotides, preferably about 15 to 20 nucleotides.

[0052] In certain embodiments, antagomirs are RNA-like oligonucleotides that harbor various modifications for RNase protection and pharmacologic properties such as enhanced tissue and cellular uptake. An antagomir can differ from normal RNA by having complete 2'- O-methylation of sugar, phosphorothioate backbone and a cholesterol-moiety at 3'-end. Phosphorothioate modifications provide protection against RNase activity and their lipophilicity contributes to enhanced tissue uptake. In a preferred embodiment, the antagomir includes six phosphorothioate backbone modifications; two phosphorothioates are located at the 5'-end and four at the 3'-end. Antagomirs of the present invention can also be modified with respect to their length or otherwise the number of nucleotides making up the antagomir. [0053] The nucleic acid sequences used to practice this invention, whether RNA, cDNA, genomic DNA, vectors, viruses or hybrids thereof, can be isolated from a variety of sources, genetically engineered, amplified, and/or expressed/ generated recombinantly. Recombinant nucleic acid sequences can be individually isolated or cloned and tested for a desired activity. Any recombinant expression system can be used, including e.g. in vitro, bacterial, fungal, mammalian, yeast, insect or plant cell expression systems.

[0054] Nucleic acid sequences of the invention can be inserted into delivery vectors and expressed from transcription units within the vectors (e.g., AAV vectors). The recombinant vectors can be DNA plasmids or viral vectors. Generation of the vector construct can be accomplished using any suitable genetic engineering techniques well known in the art, including, without limitation, the standard techniques of PCR, oligonucleotide synthesis, restriction endonuclease digestion, ligation, transformation, plasmid purification, and DNA sequencing, for example as described in Sambrook et al. Molecular Cloning: A Laboratory Manual. (1989)), Coffin et al. (Retroviruses. (1997)) and "RNA Viruses: A Practical Approach" (Alan J. Cann, Ed., Oxford University Press, (2000)). As will be apparent to one of ordinary skill in the art, a variety of suitable vectors are available for transferring nucleic acids of the invention into cells. The selection of an appropriate vector to deliver nucleic acids and optimization of the conditions for insertion of the selected expression vector into the cell, are within the scope of one of ordinary skill in the art without the need for undue experimentation. Viral vectors comprise a nucleotide sequence having sequences for the production of recombinant virus in a packaging cell. Viral vectors expressing nucleic acids of the invention can be constructed based on viral backbones including, but not limited to, a retrovirus, lentivirus, adenovirus, adeno-associated virus, pox virus or alphavirus. The recombinant vectors capable of expressing the nucleic acids of the invention can be delivered as described herein, and persist in target cells (e.g., stable transformants).

[0055] Nucleic acid sequences used to practice this invention can be synthesized in vitro by well-known chemical synthesis techniques, as described in, e.g., Adams (1983) J. Am. Chem. Soc. 105:661; Belousov (1997) Nucleic Acids Res. 25:3440-3444; Frenkel (1995) Free Radic. Biol. Med. 19:373-380; Blommers (1994) Biochemistry 33:7886-7896; Narang (1979) Meth. Enzymol. 68:90; Brown (1979) Meth. Enzymol. 68: 109; Beaucage (1981) Tetra. Lett. 22: 1859; U.S. Patent No. 4,458,066.

[0056] Nucleic acid sequences of the invention can be stabilized against nucleolytic degradation such as by the incorporation of a modification, e.g., a nucleotide modification. For example, nucleic acid sequences of the invention include a phosphorothioate at least the first, second, or third internucleotide linkage at the 5' or 3' end of the nucleotide sequence. As another example, the nucleic acid sequence can include a 2'-modified nucleotide, e.g., a 2'-deoxy, 2'-deoxy-2'-fluoro, 2'-0-methyl, 2'-0-methoxyethyl (2'-0-MOE), 2'-0-aminopropyl (2'-0-AP), 2'-0-dimethylaminoethyl (2'-0-DMAOE), 2'-0-dimethylaminopropyl (2'-0- DMAP), 2'-0-dimethylaminoethyloxyethyl (2'-0-DMAEOE), or 2'-0~N-methylacetamido (2'-0~NMA). As another example, the nucleic acid sequence can include at least one 2'-0- methyl-modified nucleotide, and in some embodiments, all of the nucleotides include a 2'-0- methyl modification.

[0057] Techniques for the manipulation of nucleic acids used to practice this invention, such as, e.g., subcloning, labeling probes (e.g., random-primer labeling using Klenow polymerase, nick translation, amplification), sequencing, hybridization and the like are well described in the scientific and patent literature, see, e.g., Sambrook, ed., MOLECULAR CLONING: A LABORATORY MANUAL (2ND ED.), Vols. 1-3, Cold Spring Harbor Laboratory, (1989); CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Ausubel, ed. John Wiley & Sons, Inc., New York (1997); LABORATORY TECHNIQUES IN BIOCHEMISTRY AND

MOLECULAR BIOLOGY: HYBRIDIZATION WITH NUCLEIC ACID PROBES, Part I. Theory and Nucleic Acid Preparation, Tijssen, ed. Elsevier, N.Y. (1993).

[0058] ABCDl vector administration provided by intravenous (IV) or intracerebroventricular (ICV) administration has recently been determined to cause cardiac toxicity. Intrathecal administration is a route of administration comprising injection of desired agents into the subarachnoid space of the spinal canal, thereby providing the agents into the cerebrospinal fluid (CSF). Using intrathecal administration, ABCDl expression from a vector within in the central nervous system is less than ABCDl expression from a vector within peripheral organs, such as the heart. Excess ABCD 1 expression in peripheral organs can result in toxicity and therefore, intrathecal administration of ABCDl vectors comprises an improved method of therapy for X-ALD, e.g., for AMN.

[0059] In some embodiments, the intrathecal administration is via a pump. The pump may be a surgically implanted osmotic pump. In certain embodiments, the osmotic pump is implanted into the subarachnoid space of the spinal canal to facilitate intrathecal administration.

[0060] In certain embodiments, human subjects receive a one-time treatment of intrathecally delivered vector (e.g., AAV9) comprising ABCDl in an amount of about lxlO 13 GC to about 10x10 13 GC over a period of about 24 hours.

[0061] The slow continuous intrathecal infusion of the AAV9-hABCD l can be scaled up to humans by using an osmotically driven pump such as the DUROS ® implant, ALZA Corporation (Mountain View, CA). See also, J.C. Wright, J. Culwell, Long-term controlled delivery of therapeutic agents by the osmotically driven DUROS® implant, in: M.J.

Rathbone, J. Hadgraft, M.S. Roberts (Eds.), Modified-Release Drug Delivery Technology, Informa Healthcare, New York, 2008, pp. 143-149.

[0062] Osmotic delivery devices and their component parts have been described, for example, in U.S. Pat. Nos. 5,609,885; 5,728,396; 5,985,305; 5,997,527; 6, 113,938;

6,132,420; 6,156,331; 6,217,906; 6,261,584; 6,270,787; 6,287,295; 6,375,978; 6,395,292; 6,508,808; 6,544,252; 6,635,268; 6,682,522; 6,923,800; 6,939,556; 6,976,981; 6,997,922; 7,014,636; 7,207,982; 7, 112,335; 7, 163,688; U.S. Patent Publication Nos. 2005-0175701, 2007-0281024, and 2008-0091176.

[0063] The DUROS® delivery device typically consists of a cylindrical reservoir which contains the osmotic engine, piston, and drug formulation. The reservoir is capped at one end by a controlled-rate water-permeable membrane and capped at the other end by a diffusion moderator through which drug formulation is released from the drug reservoir. The piston separates the drug formulation from the osmotic engine and utilizes a seal to prevent the water in the osmotic engine compartment from entering the drug reservoir. The diffusion moderator is designed, in conjunction with the drug formulation, to prevent body fluid from entering the drug reservoir through the orifice.

[0064] The DUROS® device releases a therapeutic agent at a predetermined rate based on the principle of osmosis. Extracellular fluid enters the DUROS® device through a semipermeable membrane directly into a salt engine that expands to drive the piston at a slow and even delivery rate. Movement of the piston forces the drug formulation to be released through the orifice or exit port at a predetermined sheer rate. In one embodiment of the present invention, the reservoir of the DUROS® device is load with a suspension formulation of the present invention, comprising, for example, lxl0 n gc AAV9-hABCDl, wherein the device is capable of delivering the suspension formulation to a subject over an extended period of time at a pre-determined, therapeutically effective delivery rate.

[0065] Other implantable, drug delivery devices may be used in the practice of the present invention and may include regulator-type implantable pumps that provide constant flow, adjustable flow, or programmable flow of the compound, such as those available from Codman & Shurtleff, Inc. (Raynham, Mass.), Medtronic, Inc. (Minneapolis, Minn.), and Tricumed Medinzintechnik GmbH (Germany). [0066] The present invention is additionally described by way of the following illustrative, non-limiting Examples that provide a better understanding of the present invention and of its many advantages.

EXAMPLES

[0067] The following Examples illustrate some embodiments and aspects of the invention. It will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be performed without altering the spirit or scope of the invention, and such modifications and variations are encompassed within the scope of the invention as defined in the claims which follow. The following Examples do not in any way limit the invention.

Example 1: Production of AAV9-ABCD1 in the presence of siRNA specific for ABCDl mRNA improves AAV vector titers from transfected 293T cells.

[0068] A high amount of cell death/cytopathic effects during production of AAV9-ABCD 1 has been previously observed, likely due to overexpression of ABCDl protein in producer cells. This toxicity reduced AAV vector yields. To mitigate the toxicity and improve vector yields, ABCDl mRNA was targeted using a pool of siRNAs.

[0069] AAV packing was carried out as follows. 1-1.5χ10 7 293T cells were plated on 15cm plates in and cultured overnight.

Table 1 : Packaging and Collection Media

[0070] On day 2, transfection mix was prepared as follows:

Table 2: Transfection Mix "Serotype plasmid 12ug

2M CaCl 96.9

2.5mM Hepes up to 780ul

Total Volume 780ul Total Volume 780ul

[0071] Tube A and Tube B were combined drop-wise while vortexing for 1 minute. The mix was incubated at room temperature for 20 minutes. 1.5ml of virus mix per 15cm plate was added, distributing drop-wise over the surface of the plate. Plates were tilted to mix evenly and incubated overnight. At day 3, media was replaced with DMEM 2% FBS 1% p/s. At day 5, half of the plate media volume was removed from each plate. Cells were collected from plates by washing with remaining media or gently using a cell scraper. Cells were spun down at 1300 RPM for 5 minutes. Supernatant was removed. Cells were loosened by flicking the bottom of the tube and re-suspended in lml EDTA PBS per plate of cells and spun at 1300RPM for 5 minutes. Cells were re-suspended in lml lysis buffer per plate and optionally stored at -80 C. Following gradient purification, virus was buffer exchanged into PBS, quantified by qPCR, and used for experimentation.

[0072] On the day of 293T cell plating, cells were transfected with the pool of siRNA specific for ABCDl mRNA or a non-targeting control siRNA. Another control was an AAV9 vector encoding GFP (AAV9-GFP) in the presence of the ABCDl siRNA. The following day both samples were transfected with AAV plasmids to produce AAV9-ABCD 1.

[0073] The siRNA protocol has multiple steps:

1. Prepare 5 μΜ of pooled (25% of each of 4 siRNAs) siRNA solution in 1 χ siRNA buffer (GE Healthcare) or another appropriate RNase-free solution from stock solution.

2. In separate tubes, dilute the siRNA (lOOul in tubel) and the appropriate DharmaFECT transfection reagent (30ul in tube2) with serum-free DMEM medium to 2ml volume respectively.

3. Gently mix the contents of each tube by pipetting carefully up and down. Incubate for 5 minutes at room temperature.

4. Add the contents of Tube 1 to Tube 2, for a total volume of 4ml. Mix by pipetting carefully up and down and incubate for 20 minutes at room temperature. Add 12 ml of complete DMEM (10% FBS) to the 4ml.

5. Add 4 ml of resuspended 293T cells in complete media that are at a concentration of 3.75e6 cells/ml (total 1.5E7 cells) to the 16 ml of the transfection mixture from step 4 (final siRNA concentration of 25 nM).

6. Plate into 15 cm dish and incubate 24 h. 7. Change media to complete DMEM 10% FBS 1 h before calcium phosphate transfection with AAV plasmids.

8. Proceed with standard AAV production and purification protocol.

[0074] Three days post transfection cells were harvested, lysed, and vector yields (in genome copies) was determined by qPCR as follows:

Table 3 : aPCR Materials

F2: CCTCGACTGTGCCTTCTAG (SEQ ID NO. 1)

R2: TGCGATGCAATTTCCTCAT (SEQ ID NO. 2)

Probe: 5'FAM-tgccagccatctgttgtttgcc-MGB (SEQ ID NO. 3)

Nuclease free water

F and R qPCR primers

TM FAM Probe

Tag Man Fast universal PCR Master Mix

PCR plates for 7500 (Fast) Qpcr

PCR plate film

[0075] The qPCR protocol has multiple steps:

1. Dilute vector 1 : 100-1 : 1000 in nuclease-free water and vortex. Use in qPCR.

2. Use plasmid 675.5 (5999bp) as genome copy (GC) standard. Create a standard from 10 7 - 10 2 gc/mL.

3. Prepare the master mix in an amount large enough to measure the standards and samples in triplicate. The master mix includes 2ul H20, 1.2ul primer mix (F and R= 5ul of each lOOum stock in 90ul of water), lul primer mix (F and R= 6ul of each lOOum stock in 88ul of water), 0.8ul TM FAM Probe 2.5uM, lul TM FAM Probe 2uM (4 ul of lOOuM stock

+ 196 ul water), and 5ul of TaqMan Fast universal PCR Master Mix 2x.

4. Mix and aliquot 9ul in wells of plate.

5. Add lul of template diluted in water.

6. Program 7500 machine to have thermal cycling parameters where stage 1 has reps 1, 95°C:20 and stage 2 has reps 40, 95°C:03; 60°C:30.

7. Analyze data. Slope for standard should be— 3.3.

[0076] The yield is reported as relative titer in which the AAV9-GFP sample was set arbitrarily at 100% and the other two samples normalized to this value. Production of AAV9- ABCD1 in the presence of the siRNA pool against ABCDl mRNA improved the vector titer (and yield) by approximately 4-fold compared to the control siRNA (Figure 1). Table 4: siR As Targeting ABCDl

[0077] 293T cells were transfected with control siRNA (Figure 2, lanes 1, 2), siRNA pool against ABCDl mRNA (Figure 2, lanes 3-6) or untransfected (Figure 2, lane 7, "normal" refers to endogenous levels of ABCDl protein in 293T cells). AAV-ABCD1 plasmid (Figure 2, lanes 1-4) or AAV-GFP plasmid (Figure 2, lane 5, 6) was transfected the following day. Three days later, cell lysates were electrophoresed on an SDS PAGE gel and an immunoblot for ABCDl protein was performed to assess siRNA knockdown. Actin blotting was performed for loading control. 8s and Is refers to 8 second and 1 second exposure of the radiographic film, respectively. The ABCDl siRNA reduces the level of overexpressed ABCD 1 compared to control siRNA.

[0078] 293T cells were left untransfected (no siRNA) or transfected with control siRNA or the siRNA pool against ABCDl mRNA. The following day cells were transfected with AAV plasmids to produce AAV9-ABCD 1. On day 3 post transfection of AAV plasmids, qPCR was performed to determine the amount of vector (g.c.) in cell lysate and in the media of the transfected cells. An approximate 3-4 fold increase in AAV9-ABCD1 vector yield in both cell lysate and media was observed (Figure 3).

Example 2: Intrathecal delivery of rAAV9-ABCDl by osmotic pump in a mouse model of adrenomyeloneuropathy leads to more uniform and widespread gene delivery to the CNS

[0079] Self-complementary AAV9 GFP(scAAV9GFP) and rAAV9 encoding ABCD 1

(rAAV9-ABCDl) were delivered to Abcdl-/- mice intrathecally (IT) either by bolus over a 2 minute duration or by osmotic pump over 24 hour duration with PBS injection as sham control. Two weeks after injection, mice were sacrificed and perfused with 4% PFA. Tissues were then collected, sectioned and stained for immunofluorescence analysis.

[0080] scAAV9-GFP delivered IT by osmotic pump led to widespread expression across CNS-relevant cell types and DRGs in a dose -dependent manner. Spinal cord and DRG had higher expression compared with brain, but GFP expression was also detected in peripheral organs (liver, heart and adrenal gland), with highest expression seen at 3X10 n GC.

[0081] A similar distribution pattern of ABCDl protein was detected after rAAV9-ABCDl intrathecal pump delivery. In general, higher doses (2X10 n GC and 1X10 U GC) led to more expression in CNS and peripheral organs compared with a lower dose (0.5X10 n GC). By comparison, intrathecal bolus delivery over 2 minutes led to the highest amount of ABCD 1 expression in the thoracic region, however, even a higher dose (lxlO n gc) did not lead to more widespread delivery in cervical and lumbar regions (Figure 4).

[0082] Notably, widespread expression of ABCD l across CNS was even detected after low dose, direct intrathecal bolus injection of 0.5X10 n GC (Figure 5). For example, 0.5X10 n GC bolus and pump delivery show similar expression of ABCDl in the cervical cord, while heart tissue demonstrated higher expression after bolus injection (Figure 6). It was concluded that the same dose delivered by pump led to higher expression in brain and spinal cord far from the injection site and comparatively less leakage to peripheral organs compared with bolus injection (Figure 7). Delivering rAAV9-ABCDl at 0.5X10 n GC by intracerebroventricular administration results in behavioral improvement in the Abcdl-/- mouse despite localized expression in brain. Therefore, even better performance at this dose using the outlined intrathecal pump delivery can be achieved. At a dose of 1X10 U GC administered via intrathecal pump, ABCDl expression in the central nervous system was about 3 fold higher than expression of ABCDl in the central nervous system of an untreated subject that does not have X-ALD (e.g., wild-type). Importantly, ABCDl expression in peripheral organs was about 90% less than expression of ABCD 1 expression in peripheral organs of an untreated subject that does not have X-ALD (see Figure 11, where protein expression among different tissue types in Western blots was normalized to endogenous wild-type levels).

[0083] In conclusion, rAAV9-mediated ABCD 1 gene transfer via intrathecal osmotic pump leads to more uniform and widespread gene delivery to the CNS with reduced leakage into the systemic circulation compared with intrathecal bolus injection.

Example 3: rAAV9 -mediated ABCDl gene transfer via intrathecal osmotic pump leads to a reduction in C26:0 levels in the spinal cord

[0084] C26:0 is the biochemical hallmark of adrenomyeloneuropathy. To assess for the presence of free very long chain fatty acids (VLCFA) after AAV9 gene delivery, lipidomic analysis was performed on spinal cord samples. Absolute values of C26:0 and C24:0 as well as ratios of C26:0/C22:0 are reported. It was determined that rAAV9-mediated ABCDl gene transfer via intrathecal osmotic pump (lxl0 n gc) leads to a 20% reduction in C26:0 levels in the spinal cord (Figure 13). The levels after intrathecal osmotic pump delivery are comparable to those after intrathecal bolus delivery but avoid systemic leakage. [0085] Immunofluorescence staining and confocal microscopy imaging were additionally conducted. For tissue section imaging, sections of spinal cord (16 μιη) were cut at -25 °C using cryostat (Leica) and stored at -80 °C. Sections were stained with mouse antihuman ABCD 1 antibody and then costained with rabbit anti-GFAP (Dako, Carpinteria, CA), rabbit anti-IBAl (Wako, Richmond, VA) and rabbit anti-CD31 (Abeam) respectively to localize the cell type. TOPRO-3 (Thermo Fisher Scientific) was used as fluorescent dye for nuclear counterstaining. The slides were imaged by confocal laser microscope and transduced cells counted. Estimates of ABCD 1 transduced cells of each cell type were documented in 20 χ and 40 x (for microglia) magnification images. rAAV9-mediated ABCD 1 gene transfer via intrathecal osmotic pump ( lxl0 n gc) targets mainly astrocytes, endothelial cells and a few neurons in the spinal cord (Figure 14). Within with dorsal root ganglia it targets both satellite cells and neurons (Figure 14).

OTHER EMBODIMENTS

[0086] It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.