Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTISENSE OLIGONUCLEOTIDES FOR TARGETING PROGRANULIN
Document Type and Number:
WIPO Patent Application WO/2022/129320
Kind Code:
A1
Abstract:
The present invention relates to oligonucleotides which alter the splicing pattern of progranulin in cells, and their use in the treatment of neurological disorders.

Inventors:
JAGASIA RAVI (CH)
JØNSON LARS (DK)
RASMUSSEN SØREN (DK)
RAVN JACOB (DK)
TEHLER DISA (DK)
VANG DORTHE (DK)
WORM JESPER (DK)
Application Number:
PCT/EP2021/086167
Publication Date:
June 23, 2022
Filing Date:
December 16, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (US)
HOFFMANN LA ROCHE (US)
International Classes:
C12N15/113
Domestic Patent References:
WO2020191212A12020-09-24
WO2021229036A12021-11-18
WO2020191212A12020-09-24
WO2020191912A12020-10-01
WO2011017521A22011-02-10
WO2013154798A12013-10-17
WO1999014226A21999-03-25
WO2000066604A22000-11-09
WO1998039352A11998-09-11
WO2004046160A22004-06-03
WO2000047599A12000-08-17
WO2007134181A22007-11-22
WO2010077578A12010-07-08
WO2010036698A12010-04-01
WO2007090071A22007-08-09
WO2009006478A22009-01-08
WO2011156202A12011-12-15
WO2008154401A22008-12-18
WO2009067647A12009-05-28
WO2008150729A22008-12-11
WO2001023613A12001-04-05
WO2014076195A12014-05-22
WO2015113922A12015-08-06
WO2020191212A12020-09-24
Other References:
FRYDAS ALEXANDROS ET AL.: "Genetic variants in progranulin upstream open reading frames increase downstream protein expression", NEUROBIOLOGY OF AGING, TARRYTOWN, NY, US, vol. 110, 10 September 2021 (2021-09-10), pages 113 - 121, XP086921354, ISSN: 0197-4580, [retrieved on 20210910], DOI: 10.1016/J.NEUROBIOLAGING.2021.09.007
JASON L. ERIKSEN ET AL.: "Progranulin: normal function and role in neurodegeneration", JOURNAL OF NEUROCHEMISTRY, vol. 104, no. 0, 1 January 2008 (2008-01-01), pages - ???, XP055033005, ISSN: 0022-3042, DOI: 10.1111/j.1471-4159.2007.04968.x
ZHIHENG HE ET AL.: "Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis", JOURNAL OF MOLECULAR MEDICINE, vol. 81, no. 10, 1 October 2003 (2003-10-01), pages 600 - 612, XP055120793, ISSN: 0946-2716, DOI: 10.1007/s00109-003-0474-3
TOWNLEY ET AL., NEUROLOGY, vol. 90, no. 24, 12 June 2018 (2018-06-12), pages 1127
CAPELL ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 289, no. 37, 2014, pages 25879 - 25889
HIRAO ET AL., ACCOUNTS OF CHEMICAL RESEARCH, vol. 45, 2012, pages 2055
BERGSTROM, CURRENT PROTOCOLS IN NUCLEIC ACID CHEMISTRY, 2009
MERGNYLACROIX, OLIGONUCLEOTIDES, vol. 13, 2003, pages 515 - 537
HANSEN ET AL., CHEM. COMM., vol. 36-38, 1965
HOLDGATE ET AL., DRUG DISCOV TODAY, 2005
SANTALUCIA, PROC NATL ACAD SCI USA., vol. 95, 1998, pages 1460 - 1465
SUGIMOTO ET AL., BIOCHEMISTRY, vol. 34, 1995, pages 11211 - 11216
MCTIGUE ET AL., BIOCHEMISTRY, vol. 43, 2004, pages 5388 - 5405
FREIERALTMANN, NUCL. ACID RES., vol. 25, 1997, pages 4429 - 4443
UHLMANN, CURR. OPINION IN DRUG DEVELOPMENT, vol. 3, no. 2, 2000, pages 293 - 213
DELEAVEYDAMHA, CHEMISTRY AND BIOLOGY, vol. 19, 2012, pages 937
MORITA ET AL., BIOORGANIC & MED.CHEM. LETT., vol. 12, pages 73 - 76
SETH ET AL., J. ORG. CHEM., vol. 75, no. 5, 2010, pages 1569 - 81
MITSUOKA ET AL., NUCLEIC ACIDS RESEARCH, vol. 37, no. 4, 2009, pages 1225 - 1238
WANSETH, J. MEDICAL CHEMISTRY, vol. 59, 2016, pages 9645 - 9667
"Manoharan in Antisense Drug Technology, Principles, Strategies, and Applications", 2001, MARCEL DEKKER, INC.
MANOHARAN, ANTISENSE AND NUCLEIC ACID DRUG DEVELOPMENT, vol. 12, 2002, pages 103
Attorney, Agent or Firm:
O'FARRELL, Jennifer (GB)
Download PDF:
Claims:
CLAIMS

1. An antisense oligonucleotide, wherein the antisense oligonucleotide is 8 - 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary to a splice regulation site of the human progranulin pre-mRNA transcript.

2. The antisense oligonucleotide according to claim 1 , wherein the human progranulin pre-mRNA transcript comprises the exon 1, intron 1 and exon 2 sequence of the human progranulin pre-mRNA transcript (SEQ ID NO 276).

3. The antisense oligonucleotide according to claim 1 or claim 2, wherein the contiguous nucleotide sequence is complementary to SEQ ID NO 277, SEQ ID NO 278, SEQ ID NO 279 or SEQ ID NO 280.

4. The antisense oligonucleotide according to any one of claims 1-3, wherein the contiguous nucleotide sequence is selected from SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74 and SEQ ID NO 75, or at least 8 or at least 10 contiguous nucleotides thereof.

5. The antisense oligonucleotide according to claim 1 or claim 2, wherein the contiguous nucleotide sequence is complementary to SEQ ID NO 281.

6. The antisense oligonucleotide according to claim 5, wherein the contiguous nucleotide sequence is SEQ ID NO 134, or at least 8 or at least 10 contiguous nucleotides thereof.

7. The antisense oligonucleotide according to claims 1 or claim 2, wherein the contiguous nucleotide sequence is selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71 , SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252.

8. The antisense oligonucleotide according to any one of claims 1-7, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof comprises one or more modified nucleotides or one or more modified nucleosides.

9. The antisense oligonucleotide according to any one of claims 1-7, wherein the antisense oligonucleotide is or comprises an antisense oligonucleotide mixmer or totalmer.

10. An antisense oligonucleotide having the structure:





11. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GaGctGggTcAagAAT (SEQ ID NO: 71), GgtCaaGaAtgGtgTG (SEQ ID NO: 73), CaGaAtGgtGtGgTC (SEQ ID NO 74), GaAtGgtGtGgTccC (SEQ ID NO 75) or CtcAagCtcAcAtgGC (SEQ ID NO 134) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

12. A pharmaceutical composition comprising the antisense oligonucleotide according to any one of claims 1-11 and a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.

13. An in vivo or in vitro method for enhancing the expression of the Exon1-Exon2 progranulin splice variant in a cell which is expressing progranulin, said method comprising administering an antisense oligonucleotide according to any one of claims 1-11 , or a pharmaceutical composition according to claim 12 in an effective amount to said cell.

14. The antisense oligonucleotide according to any one of claims 1-12, or the pharmaceutical composition according to claim 13 for use in the treatment of a neurological disease.

15. The antisense oligonucleotide according to any one of claims 1-12, or the pharmaceutical composition according to claim 13 for use in the treatment of progranulin haploinsufficiency or a related disorder.

Description:
ANTISENSE OLIGONUCLEOTIDES FOR TARGETING PROGRANULIN

FIELD OF INVENTION

The present invention relates to antisense oligonucleotides which alter the splicing pattern of progranulin, and their use in the treatment of neurological disorders. Such antisense oligonucleotides may up-regulate or restore expression of the Exon1-Exon2 progranulin splice variant in cells.

BACKGROUND

Progranulin (PGRN) is a highly conserved secreted protein that is expressed in multiple cell types, both in the CNS and in peripheral tissues.

Deficiency of the secreted protein progranulin in the central nervous system causes the neurodegenerative disease fronto temporal dementia (FTD). Pathogenic progranulin mutations lead to a loss of about 50% in progranulin levels through haploinsufficiency and to intraneuronal aggregation of TDP-43 protein. Progranulin plays a supportive and protective role in numerous processes within the brain, including neurite outgrowth, synapse biology, response to exogenous stressors, lysosomal function, neuroinflammation, and angiogenesis in both cell autonomous and non-autonomous manners.

Both directly and via its conversion to granulins, progranulin regulates lysosomal function, cell growth, survival, repair, and inflammation. Progranulin has a major role in regulation of lysosomal function associated microglial responses in the CNS. Autosomal dominant mutations of the progranulin gene leading to protein haploinsufficiency are linked to familial frontotemporal dementia with neuropathologic frontotemporal lobar degeneration (FTLD) associated with accumulation of TAR-DNA binding protein of 43kDA (TDP-43) inclusions (FTLD-TDP).

Homozygous GRN mutations are linked to neuronal ceroid lipofuscinosis (NCL) (Townley, et al., Neurology, 2018 June 12; 90(24): 1127).

Mutations in the progranulin gene have recently been identified as a cause of about 5% of all FTD, including some sporadic cases. Recent studies using mouse models have defined the expression of progranulin in the brain (Petkau et al., 2010). Progranulin is expressed late in neurodevelopment, localizing with markers of mature neurons. Progranulin is expressed in neurons in most brain regions, with highest expression in the thalamus, hippocampus, and cortex. Microglia cells also express progranulin, and the level of expression is upregulated by microglial activation. Around 70 different progranulin gene mutations have been identified in FTD and all reduce progranulin levels or result in loss of progranulin function.

There is therefore an urgent need for therapeutic agents which can increase the expression and/or activity of progranulin.

SUMMARY OF INVENTION

A splice variant of progranulin which retains the 5’ part of Intron 1 is expressed in the brain such as in neurons or microglia cells (Capell et al. The Journal of Biological Chemistry, 2014, 289(37), 25879-25889). This splice variant include the 5’ most 271 nucleotides of intron 1 , which totals 3823 nucleotides. The 271 nucleotide fragment of intron 1 includes two AUG sites upstream of the canonical downstream AUG (open reading frame) in exon 2. Translation from these two upstream AUG sites will not encode the progranulin protein, and due to premature termination codons the transcript may undergo non-sense mediated mRNA decay (NMD).

W02020/191212 describes specific oligonucleotides which can target the progranulin mRNA. Here the inventors have determined that reducing the splice variant which retains the 5’ part of intron 1 increases the Exonl and Exon2 splice variant and further increases progranulin protein expression.

The present invention provides antisense oligonucleotides of progranulin. These antisense oligonucleotides are capable of altering the splicing pattern of progranulin, In particular the antisense oligonucleotides may up-regulate expression of the Exon1-Exon2 progranulin splice variant, reducing production of the progranulin Intron1-Exon2 splice variant which retains the 5’ part of intron 1 , increasing the expression of the progranulin protein. These antisense oligonucleotides could be described as modulators of progranulin splicing, or as agonists of progranulin Exon1-Exon 2.

The antisense oligonucelotides of the invention may be used to restore or enhance expression of the progranulin Exon1-Exon2 splice variant in cells.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA. The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the exon 1 , intron 1 and exon 2 sequence of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a human progranulin pre-mRNA transcript that comprises the exonl, intron 1 and exon 2 sequence of the human progranulin pre- mRNA transcript (SEQ ID NO 276).

The progranulin exon 1, intron 1 and exon 2 sequence is shown below as SEQ ID NO: 276. The progranulin exonl sequence (in capital letters) corresponds to genome Ensemble (www.ensemble.org) chromosome 17 position 44,345,123; to position 44,345,334. Intron 1 corresponds to genome Ensemble chromosome 17 position 44,345,335 to 44,349,157 and Exon 2 sequence (in capital letters) corresponds to genome Ensemble chromosome 17 position 44,349,158 to position 44,349,302.

Exon 1, intron 1 and exon 2 sequence of the human progranulin pre-mRNA (SEQ ID NO: 276): GGCGAGAGGAAGCAGGGAGGAGAGTGATTTGAGTAGAAAAGAAACACAGCATTCCAGGCT G GCCCCACCTCTATATTGATAAGTAGCCAATGGGAGCGGGTAGCCCTGATCCCTGGCCAAT GG AAACTGAGGTAGGCGGGTCATCGCGCTGGGGTCTGTAGTCTGAGCGCTACCCGGTTGCTG C TGCCCAAGGACCGCGGAGTCGGACGCAGgtaggagagcggccgcgcagacctctcgcctg ctcctgcccaggggc ccgccagggccatgtgagcttgaggttcccctggagtctcagccggagacaacagaagaa ccgcttactgaaactccttgggggttctga tacactagggggagttttatgggaaagaggaagcagtaattgcagtgacgccccgttaga aggggctttctacctccccagcattccccca aagcagggaccacaccattcttgacccagctccacccctgtcggtaggtgctggcttctt cccctctcctggtggtggtgggtggttcccgcg gcggcctggagccggaggggcgcgcgaccctgggctgggagctccgagggcctgggaacg agacctgagaccttggcttctcgaag gtagtagggacttgggagtggtgactgaacctggtctggctcctccttacttcctcttgt tgcgggtgggacgagctagcttccgcctctccca gccactttttcctgctcatttgcagctaggttggctccccttttgggaatttcctctccc cttggcactcggagttggggggtgccacctagtggaa gataacggagctagggtcttgaagaggctgctgtcccctctggctgttttggcggtgtag ggtggcatgagagactgcgactcgcctcctcat ccctgtttctgtatgcgagtgcttgtattcagtagaagcatacactatactccctcaatt tagggtaaacaggaggggccacatgcacaggta attcaccagggagccgaacactcctgtgcagacagactccccttcccagcaagccatggc agcggacagcctgctgagaacacccag gaagcaggcggtgccagctgcaggtgctttgcctgggagctgtggggctgaggagagggt ccactgtccaggaccagtgaacttcatcct tatctgtccaggaggtggcctcttggggatgctgagttaggggaggggcacttgaggaaa gccaggtggagcagagaggatgtgagtg actgggtgggtgagatttcctgcccctccccccgcagtggtatccacacctagactcgtg gggtaactgaggcacagacagagagcaact tctcaggccctcacagttggcaattctaggattaggacccaagtgcgattttcaggcagt ccctgtaccctgtttctgttgtacctgttgcaccatt cccaggcactgcccatcgtgccactagtgatatgaacccaggtccaatacgctctggggc catcaaagcctgacgtcaccatgacctgat gtgtgacgtgttataggtgtcccttggtatcttcacggaactggttccaggaccccaaaa tctgtgggtgctcaagcccctgagataaaatgg tgtaatatttgcatataacctatacatactttaaatcatttctagattacttatacctaa tacaatggaaatgacatgtcggctgggcgtggtggct catgcctgtaatcccaccactttgggaggccgtggcaggtggatcacctgaggtctggag tttgagaccagcctgaccaacatggtgaaa cccccatctctactaaaaatacaaaaattagccaggtgtggtagcgcacacctataatcc cacctacttgggaggctgaggcaggagaat tgcttgaacctgggaggcggagttcgcagtaagctgagatcgcgccactgtactacagcc tgggtgacagagcaggactccatctcaaa aaaaaaagagaaaaagaaaaagaaatgccatgtaaatagttgtgatcctgaattgtttag ggaataataagaaagaactatctgtagat gttcagtatagatgcacccatcgtaagcctaactacattgtataactcagcaacgatgta acattttcaggggtttttttgttttgttttttgagacag aatctcagtctcactctgtcacccaggctggagtatgttggcgtgatctctgctcactgc aacctccacctcctgggctcaagcgattctcctgc ctcagcctcttgagtagctgggattgcaggtgtgcgctaccacgcatggctaatttttgt atttttaatagagatggggttttaccacgttggtcag gctggtcttgaactcctgaccttgggatccgcccacctgggcctcccaaagtgctgggat tacaggcgttagccaccgcgcccaatatatttt gatccctggttggatatggagggctgactgtacttaacatctctaagcttcagtttcctc ctttaaaataaaggtgtggctgggtgtggtggttca agcctgtaatcccagcacttagggaggctgaggtgggtggatcagctgaggtcaggagtt caagaccagcctgaccaatatggtgaaac cccctctctgctaaaaatacaaaaattagccaggcgtggtggcgagcgcctgtagtccca gctacttgcttgaacttgggaggcagaggtt gcagtgagctgagatcgtgccactgaactcgagcatgggcaacagagcaagactgtctca aaaaaaaaaaaaaaaagggggtgag cagacgtggtggcacgctcccacagtcccagctacttagtaggaggccaaggttggagga ttgcttgatcccaggagtctgagtccagcc tgggcaacatggcaatacctcatctctaaaaataaaataaaagtaaaggtattaattact actttggatggttgttgcaaagaaatatatata aaataatggagagtcttgtaactggctcccaagaggctcaacagacattactgtttttgc ttcttcattatgagttacctctctggccaccccact gaactagctgggctagctgagcctgggagaagagttgtttaggaagtgagaggctgctct ccacagagactcaaggctcagttcctcctg gtgactcagatgggcagcccagtgggcacacgtggtctctctccacatgtggctgagttt cacttccagaatagatggagaggcaagggc agggtttagcatgcttgaggaatctcagagggccctggtggtgtgggggaccctcagaac acaggtgtctcaagggctgacccagcttct gtgtccttttctctgggtgaggaggggacattcatgggcagatggtgacctctggggaag gcagcccagactccactggccaccatatttcc tttttcacaactttctcacccctgtggtttcccatgtcatcatgtggccgcttcccgcaa ggccttagcggggtgcaggtatgaacatagtgtcag gcaaggaggcatctggaggggaaccctggcttttcctggggggactccctccctgcaccc tagccctgtcctctcccatggctactgatgcc ttcccctcaccccagaggtggcccacatctgcacagatcagacccacaaaaatcacgtct tcctgactctcataagcctgcccagtgagg cccaggcattaggccatgtgctggggactcagacccacacatatacgcatgtcagcattc atgcttacaggtccgcacatgctggggcaa gtgtcacacacggggcgctgtaggaagctgactctcagcccctgcagatttctgcctgcc tggacagggaggtgttgagaaggctcaggc agtcctgggccaggaccttggcctggggctagggtactgagtgaccctagaatcaagggt ggcgtgggcttaagcagttgccagacgttc cttggtactttgcagGCAGACCATGTGGACCCTGGTGAGCTGGGTGGCCTTAACAGCAGG GCTGGTG GCTGGAACGCGGTGCCCAGATGGTCAGTTCTGCCCTGTGGCCTGCTGCCTGGACCCCGGA G GAGCCAGCTACAGCTGCTGCCGTCCCCTTCTG

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 —

40 nucleotides in length and comprises a contiguous nucleotide sequence of at least 12 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 12 - 16 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 12 - 16 nucleotides in length and comprises a contiguous nucleotide sequence of 12 - 16 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 12 - 18 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 12 - 18 nucleotides in length and comprises a contiguous nucleotide sequence of 12 - 18 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length which is complementary, such as fully complementary, to a splice regulation site of the human progranulin pre-mRNA.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a nucleotide sequence comprised within SEQ ID NO: 276. The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a nucleotide sequence comprised within nucleotides 441-468 of SEQ ID NO: 276.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a nucleotide sequence comprised within nucleotides 441-462 of SEQ ID NO: 276.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a sequence selected from the group consisting of: SEQ ID NO 277, SEQ ID NO 278, SEQ ID NO 279 and SEQ ID NO 280.

Target site for SEQ ID NO: 71 (SEQ ID NO 277)

ATTCTTGACCCAGCTC

Target site for SEQ ID NO: 73 (SEQ ID NO 278)

CACACCATTCTTGACC

Target site for SEQ ID NO: 74 (SEQ ID NO 279)

GACCACACCATTCTTG

Target site for SEQ ID NO: 75 (SEQ ID NO 280)

AGGGACCACACCATTC

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a nucleotide sequence comprised within nucleotides 268-283 of SEQ ID NO: 276.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to SEQ ID NO 281. Target site for SEQ ID NO: 134 (SEQ ID NO: 281)

GCCATGTGAGCTTGAG

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary, such as fully complementary, to a sequence selected from the group consisting of: SEQ ID NO: 291 and SEQ ID NO: 292.

The antisense oligonucleotide may be 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length. In some embodiments the antisense oligonucleotide is 8 - 40, 12 - 40, 12 - 20, 10-20, 14 - 18, 12 - 18 or 16 - 18 nucleotides in length.

The contiguous nucleotide sequence may be 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length. In some embodiments, the contiguous nucleotide sequence is of a length of at least 12 nucleotides in length, such as 12 - 16 or 12 - 18 nucleotides in length.

In some embodiments, the contiguous nucleotide sequence is the same length as the antisense oligonucleotide.

In some embodiments the antisense oligonucleotide consists of the contiguous nucleotide sequence.

In some embodiments the antisense oligonucleotide is the contiguous nucleotide sequence.

In some embodiments, the contiguous nucleotide sequence is fully complementary to a nucleotide sequence comprised within SEQ ID NO: 276.

In some embodiments, the contiguous nucleotide sequence is fully complementary to a nucleotide sequence comprised within nucleotides 441-468 of SEQ ID NO: 276.

In some embodiments, the contiguous nucleotide sequence is fully complementary to a nucleotide sequence comprised within nucleotides 441-462 of SEQ ID NO: 276. In some embodiments, the contiguous nucleotide sequence is fully complementary to a sequence selected from the group consisting of SEQ ID NO 277, SEQ ID NO 278, SEQ ID NO 279 and SEQ ID NO 280.

In some embodiments, the contiguous nucleotide sequence is fully complementary to SEQ ID NO

277.

In some embodiments, the contiguous nucleotide sequence is fully complementary to SEQ ID NO

278.

In some embodiments, the contiguous nucleotide sequence is fully complementary to SEQ ID NO

279.

In some embodiments, the contiguous nucleotide sequence is fully complementary to SEQ ID NO

280.

In some embodiments, the contiguous nucleotide sequence is fully complementary to a nucleotide sequence comprised within nucleotides 256-283 of SEQ ID NO: 276.

In some embodiments, the contiguous nucleotide sequence is fully complementary to SEQ ID NO

281.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 8 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 9 contiguous nucleotides thereof. In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 10 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 11 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 12 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 13 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 14 contiguous nucleotides thereof.

In some embodiments, the contiguous nucleotide sequence is a sequence selected from the group consisting of SEQ ID NO 51, SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252, or at least 15 contiguous nucleotides thereof. In some embodiments the contiguous nucleotide sequence is selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252.

In some embodiments the contiguous nucleotide sequence is SEQ ID NO 71.

In some embodiments the contiguous nucleotide sequence is SEQ ID NO 73.

In some embodiments the contiguous nucleotide sequence is SEQ ID NO 74.

In some embodiments the contiguous nucleotide sequence is SEQ ID NO 75.

In some embodiments the contiguous nucleotide sequence is SEQ ID NO 134.

The invention provides an antisense oligonucleotide, wherein the antisense oligonucleotide is 8 — 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length, wherein the contiguous nucleotide sequence is selected from the group consisting of : SEQ ID NO: 289 and SEQ ID NO: 290.

The invention provides for an antisense oligonucleotide which is isolated, purified or manufactured.

In some embodiments, the antisense oligonucleotide is or comprises an antisense oligonucleotide mixmer or totalmer. In some embodiments, the contiguous nucleotide sequence is a mixmer or a tolalmer.

The invention provides for a conjugate comprising the antisense oligonucleotide according to the invention, and at least one conjugate moiety covalently attached to said antisense oligonucleotide.

The invention provides an antisense oligonucleotide covalently attached to at least one conjugate moiety.

The invention provides for a pharmaceutically acceptable salt of the antisense oligonucleotide according to the invention, or the conjugate according to the invention. The invention provides for an antisense oligonucleotide according to the invention wherein the antisense oligonucleotide is in the form of a pharmaceutically acceptable salt. In some embodiments the pharmaceutically acceptable salt may be a sodium salt, a potassium salt or an ammonium salt.

The invention provides for a pharmaceutically acceptable sodium salt of the antisense oligonucleotide according to the invention, or the conjugate according to the invention.

The invention provides for a pharmaceutically acceptable potassium salt of the antisense oligonucleotide according to the invention, or the conjugate according to the invention.

The invention provides for a pharmaceutically acceptable ammonium salt of the antisense oligonucleotide according to the invention, or the conjugate according to the invention.

The invention provides for a pharmaceutical composition comprising the antisense oligonucleotide of the invention, or the conjugate of the invention, and a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant.

The invention provides for a pharmaceutical composition comprising the antisense oligonucleotide of the invention, or the conjugate of the invention, and a pharmaceutically acceptable salt. For example, the salt may comprise a metal cation, such as a sodium salt, a potassium salt or an ammonium salt.

The invention provides for a pharmaceutical composition according to the invention, wherein the pharmaceutical composition comprises the antisense oligonucleotide of the invention or the conjugate of the invention, or the pharmaceutically acceptable salt of the invention; and an aqueous diluent or solvent.

The invention provides for a solution, such as a phosphate buffered saline solution of the antisense oligonucleotide of the invention, or the conjugate of the invention, or the pharmaceutically acceptable salt of the invention. Suitably the solution, such as phosphate buffered saline solution, of the invention, is a sterile solution. The invention provides for a method for enhancing the expression of the Exon1-Exon2 progranulin splice variant in a cell which is expressing progranulin, said method comprising administering an antisense oligonucleotide of the invention, or a conjugate of the invention, or a salt of the invention, or a pharmaceutical composition of the invention in an effective amount to said cell. In some embodiments the method is an in vitro method. In some embodiments the method is an in vivo method.

In some embodiments, the cell is either a human cell or a mammalian cell.

The invention provides for a method for treating or preventing progranulin haploinsufficiency or a related disorder, comprising administering a therapeutically or prophylactically effective amount of an antisense oligonucleotide of the invention, or a conjugate of the invention, or a salt of the invention, or a pharmaceutical composition of the invention to a subject suffering from or susceptible to progranulin haploinsufficiency or a related disorder.

The invention provides for a method for treating or preventing neurological disease, comprising administering a therapeutically or prophylactically effective amount of an antisense oligonucleotide of the invention, or a conjugate of the invention, or a salt of the invention, or a pharmaceutical composition of the invention to a subject suffering from or susceptible to neurological disease. In one embodiment the neurological disease may be a TDP-43 pathology.

The invention provides for an antisense oligonucleotide of the invention, for use as a medicament.

The invention provides for an antisense oligonucleotide of the invention, for use in therapy.

The invention provides for the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention, for use as a medicament.

The invention provides the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention for use in therapy.

The invention provides for the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention for use in the treatment of a neurological disease. In one embodiment the neurological disease may be a TDP-43 pathology.

The invention provides for the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention for use in the treatment or prevention of progranulin haploinsufficiency or a related disorder.

The invention provides for the use of the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention, for the preparation of a medicament for treatment or prevention of a neurological disease. In one embodiment the neurological disease may be a TDP-43 pathology.

The invention provides for the use of the antisense oligonucleotide of the invention or the conjugate of the invention, or the salt of the invention, or the pharmaceutical composition of the invention, for the preparation of a medicament for treatment or prevention of progranulin haploinsufficiency or a related disorder.

In some embodiments the method, use, or antisense oligonucleotide for use, of the invention is for the treatment of fronto temporal dementia (FTD), neuropathologic frontotemporal lobar degeneration or neuroinflammation. In other embodiments the method, use, or antisense oligonucleotide for use, of the invention is for the treatment of amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, Parkinson’s disease, Autism, Hippocampal sclerosis dementia, Down syndrome, Huntington’s disease, polyglutamine diseases, spinocerebellar ataxia 3, myopathies or Chronic Traumatic Encephalopathy.

In one aspect the invention includes an oligonucleotide progranulin agonist having the structure corresponding to SEQ ID NO 71: In another aspect the invention includes an oligonucleotide progranulin agonist having the structure corresponding to SEQ ID NO 73: In another aspect the invention includes an oligonucleotide progranulin agonist having the structure corresponding to SEQ ID NO 74: In another aspect the invention includes an oligonucleotide progranulin agonist having the structure corresponding to SEQ ID NO 75: In another aspect the invention includes an oligonucleotide progranulin agonist having the structure corresponding to SEQ ID NO 134: In another aspect the invention includes an antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GaGctGggTcAagAAT (SEQ ID NO: 71) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

In another aspect the invention includes an antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GgtCaaGaAtgGtgTG (SEQ ID NO: 73) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

In another aspect the invention includes an antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound CaGaAtGgtGtGgTC (SEQ ID NO 74) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

In another aspect the invention includes an antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GaAtGgtGtGgTccC (SEQ ID NO 75) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

In another aspect the invention includes an antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound CtcAagCtcAcAtgGC (SEQ ID NO 134) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

BRIEF DESCRIPTION OF FIGURES

Figure 1 shows expression levels of the Exon1-Exon2 mRNA splice form of progranulin relative to HPRT1 and Intron1-Exon2 relative to HPRT1. SEQ ID NOs: 1-49 are shown in Figure 1a. SEQ ID NOs: 50-109 are shown in Figure 1b. SEQ ID NOs: 110-169 are shown in Figure 1c. SEQ ID NOs: 170-229 are shown in Figure 1d. SEQ ID Nos: 230-275 are shown in Figure 1e. Figure 2 shows progranulin expression levels following treatment with an oligonucleotide for 5 days.

Figure 3 shows progranulin expression levels following treatment with an oligonucleotide for 5 days compared to oligonucleotides S7 and S10 from WO 2020/191212.

Figure 4 shows sequence localization of SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75 and SEQ ID NO: 134.

Figure 5 shows progranulin expression levels following treatment with an oligonucleotide for 4 days.

Figure 6 shows progranulin expression levels following treatment with an oligonucleotide for 4 days compared to oligonucleotides S7, S10 and S37 from WO 2020/191212.

Figure 7 shows ddPCR data quantifying the abundance of the 5 UTR splice variants in GRN mRNA 48h after transfection in H4 cells relative to Mock transfected cells. Grey bars quantify the abundance of the splice variant with retention of intron 1 (Int1-Ex2) and the black bars the splice variant with the splicing of Ex1- Ex2 (Ex1-Ex2). SEQ ID#73 (Figure 7a), SEQ ID#74 (Figure 7b) and SEQ ID#75 (Figure 7c) show dose-dependent skipping of intronl retention (Int1-Ex2) and an increase in Ex1-Ex2 splice-variant. The S10 compound from WO 2020/191212 (Figure 7d) shows no/limited effects on skipping of intronl retention.

Figure 8 shows ddPCR data quantifying the abundance of the 5 UTR splice variants in GRN mRNA 48h after transfection in H4 cells relative to Mock transfected cells. Grey bars quantify the abundance of the splice variant with retention of intronl (Int1-Ex2) and the black bars the splice variant with the splicing of Ex1- Ex2 (Ex1-Ex2). SEQ ID#289 (Figure 8a) and SEQ ID#290 (Figure 8b) show dose-dependent skipping of intronl retention (Int1-Ex2) and an increase in Ex1-Ex2 splice-variant. The S10 compound from WO 2020/191212 showed no/limited effects on skipping of intronl retention (Figure 8c).

Figure 9 shows ddPCR data quantifying the abundance of the 5 UTR splice variants in GRN mRNA after 5 days gymnosis in Microglia cells relative to PBS transfected cells. Grey bars quantify the abundance of the splice variant with retention of intronl (Intron 1 retention) and the black bars the splice variant with the splicing of Exonl- Exon 2 (exon1-exon2). SEQ ID#290 showed dosedependent skipping of intronl retention and an increase in Exon1-Exon2 splice-variant. The S10 compound from WO 2020/191212 showed no/limited effects on skipping of intronl retention. The gapmer control show the expected dose-dependent knockdown of both splice variants.

Figure 10 shows a Sashimi plot corresponding to the splice-switch occurring with compound ID#290, and not occurring with compound S10 according to WO 2020/191912.

DEFINITIONS

OLIGONUCLEOTIDE

The term “oligonucleotide” as used herein is defined as it is generally understood by the skilled person as a molecule comprising two or more covalently linked nucleosides. Such covalently bound nucleosides may also be referred to as nucleic acid molecules or oligomers.

Oligonucleotides are commonly made in the laboratory by solid-phase chemical synthesis followed by purification and isolation. When referring to a sequence of the oligonucleotide, reference is made to the sequence or order of nucleobase moieties, or modifications thereof, of the covalently linked nucleotides or nucleosides. The oligonucleotides of the invention are man-made, and are chemically synthesized, and are typically purified or isolated. The oligonucleotides of the invention may comprise one or more modified nucleosides such as 2’ sugar modified nucleosides. The oligonucleotides of the invention may comprise one or more modified internucleoside linkages, such as one or more phosphorothioate internucleoside linkages.

ANTISENSE OLIGONUCLEOTIDE

The term “antisense oligonucleotide” as used herein is defined as an oligonucleotide capable of modulating expression of a target gene by hybridizing to a target nucleic acid, in particular to a contiguous sequence on a target nucleic acid. Antisense oligonucleotides are not essentially double stranded and are therefore not siRNAs or shRNAs. The antisense oligonucleotides of the present invention may be single stranded. It is understood that single stranded oligonucleotides of the present invention can form hairpins or intermolecular duplex structures (duplex between two molecules of the same oligonucleotide), as long as the degree of intra or inter self-complementarity is less than approximately 50% across of the full length of the oligonucleotide.

In certain contexts the antisense oligonucleotides of the invention may be referred to as oligonucleotides. In some embodiments, the single stranded antisense oligonucleotides of the invention may not contain RNA nucleosides.

Advantageously, the antisense oligonucleotides of the invention comprise one or more modified nucleosides or nucleotides, such as 2’ sugar modified nucleosides. Furthermore, in some antisense oligonucleotides of the invention, it may be advantageous that the nucleosides which are not modified are DNA nucleosides.

CONTIGUOUS NUCLEOTIDE SEQUENCE

The term “contiguous nucleotide sequence” refers to the region of the oligonucleotide which is complementary to a target nucleic acid, which may be or may comprise an oligonucleotide motif sequence. The term is used interchangeably herein with the term “contiguous nucleobase sequence”. In some embodiments all the nucleosides of the oligonucleotide constitute the contiguous nucleotide sequence. The contiguous nucleotide sequence is the sequence of nucleotides in the oligonucleotide of the invention which is complementary to, and in some instances fully complementary to, the target nucleic acid or target sequence, or target site sequence.

In some embodiments the target sequence is SEQ ID NO 276.

SEQ ID NO 276 is the sequence of exon 1 , intron 1 and exon 2 of the human progranulin pre- mRNA transcript.

In some embodiments the target sequence is or comprises nucleotides 441-468 of SEQ ID NO 276.

In some embodiments the target sequence is or comprises nucleotides 441-462 of SEQ ID NO 276.

In some embodiments the target sequence is or comprises SEQ ID NO 277.

In some embodiments the target sequence is or comprises SEQ ID NO 278

In some embodiments the target sequence is or comprises SEQ ID NO 279.

In some embodiments the target sequence is or comprises SEQ ID NO 280. In some embodiments the target sequence is or comprises nucleotides 268-283 of SEQ ID NO 276.

In some embodiments the target sequence is or comprises SEQ ID NO 281.

In some embodiments the target sequence is or comprises SEQ ID NO 291.

In some embodiments the target sequence is or comprises SEQ ID NO 292.

In some embodiments the oligonucleotide comprises the contiguous nucleotide sequence, and may optionally comprise further nucleotide(s), for example a nucleotide linker region which may be used to attach a functional group (e.g. a conjugate group) to the contiguous nucleotide sequence. The nucleotide linker region may or may not be complementary to the target nucleic acid. It is understood that the contiguous nucleotide sequence of the oligonucleotide cannot be longer than the oligonucleotide as such and that the oligonucleotide cannot be shorter than the contiguous nucleotide sequence.

NUCLEOTIDES AND NUCLEOSIDES

Nucleotides and nucleosides are the building blocks of oligonucleotides and polynucleotides, and for the purposes of the present invention include both naturally occurring and non-naturally occurring nucleotides and nucleosides. In nature, nucleotides, such as DNA and RNA nucleotides comprise a ribose sugar moiety, a nucleobase moiety and one or more phosphate groups (which is absent in nucleosides). Nucleosides and nucleotides may also interchangeably be referred to as “units” or “monomers”.

MODIFIED NUCLEOTIDE

Advantageously, the antisense oligonucleotide of the invention may comprise one or more modified nucleosides.

The term “modified nucleoside” or “nucleoside modification” as used herein refers to nucleosides modified as compared to the equivalent DNA or RNA nucleoside by the introduction of one or more modifications of the sugar moiety or the (nucleo)base moiety. Advantageously, one or more of the modified nucleosides of the antisense oligonucleotides of the invention may comprise a modified sugar moiety. The term modified nucleoside may also be used herein interchangeably with the term “nucleoside analogue” or modified “units” or modified “monomers”. Nucleosides with an unmodified DNA or RNA sugar moiety are termed DNA or RNA nucleosides herein. Nucleosides with modifications in the base region of the DNA or RNA nucleoside are still generally termed DNA or RNA if they allow Watson Crick base pairing. Exemplary modified nucleosides which may be used in the antisense oligonucleotides of the invention include LNA, 2’-O-MOE and morpholino nucleoside analogues.

MODIFIED INTERNUCLEOSIDE LINKAGE

Advantageously, the antisense oligonucleotide of the invention comprises one or more modified internucleoside linkage.

The term “modified internucleoside linkage” is defined as generally understood by the skilled person as linkages other than phosphodiester (PO) linkages, that covalently couple two nucleosides together. The antisense oligonucleotides of the invention may therefore comprise one or more modified internucleoside linkages such as one or more phosphorothioate internucleoside linkages.

In some embodiments at least 50% of the internucleoside linkages in the antisense oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate, such as at least 60%, such as at least 70%, such as at least 75%, such as at least 80%, such as at least 90% or more of the internucleoside linkages in the antisense oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate. In some embodiments all of the internucleoside linkages of the antisense oligonucleotide, or contiguous nucleotide sequence thereof, are phosphorothioate.

Advantageously, all the internucleoside linkages of the contiguous nucleotide sequence of the antisense oligonucleotide may be phosphorothioate, or all the internucleoside linkages of the antisense oligonucleotide may be phosphorothioate linkages.

NUCLEOBASE

The term nucleobase includes the purine (e.g. adenine and guanine) and pyrimidine (e.g. uracil, thymine and cytosine) moiety present in nucleosides and nucleotides which form hydrogen bonds in nucleic acid hybridization. In the context of the present invention the term nucleobase also encompasses modified nucleobases which may differ from naturally occurring nucleobases, but which are functional during nucleic acid hybridization. In this context “nucleobase” refers to both naturally occurring nucleobases such as adenine, guanine, cytosine, thymidine, uracil, xanthine and hypoxanthine, as well as non-naturally occurring variants. Such variants are for example described in Hirao et al. (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1.

In some embodiments the nucleobase moiety is modified by changing the purine or pyrimidine into a modified purine or pyrimidine, such as substituted purine or substituted pyrimidine, such as a nucleobase selected from isocytosine, pseudoisocytosine, 5-methyl cytosine, 5-thiozolo-cytosine, 5-propynyl-cytosine, 5-propynyl-uracil, 5-bromouracil 5-thiazolo-uracil, 2-thio-uracil, 2’thio-thymine, inosine, diaminopurine, 6-aminopurine, 2-aminopurine, 2,6-diaminopurine and 2-chloro-6- aminopurine.

The nucleobase moieties may be indicated by the letter code for each corresponding nucleobase, e.g. A, T, G, C or II, wherein each letter may optionally include modified nucleobases of equivalent function. For example, in the exemplified oligonucleotides, the nucleobase moieties are selected from A, T, G, C, and 5-methyl cytosine. Optionally, for LNA gapmers, 5-methyl cytosine LNA nucleosides may be used.

MODIFIED OLIGONUCLEOTIDE

The antisense oligonucleotide of the invention may be a modified oligonucleotide.

The term modified oligonucleotide describes an oligonucleotide comprising one or more sugar- modified nucleosides and/or modified internucleoside linkages. The term “chimeric oligonucleotide” is a term that has been used in the literature to describe oligonucleotides comprising sugar modified nucleosides and DNA nucleosides. In some embodiments, it may be advantageous for the antisense oligonucleotide of the invention to be a chimeric oligonucleotide.

Complementarity

The term “complementarity” describes the capacity for Watson-Crick base-pairing of nucleosides/nucleotides. Watson-Crick base pairs are guanine (G)-cytosine (C) and adenine (A) - thymine (T)/uracil (U).

It will be understood that oligonucleotides may comprise nucleosides with modified nucleobases, for example 5-methyl cytosine is often used in place of cytosine, and as such the term complementarity encompasses Watson Crick base-paring between non-modified and modified nucleobases (see for example Hirao et al (2012) Accounts of Chemical Research vol 45 page 2055 and Bergstrom (2009) Current Protocols in Nucleic Acid Chemistry Suppl. 37 1.4.1). The term “% complementary” as used herein, refers to the proportion of nucleotides (in percent) of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which across the contiguous nucleotide sequence, are complementary to a reference sequence (e.g. a target sequence or sequence motif). The percentage of complementarity is thus calculated by counting the number of aligned nucleobases that are complementary (from Watson Crick base pairs) between the two sequences (when aligned with the target sequence 5’-3’ and the oligonucleotide sequence from 3’-5’), dividing that number by the total number of nucleotides in the oligonucleotide and multiplying by 100. In such a comparison a nucleobase/nucleotide which does not align (form a base pair) is termed a mismatch. Insertions and deletions are not allowed in the calculation of % complementarity of a contiguous nucleotide sequence. It will be understood that in determining complementarity, chemical modifications of the nucleobases are disregarded as long as the functional capacity of the nucleobase to form Watson Crick base pairing is retained (e.g. 5’-methyl cytosine is considered identical to a cytosine for the purpose of calculating % identity).

Within the present invention the term “complementary” requires the antisense oligonucleotide to be at least about 80% complementary, or at least about 90% complementary, to a human progranulin pre-mRNA transcript. In some embodiments the antisense oligonucleotide may be at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98% or at least about 99% complementary to a human progranulin pre-mRNA transcript. Put another way, for some embodiments, an antisense oligonucleotide of the invention may include one, two, three or more mis-matches, wherein a mis-match is a nucleotide within the antisense oligonucleotide of the invention which does not base pair with its target.

The term “fully complementary” refers to 100% complementarity.

The antisense oligonucleotides of the invention are complementary to the human progranulin pre- mRNA. The antisense oligonucleotides of the invention are advantageously complementary to the intron 1 sequence of the human progranulin pre-mRNA transcript. The sequence of exon 1 , intron 1 and exon 2 of the human progranulin pre-mRNA transcript is exemplified herein as SEQ ID NO 276. SEQ ID NO 276 is provided herein as a reference sequence and it will be understood that the target progranulin nucleic acid may be an allelic variant of SEQ ID NO 276, such as an allelic variant which comprises one or more polymorphism in the human progranulin nucleic acid sequence.

Identity

The term “identity” as used herein, refers to the proportion of nucleotides (expressed in percent) of a contiguous nucleotide sequence in a nucleic acid molecule (e.g. oligonucleotide) which across the contiguous nucleotide sequence, are identical to a reference sequence (e.g. a sequence motif).

The percentage of identity is thus calculated by counting the number of aligned nucleobases that are identical (a Match) between two sequences (in the contiguous nucleotide sequence of the compound of the invention and in the reference sequence), dividing that number by the total number of nucleotides in the oligonucleotide and multiplying by 100. Therefore, Percentage of Identity = (Matches x 100)/Length of aligned region (e.g. the contiguous nucleotide sequence). Insertions and deletions are not allowed in the calculation the percentage of identity of a contiguous nucleotide sequence. It will be understood that in determining identity, chemical modifications of the nucleobases are disregarded as long as the functional capacity of the nucleobase to form Watson Crick base pairing is retained (e.g. 5-methyl cytosine is considered identical to a cytosine for the purpose of calculating % identity).

Hybridization

The terms “hybridizing” or “hybridizes” as used herein are to be understood as two nucleic acid strands (e.g. an antisense oligonucleotide and a target nucleic acid) forming hydrogen bonds between base pairs on opposite strands thereby forming a duplex. The affinity of the binding between two nucleic acid strands is the strength of the hybridization. It is often described in terms of the melting temperature (T m ) defined as the temperature at which half of the oligonucleotides are duplexed with the target nucleic acid. At physiological conditions T m is not strictly proportional to the affinity (Mergny and Lacroix, 2003, Oligonucleotides 13:515-537). The standard state Gibbs free energy AG° is a more accurate representation of binding affinity and is related to the dissociation constant (Kd) of the reaction by AG°=-RTIn(Kd), where R is the gas constant and T is the absolute temperature. Therefore, a very low AG° of the reaction between an oligonucleotide and the target nucleic acid reflects a strong hybridization between the oligonucleotide and target nucleic acid. AG° is the energy associated with a reaction where aqueous concentrations are 1M, the pH is 7, and the temperature is 37°C. The hybridization of oligonucleotides to a target nucleic acid is a spontaneous reaction and for spontaneous reactions AG° is less than zero. AG° can be measured experimentally, for example, by use of the isothermal titration calorimetry (ITC) method as described in Hansen et al., 1965, Chem. Comm. 36-38 and Holdgate et al., 2005, Drug Discov Today. The skilled person will know that commercial equipment is available for AG° measurements. AG° can also be estimated numerically by using the nearest neighbor model as described by SantaLucia, 1998, Proc Natl Acad Sci USA. 95: 1460-1465 using appropriately derived thermodynamic parameters described by Sugimoto et al., 1995, Biochemistry 34: 11211-11216 and McTigue et al., 2004, Biochemistry 43:5388-5405.

In some embodiments, antisense oligonucleotides of the present invention hybridize to a target nucleic acid with estimated AG° values below -10 kcal for oligonucleotides that are 10-30 nucleotides in length.

In some embodiments the degree or strength of hybridization is measured by the standard state Gibbs free energy AG°. The oligonucleotides may hybridize to a target nucleic acid with estimated AG° values below the range of -10 kcal, such as below -15 kcal, such as below -20 kcal and such as below -25 kcal for oligonucleotides that are 8-30 nucleotides in length. In some embodiments the oligonucleotides hybridize to a target nucleic acid with an estimated AG° value of -10 to -60 kcal, such as -12 to -40, such as from -15 to -30 kcal, or-16 to -27 kcal such as -18 to -25 kcal.

High affinity modified nucleosides

A high affinity modified nucleoside is a modified nucleotide which, when incorporated into the oligonucleotide enhances the affinity of the oligonucleotide for its complementary target, for example as measured by the melting temperature (T m ). A high affinity modified nucleoside of the present invention preferably results in an increase in melting temperature between +0.5 to +12°C, more preferably between +1.5 to +10°C and most preferably between+3 to +8°C per modified nucleoside. Numerous high affinity modified nucleosides are known in the art and include for example, many 2’ substituted nucleosides as well as locked nucleic acids (LNA) (see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213). Sugar modifications

The antisense oligonucleotides of the invention may comprise one or more nucleosides which have a modified sugar moiety, i.e. a modification of the sugar moiety when compared to the ribose sugar moiety found in DNA and RNA.

Numerous nucleosides with modification of the ribose sugar moiety have been made, primarily with the aim of improving certain properties of oligonucleotides, such as affinity and/or nuclease resistance.

Such modifications include those where the ribose ring structure is modified, e.g. by replacement with a hexose ring (HNA), or a bicyclic ring, which typically have a biradicle bridge between the C2 and C4 carbons on the ribose ring (LNA), or an unlinked ribose ring which typically lacks a bond between the C2 and C3 carbons (e.g. UNA). Other sugar modified nucleosides include, for example, bicyclohexose nucleic acids (WO2011/017521) or tricyclic nucleic acids (WO2013/154798). Modified nucleosides also include nucleosides where the sugar moiety is replaced with a non-sugar moiety, for example in the case of peptide nucleic acids (PNA), or morpholino nucleic acids.

Sugar modifications also include modifications made via altering the substituent groups on the ribose ring to groups other than hydrogen, or the 2’-OH group naturally found in DNA and RNA nucleosides. Substituents may, for example be introduced at the 2’, 3’, 4’ or 5’ positions.

2’ sugar modified nucleosides

A 2’ sugar modified nucleoside is a nucleoside which has a substituent other than H or -OH at the 2’ position (2’ substituted nucleoside) or comprises a 2’ linked biradicle capable of forming a bridge between the 2’ carbon and a second carbon in the ribose ring, such as LNA (2’ - 4’ biradicle bridged) nucleosides.

Indeed, much focus has been given to developing 2’ sugar substituted nucleosides, and numerous 2’ substituted nucleosides have been found to have beneficial properties when incorporated into oligonucleotides. For example, the 2’ modified sugar may provide enhanced binding affinity and/or increased nuclease resistance to the oligonucleotide. Examples of 2’ substituted modified nucleosides are 2’-O-alkyl-RNA, 2’-O-methyl-RNA, 2’-alkoxy-RNA, 2’-O-methoxyethyl-RNA (MOE), 2’-amino-DNA, 2’-Fluoro-RNA, and 2’-F-ANA nucleoside. For further examples, please see e.g. Freier & Altmann; Nucl. Acid Res., 1997, 25, 4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213, and Deleavey and Damha, Chemistry and Biology 2012, 19, 937. Below are illustrations of some 2’ substituted modified nucleosides.

In relation to the present invention 2’ substituted sugar modified nucleosides does not include 2’ bridged nucleosides like LNA.

Locked Nucleic Acid Nucleosides (LNA nucleoside)

A “LNA nucleoside” is a 2’- modified nucleoside which comprises a biradical linking the C2’ and C4’ of the ribose sugar ring of said nucleoside (also referred to as a “2’- 4’ bridge”), which restricts or locks the conformation of the ribose ring. These nucleosides are also termed bridged nucleic acid or bicyclic nucleic acid (BNA) in the literature. The locking of the conformation of the ribose is associated with an enhanced affinity of hybridization (duplex stabilization) when the LNA is incorporated into an oligonucleotide for a complementary RNA or DNA molecule. This can be routinely determined by measuring the melting temperature of the oligonucleotide/complement duplex.

Non limiting, exemplary LNA nucleosides are disclosed in WO 99/014226, WO 00/66604, WO 98/039352 , WO 2004/046160, WO 00/047599, WO 2007/134181, WO 2010/077578, WO 2010/036698, WO 2007/090071 , WO 2009/006478, WO 2011/156202, WO 2008/154401 , WO 2009/067647, WO 2008/150729, Morita et al., Bioorganic & Med.Chem. Lett. 12, 73-76, Seth et al. J. Org. Chem. 2010, Vol 75(5) pp. 1569-81 , and Mitsuoka et al., Nucleic Acids Research 2009, 37(4), 1225-1238, and Wan and Seth, J. Medical Chemistry 2016, 59, 9645-9667. Further non limiting, exemplary LNA nucleosides are disclosed in Scheme 1.

Scheme 1 :

Particular LNA nucleosides are beta-D-oxy-LNA, 6’-methyl-beta-D-oxy LNA such as (S)-6’-methyl- beta-D-oxy-LNA (ScET) and ENA.

A particularly advantageous LNA is beta-D-oxy-LNA.

Morpholino Oligonucleotides

In some embodiments, the antisense oligonucleotide of the invention comprises or consists of morpholino nucleosides (/.e. is a Morpholino oligomer and as a phosphorodiamidate Morpholino oligomer (PMO)). Splice modulating morpholino oligonucleotides have been approved for clinical use - see for example eteplirsen, a 30nt morpholino oligonucleotide targeting a frame shift mutation in DMD, used to treat Duchenne muscular dystrophy. Morpholino oligonucleotides have nucleobases attached to six membered morpholine rings rather ribose, such as methylenemorpholine rings linked through phosphorodiamidate groups, for example as illustrated by the following illustration of 4 consecutive morpholino nucleotides:

In some embodiments, morpholino oligonucleotides of the invention may be, for example 20 - 40 morpholino nucleotides in length, such as morpholino 25 - 35 nucleotides in length.

RNase H Activity and Recruitment

The RNase H activity of an antisense oligonucleotide refers to its ability to recruit RNase H when in a duplex with a complementary RNA molecule. WO01/23613 provides in vitro methods for determining RNaseH activity, which may be used to determine the ability to recruit RNaseH. Typically an oligonucleotide is deemed capable of recruiting RNase H if it, when provided with a complementary target nucleic acid sequence, has an initial rate, as measured in pmol/l/min, of at least 5%, such as at least 10%, at least 20% or more than 20%, of the initial rate determined when using an oligonucleotide having the same base sequence as the modified oligonucleotide being tested, but containing only DNA monomers with phosphorothioate linkages between all monomers in the oligonucleotide, and using the methodology provided by Examples 91 - 95 of WO01/23613 (hereby incorporated by reference). For use in determining RHase H activity, recombinant RNase H1 is available from Lubio Science GmbH, Lucerne, Switzerland.

DNA oligonucleotides are known to effectively recruit RNaseH, as are gapmer oligonucleotides which comprise a region of DNA nucleosides (typically at least 5 or 6 contiguous DNA nucleosides), flanked 5’ and 3’ by regions comprising 2’ sugar modified nucleosides, typically high affinity 2’ sugar modified nucleosides, such as 2-O-MOE and/or LNA. For effective modulation of splicing, degradation of the pre-mRNA is not desirable, and as such it is preferable to avoid the RNaseH degradation of the target. Therefore, the antisense oligonucleotides of the invention are not RNaseH recruiting gapmer oligonucleotide.

RNaseH recruitment may be avoided by limiting the number of contiguous DNA nucleotides in the oligonucleotide - therefore mimxes and totalmer designs may be used. Advantageously the antisense oligonucleotides of the invention, or the contiguous nucleotide sequence thereof, do not comprise more than 3 contiguous DNA nucleosides. Further, advantageously the antisense oligonucleotides of the invention, or the contiguous nucleotide sequence thereof, do not comprise more than 4 contiguous DNA nucleosides. Further advantageously, the antisense oligonucleotides of the invention, or contiguous nucleotide sequence thereof, do not comprise more than 2 contiguous DNA nucleosides.

Mixmers and Totalmers

For splice modulation it is often advantageous to use antisense oligonucleotides which do not recruit RNAaseH. As RNaseH activity requires a contiguous sequence of DNA nucleotides, RNaseH activity of antisense oligonucleotides may be achieved by designing antisense oligonucleotides which do not comprise a region of more than 3 or more than 4 contiguous DNA nucleosides. This may be achieved by using antisense oligonucleotides or contiguous nucleoside regions thereof with a mixmer design, which comprise sugar modified nucleosides, such as 2’ sugar modified nucleosides, and short regions of DNA nucleosides, such as 1 , 2 or 3 DNA nucleosides. Mixmers are exemplified herein by every second design, wherein the nucleosides alternate between 1 LNA and 1 DNA nucleoside, e.g. LDLDLDLDLDLDLDLL, with 5’ and 3’ terminal LNA nucleosides, and every third design, such as LDDLDDLDDLDDLDDL, where every third nucleoside is a LNA nucleoside. A totalmer is an antisense oligonucleotide or a contiguous nucleotide sequence thereof which does not comprise DNA or RNA nucleosides, and may for example comprise only 2’-O-MOE nucleosides, such as a fully MOE phosphorothioate, e.g. MMMMMMMMMMMMMMMMMMMM, where M = 2’-O-MOE, which are reported to be effective splice modulators for therapeutic use.

Alternatively, a mixmer may comprise a mixture of modified nucleosides, such as MLMLMLMLMLMLMLMLMLML, wherein L = LNA and M = 2’-O-MOE nucleosides. Advantageously, the internucleoside nucleosides in mixmers and totalmers may be phosphorothioate, or a majority of nucleoside linkages in mixmers may be phosphorothioate. Mixmers and totalmers may comprise other internucleoside linkages, such as phosphodiester or phosphorodithioate, by way of example.

Region D’ or D” in an oligonucleotide

The antisense oligonucleotide of the invention may in some embodiments comprise or consist of the contiguous nucleotide sequence of the oligonucleotide which is complementary to the target nucleic acid, such as a mixmer or toalmer region, and further 5’ and/or 3’ nucleosides. The further 5’ and/or 3’ nucleosides may or may not be complementary, such as fully complementary, to the target nucleic acid. Such further 5’ and/or 3’ nucleosides may be referred to as region D’ and D” herein.

The addition of region D’ or D” may be used for the purpose of joining the contiguous nucleotide sequence, such as the mixmer or totoalmer, to a conjugate moiety or another functional group. When used for joining the contiguous nucleotide sequence with a conjugate moiety it can serve as a biocleavable linker. Alternatively, it may be used to provide exonucleoase protection or for ease of synthesis or manufacture.

Region D’ or D” may independently comprise or consist of 1 , 2, 3, 4 or 5 additional nucleotides, which may be complementary or non-complementary to the target nucleic acid. The nucleotide adjacent to the F or F’ region is not a sugar-modified nucleotide, such as a DNA or RNA or base modified versions of these. The D’ or D” region may serve as a nuclease susceptible biocleavable linker (see definition of linkers). In some embodiments the additional 5’ and/or 3’ end nucleotides are linked with phosphodiester linkages, and are DNA or RNA. Nucleotide based biocleavable linkers suitable for use as region D’ or D” are disclosed in WO2014/076195, which include by way of example a phosphodiester linked DNA dinucleotide. The use of biocleavable linkers in poly- oligonucleotide constructs is disclosed in WO2015/113922, where they are used to link multiple antisense constructs within a single oligonucleotide.

In one embodiment the antisense oligonucleotide of the invention comprises a region D’ and/or D” in addition to the contiguous nucleotide sequence which constitutes a mixmer or a totalmer.

In some embodiments the internucleoside linkage positioned between region D’ or D” and the mixmer or totalmer region is a phosphodiester linkage.

Conjugate

The invention encompasses an antisense oligonucleotide covalently attached to at least one conjugate moiety. In some embodiments this may be referred to as a conjugate of the invention.

The term “conjugate” as used herein refers to an antisense oligonucleotide which is covalently linked to a non-nucleotide moiety (conjugate moiety or region C or third region). The conjugate moiety may be covalently linked to the antisense oligonucleotide, optionally via a linker group, such as region D’ or D”.

Oligonucleotide conjugates and their synthesis has also been reported in comprehensive reviews by Manoharan in Antisense Drug Technology, Principles, Strategies, and Applications, S.T. Crooke, ed., Ch. 16, Marcel Dekker, Inc., 2001 and Manoharan, Antisense and Nucleic Acid Drug Development, 2002, 12, 103.

In some embodiments, the non-nucleotide moiety (conjugate moiety) is selected from the group consisting of carbohydrates (e.g. GalNAc), cell surface receptor ligands, drug substances, hormones, lipophilic substances, polymers, proteins, peptides, toxins (e.g. bacterial toxins), vitamins, viral proteins (e.g. capsids) or combinations thereof.

Linkers

A linkage or linker is a connection between two atoms that links one chemical group or segment of interest to another chemical group or segment of interest via one or more covalent bonds. Conjugate moieties can be attached to the antisense oligonucleotide directly or through a linking moiety (e.g. linker or tether). Linkers serve to covalently connect a third region, e.g. a conjugate moiety (Region C), to a first region, e.g. an oligonucleotide or contiguous nucleotide sequence complementary to the target nucleic acid (region A). In some embodiments of the invention the conjugate or antisense oligonucleotide conjugate of the invention may optionally comprise a linker region (second region or region B and/or region Y) which is positioned between the oligonucleotide or contiguous nucleotide sequence complementary to the target nucleic acid (region A or first region) and the conjugate moiety (region C or third region).

Region B refers to biocleavable linkers comprising or consisting of a physiologically labile bond that is cleavable under conditions normally encountered or analogous to those encountered within a mammalian body. Conditions under which physiologically labile linkers undergo chemical transformation (e.g., cleavage) include chemical conditions such as pH, temperature, oxidative or reductive conditions or agents, and salt concentration found in or analogous to those encountered in mammalian cells. Mammalian intracellular conditions also include the presence of enzymatic activity normally present in a mammalian cell such as from proteolytic enzymes or hydrolytic enzymes or nucleases. In one embodiment the biocleavable linker is susceptible to S1 nuclease cleavage. In some embodiments the nuclease susceptible linker comprises between 1 and 5 nucleosides, such as DNA nucleoside(s) comprising at least two consecutive phosphodiester linkages. Phosphodiester containing biocleavable linkers are described in more detail in WO 2014/076195.

Region Y refers to linkers that are not necessarily biocleavable but primarily serve to covalently connect a conjugate moiety (region C or third region), to an oligonucleotide (region A or first region). The region Y linkers may comprise a chain structure or an oligomer of repeating units such as ethylene glycol, amino acid units or amino alkyl groups. The antisense oligonucleotide conjugates of the present invention can be constructed of the following regional elements A-C, A- B-C, A-B-Y-C, A-Y-B-C or A-Y-C. In some embodiments the linker (region Y) is an amino alkyl, such as a C2 - C36 amino alkyl group, including, for example C6 to C12 amino alkyl groups. In some embodiments the linker (region Y) is a C6 amino alkyl group.

TREATMENT

The term ’treatment’ as used herein refers to both treatment of an existing disease (e.g. a disease or disorder as herein referred to), or prevention of a disease, i.e. prophylaxis. It will therefore be recognized that treatment as referred to herein may, in some embodiments, be prophylactic. TDP-43 Pathologies

A TDP-43 pathology is a disease which is associated with reduced or aberrant expression of TDP- 43, often associated with an increase in cytoplasmic TDP-43, particularly hyper-phosphorylated and ubiquitinated TDP-43.

Diseases associated with TDP-43 pathology include amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), Alzheimer’s disease, Parkinson’s disease, Autism, Hippocampal sclerosis dementia, Down syndrome, Huntington’s disease, polyglutamine diseases, such as spinocerebellar ataxia 3, myopathies and Chronic Traumatic Encephalopathy.

DETAILED DESCRIPTION OF THE INVENTION

The inventors have identified that targeting the progranulin pre-mRNA transcript with antisense oligonucleotides can increase expression of the progranulin Exon1-Exon 2 spliced mRNA, decrease expression of the progranulin Intron1-Exon2 spliced mRNA (which retains the 271 nucleotide 5’ fragment of intron 1) and/or alter the ratio of Exon1-Exon2 vs Intronl- Exon2 mRNA. This is particularly the case when antisense oligonucleotides which comprise high affinity sugar modified nucleosides, such as high affinity 2’ sugar modified nucleosides, such as LNA nucleosides or 2’-O-methoxyethyl (MOE) nucleosides are used.

Described herein are target sites present on the human progranulin pre-mRNA which can be targeted by antisense oligonucleotides. Also described are antisense oligonucleotides which are complementary, such as fully complementary, to these target sites.

Without wishing to be bound by theory, it is considered that the antisense oligonucleotides of the invention can increase expression of the progranulin Exon1-Exon2 spliced mRNA, decrease expression of the progranulin Intron1-Exon1 spliced mRNA and/or alter the ratio of Exon1-Exon2 vs Intronl- Exon2 mRNA by binding to these regions and affecting, such as increasing, production of the Exon1-Exon2 splice variant.

Oligonucleotides, such as RNaseH recruiting single stranded antisense oligonucleotides or siRNAs are used extensively in the art to inhibit target RNAs - i.e. are used as antagonists of their complementary nucleic acid target. The antisense oligonucleotides of the present invention may be described as modulators, i.e. they alter the expression of a particular splice variant of their complementary target, progranulin pre- mRNA, and thereby increase the production of active progranulin protein.

Reduced expression of the progranulin Intron1-Exon2 splice variant is desirable because the inclusion of an intron, such as Intron 1 , within a mature mRNA sequence leads to nonsense- mediated mRNA decay (NMD).

Enhanced expression of the progranulin Exon1-Exon2 over the splice variant which retains the 5’ part of intron 1 is desirable because the Exon1-Exon2 splice variant does not include the 271 nucleotide fragment of intron 1 with two AUG sites upstream of the canonical downstream AUG in Exon 2 (open reading frame). Translation from these two upstream AUG sites will not encode the progranulin protein and due to premature termination codons the transcript may undergo nonsense mediated mRNA decay (NMD). Changing the splicing to the Exon1-Exon2 splice variant will instead lead to translation of an active version of the progranulin protein. Progranulin is a neuroprotective protein, and increasing its production can be used to treat a range of neurological disorders, such as TDP-43 pathologies.

In certain embodiments the antisense oligonucleotides of the present invention may enhance the production of the Exon1-Exon2 progranulin splice variant.

In certain embodiments the antisense oligonucleotides of the present invention may enhance the production of the Exon1-Exon2 progranulin splice variant mRNA by at least about 10% relative to the production of the Exon1-Exon2 progranulin splice variant mRNA in the absence of an antisense oligonucleotide of the invention. In other embodiments the antisense oligonucleotides of the present invention may enhance the production of the Exon1-Exon2 progranulin splice variant mRNA by at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 200%, at least about 300%, at least about 400%, at least about 500% or more relative to the production of the Exon1-Exon2 progranulin splice variant mRNA in the absence of an antisense oligonucleotide of the invention.

In certain embodiments the antisense oligonucleotides of the present invention may reduce the production of the Intron1-Exon2 progranulin splice variant mRNA. In certain embodiments the antisense oligonucleotides of the present invention may reduce the production of the Intron1-Exon2 progranulin splice variant mRNA by at least about 10% relative to the production of the Intron1-Exon2 progranulin splice variant mRNA in the absence of an antisense oligonucleotide of the invention. In other embodiments the antisense oligonucleotides of the present invention may reduce the production of the Intron1-Exon2 progranulin splice variant mRNA by at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 200%, at least about 300%, at least about 400%, at least about 500% or more relative to the production of the Intron1-Exon2 progranulin splice variant mRNA in the absence of an antisense oligonucleotide of the invention.

Enhanced expression of the progranulin Exon1-Exon2 splice variant should lead to translation of an active version of the progranulin protein. In certain embodiments the antisense oligonucleotides of the present invention may increase production of the progranulin protein by at least about 10% relative to the production of the progranulin protein in the absence of an antisense oligonucleotide of the invention. In other embodiments the antisense oligonucleotides of the present invention may increase the production of the progranulin protein by at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 200%, at least about 300%, at least about 400%, at least about 500% or more relative to the production of the progranulin protein in the absence of an antisense oligonucleotide of the invention.

In certain embodiments, the antisense oligonucleotides of the present invention may alter the ratio of Exon 1-Exon2 vs Intron-Exon2 progranulin mRNA.

In certain embodiments, the antisense oligonucleotides of the present invention may alter the ratio of Exon1-Exon2 vs Intron1-Exon2 progranulin mRNA by at least about 10% relative to the ratio of Exon1-Exon2 vs Intron1-Exon2 progranulin mRNA in the absence of an antisense oligonucleotide of the invention. In other embodiments the antisense oligonucleotides of the present invention may alter the ratio of Exon1-Exon2 vs Intron1-Exon2 progranulin mRNA by at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100% or more, relative to the ratio of Exon1-Exon2 vs Intronl- Exon2 progranulin mRNA in the absence of an antisense oligonucleotide of the invention.

In certain embodiments, the antisense oligonucleotides of the present invention may alter the ratio of Exon1-Exon2 vs Intron1-Exon2 progranulin mRNA to at least about 1.2. In certain embodiments, the antisense oligonucleotides of the present invention may alter the ratio of Exon1-Exon2 vs Intron1-Exon2 progranulin mRNA to at least about 1.3, at least about 1.4, at least about 1.5, at least about 1.6, at least about 1.7, at least about 1.8, at least about 1.9, at least about 2.0 or more.

In some embodiments, the antisense oligonucleotides of the invention or the contiguous nucleotide sequence thereof comprises or consists of 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39 or 40 contiguous nucleotides in length.

In some embodiments, the entire nucleotide sequence of the antisense oligonucleotide is the contiguous nucleotide sequence.

In one embodiment the contiguous nucleotide sequence may a sequence selected from the group consisting of SEQ ID NO 71 , SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75 and SEQ ID NO 134. The invention also contemplates fragments of these contiguous nucleotide sequences, including fragments of at least 8, at least 9, at least 10, at least 11 , at least 12, at least 13, at least 14, or at least 15 contiguous nucleotides thereof.

In some embodiments, the antisense oligonucleotide or contiguous nucleotide sequence comprises or consists of a sequence selected from the group consisting of SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75 and SEQ ID NO 134. It will be understood that the sequences shown in SEQ ID NO 71, SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75 and SEQ ID NO 134 may include modified nucleobases which function as the shown nucleobase in base pairing, for example 5-methyl cytosine may be used in place of methyl cytosine. Inosine may be used as a universal base.

In some embodiments, the antisense oligonucleotide or contiguous nucleotide sequence comprises or consists of 8 to 30 or 8 to 40 nucleotides in length with at least 90% identity, preferably 100% identity, to a sequence selected from the group consisting of SEQ ID NO 71 , SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75 and SEQ ID NO 134. In some embodiments the antisense oligonucleotide may be 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length.

In some embodiments, the antisense oligonucleotide or contiguous nucleotide sequence comprises or consists of 8 to 30 or 8 to 40 nucleotides in length with at least 90% identity, preferably 100% identity, to a sequence selected from the group consisting of SEQ ID NO 289 and SEQ ID NO 290. In some embodiments the antisense oligonucleotide may be 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length.

It is understood that the contiguous nucleobase sequences (motif sequence) can be modified to, for example, increase nuclease resistance and/or binding affinity to the target nucleic acid.

The pattern in which the modified nucleosides (such as high affinity modified nucleosides) are incorporated into the oligonucleotide sequence is generally termed oligonucleotide design.

The antisense oligonucleotides of the invention are designed with modified nucleosides and DNA nucleosides. Advantageously, high affinity modified nucleosides are used.

In an embodiment, the antisense oligonucleotide comprises at least 1 modified nucleoside, such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11 , at least 12, at least 13, at least 14, at least 15 or at least 16 modified nucleosides.

In an embodiment the antisense oligonucleotide comprises from 1 to 10 modified nucleosides, such as from 2 to 9 modified nucleosides, such as from 3 to 8 modified nucleosides, such as from 4 to 7 modified nucleosides, such as 6 or 7 modified nucleosides. Suitable modifications are described in the “Definitions” section under “modified nucleoside”, “high affinity modified nucleosides”, “sugar modifications”, “2’ sugar modifications” and Locked nucleic acids (LNA)”.

In an embodiment, the antisense oligonucleotide comprises one or more sugar modified nucleosides, such as 2’ sugar modified nucleosides. Preferably the antisense oligonucleotide of the invention comprises one or more 2’ sugar modified nucleoside independently selected from the group consisting of 2’-O-alkyl-RNA, 2’-O-methyl-RNA, 2’-alkoxy-RNA, 2’-O-methoxyethyl-RNA, 2’- amino-DNA, 2’-fluoro-DNA, arabino nucleic acid (ANA), 2’-fluoro-ANA and LNA nucleosides. It is advantageous if one or more of the modified nucleoside(s) is a locked nucleic acid (LNA). In a further embodiment the antisense oligonucleotide comprises at least one modified internucleoside linkage. Suitable internucleoside modifications are described in the “Definitions” section under “Modified internucleoside linkage”. It is advantageous if at least 75%, such as all, the internucleoside linkages within the contiguous nucleotide sequence are phosphorothioate or boranophosphate internucleoside linkages. In some embodiments all the internucleotide linkages in the contiguous sequence of the oligonucleotide are phosphorothioate linkages.

NUMBERED EMBODIMENTS OF THE INVENTION

1. An antisense oligonucleotide, wherein the antisense oligonucleotide is 8 - 40 nucleotides in length and comprises a contiguous nucleotide sequence of 8 - 40 nucleotides in length which is complementary to a splice regulation site of the human progranulin pre-mRNA transcript.

2. The antisense oligonucleotide according to embodiment 1 , wherein the human progranulin pre-mRNA transcript comprises the exon 1 , intron 1 and exon 2 sequence of the human progranulin pre-mRNA transcript (SEQ ID NO 276).

3. The antisense oligonucleotide according to embodiment 1 or embodiment 2, wherein the contiguous nucleotide sequence is of a length of at least 12 nucleotides in length.

4. The antisense oligonucleotide according to embodiment 3, wherein the contiguous nucleotide sequence is 12 - 16 nucleotides in length.

5. The antisense oligonucleotide according to embodiment 3, wherein the contiguous nucleotide sequence is 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39 or 40 nucleotides in length.

6. The antisense oligonucleotide according to any one of embodiments 1-5, wherein the contiguous nucleotide sequence is the same length as the antisense oligonucleotide.

7. The antisense oligonucleotide according to any one of embodiments 1-6, wherein the contiguous nucleotide sequence is fully complementary to the human progranulin pre-mRNA transcript. 8. The antisense oligonucleotide according to any one of embodiments 1-7, wherein the contiguous nucleotide sequence is complementary to a nucleotide sequence comprised within nucleotides 441-468 of SEQ ID NO: 276.

9. The antisense oligonucleotide according to embodiment 8, wherein the contiguous nucleotide sequence is complementary to a nucleotide sequence comprised within nucleotides 441-462 of SEQ ID NO: 276.

10. The antisense oligonucleotide according to embodiment 9, wherein the contiguous nucleotide sequence is complementary to SEQ ID NO 277, SEQ ID NO 278, SEQ ID NO 279 or SEQ ID NO 280.

11. The antisense oligonucleotide according to any one of embodiments 1-10, wherein the contiguous nucleotide sequence is fully complementary to SEQ ID NO 277, SEQ ID NO 278, SEQ ID NO 279 or SEQ ID NO 280.

12. The antisense oligonucleotide according to any one of embodiments 1-11, wherein the contiguous nucleotide sequence is selected from SEQ ID NO 71 , SEQ ID NO 73, SEQ ID NO 74 and SEQ ID NO 75, or at least 8 or at least 10 contiguous nucleotides thereof.

13. The antisense oligonucleotide according to embodiment 12, wherein the contiguous nucleotide sequence SEQ ID NO 71.

14. The antisense oligonucleotide according to embodiment 12, wherein the contiguous nucleotide sequence SEQ ID NO 73.

15. The antisense oligonucleotide according to embodiment 12, wherein the contiguous nucleotide sequence SEQ ID NO 74.

16. The antisense oligonucleotide according to embodiment 12, wherein the contiguous nucleotide sequence SEQ ID NO 75.

17. The antisense oligonucleotide according to any one of embodiments 1-7, wherein the contiguous nucleotide sequence is complementary to a nucleotide sequence comprised within nucleotides 268-283 of SEQ ID NO: 276. 18. The antisense oligonucleotide according to embodiment 17, wherein the contiguous nucleotide sequence is complementary to SEQ ID NO 281.

19. The antisense oligonucleotide according to embodiment 17 or embodiment 18, wherein the contiguous nucleotide sequence is fully complementary to SEQ ID NO 281.

20. The antisense oligonucleotide according to any one of embodiments 17-19, wherein the contiguous nucleotide sequence is SEQ ID NO 134, or at least 8 or at least 10 contiguous nucleotides thereof.

21. The antisense oligonucleotide according to any one of embodiments 1-7, wherein the contiguous nucleotide sequence is selected from the group consisting of SEQ ID NO 51 , SEQ ID NO 52, SEQ ID NO 53, SEQ ID NO 54, SEQ ID NO 55, SEQ ID NO 62, SEQ ID NO 63, SEQ ID NO 67, SEQ ID NO 71 , SEQ ID NO 73, SEQ ID NO 74, SEQ ID NO 75, SEQ ID NO 100, SEQ ID NO 134, SEQ ID NO 135, SEQ ID NO 196, SEQ ID NO 220, SEQ ID NO 228 and SEQ ID NO 252.

22. The antisense oligonucleotide according to any one of embodiments 1-21, wherein the antisense oligonucleotide is isolated, purified or manufactured.

23. The antisense oligonucleotide according to any one of embodiments 1-22, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof comprises one or more modified nucleotides or one or more modified nucleosides.

24. The antisense oligonucleotide according to any one of embodiments 1-23, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof, comprises one or more modified nucleosides, such as one or more modified nucleotides independently selected from the group consisting of 2'-O-alkyl-RNA; 2'-O-methyl RNA (2'-OMe); 2'-alkoxy-RNA; 2'-O-methoxyethyl- RNA (2'-MOE); 2'-amino-DNA; 2'-fluro-RNA; 2'-fluoro-DNA; arabino nucleic acid (ANA); 2'-fluoro- ANA; bicyclic nucleoside analog (LNA); or any combination thereof.

25. The antisense oligonucleotide according to embodiment 23 or embodiment 24, wherein one or more of the modified nucleosides is a sugar modified nucleoside. 26. The antisense oligonucleotide according to any one of embodiments 23-25, wherein one or more of the modified nucleosides comprises a bicyclic sugar.

27. The antisense oligonucleotide according to any one of embodiments 23-26, wherein one or more of the modified nucleosides is an affinity enhancing 2' sugar modified nucleoside.

28. The antisense oligonucleotide according to any one of embodiments 23-27, wherein one or more of the modified nucleosides is an LNA nucleoside, such as one or more beta-D-oxy LNA nucleosides.

29. The antisense oligonucleotide according to any one of embodiments 1-28, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof, comprises one or more 5'-methyl-cytosine nucleobases.

30. The antisense oligonucleotide according to any one of embodiments 1-29, wherein one or more of the internucleoside linkages within the contiguous nucleotide sequence of the antisense oligonucleotide is modified.

31. The antisense oligonucleotide according to embodiment 30, wherein at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95% or about 100% of the internucleoside linkages are modified.

32. The antisense oligonucleotide according to embodiment 30 or embodiment 31 , wherein the one or more modified internucleoside linkages comprise a phosphorothioate linkage.

33. The antisense oligonucleotide according to any one of embodiments 1-32, wherein the antisense oligonucleotide is a morpholino modified antisense oligonucleotide.

34. The antisense oligonucleotide according to any one of embodiments 1-22, wherein the antisense oligonucleotide is or comprises an antisense oligonucleotide mixmer or totalmer.

35. The antisense oligonucleotide of any one of embodiments 1-34, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof is 10 - 20 nucleotides in length. 36. The antisense oligonucleotide of embodiment 35, wherein the antisense oligonucleotide or contiguous nucleotide sequence thereof is 16 nucleotides in length.

37. An antisense oligonucleotide having the structure: 38. An antisense oligonucleotide having the structure: 39. An antisense oligonucleotide having the structure: 40. An antisense oligonucleotide having the structure: 41. An antisense oligonucleotide having the structure:

42. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GaGctGggTcAagAAT (SEQ ID NO: 71) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages. 43. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GgtCaaGaAtgGtgTG (SEQ ID NO: 73) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

44. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound CaGaAtGgtGtGgTC (SEQ ID NO 74) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

45. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound GaAtGgtGtGgTccC (SEQ ID NO 75) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

46. An antisense oligonucleotide wherein the oligonucleotide is the oligonucleotide compound CtcAagCtcAcAtgGC (SEQ ID NO 134) wherein capital letters represent beta-D-oxy LNA nucleosides, lowercase letters represent DNA nucleosides, all LNA C are 5-methyl cytosine, and all internucleoside linkages are phosphorothioate internucleoside linkages.

47. An antisense oligonucleotide according to any one of embodiments 1-46 covalently attached to at least one conjugate moiety.

48. An antisense oligonucleotide according to any one of embodiments 1-47 wherein the antisense oligonucleotide is in the form of a pharmaceutically acceptable salt.

49. An antisense oligonucleotide according to embodiment 48 wherein the salt is a sodium salt, a potassium salt or an ammonium salt.

50. A pharmaceutical composition comprising the antisense oligonucleotide according to any one of embodiments 1-49 and a pharmaceutically acceptable diluent, solvent, carrier, salt and/or adjuvant. 51. A pharmaceutical composition according to embodiment 50, wherein the pharmaceutical composition comprises an aqueous diluent or solvent, such as phosphate buffered saline.

52. An in vivo or in vitro method for enhancing the expression of the Exon1-Exon2 progranulin splice variant in a cell which is expressing progranulin, said method comprising administering an antisense oligonucleotide according to any one of embodiments 1-49, or a pharmaceutical composition according to embodiment 50 or embodiment 51 in an effective amount to said cell.

53. The method according to embodiment 52, wherein the cell is either a human cell or a mammalian cell.

54. A method for treating or preventing neurological disease comprising administering a therapeutically or prophylactically effective amount of an antisense oligonucleotide according to any one of embodiments 1-49 or the pharmaceutical composition according to embodiment 50 or embodiment 51 to a subject suffering from or susceptible to neurological disease.

55. A method for treating or preventing progranulin haploinsufficiency or a related disorder comprising administering a therapeutically or prophylactically effective amount of an antisense oligonucleotide according to any one of embodiments 1-49 or the pharmaceutical composition according to embodiment 50 or embodiment 51 to a subject suffering from or susceptible to progranulin haploinsufficiency or a related disorder.

56. The antisense oligonucleotide according to any one of embodiments 1-49, or the pharmaceutical composition according to embodiment 50 or embodiment 51 for use as a medicament.

57. The antisense oligonucleotide according to any one of embodiments 1-49, or the pharmaceutical composition according to embodiment 50 or embodiment 51 for use in the treatment of a neurological disease.

58. The antisense oligonucleotide or pharmaceutical composition for use in the treatment of a neurological disease according to embodiment 56, wherein the neurological disease is a TDP- 43 pathology. 59. The antisense oligonucleotide according to any one of embodiments 1-49, or the pharmaceutical composition according to embodiment 50 or embodiment 51 for use in the treatment of progranulin haploinsufficiency or a related disorder.

60. Use of the antisense oligonucleotide according to any one of embodiments 1-49, or the pharmaceutical composition according to embodiment 50 or embodiment 51 for the preparation of a medicament for treatment or prevention of a neurological disease.

61. Use of an antisense oligonucleotide, or pharmaceutical composition according to embodiment 60, wherein the neurological disease is a TDP-43 pathology.

62. Use of the antisense oligonucleotide according to any one of embodiments 1-49, or the pharmaceutical composition according to embodiment 50 or embodiment 51 for the preparation of a medicament for treatment or prevention of progranulin haploinsufficiency or a related disorder.

EXAMPLES

Example 1 : 275 oligonucleotides screened for effects on intron 1 skipping

H4 neuroglioma cells were seeded 15000 pr well in 96-well plates the day before transfection in medium (DMEM Sigma: D0819, 15% FBS, 1 mM Sodium Pyruvate, 25 pg/ml Gentamicin).

Microglia cells were chosen for analysis because these cells produce high levels of progranulin. Reduction of progranulin in microglia cells alone is sufficient to recapitulate inflammation, lysosomal dysfunction, and hyperproliferation in a cell-autonomous manner. Therefore, targeting microglial dysfunction caused by progranulin insufficiency represents a potential therapeutic strategy to manage neurodegeneration in Frontotemporal dementia. To study effects on Progranulin in cellular systems, H4 cells (ATCC HTB-148) a commercial available glial cell line derived from a cancer patient have been used for identifying oligonucleotides capable of increasing progranulin production.

To further use Microglia that exhibit functional characteristics similar to human microglia, including phagocytosis and cytokine-mediated inflammatory responses, and express relevant microglial markers, hiPSC derived microglia iCell® Microglia from FujiFilm Cellular Dynamics Inc. (Cat. no R1131) have been used for investigating effects of selected oligonucleotides on progranulin production.

Transfection was performed using Lipofectamine 2000 (Invitrogen) using the following procedure.

Medium was removed from cells and 80 pL Optimem reduced serum medium (Gibco) containing 6.25 pg/mL Lipofectamine 2000 (Invitrogen) was added, 20 pL Optimem with compounds (125 nM) were added to each well (25 nM final). As control PBS was used instead of compound. After 5 hours, transfection solution was removed from wells and full growth medium was added. The day after transfection, RNA was extracted by adding 125 pL RLT buffer (Qiagen) and using RNeasy 97 kit and protocols from Qiagen. cDNA synthesis was performed using 4 pL input RNA was performed using IScript Advanced cDNA Synthesis Kit for RT-qPCR (Bio-Rad) and 2 pL was used as input for digital droplet PCR using ddPCR supermix for probes (no dllTP) (Bio-Rad) according to Manufactor’s protocol. The following Primers and Probes (IDT) were used:

GRN Exon1-Exon2 (FAM):

Primer 1 : GCTGCTGCCCAAGGACCGCGGA (SEQ ID NO 282)

Primer 2: GCCCTGCTGTTAAGGCCACCCA (SEQ ID NO 283)

Probe /56-FAM/GGACGCAGG/ZEN/CAGACCATGTGGACCCTG/3IABkFQ/ (SEQ ID NO 284)

GRN Intron1-Exon2 (HEX):

Primer 1 : CCAAAGCAGGGACCACACCATTCTT (SEQ ID NO 285)

Primer 2: GCCCTGCTGTTAAGGCCACCCA (SEQ ID NO 286)

Probe /5HEX/CCCAGCTCC/ZEN/ACCCCTGTCGGCAGACCATG/3IABkFQ/ (SEQ ID NO 287)

HPRT1 : HPRT1 (FAM, PT.58v.45621572, IDT) and HPRT1 (HEX, Hs.PT.58v.45621572) IDT.

Exon1-Exon2 GRN mRNA and Intron1-Exon2 GRN mRNA concentrations were quantified relative to the housekeeping gene HPRT1 using QuantaSoft Software (Bio-Rad).

The compounds were profiled and ranked according to expression of Exon 1 - Exon2 mRNA splice form relative to HPRT1 and Intron1-Exon2 relative to HPRT1 and results are shown in Figure 1.

The following compounds relative to PBS transfected cells show an increased expression of Exonl - Exon 2 spliced mRNA, a reduced expression of Intron1-Exon 1 spliced mRNA and a more than 25% shift in ratio Exon 1-Exon2 vs Intronl- Exon2: Compound ID#51, I D#52, I D#53, I D#54, ID#55, ID#62, ID#63, ID#67, ID#71, ID#73, ID#74, ID#75, ID#100, ID#134, ID#135, ID#196, ID#220, ID#228 and ID#252 as shown in Figure 1.

Table 1 : All the antisense oligonucleotides are designed as 16-mers DNA-LNA mixmers with a phosphorothioate backbone, DNA-LNA mixmers with a phosphorothioate backbone, LNA at the very 5’ and 3’ position and e.g. LNA for every 2 nd or 3 rd nucleotide (see Compound table).

Example 2: Oligonucleotides tested for effect on progranulin expression

H4 neuroglioma cells seeded in 96 well plates 5000 pr well, were treated with either 3 pM or 10 pM final concentration of oligo for 5 days in 200pL medium. Progranulin expression levels were evaluated in media after dilution 1:8 by ELISA from Abeam (ab252364). Oligos #71 , #73, #74, #75 and #134 induced progranulin secretion more than 1.5 fold compared to PBS as shown in Figure 2. For comparison effects of oligonucleotides S7 and S10 from patent (WO 2020/191212A1) are shown in Figure 3.

Example 3: Sequence localization of oligos

Sequence localization of SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75 and SEQ ID NO: 134 are shown in Figure 4.

Example 4: Oligonucleotides tested for effect on progranulin expression in hiPSC derived microglia hiPSC derived microglia (iCell Microglia Kit, 01279, Cat. no R1131) were seeded (n=3) in Poly-D- lysine coated 96-well plates (Greiner Catalog # 655946) with 20000 cells pr well in 200 pL and were treated with indicated concentrations of SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75 and SEQ ID NO: 134 for 4 days. Progranulin protein expression levels were evaluated in media after dilution 1 :8 using ELISA from Abeam (ab252364). SEQ ID NO: 73, SEQ ID NO: 74, SEQ ID NO: 75 and SEQ ID NO: 134 induced progranulin secretion more than 1.5 fold compared to PBS as shown in Figure 5. For comparison effects of oligonucleotides S7, S10 and S37 from WO 2020/191212 are shown in Figure 6.

Example 5: Oligonucleotides tested for effect on progranulin expression in H4 neuroglioma cellsH4 neuroglioma cells were seeded 15000 pr well in 96-well plates the day before transfection in medium (DM EM Sigma: D0819, 15% FBS, 1 mM Sodium Pyruvate, 25 pg/ml Gentamicin). Transfection was performed using Lipofectamine 2000 (Invitrogen) using the following procedure. Medium was removed from cells and 80 pL Optimem reduced serum medium (Gibco) containing 6.25 pg/mL Lipofectamine 2000 (Invitrogen) was added, 20 pL Optimem with compounds (125 nM) were added to each well (25 nM final). As control PBS was used instead of compound. After 5 hours, transfection solution was removed from wells and full growth medium was added. The day after transfection, RNA was extracted by adding 125 pL RLT buffer (Qiagen) and using RNeasy 97 kit and protocols from Qiagen. cDNA synthesis was performed using 4 pL input RNA was performed using IScript Advanced cDNA Synthesis Kit for RT-qPCR (Bio-Rad) and 2 pL was used as input for digital droplet PCR using ddPCR supermix for probes (no dllTP) (Bio-Rad) according to Manufactor’s protocol.

The following Primers and Probes (IDT) were used

GRN Exon1-Exon2 (FAM):

Primer 1: GCTGCTGCCCAAGGACCGCGGA, Primer 2: GCCCTGCTGTTAAGGCCACCCA and Probe /56-FAM/GGACGCAGG/ZEN/CAGACCATGTGGACCCTG/3IABkFQ/

GRN Intron1-Exon2 (HEX):

Primer 1: CCAAAGCAGGGACCACACCATTCTT, Primer 2: GCCCTGCTGTTAAGGCCACCCA and Probe /5HEX/CCCAGCTCC/ZEN/ACCCCTGTCGGCAGACCATG/3IABkFQ/

HPRT1: HPRT1 (FAM, PT.58v.45621572, IDT) and HPRT1 (HEX, Hs.PT.58v.45621572) IDT.

Exon1-Exon2 GRN mRNA and Intron1-Exon2 GRN mRNA concentrations were quantified relative to the housekeeping gene HPRT1 using QuantaSoft Software (Bio-Rad).

The results for SEQ I D#73, SEQ ID#74 and SEQ ID#75 are show in Figure 7. SEQ I D#73, SEQ ID#74 and SEQ ID#75 show dose-dependent skipping of intronl retention (Int1-Ex2) and an increase in Ex1-Ex2 splice-variant. The S10 compound from WO 2020/191212 showed no/limited effects on skipping of intronl retention.

Example 6 - Oligonucleotides tested for effect on progranulin expression in H4 neuroglioma cells H4 neuroglioma cells were seeded 15000 pr well in 96-well plates the day before transfection in medium (DMEM Sigma: D0819, 15% FBS, 1 mM Sodium Pyruvate, 25 pg/ml Gentamicin). Transfection was performed using Lipofectamine 2000 (Invitrogen) using the following procedure. Medium was removed from cells and 80 pL Optimem reduced serum medium (Gibco) containing 6.25 pg/mL Lipofectamine 2000 (Invitrogen) was added, 20 pL Optimem with compounds (125 nM) were added to each well (25 nM final). As control PBS was used instead of compound. After 5 hours, transfection solution was removed from wells and full growth medium was added. The day after transfection, RNA was extracted by adding 125 pL RLT buffer (Qiagen) and using RNeasy 97 kit and protocols from Qiagen. cDNA synthesis was performed using 4 pL input RNA was performed using IScript Advanced cDNA Synthesis Kit for RT-qPCR (Bio-Rad) and 2 pL was used as input for digital droplet PCR using ddPCR supermix for probes (no dllTP) (Bio-Rad) according to Manufactor’s protocol. The following Primers and Probes (IDT) were used

GRN Exon1-Exon2 (FAM):

Primer 1: GCTGCTGCCCAAGGACCGCGGA,

Primer 2: GCCCTGCTGTTAAGGCCACCCA and

Probe /56-FAM/GGACGCAGG/ZEN/CAGACCATGTGGACCCTG/3IABkFQ/

GRN Intron1-Exon2 (HEX): Primer 1 : CCAAAGCAGGGACCACACCATTCTT, Primer 2: GCCCTGCTGTTAAGGCCACCCA and

Probe /5HEX/CCCAGCTCC/ZEN/ACCCCTGTCGGCAGACCATG/3IABkFQ/

HPRT1: HPRT1 (FAM, PT.58v.45621572, IDT) and HPRT1 (HEX, Hs.PT.58v.45621572) IDT.

Exon1-Exon2 GRN mRNA and Intron1-Exon2 GRN mRNA concentrations were quantified relative to the housekeeping gene HPRT1 using QuantaSoft Software (Bio-Rad).

The results for SEQ ID#289 and SEQ ID#290 are show in Figure 8. SEQ ID: 289 and 290 show dose-dependent skipping of intron 1 retention (Int1-Ex2) and an increase in Ex1-Ex2 splice-variant. The S10 compound from WO 2020/191212 showed no/limited effects on skipping of intron 1 retention.

Example 7 - Oligonucleotides tested for effect on progranulin expression in hiPSC derived microglia hiPSC derived microglia (iCell Microglia Kit, 01279, Cat. no R1131) were seeded (n=3) in Poly-D- lysine coated 96-well plates (Greiner Catalog # 655946) with 20000 cells pr well in 200 pL and were treated with indicated concentrations of Compound S10 and SEQ ID #290 for 5 days.

RNA was extracted by adding 125 pL RLT buffer (Qiagen) and using RNeasy 97 kit and protocols from Qiagen. cDNA synthesis was performed using 4 pL input RNA was performed using IScript Advanced cDNA Synthesis Kit for RT-qPCR (Bio-Rad) and 2 pL was used as input for digital droplet PCR using ddPCR supermix for probes (no dllTP) (Bio-Rad) according to Manufactor’s protocol. The following Primers and Probes (IDT) were used

GRN Exon1-Exon2 (FAM):

Primer 1: GCTGCTGCCCAAGGACCGCGGA,

Primer 2: GCCCTGCTGTTAAGGCCACCCA and

Probe /56-FAM/GGACGCAGG/ZEN/CAGACCATGTGGACCCTG/3IABkFQ/

GRN Intron1-Exon2 (HEX):

Primer 1: CCAAAGCAGGGACCACACCATTCTT,

Primer 2: GCCCTGCTGTTAAGGCCACCCA and

Probe /5HEX/CCCAGCTCC/ZEN/ACCCCTGTCGGCAGACCATG/3IABkFQ/

GAPDH: GAPDH (FAM, Hs.PT.39a.22214836, IDT) and GAPDH (HEX, Hs.PT.39a.22214836, IDT).

Exon1-Exon2 GRN mRNA and Intron1-Exon2 GRN mRNA concentrations were quantified relative to the housekeeping gene GAPDH using QuantaSoft Software (Bio-Rad).

The results for SEQ ID#290 are show in Figure 9. SEQ ID: 290 showed dose-dependent skipping of intronl retention (Int1-Ex2) and an increase in Ex1-Ex2 splice-variant. The S10 compound from WO 2020/191212 showed no/limited effects on skipping of intronl retention, a gapmer was included as control showing the expected dose-dependent knockdown of both splice variants.

Example 8 - Splice switch analysis using SEQ ID NO: 290

H4 neuroglioma cells were seeded 15000 pr well in 96-well plates the day before transfection in medium (DMEM Sigma: D0819, 15% FBS, 1 mM Sodium Pyruvate, 25 pg/ml Gentamicin). Transfection was performed using Lipofectamine 2000 (Invitrogen) using the following procedure. Medium was removed from cells and 80 pL Optimem reduced serum medium (Gibco) containing 6.25 pg/mL Lipofectamine 2000 (Invitrogen) was added, 20 pL Optimem with compounds (125 nM) were added to each well (25 nM final). As control PBS was used instead of compound. After 5 hours, transfection solution was removed from wells and full growth medium was added. Two days after transfection, RNA was extracted by adding 350 pL Magnapure lysis buffer (Roche) and using the Magnapure system and protocols (including DNase treatment) from Roche. Total RNA was eluted in 50 pL elution buffer. KAPA mRNA HyperPrep Kits (Roche) was used to generate next generation sequencing (NGS) libraries using 100 ng of total RNA as input. Sequencing was performed on the Illumina NextSeq 550 system to obtain more than 30 million paired end reads (2x151 bp) per sample. Reads were trimmed by removal of 1 nucleotide at the 3’end, and subsampled to 30 million reads per sample prior to RNA-Seq analysis using CLC Genomic Workbench (Qiagen). To generate the Sashimi plots (showing the number of reads spanning the exon junctions), the Bam files was exported from CLC Genomic Workbench (Qiagen) and imported into The Integrative Genomics Viewer (IGV) (version IGV_2.8.2) from Broad Institute. Sashimi plots showing the splice-switching to obtain the correct splicing from exon 1 to exon 2 with the ID#290 and not with S10 compound from WO 2020/191212. This is shown in Figure 10.

Compound Table

Helm Annotation Key:

[LR](G) is a beta-D-oxy-LNA guanine nucleoside,

[LR](T) is a beta-D-oxy-LNA thymine nucleoside,

[LR](A) is a beta-D-oxy-LNA adenine nucleoside,

[LR]([5meC] is a beta-D-oxy-LNA 5-methyl cytosine nucleoside,

[dR](G) is a DNA guanine nucleoside,

[dR](T) is a DNA thymine nucleoside,

[dR](A) is a DNA adenine nucleoside,

[dR]([C] is a DNA cytosine nucleoside,

[mR](G) is a 2’-O-methyl RNA guanine nucleoside,

[mR](U) is a 2’-O-methyl RNA DNA uracil nucleoside,

[mR](A) is a 2’-O-methyl RNA DNA adenine nucleoside,

[mR]([C] is a 2’-O-methyl RNA DNA cytosine nucleoside,

[MOE](G) is a 2’-MOE RNA guanine nucleoside,

[MOE](T) is a 2’- MOE RNA DNA thymine nucleoside,

[MOE](A) is a 2’- MOE thyl RNA DNA adenine nucleoside, [MOE]([C] is a 2’- MOE RNA DNA cytosine nucleoside.