Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MULTIPLE SCLEROSIS TREATMENT
Document Type and Number:
WIPO Patent Application WO/2016/179634
Kind Code:
A1
Abstract:
An antisense oligomer capabfe of binding to a selected target on an ITGA4 gene transcript to modify pre-mRNA splicing in an ITGA4 gene transcript or part thereof.

Inventors:
WILTON STEPHEN DONALD (AU)
FLETCHER SUE (AU)
AUNG-HTUT MAY (AU)
Application Number:
PCT/AU2016/000158
Publication Date:
November 17, 2016
Filing Date:
May 10, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV MURDOCH (AU)
International Classes:
C12N15/113
Domestic Patent References:
WO2004094636A12004-11-04
WO2014201560A12014-12-24
WO2011143274A12011-11-17
WO2000020635A12000-04-13
WO2005072340A22005-08-11
WO2010008474A22010-01-21
Foreign References:
US5149797A1992-09-22
US4458066A1984-07-03
US6287860B12001-09-11
US31529899A1999-05-20
US6965025B22005-11-15
US6969400B22005-11-29
US6887906B12005-05-03
US9315298A1998-06-08
US20030027780A12003-02-06
US20040248833A12004-12-09
US6806084B12004-10-19
US20100016215A12010-01-21
Other References:
LIMMROTH, V. ET AL.: "CD 49d antisense drug ATL1102 reduces disease activity in patients with relapsing-remitting MS", NEUROLOGY, vol. 83, no. 20, 2014, pages 1780 - 1788, XP055327974
AARTSMA-RUS, A. A. ET AL.: "Antisense-mediated exon skipping: a versatile tool with therapeutic and research applications", RNA, vol. 13, no. 10, 2007, pages 1609 - 1624, XP009144451
MARCEL VELTROP ET AL.: "Antisense-mediated exon skipping: Taking advantage of a trick from Mother Nature to treat rare genetic diseases", EXPERIMENTAL CELL RESEARCH, vol. 325, 30 January 2014 (2014-01-30), pages 50 - 55, XP055225698, DOI: 10.1016/j.yexcr.2014.01.026
KAVITHA SIVA ET AL.: "Exon-Skipping Antisense Oligonucleotides to Correct Missplicing in Neurogenetic Diseases", NUCLEIC ACID THERAPEUTICS, vol. 24, no. 1, 1 February 2014 (2014-02-01), pages 69 - 86, XP055327023, DOI: 10.1089/nat.2013.0461
ALEKSANDER TOUZNIK ET AL.: "New developments in exon skipping and splice modulation therapies for neuromuscular diseases", EXPERT OPINION ON BIOLOGICAL THERAPY, vol. 14, no. 6, 27 June 2014 (2014-06-27), pages 809 - 819, XP055492635, DOI: 10.1517/14712598.2014.896335
KOURO ET AL.: "The Novel a4B Murine a4 Integrin Protein Splicing Variant Inhibits a4 Protein-dependent Cell Adhesion", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 289, no. 23, 6 June 2014 (2014-06-06), pages 16389 - 16398, XP055514525, DOI: 10.1074/jbc.M114.553610
MYERS K. J. ET AL.: "Antisense oligonucleotide blockade of alpha 4 integrin prevents and reverses clinical symptoms in murine experimental autoimmune encephalomyelitis", JOURNAL OF NEUROIMMUNOLOGY, vol. 160, no. 1-2, 1 March 2005 (2005-03-01), pages 12 - 24, XP004745483, DOI: 10.1016/j.jneuroim.2004.10.029
LIMMROTH, V. ET AL.: "CD49d antisense drug ATL1102 reduces disease activity in patients with relapsing-remitting MS", NEUROLOGY, vol. 83, no. 20, 2014, pages 1780 - 1788, XP055327974, DOI: 10.1212/WNL.0000000000000926
HAMES ET AL.: "Nucleic Acid Hybridization", 1985, IRL PRESS, pages: 107 - 108
MIYADA C. G.WALLACE R. B.: "Oligonucleotide Hybridization Techniques", METHODS ENZYMOL., vol. 154, 1987, pages 94 - 107, XP009096793, DOI: 10.1016/0076-6879(87)54072-1
DEVERAUX ET AL., NUCLEIC ACIDS RESEARCH, vol. 12, 1984, pages 387 - 395
SUMMERTONWELLER, ANTISENSE NUCLEIC ACID DRUG DEVELOPMENT, vol. 7, pages 187 - 197
JEARAWIRIYAPAISARN ET AL., MOL. THER., vol. 16, no. 9, 2008, pages 1624 - 1629
BEAUCAGE ET AL., TETRAHEDRON LETTERS, vol. 22, 1981, pages 1859 - 1862
WOLFF ET AL., SCIENCE, vol. 247, 1990, pages 1465 - 1468
MANN CJ ET AL., PROC, NATL. ACAD. SCIENCE, vol. 98, no. 1, 2001, pages 42 - 47
GEBSKI ET AL., HUMAN MOLECULAR GENETICS, vol. 12, no. 15, 2003, pages 1801 - 1811
FRALEY ET AL., TRENDS BIOCHEM. SCI., vol. 6, 1981, pages 77
MANNINO ET AL., BIOTECHNIQUES, vol. 6, 1988, pages 682
DOKKAROJANASAKUL, ADVANCED DRUG DELIVERY REVIEWS, vol. 44, pages 35 - 49
FRIEDMANN, SCIENCE, vol. 244, 1989, pages 1275 - 1280
ROSENBERG, CANCER RESEARCH, vol. 51, no. 18, 1991, pages 5074S - 5079S
ROSENFELD ET AL., CELL, vol. 68, 1992, pages 143 - 155
ROSENFELD ET AL., SCIENCE, vol. 252, 1991, pages 431 - 434
THE BRIGHAM ET AL., AM. J. MED. SCI., vol. 298, 1989, pages 278 - 281
HAZINSKI ET AL., AM. J. RESP. CELL MOLEC. BIOL., vol. 4, 1991, pages 206 - 209
WANGHUANG, PROC. NATL. ACAD. SCI. (USA, vol. 84, 1987, pages 7851 - 7855
WUWU, J. BIOL. CHEM., vol. 263, 1988, pages 14621 - 14624
CLINICAL RESEARCH, 1991, pages 39
ANDERSON, SCIENCE, vol. 256, 1992, pages 808 - 813
BARTEAU ET AL., CURR GENE THER, vol. 8, no. 5, 2008, pages 313 - 23
MUELLER ET AL., CLIN REV ALLERGY IMMUNOL, vol. 35, no. 3, 2008, pages 164 - 78
LI ET AL., GENE THER., vol. 13, no. 18, 2006, pages 1313 - 9
SIMOES ET AL., EXPERT OPIN DRUG DELIV, vol. 2, no. 2, 2005, pages 237 - 54
MANN ET AL., J GEN MED, vol. 4, 2002, pages 644 - 654
VILLEGASMCPHAUL, CURR PROTOC MOL BIOL, 2005
COOPER ET AL., NEUROLOGY, vol. 61, 2003, pages 93 - 96
Attorney, Agent or Firm:
WRAYS PTY LTD (West Perth, Western Australia 6005, AU)
Download PDF:
Claims:
CLAIMS

1. An antisense oligomer capable of binding to a selected target on an ITGA4 gene transcript to modify pre-mRNA splicing in an ITGA4 gene transcript or part thereof.

2. The antisense oiigomer of claim 1 chosen from the list comprising: SEQ ID NO; 1- 202.

3. The antisense oligomer of claim 2 chosen from the list comprising: SEQ ID NO: 1- 22.

4. The antisense oligomer of claim 1 chosen from the antisense oligomers listed in Table 3 to 7.

5. The antisense oligomer of claim 4 chosen from the antisense oligomers listed in Table 3.

6. Th antisense oligomer of any one of claims 1-5 wherein the antisense oiigomer contains one or more nudeotide positions subject to an alternative chemistry or modification chosen from the list comprising: (i) a modified backbone structure; (ii) modified sugar moieties; (Hi) resistance to RNase H; (ίν) oligomeric mimetic chemistry; (v) chemical conjugation to a moiety; (vi) tagging with a cell penetrating peptide.

7. The antisense oligomer of cfairn 6 wherein the uracii (U) of the antisense oiigomer is replaced by a thymine (T).

8. Th antisense oiigomer of claim 1 wherein the modification of pre-mRNA splicing results in skipping of one or more exons of the ITGA4 RNA.

9. A method for manipulating splicing in an ITGA4 gene transcript, the method including the step of: a) providing one or more of the antisense oligomers according to any one of claims 1 to 7 and allowing the oligomers) to bind to a target nucleic acid site.

10. A pharmaceutical, prophylactic, or therapeutic composition to treat, prevent or ameliorate the effects of a disease related to ITGA4 expression in a patient, th composition comprising: a) one or more antisense oligomers according to any one of claims 1 to 7, and b) one or more pharmaceutically acceptable carriers and/or diluents.

1 1. A method to treat, prevent or ameiiorate the effects of a disease associated with ITGA4 expression, comprising the step of: a) administering to the patient an effective amount of one or more antisense oligomers or pharmaceutical composition comprising one or more antisense oligomers according to any one of claims 1 to 7.

12. The use of purified and isolated antisense oligomers according to any one of claims 1 to 7, for the manufacture of a medicament to treat, prevent or ameliorat the effects of a disease associated with ITGA4 expression.

13. A kit to treat, prevent or ameiiorate the effects of a disease associated with 1TGA4 expression in a patient, which kit comprises at least an antisense oligomer according to any one of claims 1 to 7 and combinations or cocktails thereof, packaged in a suitable container, together with instructions for its use

The composition of claim 9, method of claim 10, use of claim 11 or kit of claim 12 wherein the iTGA4 expression related disease is multiple sclerosis.

Description:
Multiple Scierosis Treatment TECHNICAL FIELD

[00013 The present invention relates to antisense oligomers to facilitate splice modification and induce exon skipping in the integrin a!pha-4 (1TGA4) gene. The invention further provides methods to treat, prevent or ameliorate the effects of multiple sclerosis b administration of antisense oligomers and therapeutic compositions comprising antisense oligomers to the integrin aipha-4 (ITGA4) gene.

BACKGROUND ART

[0002] The following discussion of the background art is intended to facilitate an understanding of the present invention only. The discussio is not an acknowledgement or admission that any of the material referred to is or was part of the common general knowledge as at the priority date of the application.

[00033 Multiple Sclerosis (MS) is a neurodegenerative disease affecting approximately 2,5 million people around the world, with a higher risk in women. It is an autoimmune disease that attacks the protective myelin sheath of the neurons in the Central Nervous System (CNS) and results in inflammation and lesions. Patients with MS show one or more symptoms affecting sensation, movement and strength, and feeling and thinking. Symptoms are variable and some may be hidden for years before being diagnosed. MS can be either relapsing or progressive form and 65% of the patients are in the former condition.

[0004] The disease onset is stili unclear and there is no cure for MS yet. There are several approved disease modifying drugs to modulate the immune system and decrease the frequency and severity of attacks or relapses. To date, no therapeutic drug is available to stop or reverse neurodegeneration. Of all the anti-inflammatory drugs, humanized monoclonal antibody targeting the adhesion molecule integrin alpha- 4 (ITGA4) has shown to be effective in treating Relapsing-Rernitfing MS (RRMS). However, the side effects associated with the use of monoclonal antibodies such as infusion reaction, hypersensitivity and presence of neutralizing antibodies still persist in patients.

[0005] The present invention seeks to provide an alternative method to treat, prevent or ameliorate the effects of multiple sclerosis.

SUMMARY OF INVENTION

[0006] Broadly, according to one aspect of the invention, there is provided an isolated or purified antisense oiigomer for modifying pre-mRNA splicing in the integrin alpha-4 (ITGA4) gene transcript or part thereof. Preferably, there is provided an isolated or purified antisense oiigomer for inducing exon exclusion in the ITGA4 gene transcript or part thereof.

[0007] For example, in one aspect of the invention, there is provided an antisense oligomer of 10 to 50 nucleotides comprising a targeting sequence complementary to a region near or within an tntron of the ITGA4 gene pre-mRNA.

[0008] Preferably, the antisense oiigomer is a phosphorodiamidat morphofino oligomer,

[00093 Preferably, the antisense oligomer is selected from the group comprising the sequences set forth in Tables 3 to 7.

[0010] Preferably, the antisense oiigomer is seiected from the list comprising: SEQ ID NO: 1-202.

[0011] The antisense oligomer preferably operates to induce skipping of one or more of the exons of the 1TGA4 gene.

[0012] The antisense oiigomer of the invention may be selected to be an antisense oligomer capable of binding to a selected ITGA4 target site, wherein the target site is an mRNA splicing site selected from a splice donor site, splice acceptor sites, or exonic splicing elements. The target site may also include some flanking intronic sequences when the donor or acceptor splice sites are targeted.

[0013] More specifically, the antisense oligomer may be selected from the group comprising of any one or more of SEQ ID NOs: 1-202 and/or the sequences set forth in Tables 3 to 7, and combinations or cocktails thereof. This includes sequences which can hybridis to suc sequences under stringent hybridisation conditions, sequences complementary thereto, sequences containing modified bases, modified backbones, and functional truncations or extensions thereof which possess or modulate pre-mRNA processing activity in an 1TGA4 gene transcript in certain embodiments, antisense oligomers may be 100% complementary to the target sequence, or may include mismatches, e.g., to accommodate variants, as long as a heteroduplex formed between the oligonucleotide and target sequence is sufficiently stable to withstand the action of cellular nucleases and other modes of degradation which may occur in vivo. Hence, certain oligonucleotides may have about or at least about 70% sequence complementarity, e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%. 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 9S%, 99% or 100% sequence complementarity, between the oligonucleotide and the target sequence.

[0014] The invention extends also to a combination of two or more antisense oligomers capable of binding to a selected target t induce exon exclusion in an 1TGA4 gene transcript, including a construct comprising two or more such antisense oligomers. The construct may be used for an antisense oiigorner-based therapy.

[00 5] The invention extends, according to a still further aspect thereof, to cDNA or cloned copies of the antisense oligomer sequences of the invention, as well as to vectors containing the antisense oligomer sequences of the invention. The invention extends further also to ceils containing such sequences and/or vectors.

[0016] There is also provided a method fo manipulating splicing in an ITGA4 gene transcript, the method including the step of: a) providing one or more of the antisense oligomers as described herein and allowing the oligomer(s) to bind to a target nucleic acid site.

[0017] There is also provided a pharmaceutical, prophylactic, or therapeutic composition to treat, prevent or ameliorate th effects of a disease related to ITGA4 expression in a patient, the composition comprising: a) one or more antisense oligomers, as described herein and b) one or more pharmaceutically acceptable carriers and/or diluents.

[0018] The composition may comprise about 1 nlVI to 1000 nM of each of the desired antisense oligomers) of the invention. Preferably, the composition may comprise about 10 nM to S00 nM, most preferably between 1 M and 10 nM of each of the antisense oligomers) of the invention.

[0019] There is also provided a method to treat, prevent or ameliorate the effects of a disease associated with 1TGA4 expression, comprising the step of: a) administering to the patient an effective amount of one or more antisense oligomers or pharmaceutical composition comprising one or more antisense oligomers as described herein.

[0020] There is also provided the use of purified and isolated antisense oligomers as described herein, for the manufacture of a medicament to treat, prevent or ameliorate the effects of a disease associated with 1TGA4 expression.

[0021] There is also provided a kit to treat, prevent or ameliorate the effects of a disease associated with ITGA4 expression in a patient, which kit comprises at least an antisense oligomer as described herein and combinations or cocktails thereof, packaged in a suitable container, together with instructions for its use.

[0022] Preferably the disease associated with ITGA4 expression in a patient is multiple sclerosis (MS). The subject with the disease associated with 1TGA4 expression may be a mammal, including a human.

[00233 Further aspects of the invention will now be described with reference to the accompanying non-limiting examples and drawings. BRIEF DESCRIPTION OF THE DRAWINGS

[0024] in the drawings:

Figure 1 shows RT-PGR studies indicating changes n the amounts of ITGA4-FL and ΙΤΘΑ4Δ3 and 4 RNA (indicated with arrows) (top panel), ITGA4-FL and ΙΤΘΑ4Δ4 RNA (middle panel) and ITQA4-FL and ΓΠ3Α4Δ19 RNA (bottom panel) in normal human fibroblasts after transfection with oligomers directed to Exon 3 (antisense oligomer SEQ ID NO: 1 ), Exon 4 (antisense oligomer SEQ ID NO: 21) and Exon 19 (antisense oligomer SEQ ID NO: 47) at 100, 50 and 5nM. A clear dose dependent response is seen with antisense oligomer SEQ ID NOs: 1 , 21 and 47. An oligomer of scrambled sequence was used as a control (Scrambled antisense oligomer) for nonspecific exon skipping.

Figure 2 shows Western blotting data and a graph indicating a knockdown of the [TGA4 protein in normal fibroblasts following treatment with exon skipping antisense oligomers targeting exons 3 (H3A), 4 (H4A) and 19 (H19A) and an oligomer of scrambled sequence. Antisense oligomers were tested at 100 nIVL Levels of ITGA4 expression are normalised against β-tubuli expression.

Figure 3 provides graphs showing the effect of treatment with exon skipping antisense oligomers targeting exons 3 (AG1), 4 (AO21 ) and 19 (A047) and an oligomer of scrambled sequence (at 100 nM) on cell adhesion property in normal fibroblasts. The results were an average of three experiments performed independently and data were normalised to the sample treated with scrambled oligomer. The error bars represent standard error mean (SEIVl). Reduced cell adhesion to the microplate pre~coated with VCAM- was observed for the cells pre-treated with antisense oligomers directed to exon 4 (A021). p<Q.DQ5 compared to the scrambled oligomer treated samples.

Figure 4 provides a bar graph showing the effect of treatment with exon skipping antisense oligomers targeting exons 3 (AOI), 4 (A021 ) and 19 (A047) and an oligomer of scrambled sequence (at 100 nM) on cell migration property in norma! fibroblasts. The bar graph shows an average cell migration after 8 h of wounding from three experiments performed independently. All data were normalised to the sample treated with scrambled oligomer. The error bar represents SE ' M. Celis treated with antisense oligomers targeting exon 4 (A021 ) shows slower migration than the rest of the samples. p<Q.05 compared to the scrambled oligomer treated samples.

Figure 5A-C sho ITGA4 expression in jurkat cells after nucelofection with different PMOs [H3A (+41 +65). H3A (+30+49), H4A (+51 +75), H 9A (+30+49) and H27A (+20+44)] and scrambled PMO (Scramble) at 500 nM for 3 days. Figure 5A is a gel of the changes in ITGA4 transcript analysed by RT-PCR. Full length (FL) and exon deleted (Δ) products of ITGA4 transcript are indicated; Untreated (UT). Figure 5B is a Western blot of 1TGA4 expression. Figure 5C is flow cytometry of the jurkat cells. Among the PMOs tested, the PMOs H3A (+41+65), H4A (+51+75) and H27A (+20+44) targeting exon 3, 4 and 27 produced >50 % of transcripts with the targeted exon being skipped. A significant knockdown of th ITGA4 protein was observed in Western blotting and flow cytometry analysis following treatment with H3A (+41 +65) and H4A (+51+75). These result correlate with the transcript analysis. For the cells treated with PMO H27A (+2:0+44), instead of downreguSating ITGA4 expression, increased expression of truncated ITGA4 was observed in Western blotting. However, the flow cytometry analysis shows that ceil surface expression of ITGA4 was reduced in these cells, indicating the truncated ITGA4 are accumulating within the cells.

Figure 6A is a diagram of the setu of the migration test. Figure 6B is a graph of the migration of jurkat cells treated with different PMOs [H3A (+41+65), H4A (+51+75), H19A (+30+49) and H27A (+20+44)] and scrambled PMO (Scramble) at 500 nM for 2 days. The top panel shows the experimental set up for the migration assay and the jurkat cells nucteofected with PMO were allowed to migrate for 5 h. The assay was performed in duplicates, jurkat cells treated with PMO H4A (+51+75) and H27A (+20+44) show slower migration to the bottom chamber compared to the scrambled treated cells. DESCRIPTION OF INVENTION

Detailed Description of the Invention

[0025] integrin alpha-4 is the alpha chain of a hetero-dirneric membrane protein integrin receptor. Together with integrin beta-1 , they recognize alternatively spliced CS-1 and CS-5 regions of fibronectin, vascular cell adhesion molecule 1 (VCAM-1) and mucosas vaseuiar addressin ceil adhesion moiecule 1 (j ADCAM-1). A!pha-4/beta-1 integrin (also known as Very Late Antigen (VLA4)) facilitates leucocytes migration across the blood brain barrier through interaction with VCAM-1. The expression level of integrin alpha-4 (ITGA4) has been found to be elevated in MS patients and 1ΤΘΑ4 has been show to be an effective therapeutic target.

[0028] The present invention provides an alternative method for the treatment, prevention or amelioration of the effects of MS by developing anfisense oligomers to alter the activity of ITGA4 by reducing the transcripts level through modifying pre-mRNA splicing in the integrin alpha-4 (1TGA4) gene transcript or part thereof.

[0027] 1TGA4 is encoded by ITGA4 gene located on chromosome 2. It consists of 28 exons and generates 11 transcript variants during R A maturation and processing. Of these, one transcript codes for a full-length protein and two other transcripts code for truncated proteins. The functional full-length protein contains an extracellular domain encoded by exon 1-26, a trans-membrane domain and a cytoplasmic domain encoded by exon 27 and exon 28 respectively {Table 1).

Table 1. A summary of exorts encoding for different structures in ITGA4

[0028] in contrast to other antisense oligomer based therapies, the present invention does not induce increased degradation of RNA via recruitment of RNase H, wherein the RNase H preferentially binds and degraded RNA bound in duplex to DNA of the ITGA4 gene. Nor does it rely on hybridization of the antisense oligomer to the 1TGA4 genomic DNA or the binding of antisense oligomers to mRNA to modulate the amount of ITGA4 protein produced by interfering with norma! functions such as replication, transcription, translocation and translation.

[0029] Rather, the antisense oligomers are used to modify pre-mRNA splicing in an ITGA4 gene transcript or part thereof and induce exon "skipping". The strategy preferably reduces total protein expression or generates proteins which lack functional domains involved in cell migration,

[0030] According to a first aspect of the invention, there is provided antisense oligomers capable of binding to a selected target on an ITGA4 gene transcript to modify pre- mRNA splicing in an ITGA4 gene transcript or part thereof. Broadly, there is provided an isolated or purified antisense oligomer for inducing targeted exon exclusion in an ITGA4 gene transcript or part thereof.

[0031] By "isolated" is meant material that is substantially or essentially free from components that normally accompany it in its native state. For example, an isolated polynucleotide" or "isolated oligonucleotide," as used herein, may refer to a polynucleotide that has been purified or removed from the sequences that flank it in a naturally-occurring state, e.g., a DNA fragment that is removed from the sequences that are adjacent to the fragment in the genome. The term "isolating" as it relates to cells refers to the purification of cells (e.g., fibroblasts, Iymphoblasts) from a source subject (e.g., a subject with a polynucleotide repeat disease). In the context of mRNA or protein. solating" refers to the recovery of mRNA or protein from a source, e.g., cells.

[0032] An antisense oligomer can be said to be "directed to" or "targeted against" a target sequence with which if hybridizes, In certain embodiments, the target sequence includes a region including a 3' or 5' splice site of a pre-processed mRNA, a branch point, or other sequences involved in the regulation of splicing. The target sequence may be within an exon or within an infron or spanning an intron/exon junction.

[0033] In certain embodiments, the antisense oligomer has sufficient sequence complementarity to a target RNA (i.e., the RNA for which splice site selection is modulated) to block a region of a target RNA (e.g., pre-mRNA) in an effective manner. In exemplary embodiments, such blocking of JTGA4 pre-mRNA serves to modulate splicing, either by masking a binding site for a native protein that would otherwise modulate splicing and/or by altering the structure of the targeted RNA. In some embodiments, the target RNA is target pre-mRNA (e.g., ITGA4 gene pre-mRNA).

[0034] An antisens oligomer having a sufficient .sequence complementarity to a target RNA sequence to modulate splicing of the target RNA means that th antisense oligomer ha a sequence sufficient to trigger the masking of a binding site for a native protein that would otherwise modulate splicing and/or alters the three-dimensional structure of the targeted RNA. [0035] Selected aniisense oligomers can be made shorter, e.g., about 12 bases, or longer, e.g., about 50 bases, and include a small number of mismatches, as long as the sequence is sufficiently complementary to effect splice modulation upon hybridization to the target sequence, and optionally forms with the RNA a heterodupiex having a Tm of 45°C or greater.

{0036] Preferably, th antisense oligomer is selected from the group comprising the sequences set forth in Tables 3 to 7.

[0037] in certain embodiments, the degree of complementarity between the target sequence and antisense oligomer is sufficient to form a stable duplex. The region of complementarity of the antisense oligomers with the target RNA sequence may be as short as 8-11 bases, but can be 12-15 bases or more, e.g., 10-50 bases, 10-40 bases, 12-30 bases, 12-25 bases, 15-25 bases, 12-20 bases, or 15-20 bases, including all integers in between these ranges. An antisense oligomer of about 16-17 bases is generally long enough to have a unique complementary sequence. In certain embodiments, a minimum length of complementary bases may be required to achieve the requisite binding Tm, as discussed herein.

[0038] in certain embodiments, oligonucleotides as long as 50 bases may be suitable, where at least a minimum number of bases, e,g,, 10-12 bases, are complementary to the target sequence. In general, however, facilitated or active uptake in cells is optimized at oligonucleotide lengths of less than about 30 bases. For phosphorodiamidaie morpholino oligomer (PMO) antisense oligomers described further herein, an optimum balance of binding stabilit and uptake generally occurs at lengths of 18-25 bases. Included are antisense oligomers (e.g., PMOs, PMD-X, PNAs, LNAs, 2 ! -OMe) that consist of about 10, 11 , 12, 13, 14, 15, 18, 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, 4 or 50 bases.

[0039] In certain embodiments, antisense oligomers may be 100% complementary to the target sequence, or may include mismatches, e.g., to accommodate variants, as long as a heieroduplex formed between the oligonucleotide and target sequence is sufficiently stable to withstand the action of cellular nucleases and other modes of degradation which may occur in vivo. Hence, certain oiigonucteottdes may have about or at least about 70% sequence complementarity, e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 88%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence complementarity, between the oligonucleotide and the target sequence.

[0040] Mismatches, if present, are typically less destabilizing toward the end regions of the hybrid duplex than in the middle. The number of mismatches allowed will depend on the length of the oligonucleotide, the percentage of G;C base pairs in the duplex and the position of the mismatch(es) in the duplex, according to well understood principles of duplex stability. Although such an antisense oligomer is not necessarily 100% complementary to the target sequence, it is effective to stably and specifically bind to the target sequence, such that splicing of the target pre-RNA is modulated.

[0041] The stability of the duplex formed between an antisense oligomer and a target sequence is a function of the binding Tm and the susceptibility of the duplex to cellular enzymatic- cleavage. The Tm of an oligonucleotide with respect to compiementary- sequen.ce RNA may be measured by conventional methods, such as those described by Hames et a!., Nucleic Acid Hybridization, IRL Press, 1985, pp. 107-108 or as described in Miyada C. G. and Wallace R. EL, 1987, Oligonucleotide Hybridization Techniques, Methods Enzymol. Vol. 154 pp. 94-107. In certain embodiments, antisense oligomers may have a binding Tm, with respect to a complementary-sequence RNA, of greater than body temperature and preferably greater than about 45 ti G o 50°C. Tnf s in the range 60-8Q°C or greater are also included.

[0042] Additional examples of variants include antisense oligomers having about or at least about 70% sequence identity or homology, e.g., 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity or homology, over the entire length of any of SEQ ID OS: 1-202. [0043] More specifically, there is provided an antisense oligomer capable of binding to a selected target site to modify pre-mRNA splicing in an ITGA4 gene transcript or part ihereof. The antisense oligomer is preferably selected from those provided i Tables 3 to 7,

[0044] The modification of pre-mRNA splicing preferably induces "skipping", or the removal of one or more exons or introns of the mRNA. The resultant protein is preferably of a shorter length when compared to the parent full-length ITGA4 protein due to either internal truncation or premature termination. These truncated ITGA4 proteins may be termed isoforms of the full length ITGA4 protein.

[0045] The remaining exons of the mRNA generated ma be in-frame and produce a shorter protein with a sequence that is similar to that of the parent full length protein, except that It has an internal truncation in a region between the original 3' and 5 * ends. In another possibility, the exon skipping may induce a frame shift that results in a protein wherein the first part of the protein is substantially identical to the parent full length protein, but wherein the second part of the protein has a different sequence (eg a nonsense sequence) due to a frame-shift. Alternatively, the exon skipping may induce the production of a prematurel terminated protein due to a disruption of the reading frame and presence of a premature termination of translation.

[0046] Skipping individual exons of exons 3-7, 10-15, 17, 18, 20 and 22 will preferably disrupt the reading frame of the ITGA4 transcript. This will lead to increased degradation of RNA through nonsense mediated decay.

[0047] Skipping individual exons of exons 2, 8, 9, 16, 19, 21 and 23-17 will preferably keep the reading frame intact. Skipping a combination of exons 3 and 4 will also preferably keep the reading frame intact. This will preferably lead to translation into art internall truncated protein. The truncated protein or ITGA 4 isoform may have a completely ablated function, or may have a reduced function. Table 2. A summary of framesHtft exons

[0048] Preferably, these truncated, nonsense or prematurely terminated proteins are lacking one or more functional domains involved in the cell migration process. For example, exon 16 encodes a cell adhesion motif and translated proteins lacking this domai may have slow or disrupted cell migratton. Exon 27 encodes a transmembrane domain and removing this exon may generate a soluble ITGA4 protein, which could potentially act as a decoy. The truncated, nonsense or prematureiy terminated proteins may further lack an attachment or binding site for other factors, removal of which may lead to a reduction in interaction of the ITGA4 protein with relevant pathways. For example, exerts 1 to 26 are extracellular and exon 28 is cytoplasmic. Removal of one or more of these axons may remove a binding site.

[0049] Alternatively, the removal of one or more exons may lead to misfolding of the ΙΤΘΑ4 protein and a reduction in the ability of the protein to be successfully transported through the membrane.

[0050] The presence of internally truncated proteins (ie proteins lacking the amino acids encoded by one or more exons) is preferable. If the ITGA4 protein is knocked out, there may be problems with elevation of ITGA4 transcription as the body tries to compensate for the reduction in the total amount of 1TGA4 protein. In contrast, the presence of an ' internally truncated protein (preferably lacking one o more of the features of the complete ITGA4 protein), should be sufficient to prevent elevated transcription, but still provide a therapeutic advantage due to a reduction in the total amount of functional ITGA4 protein.

{0051] The antisense oligomer induced exon skipping of th present invention need not completely or even substantially ablate the function of the ITGA4 protein. Preferably, the exon skipping process results in a reduced or compromised functionality of the 1TGA4 protein >

[0052] The different isoforms of ITGA4 produced using different skipping strategies could result in proteins with ablated or reduced activity that could preferably be used to treat or prevent different forms of MS, For example, MS may be in the form of relapsing-remifting MS (R MS), secondary progressive MS {SPMS), primary progressive MS (PPMS) or progressive relapsing MS. There are also many different symptoms and syndromes associated with MS and its progression. Alternative splicing strategies may form truncated proteins or proteins with reduced functions that can be preferably used as treatments for specific aspects, forms or progression of the disease.

[0053] The skipping process of the present invention, using antisense oligomers, may skip an individual exon, or may result in skipping two or more exons at once.

[00541 The antisens oligomers of the invention may be a combination of two or more antisense oligomers capable of binding to a selected target to induce exon exclusion in an 1TGA4 gene transcript. The combination may be a cocktail of two or more antisense oligomers and/or a construct comprising two or more or two o more antisense oligomers joined together.

Tab!© 4 SEQ ID listing of antisense oligomers (other chemistry) for inducing human ITGA4 Exon 6,7,8, 1,12,13,15,16,20,22 skipping from transcript

Tab!© 5. SEQ ID iisting of antisense oligomers fo inducing human STGA4 Exon 2, 3, 4, 5, 8, 9, 10, 1 , 6, 17, 18, 19, 21, 23, 24, 25, 26 and 27 skipping from transcript.

Table 6. SEQ ID listing of antisense oligomers for inducing human ITGA4 Exon 8,

1.9, 25 and 27 skipping from transcript

% exon

SEQ

Length skipping

ID Co-ordinates Sequence

(bases) ITGA4 at

NO

100 n

Exo n 8

133 H8A (-1Q+15) GCU GAA UAU AUA UGC CUG UAA UUA G 25 33

J 134 H8A (-9+18) UGG UGA AUA UAU AUG GCU UA AUU A 25 36

135 H8A (-8+l7 AUG CUG AAU AUA UAU GCC UGU AAU U 25 44

136 HSA (-7+1S) AAU GCU GAA UAU AUA UGC CUG UAA U 25 36

13? H8A (-6+19) CAA UGC UGA AUA UAU AUG CCU GUA A 25 42

i 138 H8A (-4+21 ) AUC AAU GCU GAA UAU AUA UGC CUG U 25 40

j 139 H8A (-3+22) GAU CAA UGC UGA AUA UAU AUG CCU G 25 45

140 H8A (-2+23) UC UGA AUG CUG AAU AUA UAU GCC U 25 13

141 H8A (-1+24) UUG AUC AAU GCU GAA UAU AUA UGC C 25 42

H8A (+1 +25) UUU CAU CAA UGC UGA AUA UAU AUG C 25 40

Exon 19

143 H19A (+20+44) ACG GGA GAG UUA UCU GUG ACU UCA C 25 37

[ 144 H19A (+25+44) ACG GGA GAG UUA UCU GUG AC 20 28

145 H19A (+30+49) GUA CCA CGC CAG AGU UAU CU 20 40

J 146 H10A (+32+49) GUA CCA CGC CAG AGU UAU 18

147 H19A (+30+47) A CCA CGC CAG AGU UAU CU 18

Exon 25

148 H25A(+55+S0) ACA CU GCU UCU UUU CCA CUU UCC AU 26 48

[ 149 H25A(+56+81) AAC ACU GGC UUC UUU UGC ACU UUC CA 26 49

150 H25A(+57+82) GAA CAG UGG CUU CUU UUC CAC UUU CC 26 49

[ 15 H25A(+58+83) UGA ACA CUG GCU UCU UUU CCA CUU UC 26 47

S 152 H25A(+59+S4) AUG AAC ACU GGC UUC UUU UCC ACU UU 26 48

153 H25A(+61 +86) AUA UGA ACA CUG GCU UCU UUU CCA CU 26 48

15 H25A(+62+87) GAU AUG AAC ACU GGC UUC UUU UCC AC 26 43

155 H25A(+63+88) GGA UAU GAA CAC UGG GUU CUU UUG GA 28 43

156 H25A{+64+89) UGG AUA UGA ACA CUG GCU UCU UUU CC 28 46

157 H25A(+65+90) UUG GAU AUG AAC ACU GGC UUC UUU UG 28 44

Exon 27

158 H27A (-15+10) CCA GUA GAA GCU ACG UGA GUU AAG A 25 28

j 159 H27A (-14+11) UGC AGU AGA ACC UAC GUG AGU UAA G 25 36

1 160 H27A (-13+12) UUC CAG UAG AAC CUA CGU GAG UUA A 25 40

j 161 H27A (-12+13) GUU CCA GUA GAA CGU ACG UGA GUU A 2:5 41

162 H27A (-11 +14) GCU UCC AGU AGA ACC UAC GUG AGU U 25 40

i 163 H27A (-9+16) GUC GUU CCA GUA GAA CGU ACG UGA G 25 42

[ 164 H27A (-8+17) AGU CCU UCC AGU AGA ACC UAC GUG A 25 44

165 H27A (-7+18) UAG UCC UUC CAG UAG AAC CUA CGU G 25 45

j 166 H27A (-6+19) GUA GUC CUU CCA GUA GAA CCU ACG U 25 48

167 H27A (-5+20) UGU AGU GCU UCC AGU AGA ACG U AC G 25 44

168 H27D(+1 -8) AUG GUU ACC UUC CAC AUA ACA UAU G 25 22

I 169 H27D(+18-7) UGC UUA GCU UCG ACA UAA CAU AUG A 25 22 170 M27D(+19-6) GCU UAC GUU CCA CAU AAC AUA UGA G 25 34

171 H2?D(+2 -4} UUA GCU UCC ACA UAA CAU AUG AGA U 25 38

172 H27D{+22~3) UAC GUU CCA CAU AAC AUA UGA GAU C 25 34

i 173 H27D{+23-2) ACC UUC CAC AUA ACA UAU GAG AUG A 25 37

174 H27P(+24-1) CCU UCC ACA UAA CAU AUG AGA UCA A 25 38

Table 7. SEQ ID listing of antisense oligomers for inducing human ITGA4 Exon 27 skipping from transcript

[0055] The invention further provides a method for manipulating splicing in an ITGA4 gene transcript, the method including the step of. a) providing one or more of the antisense oligomers as described herein and allowing the oligomer(s) to bind to a target nucleic add site.

[0056] According to yet another aspec of the invention, there is provided a splice manipulation target nucleic acid sequence for ITGA4 comprising the DNA equivalents of the nucleic acid sequences selected from the group consisting of SEG ID NOs: 1-202, and sequences complementary thereto.

[0057] Designing antisense oligomers to completely mask consensus splice sites may not necessarily generate a change in splicing of the targeted exon. Furthermore, the inventors have discovered that size or length of the antisense oligomer itself is not always a primary factor when designing antisense oligomers. With some targets such as IGTA4 exon 3, antisense oligomers as short as 20 bases were able to induce some exon inclusion, in certain cases more efficiently than other longer (eg 25 bases) oligomers directed to the same exon.

[0058] The inventors have also discovered that there does not appear to be any standard motif that can be blocked or masked by antisense oligomers to redirect splicing. It has been found that antisense oligomers must be designed and their individual efficacy evaluated empirically,

[0059] More specifically, the antisense oligomer may be selected from those set forth in Tables 3 to 7, The sequences are preferably selected from the group consisting of any one or more of any one or more of SEG ID NOs: 1-202, more specifically SEQ ID NOs: 1-132, and combinations or cocktails thereof. This includes sequences which can hybridise to such sequences under stringent hybridisation conditions, sequences complementary thereto, sequences containing modified bases, modified backbones, and functional truncations or extensions thereof which possess or modulate pre-mRNA processing activity in an fTGA4 gene transcript.

[0060] The oligomer and the DNA, cDNA o RNA are complementary to each othe whe a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, "specifically hybridisable" and "complementary" are terms which are used to indicate a sufficient degree of complementarity or pairing such that stable and specific binding occurs between the oligomer and the DNA, cDNA or RNA target, it is understood in the art that the sequence of an antisens oligomer need not be 100% complementary to that of its target sequence to be specifically hybridisable. An antisense oligomer is specifically hybridisable when binding of th compound to the target DNA or RNA molecule interferes with the norma! function of the target DNA or RNA product, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense oligomer 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.

[0061] Selective hybridisation may be under low, moderate or high stringency conditions, but is preferably under high stringency. Those skilled in the art will recognise that the stringency of hybridisation will be affected by such conditions as salt concentration, temperature, or organic solvents, in addition to the base composition, iength of the complementary strands and the number of nucieotide base mismatches between the hybridising nucleic acids. Stringent temperature conditions will generally include temperatures in excess of 3G°C ; typically in excess of 37°C, and preferably in excess of 45 Q C, preferably at least 50 Q C, and typically 60°C-80°C or higher. Stringent salt conditions will ordinarily be less than 1000 mM, typically less than 500 mM, and preferably less than 200 mM. However, the combination of parameters is much more important than the measure of any singl parameter. An example of stringent hybridisation conditions is 65°C and 0.1 x SSC (1 x SSC = 0.15 M NaCi, 0.015 M sodium citrate pH 7.0). Thus, the antisense oligomers of the present invention may include oligomers that selectively hybridise to the sequences provided in Tables 3 to 7, or SEQ ID NOs: 1-202.

[0062] It will be appreciated that th codon arrangements at the end of exons in structural proteins may not always break at the end of a codon, consequently there may be a need to delete more than one exon from the pre-mRNA to ensure in-frame reading of the mRNA. In such circumstances, a plurality of antisense oligomers may need to be selected by the method of the invention wherein each is directed to a different region responsible for inducing inclusion of the desired exon and/or tntron. At a given ionic strength and pH, the Tm is the temperature at which 50% of a target sequence hybridizes to a complementary polynucleotide. Such hybridization may occur with "near" or "substantia!" complementarity of the antisense oligomer to the target sequence, as well as with exact complementarity,

[0083] Typically, selective hybridisation will occur when there is at least about 55% identit over a stretch of at least about 14 nucleotides, preferabl at least about 85%, more preferably at feast about 75% arid most preferably at least about 90%, 95%, 98% o 99% identity with the nucleotides of the antisense oligomer. The length of homolog comparison, as described, may be over longer stretches, and in certain embodiments will often be over a stretch of at least about nine nucleotides, usually at least about 12 nucleotides, mor usually at least about 20, often at least about 21 , 22, 23 or 24 nucleotides, at least about 25, 26, 27 or 28 nucleotides, at least about 29, 30, 31 or 32 nucleotides, at least about 36 or more nucleotides.

[0064] Thus, the antisense oligomer sequences of the invention preferably have at least 75%, more preferably at least 85%, more preferably at least 86, 87, 88, 89 or 90% homology to the sequences shown in the sequenc listings herein. More preferably there is at least 91, 92, 93 94, or 95%, more preferably at least 96, 97, 98% o 99%, homology. Generally, the shorter the length of the antisense oligomer, the greater the homology required to obtain selective hybridisation, Consequently, where an antisense oligomer of the invention consists of less than about 30 nucleotides, it is preferred that the percentage identity is greater tha 75%, preferably greater than 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95%, 98, 97, 98% or 99% compared with th antisense oligomers set out in the sequence listings herein. Nucleotide homology comparisons may be conducted by sequence comparison programs such as the GCG Wisconsin Bestfit program or GAP (Deveraux et ai. f 1984, Nucleic Acids Research 12, 387-395). In this way sequences of a similar or substantially different length to those cited herein could be compared by insertion of gaps into the alignment, such gaps being determined, for example, by the comparison algorithm used by GAP,

[0065] The antisense oligomer of th present invention may have regions of reduced homology, and regions of exact homology with the target sequence. It is not necessary for an oligomer to have exact homology for its entire length. For example, the oligomer may have continuous stretches of at least 4 or 5 bases that are identical to the target sequence, preferably continuous stretches of at least 6 or 7 bases that are identical to the target sequence, more preferably continuous stretches of at least 8 or 9 bases that are identical to the target sequence. The oligome may have stretches of at least 10, 11 , 12, 13, 14, 15, 16, 17, 1 ' 8, 19, 20, 21, 22, 23, 24, 25 or 26 bases that are identical to the target sequence. The remaining stretches of oligomer sequence may be intermittently identical with the target sequence; for example, the remaining sequence may have an identical base, followed b a non-identical base, followed by an identical base. Alternatively (or as well) the oligomer sequence may have several stretches of identical sequence (for example 3, 4, 5 or 6 bases) interspersed with stretches of less than perfect homology. Such sequence mismatches will preferably have no or very little loss of splice switching activity.

[0066] The term "modulate" or "modulates" includes to "increase" or "decrease" one or more quantifiable parameters, optionally by a defined and/or statisticall significant amount. The terms "increase" or "increasing," "enhance" or "enhancing," or "stimulate" or "stimulating *' ' refer generally to the ability of one or antisense oligomers or compositions to produce or cause a greater physiological response (i.e., downstream effects) in a eel! or a subject relative to the response caused by either no antisense oligomer or a control compound. The terms "decreasing" or ¾βθΓβ33β" refer generally to the ability of one or antisense oligomers or compositions to produce or cause a reduced physiological response (i.e., downstream effects) in a cell or a subject relative to the response caused by either no antisens oligomer or a control compound.

[0067] Relevant physiological or cellular responses (in vivo or in vitro) will be apparent to persons skilled in the art, and may include increases in the exclusion of specific exons in a I TA4-coding pre-mRNA, decreases in the amount of IGTA4~coding pre- mRNA or decreases in the expression of functional IGTA4 protein in a cell, tissue, or subject in need thereof. An "increased" or "enhanced" amount is typically a statistically significant amount, and may include an increase that is 1.1 , 1.2, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1.5, 20, 30, 40, 50 or more times (e.g., 500, 1000 times) (including all integers and decimal points in between and above 1 , e.g., 1.5, 1.6, 1.7. 1.8} the amount produced by no antisense oligomer (the absence of an agent) or a control compound. The term "reduce" or "inhibit" may relate generally to the ability of one or more antisens oligomers o compositions to "decrease" a relevant physiological or cellular response, such as a symptom of a disease or condition described herein, as measured according to routine techniques in the diagnostic art. Relevant physiological or cellular responses {in vivo or in vitro) will be apparent to persons skilled in the art, and ma include reductions in the symptoms or pathology of a disease such as MS. A "decrease" in a response may be statistically significant as compared to the response produced by no antisense oiigomer or a contra! composition, and may include a 1 %, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%.. 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% decrease, including ail integers in between.

[0068] The length of an antisense oligomer may vary, as Song as it is capable of binding selectively to the intended location within the pre-mRNA molecule. The Iength of such sequences ca be determined in accordance with selection procedures described herein. Generally, the antisense oligomer will be from about 10 nucleotides in length, up to about 50 nucleotides in Iength. it will be appreciated, however, that any length of nucleotides within this range may be used in the method. Preferably, the Iength of the antisense oligomer is between 10 and 40, 10 and 35, 15 to 30 nucleotides in Iength or 20 to 30 nucleotides in Iength, most preferably about 25 to 30 nucleotides in Iength. For example, the oligomer may be 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 nucleotides in Iength.

[0069] As used herein, an "antisense oligomer" refers to a linear sequence of nucleotides, or nucleotide analogs, that allows the nucieobase to hybridize to a target sequence in an RNA b Watson-Crick base pairing, to form an oligonucleotide:RNA heteroduplex within the target sequence. The terms "antisense oligomer", "antisense oligonucleotide", "oligomer" and "antisense compound" may be used interchangeably to refer to an oligonucleotide. The cyclic subunits may be based o ribose or another pentose sugar or, in certain embodiments, a morpholino group (see description of morphoiino oligonucleotides below). Also contemplated are peptide nucleic acids (PNAs), locked nucleic acids (LNAs), and 2'-0-iV1ethyl oligonucleotides, among other antisense agents known in the art.

[0070] Included are non-natura!iy-occurririg antisense oligomers, or "oligonucleotide analogs" including antisense oligomers or oligonucleotides having (i) a modified backbone structure, e.g., a backbone other than th standard phosphodiester linkage found in naturally-occurring oiigo- and polynucleotides, and/or (ii) modified sugar moieties, e.g., morphoiino moieties rather than ribose or deoxyribose moieties. Oligonucleotide analogs support bases capable of hydrogen bonding by Watson-Crick base pairing to standard polynucleotide bases, where the analog backbone presents the bases in a manner to permit such hydrogen bonding in a sequence-specific fashion between the oligonucleotide analog molecule and bases in a standard polynucleotide (e.g., single-stranded RNA or single-stranded D ' NA). Preferred analogs are those having a substantially uncharged, phosphorus containing backbone,

[0071] One method for producing antisense oligomers is the methylation of the 2' hydroxyribose position and the incorporation of a phosphorothioate backbone produces molecules that superficially resemble RNA but that are much more resistant to nuclease degradation, although persons skilled i the art of the invention will be aware of other forms of suitable backbones that may be useable in the objectives of the invention.

[0072] To avoid degradation of pre-mRNA during duplex formation with the antisense oligomers, the antisense oligomers used in the method may be adapted to minimise or prevent cleavage by endogenous RNase H. This propert is highly preferred, as the treatment of the RNA with the unmethylated oligomers, either intracellular or in crude extracts that contain RNase H, leads to degradation of the pre-mRNA:antisense oligomer duplexes. Any form of modified antisense oligomers that is capable of bypassing or not inducing such degradation may be used in the present method. Th nuclease resistance may be achieved by modifying the antisense oligomers of the invention so that it comprises partially unsaturated aliphatic hydrocarbon chain and one or more polar or charged groups including carboxylic acid groups, ester groups, and alcohol groups.

[0073] An example of antisense oligomers which when duplexed with RNA are not cleaved by cellular RNase H is 2'-0-methy! derivatives. Such 2'-G-methyj- oligoribonucleotides are stable in a cellular environment and in animal tissues, and their duplexes with RNA have higher Tm values than their tibo- or deoxyribo- counterparts. Alternatively, the nuclease reststant antisense oligomers of the invention may have at least one of the last 3'~terminus nucleotides fluoridated. Still alternatively, the nuclease resistant antisense oligomers of the invention have phosphorothioate bonds linking between at least two of the last 3-terminus nucleotide bases, preferably having phosphorothioate bonds linking between the last four 3'-terminal nucleotide bases.

[0074] increased splice-switching may also be achieved with alternative oligonucleotide chemistry. For example, the antisense oligomer may be chosen from the list comprising: phosphoramidate or phosphorodiamidate morpholino oligomer (Pr IO); PMQ-X; PPMO; peptide nucleic acid (PNA); a locked nucleic acid (LNA) and derivatives including a!pha-L-LNA, 2 -amino LNA, 4 ! -methyl LNA and 4'-G-methyl LNA; ethylene bridged nucleic acids (ENA) and their derivatives; phosphorothioate oligomer; tricyclo- DNA oligomer (tcDNA); irtcyclophosphoroihioaie oligomer; 2'0-Methyl-modified oligomer (2'-O e); 2 -O-methoxy ethyl (2'-MOE); 2 i -fluoro, 2 -fluroarabino (FANA); unlocked nucleic acid (UNA); hexitoi nucleic acid (UNA); cyeJohexenyl nucleic acid (CeNA); 2'-amino (2 -NH2); 2'-Q-ethyieneaml.ne or any combination of the foregoing as mixmers or as gapmers. To further improve the delivery efficacy, the above mentioned modified nucleotides are often conjugated with fatty acids/iipfd/cholesterol/amino acids/carbohydraies/polysaccharides/nanopartieles etc. to the sugar or nucleobase moieties. These conjugated nucleotide derivatives can also be used to construct exon skipping antisense oligomers, Antisense oligomer-induced splice modification of the human ITGA4 gene transcripts have generally used either oligoribonucleotides, PNAs, 20iv1e or OE modified bases on a phosphorothioate backbone. Although 2Oiv1eA0s are used for oiigo design, due to their efficient uptake in vitro when delivered as cationic iipoplexes, these compounds are susceptible to nuclease degradation and are not considered ideal for in vivo or clinical applications. When alternative chemistries are used to generate the antisense oligomers of th present invention, the uracil (U) of the sequences provided herein may be replaced by a thymine (T).

[0075] Included within the antisense oligomers of the present invention are non- naturally-occurring oligomers, or "oligonucleotide analogues," including oligomers having (i) a modified backbone structure, e.g., a backbone other than the standard phosphodi ester linkage found in naturally-occurring oiigo- and polynucleotides, and/or (ii) modified sugar moieties, e.g., morpholino moieties rather tha ribose or deoxyribose moieties. Oligomer analogues support bases capable of hydrogen bonding by Watson- Crick base pairing to standard polynucleotide bases, wher the analogue backbone presents the bases in a manner to permit such hydrogen bonding in a sequence- specific fashio between th oligomer analog molecule and bases in a standard polynucleotide (e.g., . single-stranded RNA or single-stranded DNA}, Preferred analogues are thos having a substantially uncharged, phosphorus containing backbone.

[0076] Antisense oligomers that do not activate RNase H can be made in accordance with known techniques (see, e.g., U.S. Pat. 5,149,797). Such antisense oligomers, which may be deoxyribonucleotide or ribonucleotide sequences, simply contain an structural modification which sterically hinders or prevents binding of RNase H to a duplex molecule containing th oligomer as one member thereof, which structural modification does not substantially hinder or disrupt duplex formation. Because the portions of the oligomer involved in duplex formation are substantially different from those portions involved in RNase H binding thereto, numerous antisense oligomers that do not activate RNase H are available. For example, such antisense oligomers may be oligomers wherein at least one, or all, of the inter-nucSeotide bridging phosphate residues are modified phosphates, such as methyl phpsphonates, methyl phosphorothioates, phosphoromorpholidates, phosphoropiperazidates boranophosphates, amide linkages and phosphoramidates. For example, every other one of the internucleotide bridging phosphate residues may be modified as described, in another non-limiting example, such antisense oligomers are molecules wherein at least one, or all, of the nucleotides contain a 2' lower a!kyl moiety {such as, for example, C1-C4, linear or branched, saturated or unsaturated aikyl, such as methyl, ethyl, ethenyS, propyl, 1 -propeny!, 2-propenyl, and isopropyi). For example, ever other one of the nucleotides may be modified as described.

[0077] While the antisense oligomers described above are a preferred form of the aniisense Giigomers of the present invention, the present invention includes other oligomeric antisense molecules, including but not limited to oligomer mimetics such as are described below.

[0078] Specific examples of preferred antisense oligomers useful in this invention include oligomers containing modified backbones or non-natural inter-nucleoside linkages. As defined in this specification, oligomers having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligomers that do not have a phosphorus atom in their inter-nucieoside backbone can also be considered to be antisense oligomers.

[0079] In other preferred oligomer mimetics, both the sugar and the inter-nucieoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligomer mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid {PNA}. In PNA compounds, the sugar-backbone of an oligomer is replaced with an amide containing backbone, in particular an aminoethySglycine backbone. The nucleo-bases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.

[0080] Another preferred chemistry is the phosphorodiamidate morphoSino oligomer (PMO) oligomeric compounds, which are not degraded by any known nuclease or protease. These compounds are uncharged, do not activate RNase H activity when bound to a R A strand and have been shown to exert sustained splice modulation after in vivo administration (Summertqrt and Welter, Antisense Nucleic Acid Drug Development, 7, 187-197).

[0081] Modified oligomers may also contain one or more substituted sugar moieties. Oligomers may also include nucleobase (often referred to in the art simply as "base") modifications or substitutions. Certain nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5- substituted pyrimidines, 6-azapynmidmes, and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5- propynyluracil and 5-propynylcytosine. 5- methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°G, even more particularly when combined with 2'-O-methoxyethyl sugar modifications.

[0082] Another modification of the oligomers of the invention involves chemically linking to the oligomer one or more moieties or conjugates that enhance the activity, cellular distribution or cellular uptake of the oligomer. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-S- trity!thiol, a thiocholesterol, an aliphatic chain, e.g., dodeeandioi or undeeyl residues, a phospholipid, e.g., di-hexadecyl- rac-glycerol or triethylammonium 1 ,2-di-O-hexadecyi- rac-glycero-3-H- phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety l myristyS, or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety.

[0083] Ceil penetrating peptides have been added to phosphorodiamidate morpholino oligomers to enhance cellular uptake and nuclear localization. Different peptide fags have been shown to influence efficiency of uptake and target tissue specificity, as shown in Jearawinyapaisam et al. (2008), Mol. Ther. 6 9, 1824-1629.

[0OB4 It is not necessary for all positions in a given compound io be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligomer. The present invention also includes antisense oligomers that are chimeric compounds. "Chimeric" antisense oligomers or "chimeras," in the context of this invention, are antisense oligomers, particularly oligomers, which contain two or more chemicall distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligomer compound. These oligomers typically contain at least one region wherein the oligomer is modified so as to confer upon the oligomer or antisense oligomer increased resistance to nuclease degradation, increased cellular uptake, and an additional region for increased binding affinity for the target nucleic acid.

[0G85] The activity of antisense oligomers and variants thereof can be assayed according to routine techniques in the art. For example, splice forms and expression levels of surveyed RNAs and proteins may b assessed by any of a wide variety of well- known methods for detecting splice forms and/or expression of a transcribed nucleic acid o protein. Non-limiting examples of such methods include RT-PCR of spliced forms of RNA followed by size separation of PCR products, nucleic acid hybridization methods e.g., Northern blots and/or use of nucleic acid arrays; nucleic acid amplification methods: immunological methods for detection of proteins; protein purification methods; and protein functio or activity assays.

[0086] RNA expression levels can be assessed by preparing mRNA/cDNA (i.e., a transcribed polynucleotide) from a cell, tissue or organism, and by hybridizing the mRNA cDNA with a reference polynucleotide, which is a complement of the assayed nucleic acid, or a fragment thereof. cDNA can, optionally, be amplified using any of a variety of poiymerase chain reaction or in vitro transcription methods prior to hybridization with the complementary polynucleotide; preferably, it is not amplified. Expression of one or more transcripts can also be detected using quantitative PG to assess the level of expression of the transcript(s),

[0087] The present invention provides antisense oligomer induced splice-switching of the 1TGA4 gene transcript, clinically relevant oligomer chemistries and delivery systems to direct ITGA4 splice manipulatio to therapeutic levels. Substantial decreases in the amount of full length 1TGA4 mRNA, and hence ITGA4 protein from ITGA4 gene transcription, ar achieved by:

1) oligomer refinement in vitro using fibroblast cell lines, through experimental assessment of (i) irttronic -enhancer target motifs, (ii) antisense oligomer length and development of oligomer cocktails, (iii) choice of chemistry, and (iv) the addition of cell-penetrating peptides (CPP) to enhance oligomer delivery; and

2) detailed evaluation of a novel approach to generate ITGA4 transcripts with one or more missing exons.

[0088] As such, it is demonstrated herein that processing of ITGA4 pre-mRNA can be manipulated with specific antisense oiigomers. in this way functionally significant decreases in the amount of ITGA4 protein can be obtained, thereby reducing the severe pathology associated with MS.

[0089] The antisense oligomers used in accordance with this invention may be conveniently made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosysfems (Foster City, Calif.). One method for synthesising oligomers on a modified solid support is described in U.S. Pat. No. 4,458,086.

[0090] Any other means for such synthesis known in the art may additionally or alternativel be employed, it is well known to use similar techniques to prepare oligomers such as the phosphorothioates and alkylated derivatives, in one such automated embodiment, diethyl-phosphoramidites are used as starting materials and may be synthesized as described by Beaucage, et al., (1981) Tetrahedron tetters, 22:1859-1882.

[0091] The antisense oligomers of the invention are synthesised in vitr and do not include antisense compositions of biological origin, or genetic vector constructs designed to direct the in vivo synthesis of antisense oligomers. The molecules of the invention may als be mixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption.

[0092] The antisense oligomers of the present invention also can be used as a prophylactic or therapeutic, which may be utilised for the purpose of treatment of a disease. Accordingly, i one embodiment the present invention provides antisense oligomers that bind to a selected target in the ITGA4 pre-mRNA to induce efficient and consistent exon skipping as described herein, in a therapeutically effective amount, admixed with a pharmaceutically acceptable carrier, diluent, or excipient. [0093] The invention therefore provides a pharmaceutical, prophylactic, or therapeutic composition to treat, prevent or ameliorate the effects of a disease associated with ITGA4 expression in a patient, the composition comprising: a) one or more antisense oligomers as described herein, and b) one or more pharmaceutically acceptable carriers and/or diluents.

[0094] Preferably the disease associated with ITGA4 expression is MS.

[0095] The composition may comprise about 1 nM to 1000 nM of each of the desired antisense oligomers) of the invention. Preferably, the composition may comprise about 1 nM to 500 nM, 10 nM to 500 nM, 50 nM to 750 nM, 10 nM to 500 nM, 1 nM to 100 nM, 1 nM to 50 nM, 1 nM to 40 nM, 1 nM to 30 nM, 1 nM to 20 nM, most preferably between 1 nM and 10 nM of each of the antisense oligomer(s) of the invention.

[0096] The composition may comprise about 1 nm, 2nm, 3nm, 4nm, 5nm, 6nm, 7nm, 8nm, 8nm t 10nm, 20nm, 50nm, 75nm, 100nm, 150nm, 20Onm, 250nm, 300nm, 35Gn , 400nm, 450nm, SOOnm, 550nm, 600nm, 650nm, 700nm, 750nm, 800nm, 850nm, 900nm, 950nm or 1000nm of each of the desired antisense oiigomer(s) of the invention.

[0097] The present invention further provides one or more antisense oligomers adapted to aid in the prophylactic or therapeutic treatment, prevention or amelioration of symptoms of a disease such as an ITGA4 expression related disease or pathology in a form suitable for delivery to a patient.

[0098] The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable and do not typically produce an allergic or similarly untoward reaction, such as gastric upset and the like, when administered to a patient. Th term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including thos of petroleum, animal, vegetable or synthetic origin, suc as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in Martin, Remington's Pharmaceutics! Sciences, 18th Ed., Mack Publishing Co., Easton, PA, (1990). [0099] In a more specific form of the invention there are provided pharmaceutical composition comprising therapeutical!y effective amounts of one or more antisense oligomers of the invention together with pharmaceutically acceptable diluents, preservatives, soiubiiizers, emuisiflers, adjuvants, and/or carriers. Such compositions include diluents of various buffe content (e.g. Tris-HCl, acetate, phosphate), pH and ionic strength and additives such as detergents and solubi!iz ng agents (e.g. Tween 80, Poiysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g. Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol). The materiai may be incorporated info particulate preparations of polymeric compounds such as poly!actic acid, polyglyco!ic acid, etc. or into liposomes. Hylauronic acid may also be used. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present proteins and derivatives. See, for example, Martin, Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA 18042) pages 1435-1712 that are herein incorporated by- reference. The compositions may be prepared in liquid form, or may be in dried powder, such as a lyophilised form.

[001003 & will be appreciated that pharmaceutical compositions provided according to the present inventio may be administered by any means known in the art. Preferably, the pharmaceutical compositions for administration are administered by injection, orally, topically or by the pulmonar or nasal route. The antisense oligomers are more preferably delivered by intravenous, intra-arterial, intraperitoneal, intramuscular or subcutaneous routes of administration. The appropriate route may be determined by one of skill in th art, as appropriate to the condition of the subject under treatment. Vascular or extravascutar circulation, the blood or lymph system, and the cerebrospinal fluid are some non-limiting sites where the antisense oligomer may be introduced. Direct CNS deliver)/ may be employed, for instance, intracerebral ventribuSar or intrathecal administration may be used as routes of administration.

[00101] Formulations for topical administration include those in which the oligomers of the disclosure are i admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Lipids and liposomes include neutral (e.g. dioleoylphosphatidyi DOPE ethanoiamine, dimyristoyiphosphafidyl choline DMPC, distearoly phosphatidyl choline) negative (e.g. dimyristoylphosphatidyS glycerol D1V1PG) and cationic (e.g. dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyi ethanoiamine DOTMA). For topical or other administration, oligomers of the disclosure may be encapsulated within liposomes or may form complexes thereto, in particular to cationsc liposomes. Alternatively, oligomers may be complexed to lipids, in particular to cationic lipids. Fatty acids and esters, pharmaceutically acceptable salts thereof, and their uses are further described in U.S. Pat. No. 6,287,860 and/or U.S. patent application Ser. No. 09/315,298 filed o May 20, 1999.

[00102] Sn certain embodiments, the antisense oligomers of the disclosure can be delivered by transdermal methods (e.g., via incorporation of the antisense oligomers into, e.g., emulsions, with such antisense oligomers optionally packaged into liposomes). Such transdermal and emulsion/liposome-mediated methods of delivery are described for delivery of antisense oligomers in the art, e.g., in U.S. Pat. No. 6,965,025.

[001.033 The antisense oligomers described herein may also be delivered via an implantable device. Design of such a device is an art-recognized process, with, e.g., synthetic implant design described in, e.g., U.S. Pal No. 6,969,400.

[001043 Compositions and formulations for oral administration include powders or granules, microparticuiates, nanoparticulates, suspensions or solutions in water or nonaqueous media, capsules, gel capsules, sachets, tablet or minitablets. Thickeners, flavoring agents, diluents, emulssfiers, dispersing aids or binders ma be .desirable. Oral formulations are those in which oligomers of the disclosure are administered in conjunction with one or more penetration enhancers surfactants and chelators. Surfactants include fatt acids and/or esters or salts thereof, bile acids and/or salts thereof. Bile acids/salts and fatty acids and their uses are further described in U.S. Pat. No. 6,287,860. In some embodiments, the present disclosure provides combinations of penetration enhancers, for example, fatty acids/salts in combination with bile acids/salts. An exemplar combination is the sodium salt of lauric acid, capric acid and UDCA. Further penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene- 20-cetyl ether. Oligomers of the disclosure may be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or nanoparticles. Oligomer comp!exing agents and their uses are further described in U.S. Pat. No. 6,287,860, Oral formulations for oligomers and their preparation are described in detail in U.S. 6,887,906, 09/315,298 filed May 20, 1999 and/or US2003Q02778Q.

[001053 Compositions and formulations for parenteral, intrathecal or intraventricular administration ma include sterile aqueous solutions which may also contain buffers, diiuents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or exeipients.

[00106] The delivery of a therapeutically useful amount of antisense oligomers may be achieved by methods previously published. For example, intracellular delivery of the antisense oligomer ma be via a composition comprising an admixture of the antisense oligomer and an effective amount of a block copolymer. An example of this method is described in US patent application US20040248833- Other methods of delivery of antisense oligomers to the nucleus are described in Mann CJ et al. (2001) Proc, Natl. Acad. Science, 98(1) 42-47, and in Gebski et al. (2003) Human Molecular Genetics, 12(15): 1801-181 A method for introducing a nucleic acid molecule into a cell by way of an expression vector either as naked DMA or complexed to lipid carriers, is described in US 6,806,084.

[00107] In certain embodiments, the antisense oligomers of th invention and therapeutic compositions comprising the same can be delivered by transdermal methods (e.g., via incorporation of the antisense oligomers info, e.g., emulsions, with such antisense oligomers optionally packaged into liposomes). Such transdermal and emuision/liposome-mediated methods of delivery are described for delivery of antisense oligomers in the art, e.g., in U.S. Pat No. 6,965,025.

[001083 ft ma ^ e desirable to deliver the antisense oligomer in a colloidal dispersion system. Colloidal dispersio systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes or liposome formulations. These colloidal dispersion systems can be used in the manufacture of therapeutic pharmaceutical compositions.

[00109] Liposomes are artificial membrane vesicles, which are useful as delivery vehicles in vitro and in. vivo. These formulations may have net cationic, anionic, or neutral charge characteristics and have useful characteristics for in vitro, in vivo and ex vivo delivery methods. It has been shown that large unilamellar vesicles can encapsulate a substantial percentage of an aqueous buffer containing large macromolecuies. RNA and DNA can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al., Trends Biochem. Sci. 6:77, 1981). [001 ID] in order for a liposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1 ) encapsulation of the antisense oligomer of interest at high efficiency while not compromising their biological activity; (2) preferential and -substantia! binding to a target cell in comparison to non-target cells; {3} delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic Information (Mannino, et a!., Biotechniques, 6:882, 1988). The composition of the liposome is usuall a combination of phospholipids, particularly high phase-transition-temperature phospholipids, usually in combination with steroids, especiall cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and the presence of divaient cations. Cationic liposomes are positively charged liposomes which are believed to interact with negatively charged DNA molecules to form a stable complex. Liposomes that are pH- sensitive or negatively-charged are believed to entrap DNA rather than complex with it. Both cationic and noncationic liposomes have been used to deliver DNA to cells.

[00111] Liposomes also include "sterically stabilized" liposomes, a term which, as used herein, refers to liposomes comprising one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are those in which part of the vesicle-forming lipid portion of the liposome comprises one or more glycolipids or is derivatized with one or more hydrophilic polymers, such as a polyethylene glycol (PEG) moiety. Liposomes and their uses are further described in U.S. 6,287,860.

[00112] The antisense oligomers described herein may also be delivered via an implantable device. Design of such a device is an art-recognized process, with, e.g., synthetic implant design described in, e.g., U.S. Pat. No. 6,969,400, the contents of which are incorporated in their entirety b reference herein.

[00113] Antisense oligomers can be introduced into cells using art-recognized techniques (e.g., transfection, e!ectroporation, fusion, liposomes, colloidal polymeric particles and viral and non-viral vectors as well as other means known in the art). Th method of delivery selected will depend at least on the cells to be treated and the location of the cells and will be apparent to the skilled artisan. For instance, localization can be achieved by liposomes with specific markers on the surface to direct the liposome, direct injection into tissue containing target cells, specific receptor-mediated uptake, or the like.

[00114] As known in the art, antisense oligomers may be delivered using, for example, methods involving !iposome-mediated uptake, lipid conjugates, polylysine- rnediated uptake, nanoparticie-mediated uptake, arid receptor-mediated endocytosis, as well as additional non-endocytic modes of delivery, such as microinjection, permeabilizafion (e.g., streptolysin-0 permeabilization, anionic peptide permeabilization), electroporation, and various non-invasive non-endocytic methods of delivery that ar known in the art (refer to Dokka and Rojanasakul, Advanced Drug Delivery Reviews 44, 35-49, incorporated by reference in its entirety).

[OOl iSj The antisense oligomer may also be combined with other pharmaceutically acceptable carriers or diluents to produce a pharmaceutical composition. Suitable carriers an diluents include isotonic saline solutions, for example phosphate-buffered saline. The composition may be formulated for parenteral, intramuscular, intravenous, subcutaneous, intraocular, oral, or transdermal administration.

[00116] The routes of administration described are intended only as a guide sinc a skilled practitioner will be able to readily determine the optimum route of administration and any dosage for any particular animal and condition.

[00117] Multiple approaches for introducing functional new genetic material into cells, bot in vitro and in vivo have been attempted (Friedmann (1989) Science, 244:1275-1280). These approaches include integration of the gene to be expressed into modified retroviruses (Friedmann (1989) supra; Rosenberg (1991) Cancer Research 51 (18), suppl.: 5Q74S-5079S); integration into non-retrovirus vectors (Rosenfeid, et al. (1992) Cell, 88:143-155; Rosenfeid, et al. (1991 ) Science, 252:431- 434); or delivery of a transgene linked to a heterologous promoter-enhancer element via liposomes (Friedmann (1989), supra; Brigham, et al. (1989) Am, J. Med. ScL, 298:278- 281 ; abel, et al. (1990) Science, 249:1285-1288; Hazinski, et al. (1991) Am. J. Resp. Cell Molec. BioL, 4:206-209; and Wang and Huang (1987) Proc. Natl. Acad. ScL (USA), 84:7851-7855); coupled to ligand-specific, cation-based transport systems (Wu and Wu (1988) J, Bioi. Chem., 263:14621-14624) or the use of naked DNA, expression vectors (Nabel et al. (1990), supra); Wolff et al. (1990) Science, 247:1465-1468). Direct injection of transgenes into tissue produces only localized expression (Rosenfeid (1992) supra); Rosenfefd et al. (1991) supra; Brigham et al. (1989) supra; Mabel (1990) supra; and Hazinskj et ai. (1991 ) supra). The Brigham et ai. group (Am. J. Med, Set (1989) 298:278-281 and Clinical Research (1991 ) 39 (abstract)) have reported in vivo transaction only of lungs of mice following either intravenous or intratracheal administration of a DNA liposome complex. An example of a review article of human gene therapy procedures Is: Anderson, Science (1992) 256:808-813; Barteau et a I, (2008), Curr Gene Thar; 8(5):313-23; Mueller et al. (2008). Clin Rev Allergy Immunol; 35(3):164-78; Li et al. (2006) Gene Ther., 13(18):1313-9; Simoes et al. (2005) Expert Opin Drug Deliv; 2(2):237-54,

[00118} The antisense oligomers of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal inciuding a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, as an example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the compounds of the invention, pharmaceutically acceptable salts of such pro-drugs, and other bioequivalents.

[00119] The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the compounds of the invention: i.e. salts that retain the desired biological activity of the parent compound and do not impart undesired toxicoSogical effects thereto. Fo oligomers, preferred examples of pharmaceutically acceptabl salts include but are not limited to (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.; (b) add addition salts formed with inorganic acids, for example hydrochloric acid, bydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; (c) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic add, ma!eic acid, fu marie acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, algintc acid, po!ygiutamic acid, naphthalenesuifonie acid, methanesuifonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine. The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and mucous membranes, as well as rectal delivery), pulmonary, e.g., by inhalation or insufflation of powders or aerosols (including by nebulizer, intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injectio or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Oligomers with at least one 2'-0-methoxyethyl modification are believed to be particularly useful for oral administration. Preferably, th antisense oligomer i delivered via the subcutaneous or intravenous route.

[00120] The pharmaceutical formulations of the present invention, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the phamiaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

[001213 In one embodiment, the antisense oligomer is administered in an amount and manner effective to result in a peak blood concentration of at least 200-400 nM antisense oligomer. Typically, one or more doses of antisense oligomer are administered, generally at regular intervals, for a period of about one to two weeks. Preferred doses for oral administration are from about 1 mg to 1000 mg oligomer per 70 kg. in some cases, doses of greater than 1 00 mg oiigomer/paiient may be necessary. For i.v. administration, preferred doses are from about 0.5 mg to 1000 mg oligomer per 70 kg. For intra venous or sub cutaneous administration, the antisense oligomer may be administered at a dosage of about 120 mg/kg daily or weekly.

[00122] The antisense oligomer may be administered at regular intervals for short time period, e.g., daily for two weeks or less. However, in some cases the oligomer is administered intermittently over a longer period of time. Administration may be followed by, or concurrent with, administration of an antibiotic or other therapeutic treatment. The treatment regimen may be adjusted (dose, frequency, route, etc.) as indicated, based on the results of immunoassays, other biochemical tests and physiological examination of the subject under treatment.

[001233 Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements df drug accumulation in the body of the patient. Persons of ordinar skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual oligomers, and can generally be estimated based on ECSOs found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 pg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligomer is administered in maintenance doses, ranging from 0.01 § to 100 g per kg of body weight, once or more daily, to once every 20 years.

[00124] An effective in viva treatment regimen using the antisense oligomers of the invention may vary according to the duration, dose, frequency and route of administration, as well as the condition of the subject under treatment {i.e., prophylactic administration versus administratio in response to localized or systemic infection). Accordingly, such in vivo therapy will often require monitoring by tests appropriate to the particular type of disorder under treatment, and corresponding adjustments in the dos or treatment regimen, in order to achieve an optimal therapeutic outcome.

[001253 Treatment may be monitored, e.g., by general indicators of diseas known in the art. The efficacy of an in vivo administered antisense oligomers of the invention ma be determined from biological samples (tissue, blood, urine etc.) taken from a subject prior to, during and subsequent to administration of the antisense oligomer. Assays of such samples include (1 ) monitoring the presence or absence of heteroduplex formation with target and non-target sequences, using procedures known to those skilled in the art, e.g., an electrophoretic gel mobility assay; (2) monitoring the amount of a mutant mRNA in relation to a reference normal mRNA or protein as determined by standard techniques such as RT-PCR, Northern blotting, ELISA or Western blotting,

[00126] Intranuclear oligomer delivery is a major challenge for antisense oligomers. Different cell-penetrating peptides (CPP) localize PMOs to varying degrees in different conditions and ceil Sines, and novel CPPs have been evaluated by the inventors for their ability to deliver PMOs to the target cells. The terms CP or "a peptide moiety which enhances cellular uptake" are used interchangeably and refer to cationic cell penetrating peptides, also called "transport peptides", "carrier peptides", or "peptide transduction domains". The peptides, as shown herein, have the capability of inducing cell penetration within about or at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of cells of a given ceil culture population and allow macromolecular translocation within multiple tissues in vivo upon systemic administration. CPPs are well-know in the art and are disclosed, for example in U.S. Application No. 2010/0016215, which is incorporated by reference in its entirety.

[00127] The present invention therefore provides antisense oligomers of the present invention win combination with cell-penetrating peptides for manufacturing therapeutic pharmaceutical compositions.

[00128] According to a still further aspect of the invention, there is provided one or more antisense oligomers as described herei for use in an antisense oligomer-based therapy. Preferably, the therapy is for a condition related to ITGA4 expression. More preferably, the therapy for a condition related to 1TGA4 expression is therapy for MS.

[00129] More specifically, the antisense oligomer may be selected from the group consisting of any one or more of SEQ ID NOs; 1-202, and combinations or cocktails thereof. This includes sequences which can hybridise to such sequences under stringent hybridisation conditions, sequences complementary thereto, sequences containing modified bases, modified backbones, and functional truncations or extensions thereof which possess or modulate pre-mRNA processing activity in an ITGA4 gene transcript.

[00130] The invention extends also to a combination of two or more antisense oligomers capable of binding to a selected target to induce exon exclusion in an ITGA4 gene transcript. The combination may be a cocktail of two or more antisense oligomers, a construct comprising two or more or two or more antisense oligomers joined togethe for use in an antisense oligomer-based therapy.

[00131] The invention provides a method to treat, prevent or ameliorate the effects of a disease associated with ITGA4 expression, comprising the step of; a) administering to the patient an effective amount of one or more antisense oligomers or pharmaceutical composition comprising one or more antisense oligomers as described herein.

[00132] Furthermore, the invention provides a method to treat, prevent or ameliorate the effects of multiple sclerosis, comprising the step of: a) administering to the patient an effective amount of one or more antisense oligomers or pharrnaceuticai composition comprising one or more antisens oiigomers as described herein.

[00133] Preferably, the therapy is used to reduce the levels of functional 1GTA4 protein via an exon skipping strategy. The reduction in levels of ITGA4 is preferably achieved by reducing the transcripts level through modifying pre-mRNA splicing in the integrin alpha-4 (ITGA4) gene transcript or part thereof.

[00134] The reduction in STGA4 will preferably lead to a reduction in the quantity, duration or severity of the symptoms of an ITGA4-related condition or pathology, such as MS.

[00135] As used herein, "treatment" of a subject (e.g. a mammal, such as a human) or a cell is any type of intervention used in an attempt to alter the natural course of the individual or ceil. Treatment includes, but is not limited to, administration of a pharmaceutical composition, and may be performed either prophylactica!ly or subsequent to th initiation of a pathologic event or contact with an etiologic agent. Also included are "prophylactic" treatments, which can be directed to reducing the rate of progression of the disease or condition being treated, delaying the onset of that disease or condition, or reducing the severity of its onset. Treatment" o "prophylaxis" does not necessaril indicate complete eradication, cure, or prevention of the disease or condition, or associated symptoms thereof.

[00136] According to another aspect of the invention there is provided the use of one or more antisense oligomers as described herein in the manufacture of a medicament for the modulation or control of a disease associated with 1TGA4 expression. [001373 The invention also provides for the use of purified and isolated antisense oligomers as described herein, for the manufacture of a medicament for treatment of a disease associated with ITGA4 expression.

[00138] There is provided the use of purified an isolated antisense oligomers as described herein for the manufacture of a medicament to treat, prevent or ameliorate the effects of a disease associated with ITGA4 expression.

[00139] Preferably, the ITGA4-reiated pathology or disease is MS.

[001403 The invention extends, according to a still further aspect thereof, to cDNA or cloned copies of the antisense oligomer sequences of the invention, as well as to vectors containing the antisense oligomer sequences of the invention. The invention extends further also to cells containing such sequences and/or vectors.

[001413 Tti invention also provides kits to treat, prevent or ameliorate a disease or condition associated with ITGA4 expression in a patient, which kit comprises at least an isolated or purified antisense oligomer for modifying pre-mRNA splicing in an 1TGA4 gene transcript or part thereof, packaged in a suitable container, together with instructions for its use.

[00142] In a preferred embodiment, the kits will contain at least one antisense oligomer as described herein or as shown in Tables 3 to 7, or a cocktail of antisense oligomers, as described herein. The kits ma also contai peripheral reagents such as buffers, stabilizers, etc.

[001433 There is therefore provided a kit to treat, prevent or ameliorate a disease or condition associated with ITGA4 expression in a patient, which kit comprises at leas an antisense oligomer described herein or as shown in Tables 3 to 7 and combinations or cocktails thereof, packaged in a suitable container, together with instructions for its use.

[001443 There is also provided a kit to treat, prevent or ameliorate a disease or condition associated with ITGA4 expression in a patient which kit comprises at least an antisense oligomer selected from the group consisting of any one or more of SEQ ID NOs: 1-202, and combinations or cocktails thereof, packaged in a suitable container, together with instructions for its use.

[00145] Preferably, the disease or condition is multipl sclerosis. [001463 The contents of the kit can be iyophi!ized and the kit can additionally contain a suitable soivent for reconstitution of the lyophilized components. Individual components of the kit would be packaged in separate containers and, associated with such containers, can be a notice in the form prescribed by a governmental agenc regulating the manufacture, use or safe of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.

[00147] When the components of the kit are provided in one or more liquid solutions, the liquid solution can be an aqueous solution, for example a sterile aqueous solution. For in vivo use, the expression construct may be formulated into a pharmaceutically acceptable syrirtgeable composition. In this case the container means may itself be an inhalant, syringe, pipette, eye dropper, or other such like apparatus, from which the formulation may be applied to an affected area of the animal, such as the lungs, injected into an animal, or even applied to and mixed with the other components of th kit.

[00148] The components of the kit may also be provided in dried or lyophilized forms. When reagents or components are provided as a dried form, reconstitution generally is by the addition of a suitable soivent it is envisioned that the solvent also may be provided in another container means. Irrespective of the number or type of containers, the kits of the invention also may comprise, or be packaged with, an instrument fo assisting with the injeciion ' administration or placement of the ultimate complex composition within the body of an animal. Such an instrument may be an inhalant, syringe, pipette, forceps, measured spoon, eye dropper or any such medically approved delivery vehicle.

[00149] Those of ordinary skili in the field should appreciate that applications of the above method has wide application for identifying antisense oligomers suitable for use in the treatment of many other diseases.

[001503 The antisense oligomers of the present invention may also be used in conjunction with alternative therapies, such as drug therapies.

[001513 The present invention therefore provides a method of treating, preventing or ameliorating the effects of a disease or condition associated with ITGA4 expression, wherein the antisense oligomers of the present invention and administered sequentially or concurrently with another afternative therapy associated with treating, preventing or ameliorating the effects of a disease or condition associated with ITGA4 expression. Preferably, the disease or condition is MS.

[00152] The alternative therapy may be chosen from the list comprising interferon beta-la, interferon beta-1 b, glattramer acetate, mito antrone, natalizumab, fingoiimod, teriflunomide, dimethyl fumarate and alemtuzumab.

General

[00153] Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes ail such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in the specification, individually or collectively and any and all combinations or an two or more of the steps or features.

[001543 The present invention is not to be limited in scope by the specific embodiments described herein, which are intended for the purpose of exemplification only. Functionally equivalent products, compositions and methods are clearly within the scope of the invention as described herein,

[00155] The entire disclosures of all publications (including patents, patent applications, journal articles, laboratory manuals, books, or other documents) cited herein are hereby incorporated by reference. No admission is made that any of the references constitut prior art or are part of the common general knowledge of those working in the field to which this invention relates.

[001563 Each document, reference, patent application or patent cited in this text is expressly incorporated herein in their entirety by reference, which means that it should be read and considered b the reader as part of this text. That the document, reference, patent application or patent cited in this text is not repeated in this text is merely for reasons of conciseness.

[00157] An manufacturer's instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. [001583 As used herein the term "derived" and "derived from" shall b taken to indicate that specific integer may be obtained from a particular source albeit not necessarily directly from that source.

[00159] As used herein, the singular forms "a," "an" and "the" include plural references unless the context clearly dictates otherwise.

[00 60] Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.

[00161] Other than in the operating example, or where otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about". Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specificatio and claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. Hence "about 80 %" means "about 80 %" and also "80 %". At the very least, each numerical parameter should be construed in Sight of the number of significant digits and ordinary rounding approaches.

[00162] Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value; however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements

[00163] Other definitions for selected terms used herein may be found within the detailed description of the invention and apply throughout. Unless otherwise defined, all other scientific and technical terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which the invention belongs.

[00164] Sequence identity numbers ("SEG ID NO:"} containing nucleotide and amino acid sequence information included in this specification are collected at the end of the description and have been prepared using the program Patentln Version 3.0. Each nucleotide or amino acid sequence is identified in the sequence listing by the numeric indicator <210> followed by the sequence identifier (e.g. <210>1 , <2T0>2, etc.). The length, type of sequence and source organism for each nucleotide or amino acid sequence are indicated by information provided in the numeric indicator fields <211>, <212> and <213>, respectively. Nucleotide and amino acid sequences referred to in the specification are defined by the information provided in numeric indicator field <40Q> followed b the sequence identifier (e.g. <400>1 , <40Q>2, etc.).

[00165] An antisense oligomer nomenclature system was proposed and published to distinguish between the different antisense oligomers (see Mann et al, (2002) J Gen Med 4, 644-654). This nomenclature became especially relevant when testing several slightly different antisense oligomers, all directed at the same target region, as shown below:

H # A/D (x:y) the first letter designates the species (e.g. H: human, ; murine) "#" designates target exon number

"A/D" indicates acceptor or donor splice site at the beginning and end of the exon, respectively

(x y) represents the annealing coordinates where *'-" or indicate in.tronic or exonic sequences respectively. As an example, A(-6+18) would indicate the last 6 bases of the intron. preceding the target exon and the first 18 bases of the target exon. The closest splice site would be the acceptor so these coordinates would be preceded with a "A". Describing annealing coordinates at th donor splice site could be D(+2-18) where the last 2 exonic bases and the first 18 intronic bases correspond to the annealing site of the antisense oligomer. Entirely exonic annealing coordinates that would be represented by Αί+βδ+δδ), that is the site between the 65th and 85th nucleotide, inclusive, from the start of that exon.

[00166] The following examples serve to more fully describe the manner of using the above-described invention, as well as to set forth the best modes contemplated for carrying out various aspects of the invention. It is understood that these methods in no way serve to limit the true scope of this invention, but rather are presented for illustrative purposes. EXAMPLES

Example 1

Oligomer nomenclature

[00167] The nomenclature system defines species, exon number, acceptor or donor targeting and annealing coordinates, where "-" indicates intronic position and "+" specifies exonic location from the splice site, as described herein. Some detailed oligomer annealing coordinates are shown in Tables 3 to 7.

Antisense oligomers

Antisense oligomers with 2-0'fV1 modification were either synthesized in-house or ordered from Tri Link BioTechnologies, Inc [San Diego, CA, USA].

Cell propagation and transfection

[00188] Primary normal dermal fibroblasts were propagated using well established techniques [Villegas and McPhaul, Gurr Protoc ol Biol (2005) 28.3.1-28.3.9]. Cells wer seeded and propagated in 75 cm 2 tissue culture fiasks and transfeetson with 20MeAO was performed in 24 well plates. One day before transfection, 15-17,000 ceils were seeded in 24 well plates and transfected with a range of concentrations (5-100 nEVI) of 20MeAO using Lipofectin® transfection reagent [Life Technologies: Carlsbad, CA, USA] (Lipofectin.oligo ratio of 2:1 ) according to the manufacturer protocol.

[00169] For PMO transfection, P O were annealed to oligonucleotides with reverse complement sequences and the resulting duplex was allowed to form complexes with Lipofecti ® (Lipofectin.Qligo ratio of 2:1 ).

[00170] Transfected cells were typically incubated for 24 hours, unless otherwise indicated, before RNA was extracted for analysis using T Izo!® acid phenol extraction [Life Technologies: Carlsbad, CA, USA], RNA samples were treated with RNase free DNAs 1 , although minor DNA contamination is riot problematic.

RT-PCR analysis

[00171] One step RT-PCR using Superscript® ill [Life Technologies Carisbad, CA, USA]: 10G ng of total RNA was used as a template and incubated for 30 min at 55°C, and at 94°C for 2 min, before 30 rounds of 94°C for 30 sec, 55°C for 30 sec and 68°C for 1 min 30 sec using exon 1 F and 10R primers for exon 2-9 skipping, exon 9F and 20R primers for exon 10-19 skipping and exon 10F and 28R primers for exon 20-27 skipping.

[00172] PCR products were fractionated on 2% agarose gels in Tris-Acetate-EDTA buffer and the images captured on gel documentation system and analysed with Bio D software Vi!ber Lourmat, Eberhardzell, Germany] to quantitafe band weight and estimate ratios of full length ITGA4 and exon skipped products. Product identity was confirmed by hand purification and DNA sequencing as necessary. The efficiency of exon skipping was determined by calculating th percentage of the transcripts with exon(s) skipped compared to the total product generated b RT-PCR.

[00173] Among the aniisense oligomers tested, the antisen.se oligomer targeting exon 3, 4 and 19 produced 30-40 % of transcripts with the targeted exon being skipped (Table 3). Based on the size of the transcripts, antisense oligomers targeting exon 3 hav induced skipping of both exon 3 and 4, leaving the reading fram intact.

Western blotting

[00174] Proteins were extracted from treated cultures after two days and prepared according to Cooper et ai., [Neurology (2003) 61 , 93-96], but with 15% SDS. SDS- PAGE electrophoresis was performed using NuPAGE® Novex® 4-12% BiS Tris gels [Life Technologies: Carlsbad, CA t USA] run at 200V for 55 mi s. Proteins were transferred to Pall Fluorotrans® W PVDF membranes [Pal! Corporation, USA] at 290 mA for overnight at 18°C. ITGA4 antibody [Cell Signaling Technology, Inc., USA Cat. No. 4800], which recognizes residues surrounding Ser1027 encoded by exon18, was applied at 1:1000 dilution overnight at 4°C and β-tubu!in was detected by a rabbit polyclonal antibody (1 :3000 dilution) overnight at 4°C (Thermo Scientific Pierce Antibody Products, Rockford IL USA Cat. No, PIEPA1-41331), as a reference loading protein, with loadings normalized to the β-tubuiin.

[00175] For immunodetection, polyclonal goat anti-rabbit immunoglobulins HRP (DakoiS), Cat. no P0448) at a dilution of 1:10, 000 and Luminata™ Crescendo Western HRP substrate (Merck MiliiporeS), Cat. No. WBLUR0100) were used. Quantification was performed using Bio~ P software [Vilber Lourmat, Eberhardzell,, Germany] for image analysis. [001763 Results are provided in Figure 2, Th results indicate a knockdown of the ITGA4 protein in normal fibroblasts following treatment with exon skipping antisense oligomers targeting exon 3 (AG ), 4 (A021), and 19 (A047).

Cell adhesion assay

[00177] 96-weil mieropiates were coated with fibronectin 3 pg weil in PBS (Msllspore® Cat. No. FC010), laminin 0.75 Mg/well in PBS (Miliipore® Cat, No, AG56P) or recombinant human ¥CAM~1 0.5 ug/well in PBS (R&D Systems™, Cat. No. ADP5-050). Cells were washed twice in PBS and labelled with 2 μίνΐ calcein AM fluorescent dye (invitrogen®, Cat No. C1430) for 30 min in serum free Duibecco's modified Eagle's medium (DfvlEM}. After two washes with D EM, the cells were resuspended in serum free DMEM and plated (30,000 cells/well) and incubated at 37°C for 30-45 min. The rnicroplate was washed four times with PBS to remove non-adherent cells, and the remaining adherent cells were measured using a fluorescence plate reader, Beckman Coulter DTX-880 Muitimode Detector (NSW, Australia) with excitation wavelength of 488 nm and emission detected at 512 nm. In order to calculate the percentage of adherent cells, the fluorescent signals of total cells were analysed in a separate rnicroplate, omitting the wash steps. Background signals were subtracted from all samples and results were normalised to the sample treated with scrambled oligomers which serves as an experimental control. The experiment was independently repeated three times and a Student s r test was performed to determine the p value.

[00178] As shown in Figure 3, transfectio with oligomers targeting exon 4 (A021) resulted in a reduction in ceils adhering to a rnicroplate pre-coated with VCAM-1. This result correlates wit a Western analysis for protein expression which shows that the most reductio i ITGA4 protein expression was observed after treatment with A021. However, no changes in cell adhesion were observed when these cells were allowed to attach to the piates pre-coated with either fibronectin or laminin. These resuits indicate that lowering STGA4 expression has no effect on ceiSuiar interaction with fibronectin and laminin. These resuits are as expected since ITGA4 is not required for interaction with laminin, and several other integrin receptors apart from ITGA4 can interact with fibronectin. On the other hand, 1TGA4 is a major integrin receptor for interaction with VCAMi-1 and towering ITGA4 expression significantly affect cellular attachment to VCAIV1-1. Cell migration assay or wound-heating assay

[00179] A cell migration assay was performed 48 hours after transfeciion with an antisense oligomer. Normal human fibroblasts were transfeeted with oligomers and allowed to form a monolayer. A "wound" was then created by scraping the monolayer with a pipet tip arid images of the wound (gap) was taken using a microscope (Nikkon TS100) at 0 and 8 hour time points. Cells migration was analysed by comparing the size of the gap at 0 and 8 hour images using Image J (Rasband, W. S., Imaged, U. S, National Institutes of Health, Bethesda, Maryland, USA). Three independent experiments wer performed and an average cell migration was calculated. Data were normalised to scrambled oiigomer treated samples and student t test was performed for p value.

[00180] As shown in Figure 4, cells transfeeted with A021 targeting exon 4 shows slower ceil migration compared to the cells transfeeted with other oligomers including scrambled oligomers.

Example 2

Jurkat cells propagation and Nucleofection

[001813 Acute T lymphocytes leukemia cell line, Jurkat cells were maintained in RPMi-1640 medium supplemented with 10% fetal bovine serum according to the instruction from ATCC. Approximately 500, 000 cells were nucleofected at 500 nM using P2 Primary Cell 4D-nucieofactor X kit S (32 RCT) (Lonza, Australia) according to the manufacturer's instructions and incubated for 2-3 days before harvesting the ceils for RNA transcript analysis, Western blotting and flow cytometry.

Flow cytometry

[00182] Jurkat cells were collected 3 days after nucleofection and washed twic with cold PBS before incubating with PE fluorophore labelled anti-human ITGA4 antibody (BO Pharmingen, Australia) at a concentration recommended by the manufacturer for 20-30 min on ice. Cells were washed once with cold PBS and analysed using Beckman Coulter GaSlios fiow cytometer. Immunostain g

[00183] Jurkat cells were allowed to attach to the poly D coated coverslips for 30 min at 37°C and fixed with iced cold methanol: acetone (1 :1 ) for 5 min on ice. The coverslips with fixed ceils were washed once with 0.2% Triton in Tris Buffered Saline (TBS-T) pH 7.6 and blocked with 10% goat serum in TBS-T for 10 min. The ceils were then incubated with anti-lTGA4 antibody (Ceil Signalling Technology, inc., USA cat. No. 8440) diluted 1 :200 in TBS-T for 1 h at room temperature. The coverslips wer then washed 5 min three times wit TBS-T and incubated with ASexa fluor® 488 conjugated goat anti-rabbit antibody (Thermo Fisher Scientific, cat. No. A-11008) diluted 1:400 in TBS-T for 1 h at room temperature. Finaily, the ceils were washed twice with TBS-T for 5 min each and incubated with Hoechst 50 pg/ml in TBS-T for 5 min at room temperature before the final wash with phosphate buffered saline (PBS).

Jurkat cells migration

[001843 The upper side of transwe!l migration inserts (Polyester membrane, 3 pm pore size, 6.5 mm diameter, Coming®) were coated with 50 μΙ of 0.5 pg/μΙ recombinant human VCAM-I (R&D systems, cat no. CD-106) for overnight at room temperature and pre-equilibrated with RPlVl!-1640 for 1 -2 h at 37°C. Jurkat cell nucleofected with PMO fo 2 days were resuspended in 100 μΐ of serum free PRMI-1640 medium and added to the upper compartment of the insert and 600 μ! of RPSVH-1640 medium supplemented with 10% FBS were added to the lower compartment. The cells were allowed to migrate for 5h at 37°C. Ceils from both top and bottom chambers were collected and stained with 2 μ calcein AM fluorescent dye (Invitrogen®, Cat. No. C1430) for 30 min and fluorescent signals were measured using a fluorescence plate reader, Beckman Coulter DTX-880 Multimode Detector (NSW, Australia) with excitation wavelength of 488 nm and emission defected at 512 nm. The percentage of cell migrated was calculated from the total fluorescent signals generated by top and bottom chamber.

[001853 Modifications of the above-described modes of carrying out the various embodiments of this invention will be apparent to those skilled in the art based on the above teachings related to the disclosed invention. The above embodiments of th invention are merely exemplary and should not be construed to be in any way limiting.




 
Previous Patent: NUTRIENT SYSTEM

Next Patent: CROSS DOMAIN DESKTOP COMPOSITOR