WO2012065051A1 - Compositions et procédés pour le traitement de troubles dépendant des récepteurs androgènes y compris les cancers - Google Patents

Compositions et procédés pour le traitement de troubles dépendant des récepteurs androgènes y compris les cancers Download PDF

Info

Publication number
WO2012065051A1
WO2012065051A1 PCT/US2011/060352 US2011060352W WO2012065051A1 WO 2012065051 A1 WO2012065051 A1 WO 2012065051A1 US 2011060352 W US2011060352 W US 2011060352W WO 2012065051 A1 WO2012065051 A1 WO 2012065051A1
Authority
WO
WIPO (PCT)
Prior art keywords
androgen receptor
pharmaceutically acceptable
oligomer
acceptable salt
conjugate
Prior art date
Application number
PCT/US2011/060352
Other languages
English (en)
Inventor
Jesper Worm
Yixian Zhang
Original Assignee
Enzon Pharmaceuticals, Inc.
Santaris Pharma A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/945,551 external-priority patent/US8450290B2/en
Priority claimed from GBGB1101969.2A external-priority patent/GB201101969D0/en
Application filed by Enzon Pharmaceuticals, Inc., Santaris Pharma A/S filed Critical Enzon Pharmaceuticals, Inc.
Priority to EP11839515.1A priority Critical patent/EP2646553A4/fr
Priority to AU2011326034A priority patent/AU2011326034B2/en
Publication of WO2012065051A1 publication Critical patent/WO2012065051A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy

Definitions

  • the invention relates to the field of androgen receptor targeted therapies and methods for treating androgen receptor dependent disorders including cancers.
  • the androgen receptor is a type of nuclear receptor which is activated by binding either of the androgenic hormones testosterone or dihydrotestosterone.
  • the main function of the androgen receptor is as a DNA binding transcription factor which regulates gene expression.
  • the androgen receptor also has additional functions independent of DNA binding.
  • the androgen receptor is most closely related to the progesterone receptor, and progestins in higher dosages can block the androgen receptor.
  • AR-A is an 87kDa protein which lacks the first 187 amino acids (N-terminal truncation).
  • Isoform AR-B is the full length 1 10 kDa version.
  • the binding of androgen to the androgen receptor induces a conformational change to the receptor, resulting in a dissociation of heat shock proteins, dimerization and transport from the cytosol to the cell nucleus where the androgen receptor dimer binds to specific DNA sequences - referred to as hormone response elements.
  • the AR controls gene expression, either increasing or decreasing transcription of specific genes, such as insulin-like growth factor I (IGF- 1 ).
  • Androgen receptors can also have cytoplasmic activities though with signal transduction proteins in the cytoplasm. Androgen binding to cytoplasmic androgen receptors , can cause rapid changes in cell function independent of gene transcription, for example ion transport, as well as indirect influence of gene transcription, for example via mediating other signal transduction pathways, thereby influencing the activity of other transcription factors.
  • T e over-expression of androgen receptor, or expression of mutated androgen receptor genes has been indicated in several diseases, such as cancer, including prostate cancer and breast cancer, as well as other disorders such as polyglutamate disease (Monks et al., PNAS November 2 2007, published on line) alopecia, benign prostatic hyperplasia, spinal and muscular atrophy and Kennedy disease.
  • W097/1 1 170 reports on a method of treating a patient diagnosed as having benign prostatic hyperplasia or a prostate cancer comprising administering an antisense
  • oligonucleotide which selectively hybridises to the androgen receptor mRNA.
  • Three antisense oligonucleotide sequences of between 27 - 29 nucleotides are disclosed.
  • US 6,733,776 and EP 0 692 972 report on a method for treating androgenic alopecia by applying liposomes comprising an antisense nucleic acid that hybridises to an androgen receptor gene. No antisense molecules with specific sequences and targeting the androgen receptor are provided.
  • US 2005/0164970 reports on a method of treating prostate cancer using siRNA complexes targeting the androgen receptor mRNA.
  • WO 2005/027833 reports on a method of treating prostate cancer comprising of administering a morpholino oligonucleotide of between 12-40 morpholino sub-units in length to the patient.
  • WO 2001/083740 reports on an antisense compound having an uncharged morpholino backbone of between 18 to 20 contiguous units which targets the human androgen receptor.
  • Morpholino antisense compounds work via binding to the nucleic acid target to block access to the mRNA by other molecules, such as molecules involved in mRNA splicing or translation initiation.
  • compositions and methods for the treatment of androgen receptor (AR) dependent medical disorders including AR-dependent cancers.
  • the present invention provides the use in combination of a new class of LNA antisense oligomer androgen receptor antagonists and diarylhydantoin androgen receptor binding inhibitors, such as 4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin- 1 -yl)-2-fluoro-N-methylbenzamide ("MDV3 100”), for the treatment of androgen receptor dependent medical disorders in mammals such as humans.
  • a new class of LNA antisense oligomer androgen receptor antagonists and diarylhydantoin androgen receptor binding inhibitors such as 4-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5- dimethyl-4-oxo-2-thioxoimidazolidin- 1 -yl)-2-fluoro-N-methylbenzamide (“MDV3 100")
  • the condition treated may, for example, be an androgen receptor-dependent cancer such as a breast cancer or a prostate cancer, for example, advanced prostate cancer, castration-resistant prostate cancer (CRPC), hormone refractory prostate cancer, alopecia, benign prostatic hyperplasia, spinal and muscular atrophy, Kennedy disease and polyglutamate disease.
  • CRPC castration-resistant prostate cancer
  • antisense oligomers targeting the androgen receptor strongly potentiate the effect of diarylhydantoin androgen receptor blockers.
  • the present invention provides an antisense oligomer or conjugate thereof, or pharmaceutically acceptable salt thereof, that affects, preferably decreases, expression of an androgen receptor for use in the treatment of an androgen receptor dependent
  • oligomer is used in combination with a diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof.
  • the present invention provides an antisense oligomer or conjugate thereof, or pharmaceutically acceptable salt thereof, for use in the treatment of an androgen receptor dependent hyperproliferative disorder in a mammal ; wherein said oligomer is used in combination with a diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof; and/or bicalutamide or a pharmaceutically acceptable salt thereof.
  • the invention provides the use in combination of a (i) diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof and (ii) an antisense androgen receptor oligomer or a pharmaceutically acceptable salt thereof or a conjugate of the oligomer or a pharmaceutically acceptable salt of the conjugate, such as those described herein, that decreases androgen receptor expression, for the treatment and/or prevention of androgen-dependent medical conditions and disorders including but not limited to hyperproliferative disorders.
  • the present invention provides a kit comprising:
  • component (a) can be administered to a mammal as a single component or as different component parts.
  • component (a) may be delivered simultaneously with component (b) [wherein component (a) is the antisense oligomer and component (b) is the diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof, and/or bicalutamide or a
  • component (a) - may be delivered before and/or after component (b) [wherein component (a) is the antisense oligomer and component (b) is the diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof, and/or bicalutamide or a pharmaceutically acceptable salt thereof] ; in other words components (a) and (b) are administered sequentially.
  • component (b) - may be delivered before and/or after component (a) [wherein component (a) is the antisense oligomer and component (b) is the diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof, and/or bicalutamide or a pharmaceutically acceptable salt thereof]; in other words components (b) and (a) are administered sequentially.
  • components (a) and (b) are in the same composition.
  • the present invention provides the use of an antisense oligomer or conjugate thereof, or pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of an androgen receptor dependent hyperproliferative disorder in a mammal; wherein said medicament is for use in combination with either i) a
  • diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof and/or ii) bicalutamide or a pharmaceutically acceptable salt thereof.
  • the antisense oligomer and (i) the diarylhydantoin type androgen receptor ligand binding inhibitor or a pharmaceutically acceptable salt thereof and/or (ii) bicalutamide or a pharmaceutically acceptable salt thereof may be in the same composition.
  • the present invention provides in another aspect a method for treating an androgen receptor dependent hyperproliferative disorder in a mammal, comprising administering to said mammal, optionally in amounts therapeutically effective in combination:
  • the present invention provides the combination use of (i) an antisense oligomer or conjugate thereof, or pharmaceutically acceptable salt thereof, and (ii) a diarylhydantoin type androgen receptor ligand binding inhibitor for the treatment of an androgen receptor dependent hyperproliferative disorder in a mammal.
  • One embodiment provides a method for treating a disease or a medical disorder as disclosed herein, such as a hyperproliferative disorder, such as cancer, said method comprising administering to a mammal, such as a human, in need of treatment for the disorder ( 1 ) an antisense oligomer, a conjugate or a pharmaceutical composition according to the invention, in an amount effective to decrease expression of AR in the mammal and (2) a diarylhydantoin androgen receptor binding inhibitor, such as 4-(3-(4-cyano-3- (trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin- l -yl)-2-fluoro-N- methylbenzamide, in an amount effective to decrease the activity of AR in the mammal, such that the effect of the oligomer, conjugate thereof or composition is temporally overlapping with the effect of the diarylhydantoin AR inhibitor.
  • a disease or a medical disorder as disclosed
  • a related embodiment provides a method for decreasing AR activity in a cell, such as a mammalian cell, such as a human cell, said method comprising contacting the cell with ( 1 ) an antisense oligomer, a conjugate or a pharmaceutical composition according to the invention in an amount effective to decrease expression of AR and (2) a diarylhydantoin androgen receptor binding inhibitor, such as 4-(3-(4-cyano-3-(triflu6romethyl)phenyl)-5,5-dimethyl-4- oxo-2-thioxoimidazolidin- l -yl)-2-fluoro-N-methylbenzamide, in an amount effective to decrease the activity of AR in the mammal, such that the effect of the oligomer, conjugate thereof or composition is temporally overlapping with the effect of the diarylhydantoin AR inhibitor.
  • a diarylhydantoin androgen receptor binding inhibitor such as 4-(3-(4-cyano-3-(triflu6romethyl
  • the oligomer used may, for example, be 10 - 50, such as 10 - 30 nucleotides in length which comprises a contiguous nucleotide sequence of a total of 10 - 50, such as 10 - 30 nucleotides, wherein said contiguous nucleotide sequence is at least 80% (e.g., 85%, 90%, 95%, 98%, or 99%) homologous to a region corresponding to the reverse complement of a nucleic acid which encodes a mammalian androgen receptor, such as a mammalian Androgen Receptor gene or mRNA, such as SEQ ID NO: 1 or naturally occurring variants thereof.
  • the oligomer hybridizes to a single stranded nucleic acid molecule having the sequence of a (corresponding) portion of SEQ ID NO: 1.
  • the oligomer used may, for example, be 10-50 nucleobases in length which comprises a contiguous nucleobase sequence of a total of 10-50 nucleobases, wherein said contiguous nucleobase sequence is at least 80% homologous to a corresponding region of a nucleic acid which encodes a mammalian androgen receptor.
  • the oligomer used may, for example, be 10-50 nucleobases in length which comprises a contiguous nucleobase sequence of a total of 10-50 nucleobases, wherein said contiguous nucleobase sequence is at least 80% identical to the reverse complement of a target region of a nucleic acid which encodes a mammalian androgen receptor.
  • T e oligomer used may, for example, consist of 10 to 30 contiguous monomers wherein adjacent monomers are covalently linked by a phosphate group or a
  • said oligomer comprises a first region of at least 10 contiguous monomers; wherein at least one monomer of said first region is a nucleoside analogue; wherein the sequence of said first region is at least 80% identical to the reverse complement of the best-aligned target region of a mammalian androgen receptor gene or a mammalian androgen receptor mRNA.
  • an antisense oligomer selected from the group consisting of:
  • these target regions may comprise an additional 4 bases
  • SEQ ID NO: 1 Homo sapiens androgen receptor (dihydrotestosterone receptor; testicular feminization; spinal and bulbar muscular atrophy; Kennedy disease) (AR), transcript variant 1 , mRNA. (Genbank Accession number NM_000044).
  • SEQ ID NO 81 Mouse androgen receptor mRNA sequence.
  • SEQ ID NO 82 Rhesus monkey androgen receptor mRNA sequence.
  • SEQ ID NO 83 Homo sapiens androgen receptor protein amino acid sequence.
  • SEQ ID NO 84 Mouse androgen receptor protein amino acid sequence.
  • SEQ ID NO 85 Rhesus monkey androgen receptor protein amino acid sequence.
  • Figure 14 Cell proliferation assay - time course post-transfection.
  • FIG. 1 Caspase 3/7 activity in LNCaP cells, 24, 48 or 72 hours post-transfection.
  • Figure 17 Average PSA in plasma after in vivo oligomer treatment.
  • SEQ ID NO: 58 potentiates the cell growth inhibitory effect of MDV3100.
  • SEQ ID NO: 58 potentiates MDV3100 inhibition of AR transcriptional activity.
  • SEQ ID NO: 58 potentiates the effect of MDV3100 on AR transcriptional activity.
  • N-methylbenzamide significantly down-modulate AR (androgen receptor) mRNA.
  • SEQ ID NO: 58 potentiates the effect of MDV3100 on PSA transcriptional activity.
  • SEQ ID NO: 58 (EZN-4176) in combination with 4-(3-(4-cyano-3- (trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin- l-yl)-2-fluoro-N- methylbenzamide (MDV3100) significantly down-modulate PSA (prostate specific antigen) mRNA.
  • oligomeric compounds for use in modulating the function of nucleic acid molecules encoding mammalian Androgen Receptor, such as the Androgen Receptor nucleic acid shown in SEQ ID NO 1 , and naturally occurring variants of such nucleic acid molecules encoding mammalian Androgen Receptor.
  • oligomer in the context of the present invention, refers to a molecule formed by covalent linkage of two or more nucleotides (i.e. an oligonucleotide).
  • each single nucleotide such as the nucleotides present in the oligomer of the invention, may also be referred to as a "monomer” or "unit".
  • the oligomer consists or comprises of a contiguous nucleotide sequence of 10-50 nucleotides in length, such as 10 - 30 nucleotides in length (i.e. comprises or consists of from 10 - 30 covalently linked monomers).
  • the antisense oligomer targeting the androgen receptor does not comprise RNA (units).
  • the antisense oligomers according to the invention are linear molecules or are synthesised as a linear molecule.
  • the oligomer in such embodiments, is a single stranded molecule, and typically does not comprise short regions of, for example, at least 3, 4 or 5 contiguous nucleotides, which are complementary to another region within the same oligomer (i.e. duplexes)
  • the oligomer is not (essentially) double stranded.
  • the oligomer is essentially not double stranded, such as is not a siRNA.
  • the oligomer of the invention may consist entirely of the contiguous nucleotide region.
  • the oligomer is not substantially self-complementary.
  • siRNAs comprise of 2 complementary short RNA (or equivalent nucleobase units) sequences, such as 21 and 23nts long, with, typically a 2nt 3' overhang on either end.
  • the siRNAs may be conjugated, such as conjugated to a sterol, such as a cholesterol group (typically at the 3' or 5' termini of one or both of the strands).
  • the invention further provides target sequences in the AR mRNA or gene, or an allelic variant thereof, in particular those corresponding to SEQ ID NOS: 2 - 22, wherein antisense oligonucleotides corresponding to said target sequences are capable of down-regulating AR.
  • target sequences which correspond to the antisense oligonucleotide sequences SEQ ID NOS: 2 - 22, respectively, are shown in Figure 4 (bold and underlined, with the corresponding oligo SEQ ID NOS indicated above).
  • Variant sequences for example but not limited to allelic variants (such as a (AR) gene present at gene locus Xq l 1 - 12) of such target sequences are also within the scope of the invention.
  • a variant sequence may have at least 60%, more preferably at least 70%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91 %, at least 92%, at least 93%, at least 94%, at least 95% sequence homology to a target sequence in AR.
  • an oligomer of the invention corresponding to said variant sequences is still capable of down- regulating AR.
  • LNA oligonucleotides are also disclosed, for example those shown in SEQ ID NOS 44 - 80.
  • the oligomers of the invention are considered to be potent inhibitors of androgen receptor mRNA and protein expression.
  • the oligomer may be an antisense oligonucleotide may or may not comprise LNA.
  • the oligomer may be an oligomer, such as a gapmer oligomer, of 10 - 50 nucleotides in length, such as 10 - 30 nucleotides in length, which comprises a contiguous nucleotide sequence of a total of between 10 - 50, such as 10 - 30 nucleotides, wherein said contiguous nucleotide sequence is at least 80% (e.g., 85%, 90%, 95%, 98%, or 99%) homologous to a region corresponding to the reverse complement of a nucleic acid which encodes a mammalian androgen receptor, such as a mammalian androgen Receptor gene or mRNA, such as SEQ ID NO: 1 or naturally occurring variants thereof.
  • the oligomer hybridizes to a single stranded nucleic acid molecule having the sequence of a (corresponding) portion of SEQ ID NO: 1 may be a gapmer.
  • the mammalian androgen receptor is preferably selected from the group consisting of human or mouse androgen receptor.
  • the mammalian androgen receptor is human androgen receptor, such as the Androgen receptor encoded by the nucleic acid as shown in SEQ ID NO 1.
  • GenBank Accession numbers for nucleic acids refer to cDNA sequences and not to mRNA sequences per se.
  • sequence of a mature mRNA can be derived directly from the corresponding cDNA sequence with thymine bases (T) being replaced by uracil bases (U).
  • the Androgen Receptor gene in humans does show a degree of allelic variation, and several of the polymorphisms are associated with disease phenotypes (Mooney et al, NAR 15; 31(8) 2003).
  • a CAG repeat expansion is associated with polyglutamine expansion disorder
  • other characterised allelic variants include a (GGC)n trinucleotide repeat and R726L, T887A and L710H single nucleotide
  • n may range from 5 and 31.
  • CAG repeats of less than 22 have been associated with an enhanced risk of prostate cancer in African American males, and this may therefore be a preferred allelic variant.
  • the target nucleic acid is an AR allelic variant which comprises one or more single nucleotide polymorphisms, including R726L, T887A and L710H.
  • the target is an AR allelic variant which comprises a (CAG)n trinucleotide repeat, or (GGC)n trinucleotide repeat.
  • the nucleic acid which encodes the mammalian androgen receptor is, in a preferable embodiment, the human androgen receptor cDNA sequence is shown as SEQ ID NO 1 and/or the mouse androgen receptor cDNA sequence is shown as SEQ ID NO 81 , or allelic variants thereof.
  • the oligomer according to the invention is an antisense oligonucleotide.
  • the target' of the oligomer according to the invention is the androgen receptor mRNA.
  • the oligomer when introduced into the cell which is expressing the androgen receptor gene, results in reduction of the androgen receptor mRNA level, resulting in reduction in the level of expression of the androgen receptor in the cell.
  • the androgen receptor is known to regulate the expression of several genes, such as a gene selected from the group consisting of Protein kinase C delta (PRKCD), Glutathione S- transferase theta 2 (GSTT2), transient receptor potential cation channel subfamily V member 3 (TRPV3), Pyrroline-5-carboxylate reductase 1 (PYCR 1 ) or ornithine aminotransferase (OAT) - such genes are referred to as androgen receptor targets herein, and as such, in some embodiments, the oligomers according to the invention may be used to modulate the expression of one or more androgen receptor targets in a cell which is expressing, or is capable of expressing (i. e. in the case of alleviation of repression (by the AR) of the androgen receptor target in a cell) said androgen receptor target.
  • PRKCD Protein kinase C delta
  • GSTT2 Glutathione S- transferase
  • the oligomers which target the androgen receptor mRNA may hybridize to any site along the target mRNA nucleic acid, such as the 5 ' untranslated leader, exons, introns and 3 ' untranslated tail. However, it is preferred that the oligomers which target the androgen receptor mRNA hybridise to the mature mRNA form of the target nucleic acid.
  • the oligomer of the invention is capable of down-regulating expression of the Androgen Receptor gene.
  • the oligomer of the invention can effect the inhibition of Androgen Receptor, typically in a mammalian such as a human cell.
  • the oligomers of the invention bind to the target nucleic acid and effect inhibition of expression of at least 10% or 20% compared to the normal expression level, more preferably at least a 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% inhibition as compared to the normal expression level (such as immediately prior to dosing of the oligomer).
  • such modulation is seen when using between 0.04 and 25nM, such as between 0.8 and 20nM concentration of the compound of the invention.
  • the inhibition of expression is less than 100%, such as less than 98% inhibition, less than 95% inhibition, less than 90% inhibition, less than 80% inhibition, such as less than 70% inhibition.
  • Modulation of expression level may be determined by measuring protein levels, e.g. by the methods such as SDS-PAGE followed by western blotting using suitable antibodies raised against the target protein.
  • modulation of expression levels can be determined by measuring levels of mRNA, e.g. by northern blotting or quantitative RT-PCR.
  • the level of down-regulation when using an appropriate dosage is, in some embodiments, typically to a level of between 10-20% the normal levels in the absence of the compound of the invention.
  • the invention therefore provides a method of down-regulating or inhibiting the expression of the androgen receptor protein and/or mRNA in a cell which is expressing the androgen receptor protein and/or mRNA, said method comprising administering or contacting the oligomer or conjugate according to the invention (suitably in an effective amount) to said cell to down-regulating or inhibiting the expression of the androgen receptor protein and/or mRNA in said cell.
  • the cell is a mammalian cell such as a human cell.
  • the administration or contacting may occur, in some embodiments, in vitro.
  • the administration or contacting may occur, in some embodiments, in vivo.
  • target nucleic acid refers to the DNA or RNA encoding mammalian androgen receptor polypeptide, such as human androgen receptor, such as SEQ ID NO: 1. Androgen receptor encoding nucleic acids or naturally occurring variants thereof, and RNA nucleic acids derived therefrom, preferably mRNA, such as pre-mRNA, although preferably mature mRNA.
  • the "target nucleic acid” may be a cDNA or a synthetic oligonucleotide derived from the above DNA or RNA nucleic acid targets.
  • the oligomer according to the invention is preferably capable of hybridising to the target nucleic acid. It will be recognised that SEQ ID NO: 1 is a cDNA sequences, and as such, corresponds to the mature mRNA target sequence, although uracil is replaced with thymidine in the cDNA sequences.
  • naturally occurring variant thereof refers to variants of the androgen receptor polypeptide of nucleic acid sequence which exist naturally within the defined taxonomic group, such as mammalian, such as mouse, monkey, and preferably human.
  • Naturally occurring variants of a polynucleotide the term also may encompass any allelic variant of the androgen receptor encoding genomic DNA which are found at the Chromosome X: 66.68 - 66.87 Mb by chromosomal translocation or duplication, and the RNA, such as mRNA derived therefrom. "Naturally occurring variants” may also include variants derived from alternative splicing of the androgen receptor mRNA. When referenced to a specific polypeptide sequence, e.g. , the term also includes naturally occurring forms of the protein which may therefore be processed, e.g. by co- or post- translational modifications, such as signal peptide cleavage, proteolytic cleavage, glycosylation, etc.
  • the oligomers comprise or consist of a contiguous nucleotide sequence which corresponds to the reverse complement of a nucleotide sequence present in SEQ ID NO: 1.
  • the oligomer can comprise or consist of, or a sequence selected from the group consisting of SEQ ID NOS: 2 - 22 and 86 - 106, wherein said oligomer (or contiguous nucleotide portion thereof) may optionally have one, two, or three mismatches against said selected sequence.
  • the oligomer may comprise or consist of a contiguous nucleotide sequence which is fully complementary (perfectly complementary) to the equivalent region of a nucleic acid which encodes a mammalian androgen receptor (e.g., SEQ ID NO: 1 ).
  • the oligomer can comprise or consist of an antisense nucleotide sequence.
  • the oligomer may tolerate I , 2, 3, or 4 (or more) mismatches, when hybridising to the target sequence and still sufficiently bind to the target to show the desired effect, i.e. down-regulation of the target.
  • Mismatches may, for example, be compensated by increased length of the oligomer nucleotide sequence and/or an increased number of nucleotide analogues, such as LNA, present within the nucleotide sequence.
  • the contiguous nucleotide sequence comprises no more than 3, such as no more than 2 mismatches when hybridizing to the target sequence, such as to the corresponding region of a nucleic acid which encodes a mammalian androgen receptor.
  • the contiguous nucleotide sequence comprises no more than a single mismatch when hybridizing to the target sequence, such as the corresponding region of a nucleic acid which encodes a mammalian androgen receptor.
  • the nucleotide sequence of the oligomers of the invention or the contiguous nucleotide sequence is preferably at least 80% homologous to a corresponding sequence selected from the group consisting of SEQ ID NOS: 2 - 22 and 86 - 106, such as at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96% homologous, such as 100% homologous (identical).
  • nucleotide sequence of the oligomers of the invention or the contiguous nucleotide sequence is preferably at least 80% homologous to the reverse complement of a
  • SEQ ID NO: 1 corresponding sequence present in SEQ ID NO: 1 , such as at least 85%, at least 90%, at least 91 %, at least 92%at least 93%, at least 94%, at least 95%, at least 96% homologous, at least 97% homologous, at least 98% homologous, at least 99% homologous, such as 100% homologous (identical).
  • the nucleotide sequence of the oligomers of the invention or the contiguous nucleotide sequence is preferably at least 80% complementary to a sub-sequence present in SEQ ID NO: 1 , such as at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96% complementary, at least 97% complementary, at least 98% complementary, at least 99% complementary, such as 100% complementary (perfectly complementary).
  • the oligomer (or contiguous nucleotide portion thereof) is selected from, or comprises, one of the sequences selected from the group consisting of SEQ ID NOS: 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , and 22, or a subsequence of at least 10 contiguous nucleotides thereof, wherein said oligomer (or contiguous nucleotide portion thereof) may optionally comprise one, two, or three mismatches when compared to the sequence.
  • the oligomer (or contiguous nucleotide portion thereof) is selected from, or comprises, one of the sequences selected from the group consisting of SEQ ID NOS: 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 100, 101 , 102, 103, 104, 105 and 106, or a sub-sequence of at least 10 contiguous nucleotides thereof, wherein said oligomer (or contiguous nucleotide portion thereof) may optionally comprise one, two, or three mismatches when compared to the sequence.
  • oligomers include a (contiguous) nucleotide sequence, such as a sequence of 12, 13, 14, 15 or 16 contiguous nucleotides in length, which have a nucleotide sequence selected from a sequence from the group consisting of SEQ ID No 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 1 2, 13, 14, 15, 16, 17, 18, 19, 20, 2 1 , and 22, wherein said oligomer (or contiguous nucleotide portion thereof) may optionally comprise one, two, or three mismatches against said selected sequence.
  • a (contiguous) nucleotide sequence such as a sequence of 12, 13, 14, 15 or 16 contiguous nucleotides in length, which have a nucleotide sequence selected from a sequence from the group consisting of SEQ ID No 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 1 2, 13, 14, 15, 16, 17, 18, 19, 20, 2 1 , and 22, wherein said oligomer (or contiguous nucleotide portion thereof) may optionally comprise one, two
  • the sub-sequence may consist of 1 1 , 12, 13, 14, 15, 16, 17, 1 8, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, or 29 contiguous nucleotides, such as 12 -22, such as
  • the sub-sequence is of the same length as the contiguous nucleotide sequence of the oligomer of the invention.
  • the nucleotide sequence of the oligomer may comprise additional 5' or 3' nucleotides, such as, independently, 1 , 2, 3, 4 or 5 additional nucleotides 5' and/or 3' , which are non-complementary to the target sequence.
  • the oligomer of the invention may, in some embodiments, comprise a contiguous nucleotide sequence which is flanked 5' and or 3' by additional nucleotides.
  • the additional 5' or 3' nucleotides are naturally occurring nucleotides, such as DNA or RNA.
  • the additional 5' or 3' nucleotides may represent region D as referred to in the context of gapmer oligomers herein.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID NO 2, such as SEQ ID NO 44, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 3, such as SEQ ID 45, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 4, such as SEQ ID NO 46, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 5, such as SEQ ID NO 47, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 6, such as SEQ ID NO 48, 49 or 50, or a subsequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 7, such as SEQ ID NO 51 , 52, or 53, or a subsequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 8, such as SEQ ID 54, 55 or 56, or a sub- sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 9, such as SEQ ID 57, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguousnucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 10, such as SEQ ID 58, 59, or 60, or a sub- sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 1 1 , such as SEQ ID 61 , or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 12, such as SEQ ID 62, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 13, such as SEQ ID 63, 64 or 65 or a subsequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 14, such as SEQ ID 66, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 15, such as SEQ ID 67, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 16, such as SEQ ID 68, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 17, such as SEQ ID 69, 70 or 71 , or a subsequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 18, such as SEQ ID 72, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 19, such as SEQ ID 73, 74 or 75, or a sub- sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 20, such as SEQ ID 76, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 21 , such as SEQ ID 77, 78 or 79, or a subsequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the oligomer according to the invention consists or comprises of a nucleotide sequence according to SEQ ID 22, such as SEQ ID 80, or a sub-sequence of at least 10 contiguous nucleotides thereof, such as 1 1 , 12, 13, 14, 15 or 16 contiguous nucleotides thereof.
  • the determination of homology may be made by a simple alignment with the corresponding nucleotide sequence of the compound of the invention and the corresponding region of the nucleic acid which encodes the mammalian androgen receptor (or target nucleic acid), or the reverse complement thereof, and the homology is determined by counting the number of bases which align and dividing by the total number of contiguous nucleotides in the compound of the invention, and multiplying by 100. In such a comparison, if gaps exist, it is preferable that such gaps are merely mismatches rather than areas where the number of nucleotides within the gap differ between the nucleotide sequence of the invention and the target nucleic acid.
  • nucleotide sequence of the oligomer or contiguous nucleotide sequence (a first sequence) and the equivalent contiguous nucleotide sequence of a further sequence selected from either i) a sub-sequence of the reverse complement of the nucleic acid target, such as the mRNA which encodes the androgen receptor protein, such as SEQ ID NO: 1 , and/or ii) the sequence of nucleotides provided herein such as the group consisting of SEQ ID NOS: 2 - 22 and 86 - 106, or sub-sequence thereof. Nucleotide analogues are compared directly to their equivalent or corresponding nucleotides.
  • a First sequence which corresponds to a further sequence under i) or ii) typically is identical to that sequence over the length of the first sequence (such as the contiguous nucleotide sequence) or, as described herein may, in some embodiments, is at least 80% homologous to a corresponding sequence, such as at least 85%, at least 90%, at least 91 %, at least 92%at least 93%, at least 94%, at least 95%, at least 96% homologous, at least 97% homologous, at least 98% homologous, at least 99% homologous, such as 100% homologous (identical).
  • nucleotide analogue and “corresponding nucleotide” are intended to indicate that the nucleotide in the nucleotide analogue and the naturally occurring nucleotide are identical.
  • the "corresponding nucleotide analogue” contains a pentose unit (different from 2-deoxyribose) linked to an adenine.
  • the oligomers comprise or consist of a contiguous nucleotide sequence of a total of 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 contiguous nucleotides in length.
  • the oligomers comprise or consist of a contiguous nucleotide sequence of a total of 10 - 22, such as 12 - 18, such as 13 - 17 or 12 - 16, such as 13, 14, 15, 16 contiguous nucleotides in length.
  • the oligomers comprise or consist of a contiguous nucleotide sequence of a total of 10, 1 1 , 12, 13, or 14 contiguous nucleotides in length.
  • the oligomer according to the invention consists of no more than 22 nucleotides, such as no more than 20 nucleotides, such as no more than 18 nucleotides, such as 15, 16 or 17 nucleotides. In some embodiments the oligomer of the invention comprises less than 20 nucleotides.
  • nucleotide refers to a glycoside comprising a sugar moiety, a base moiety and a covalently linked phosphate group and covers both naturally occurring nucleotides, such as DNA or RNA, preferably DNA, and non-naturally occurring nucleotides comprising modified sugar and/or base moieties, which are also referred to as “nucleotide analogues" herein.
  • Non-naturally occurring nucleotides include nucleotides which have modified sugar moieties, such as bicyclic nucleotides or 2' modified nucleotides, such as 2' substituted nucleotides.
  • Nucleotide analogues are variants of natural nucleotides, such as DNA or RNA nucleotides, by virtue of modifications in the sugar and/or base moieties. Analogues could in principle be merely "silent” or “equivalent” to the natural nucleotides in the context of the oligonucleotide, i. e. have no functional effect on the way the oligonucleotide works to inhibit target gene expression. Such "equivalent” analogues may nevertheless be useful if, for example, they are easier or cheaper to manufacture, or are more stable to storage or manufacturing conditions, or represent a tag or label.
  • the analogues will have a functional effect on the way in which the oligomer works to inhibit expression; for example by producing increased binding affinity to the target and/or increased resistance to intracellular nucleases and/or increased ease of transport into the cell.
  • nucleoside analogues are described by e.g. Freier & Altmann; Nucl. Acid Res. , 1997, 25,4429-4443 and Uhlmann; Curr. Opinion in Drug Development, 2000, 3(2), 293-213, and in Scheme 1 :
  • the oligomer may thus comprise or consist of a simple sequence of natural occurring nucleotides - preferably 2' -deoxynucleotides (referred to here generally as "DNA”), but also possibly ribonucleotides (referred to here generally as "RNA”), or a combination of such naturally occurring nucleotides and one or more non-naturally occurring nucleotides, i. e. nucleotide analogues.
  • nucleotide analogues may suitably enhance the affinity of the oligomer for the target sequence.
  • affinity-enhancing nucleotide analogues in the oligomer can allow the size of the specifically binding oligomer to be reduced, and may also reduce the upper limit to the size of the oligomer before non-specific or aberrant binding takes place.
  • the oligomer comprises at least 2 nucleotide analogues. In some embodiments, the oligomer comprises from 3-8 nucleotide analogues, e.g. 6 or 7 nucleotide analogues. In the by far most preferred embodiments, at least one of said nucleotide analogues is a locked nucleic acid (LNA); for example at least 3 or at least 4, or at least 5, or at least 6, or at least 7, or 8, of the nucleotide analogues may be LNA. In some embodiments all the nucleotides analogues may be LNA.
  • LNA locked nucleic acid
  • the oligomers of the invention which are defined by that sequence may comprise a corresponding nucleotide analogue in place of one or more of the nucleotides present in said sequence, such as LNA units or other nucleotide analogues, which raise the duplex stability/T m of the oligomer/target duplex (i.e. affinity enhancing nucleotide analogues).
  • any mismatches between the nucleotide sequence of the oligomer and the target sequence are preferably found in regions outside the affinity enhancing nucleotide analogues, such as region B as referred to herein, and/or region D as referred to herein, and/or at the site of non modified such as DNA nucleotides in the oligonucleotide, and/or in regions which are 5' or 3' to the contiguous nucleotide sequence.
  • modification of the nucleotide include modifying the sugar moiety to provide a 2'-substituent group or to produce a bridged (locked nucleic acid) structure which enhances binding affinity and may also provide increased nuclease resistance.
  • a preferred nucleotide analogue is LNA, such as oxy-LNA (such as beta-D-oxy-LNA, and alpha-L-oxy-LNA), and/or amino-LNA (such as beta-D-amino-LNA and alpha-L-amino- LNA) and/or thio-LNA (such as beta-D-thio-LNA and alpha-L-thio-LNA) and/or ENA (such as beta-D-ENA and alpha-L-ENA). Most preferred is beta-D-oxy-LNA.
  • oxy-LNA such as beta-D-oxy-LNA, and alpha-L-oxy-LNA
  • amino-LNA such as beta-D-amino-LNA and alpha-L-amino- LNA
  • thio-LNA such as beta-D-thio-LNA and alpha-L-thio-LNA
  • ENA such as beta-D-ENA and alpha-L-ENA
  • nucleotide analogues present within the oligomer of the invention are independently selected from, for example: 2'-0-alkyl-RNA units, 2'-amino-DNA units, 2'-fluoro-DNA units, LNA units, arabino nucleic acid (ANA) units, 2'-fluoro-ANA units, HNA units, 3'fluoro hexitol nucleic acid (3'F-HNA).
  • INA intercalating nucleic acid -Christensen, 2002. Nucl. Acids. Res. 2002 30: 4918-4925, hereby incorporated by reference
  • the nucleotide analogues are 2'-0-methoxyethyl-RNA (2'MOE), 2'-fluoro-DNA monomers or LNA nucleotide analogues, and as such the oligonucleotide of the invention may comprise nucleotide analogues which are independently selected from these three types of analogue, or may comprise only one type of analogue selected from the three types.
  • at least one of said nucleotide analogues is 2'-MOE-RNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2'-MOE-RNA nucleotide units.
  • at least one of said nucleotide analogues is 2'-fluoro DNA, such as 2, 3, 4, 5, 6, 7, 8, 9 or 10 2'- fluoro-DNA nucleotide units.
  • the oligomer according to the invention comprises at least one Locked Nucleic Acid (LNA) unit, such as 1 , 2, 3, 4, 5, 6, 7, or 8 LNA units, such as 3 - 7 or. 4 to 8 LNA units, or 3, 4, 5, 6 or 7 LNA units.
  • LNA Locked Nucleic Acid
  • all the nucleotide analogues are LNA.
  • the oligomer may comprise both beta-D-oxy- LNA, and one or more of the following LNA units: thio-LNA, amino-LNA, oxy-LNA, and/or ENA in either the beta-D or alpha-L configurations or combinations thereof.
  • all LNA cytosine units are 5'methyl-Cytosine.
  • the oligomer may comprise both LNA and DNA units.
  • the combined total of LNA and DNA units is 10-25, preferably 10-20, even more preferably 12- 16.
  • the nucleotide sequence of the oligomer such as the contiguous nucleotide sequence consists of at least one LNA and the remaining nucleotide units are DNA units.
  • the oligomer comprises only LNA nucleotide analogues and naturally occurring nucleotides (such as RNA or DNA, most preferably DNA nucleotides), optionally with modified internucleotide linkages such as phosphorothioate.
  • nucleobase refers to the base moiety of a nucleotide and covers both naturally occurring a well as non-naturally occurring variants. Thus, “nucleobase” covers not only the known purine and pyrimidine heterocycles but also heterocyclic analogues and tautomeres thereof.
  • nucleobases include, but are not limited to adenine, guanine, cytosine, thymidine, uracil, xanthine, hypoxanthine, 5-methylcytosine, isocytosine pseudoisocytosine, 5-bromouracil, 5-propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopunne, and 2-chloro-6-aminopurine.
  • At least one of the nucleobases present in the oligomer is a modified nucleobase selected from the group consisting of 5-methylcytosine, isocytosine, pseudoisocytosine, 5-bromouracil, 5-propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, and 2-chloro-6-aminopurine.
  • LNA LNA
  • LNA refers to a bicyciic nucleotide analogue, known as "Locked Nucleic Acid”. It may refer to an LNA monomer, or, when used in the context of an "LNA oligonucleotide” refers to an oligonucleotide containing one or more such bicyciic nucleotide analogues.
  • the LNA used in the oligonucleotide compounds of the invention preferably has the struct
  • X is selected from -0-, -S-, -N(R N *)-, -C(R 6 R 6* )-;
  • B is selected from hydrogen, optionally substituted C alkoxy, optionally substituted
  • _4-alkyl optionally substituted C acyloxy, nucleobases, DNA intercalators,
  • thermochemically active groups thermochemically active groups, chelating groups, reporter groups, and ligands
  • P designates the radical position for an intemucleoside linkage to a succeeding monomer, or a 5'-terminal group, such intemucleoside linkage or 5'-terminal group optionally including the substituent R 5 or equally applicable the substituent R 5 *;
  • P* designates an intemucleoside linkage to a preceding monomer, or a 3'-terminal group
  • Z is selected from -0-, -S-, and -N(R a )-
  • R a and R b each is independently selected from hydrogen, optionally substituted Ci_i 2 -alkyl, optionally substituted CM 2 - alkenyl, optionally substituted C2-i 2 -alkynyl, hydroxy, Ci_i 2 -alkoxy, C 2 .
  • 6-alkyl-aminocarbonyl C alkyl-carbonylamino, carbamido, C alkanoyloxy, sulphono, Ci-6-alkylsulphonyloxy, nitro, azido, sulphanyl, C
  • ⁇ -alkylthio, halogen, DNA intercalators, photochemically active groups, thermochemically active groups, chelating groups, reporter groups, and ligands, where aryl and heteroaryl may be optionally substituted and where two geminal substituents R a and R b together may designate optionally substituted methylene ( CH 2 ), and each of the substituents R 1 * , R 2 , R 3 , R 5 , R 5 *, R 6 and R 6 *, which are present is independently selected from hydrogen, optionally substituted C
  • 6-alkyl)-amino-carbonyl amino-Ci.6-alkyl-aminocarbonyl, mono- and di(C
  • R 4 * and R 2* together designate a biradical selected from -
  • asymmetric groups may be found in either R or 5 orientation.
  • the LNA used in the oligomer of the invention comprises at least one LNA unit according to any of the formulas
  • Y is -0-, -0-CH 2 - ,-S-, -NH-, or N(R H );
  • Z and Z* are independently selected among an intemucleotide linkage, a terminal group or a protecting group; B constitutes a natural or non-natural nucleotide base moiety, and R H is selected from hydrogen and C
  • thio-LNA comprises a locked nucleotide in which Y in the general formula above is selected from S or -CH 2 -S-.
  • Thio-LNA can be in either the beta-D and alpha-L- configuration.
  • amino-LNA refers to a locked nucleotide in which Y in the general formula above is selected from -N(H)-, N(R)-, CH 2 -N(H)-, and -CH 2 -N(R)- where R is selected from hydrogen and C
  • Amino-LNA can be in either the beta-D and alpha-L-configuration.
  • Oxy-LNA refers to a locked nucleotide in which Y in the general formula above represents -O- or -CH 2 -0-. Oxy-LNA can be in either the beta-D and alpha-L- configuration.
  • ENA refers to a locked nucleotide in which Y in the general formula above is -CH2-O- (where the oxygen atom of -CH2-O- is attached to the 2'-position relative to the base B).
  • LNA is selected from beta-D-oxy-LNA, alpha-L-oxy-LNA, beta-D-amino-LNA and beta-D-thio-LNA, in particular beta-D-oxy-LNA.
  • an oligomeric compound may function via non RNase mediated degradation of target mRNA, such as by steric hindrance of translation, or other methods, however, the preferred oligomers of the invention are capable of recruiting an
  • RNase endoribonuclease
  • the oligomer, or contiguous nucleotide sequence comprises of a region of at least 6, such as at least 7 consecutive nucleotide units, such as at least 8 or at least 9 consecutive nucleotide units (residues), including 7, 8, 9, 10, 1 1 , 12, 1 3, 14, 15 or 16 consecutive nucleotides, which, when formed in a duplex with the complementary target RNA is capable of recruiting RNase.
  • the contiguous sequence which is capable of recruiting RNAse may be region B as referred to in the context of a gapmer as described herein.
  • the size of the contiguous sequence which is capable of recruiting RNAse, such as region B may be higher, such as 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19 or 20 nucleotide units.
  • EP 1 222 309 provides in vitro methods for determining RNaseH activity, which may be used to determine the ability to recruit RNaseH.
  • a oligomer is deemed capable of recruiting RNase H if, when provided with the complementary RNA target, it has an initial rate, as measured in pmol/l/min, of at least 1 %, such as at least 5%, such as at least 10% or less than 20% of the equivalent DNA only oligonucleotide, with no 2' substitutions, with phosphorothioate linkage groups between all nucleotides in the oligonucleotide, using the methodology provided by Example 91 - 95 of EP 1 222 309.
  • an oligomer is deemed essentially incapable of recruiting RNaseH if, when provided with the complementary RNA target, and RNaseH, the RNaseH initial rate, as measured in pmol/l/min, is less than 1 %, such as less than 5%,such as less than 10% or less than 20% of the initial rate determined using the equivalent DNA only oligonucleotide, with no 2' substitutions, with phosphorothioate linkage groups between all nucleotides in the oligonucleotide, using the methodology provided by Example 91 - 95 of EP 1 222 309.
  • an oligomer is deemed capable of recruiting RNaseH if, when provided with the complementary RNA target, and RNaseH, the RNaseH initial rate, as measured in pmol/l/min, is at least 20%, such as at least 40 %, such as at least 60 %, such as at least 80 % of the initial rate determined using the equivalent DNA only oligonucleotide, with no 2' substitutions, with phosphorothioate linkage groups between all nucleotides in the oligonucleotide, using the methodology provided by Example 91 - 95 of EP 1 222 309.
  • the region of the oligomer which forms the consecutive nucleotide units which, when formed in a duplex with the complementary target RNA is capable of recruiting RNase consists of nucleotide units which form a DNA/RNA like duplex with the RNA target - and include both DNA units and LNA units which are in the alpha-L configuration, particularly preferred being alpha-L-oxy LNA.
  • the oligomer of the invention may comprise a nucleotide sequence which comprises both nucleotides and nucleotide analogues, and may be in the form of a gapmer, a headmer or a mixmer.
  • a headmer is defined by a contiguous stretch of non-RNase recruiting nucleotide analogues at the 5'-end followed by a contiguous stretch of DNA or modified nucleotide units recognizable and cleavable by the RNase towards the 3'-end (such as at least 7 such nucleotides), and a tailmer is defined by a contiguous stretch of DNA or modified nucleotides recognizable and cleavable by the RNase at the 5'-end (such as at least 7 such nucleotides), followed by a contiguous stretch of non-RNase recruiting nucleotide analogues towards the 3'-end.
  • mixmers consisting of an alternate composition of DNA or modified nucleotides recognizable and cleavable by RNase and non- RNase recruiting nucleotide analogues.
  • Some nucleotide analogues may also be able to mediate RNaseH binding and cleavage. Since a-L-LNA recruits RNaseH activity to a certain extent, smaller gaps of DNA or modified nucleotides recognizable and cleavable by the RNaseH for the gapmer construct might be required, and more flexibility in the mixmer construction might be introduced.
  • the oligomer of the invention is a gapmer.
  • a gapmer oligomer is an oligomer which comprises a contiguous stretch of nucleotides which is capable of recruiting an RNAse, such as RNAseH, such as a region of at least 6 or 7 DNA nucleotides, referred to herein in as region B, wherein region B is flanked both 5' and 3 ' by regions of affinity enhancing nucleotide analogues, such as 1 - 6 nucleotide analogues 5' and 3' to the contiguous stretch of nucleotides which is capable of recruiting RNAse - these regions are referred to as regions A and G respectively.
  • the gapmer comprises a (poly)nucleotide sequence of formula (5' to 3'), A- B-C, or optionally A-B-C-D or D-A-B-C, wherein; region A (5' region) consists or comprises of at least one nucleotide analogue, such as at least one LNA unit, such as 1 -6 nucleotide analogues, such as LNA units, and; region B consists or comprises of at least five consecutive nucleotides which are capable of recruiting RNAse (when formed in a duplex with a complementary RNA molecule, such as the mRNA target), such as DNA nucleotides, and; region C (3' region) consists or comprises of at least one nucleotide analogue, such as at least one LNA unit, such as 1 -6 nucleotide analogues, such as LNA units, and; region D, when present consists or comprises of 1 , 2 or 3 nucleotide units, such as DNA nucle
  • region A consists of 1 , 2, 3, 4, 5 or 6 nucleotide analogues, such as LNA units, such as 2-5 nucleotide analogues, such as 2-5 LNA units, such as 3 or 4 nucleotide analogues, such as 3 or 4 LNA units; and/or region C consists of 1 , 2, 3, 4, 5 or 6 nucleotide analogues, such as LNA units, such as 2-5 nucleotide analogues, such as 2-5 LNA units, such as 3 or 4 nucleotide analogues, such as 3 or 4 LNA units.
  • B consists or comprises of 5, 6, 7, 8, 9, 10, 1 1 or 12 consecutive nucleotides which are capable of recruiting RNAse, or 6- 10, or 7-9, such as 8 consecutive nucleotides which are capable of recruiting RNAse.
  • region B consists or comprises at least one DNA nucleotide unit, such as 1 - 12 DNA units, preferably 4- 12 DNA units, more preferably 6- 10 DNA units, such as 7- 10 DNA units, most preferably 8, 9 or 10 DNA units.
  • region A consist of 3 or 4 nucleotide analogues, such as LNA
  • region B consists of 7, 8, 9 or 10 DNA units
  • region C consists of 3 or 4 nucleotide analogues, such as LNA.
  • Such designs include (A-B-C) 3- 10-3, 3- 10-4, 4- 10-3, 3-9-3, 3-9-4, 4-9-3, 3-8-3, 3-8-4, 4-8-3, 3-7-3, 3-7-4, 4-7-3, and may further include region D, which may have one or 2 nucleotide units, such as DNA units.
  • the oligomer is consisting of a contiguous nucleotide sequence of a total of 10, 1 1 , 12, 13 or 14 nucleotide units, wherein the contiguous nucleotide, sequence is of formula (5' - 3' ), A-B-C, or optionally A-B-C-D or D-A-B-C, wherein; A consists of 1 , 2 or 3 nucleotide analogue units, such as LNA units; B consists of 7, 8 or 9 contiguous nucleotide units which are capable of recruiting RNAse when formed in a duplex with a complementary RNA molecule (such as a mRNA target); and C consists of 1 , 2 or 3 nucleotide analogue units, such as LNA units.
  • D consists of a single DNA unit.
  • A consists of 1 LNA unit.
  • A consists of 2 LNA units.
  • A consists of 3 LNA units.
  • C consists of 1 LNA unit.
  • C consists of 2 LNA units.
  • C consists of 3 LNA units.
  • B consists of 7 nucleotide units.
  • B consists of 8 nucleotide units.
  • B consists of 9 nucleotide units.
  • B comprises of 1 - 9 DNA units, such as 2, 3, 4, 5, 6, 7 or 8 DNA units.
  • B consists of DNA units.
  • B comprises of at least one LNA unit which is in the alpha-L configuration, such as 2, 3, 4, 5, 6, 7, 8 or 9 LNA units in the alpha-L-configuration. In some embodiments B comprises of at least one alpha-L-oxy LNA unit or wherein all the LNA units in the alpha- L- configuration are alpha-L-oxy LNA units.
  • the number of nucleotides present in A-B-C are selected from the group consisting of (nucleotide analogue units - region B - nucleotide analogue units): 1 -8- 1 , 1 -8-2, 2-8- 1 , 2-8-2, 3-8-3, 2-8-3, 3-8-2, 4-8- 1 , 4-8-2, 1 -8-4, 2-8-4, or; 1 -9- 1 , 1 -9-2, 2-9- 1 , 2-9-2, 2-9-3, 3-9-2, 1 -9-3, 3-9- 1 , 4-9- 1 , 1 -9- 4, or; 1 - 10- 1 , 1 - 10-2, 2- 10- 1 , 2- 10-2, 1 - 10-3, 3- 10-1 .
  • the number of nucleotides in A-B-C are selected from the group consisting of: 2-7- 1 , 1 -7-2, 2-7-2, 3-7-3, 2- 7-3, 3-7-2, 3-7-4, and 4-7-3.
  • both A and C consists of two LNA units each, and B consists of 8 or 9 nucleotide units, preferably DNA units.
  • linkage group or "internucleotide linkage” are intended to mean a group capable of covalently coupling together two nucleotides, two nucleotide analogues, and a nucleotide and a nucleotide analogue, etc.
  • Specific and preferred examples include phosphate groups and phosphorothioate groups.
  • nucleotides of the oligomer of the invention or contiguous nucleotides sequence thereof are coupled together via linkage groups.
  • each nucleotide is linked to the 3' adjacent nucleotide via a linkage group.
  • Suitable internucleotide linkages include those listed within PCT D 2006/000512, for example the internucleotide linkages listed on the first paragraph of page 34 of
  • internucleotide linkage from its normal phosphodiester to one that is more resistant to nuclease attack, such as phosphorothioate or boranophosphate - these two, being cleavable by RNase H, also allow that route of antisense inhibition in reducing the expression of the target gene.
  • Suitable sulphur (S) containing intemucleotide linkages as provided herein may be preferred.
  • Phosphorothioate intemucleotide linkages are also preferred, particularly for the gap region (B) of gapmers.
  • Phosphorothioate linkages may also be used for the flanking regions (A and C, and for linking A or C to D, and within region D, as appropriate).
  • Regions A, B and C may however comprise intemucleotide linkages other than phosphorothioate, such as phosphodiester linkages, particularly, for instance when the use of nucleotide analogues protects the intemucleotide linkages within regions A and C from endo- nuclease degradation - such as when regions A and C comprise LNA nucleotides.
  • the intemucleotide linkages in the oligomer may be phosphodiester, phosphorothioate or boranophosphate so as to allow RNase H cleavage of targeted RNA.
  • Phosphorothioate is preferred, for improved nuclease resistance and other reasons, such as ease of manufacture.
  • nucleotides and/or nucleotide analogues are linked to each other by means of phosphorothioate groups.
  • all remaining linkage groups are either phosphodiester or phosphorothioate, or a mixture thereof.
  • all the intemucleotide linkage groups are phosphorothioate.
  • linkages are phosphorothioate linkages
  • alternative linkages such as those disclosed herein may be used, for example phosphate (phosphodiester) linkages may be used, particularly for linkages between nucleotide analogues, such as LNA, units.
  • the oligomers of the invention may, for example, be selected from the group consisting of: 22 - 43 and 44 to 80. Conjugates
  • conjugate indicates a heterogenous molecule formed by the covalent attachment (“conjugation") of the oligomer as described herein to one or more non-nucleotide, or non-polynucleotide moieties.
  • non- nucleotide or non- polynucleotide moieties include macromolecular agents such as proteins, fatty acid chains, sugar residues, glycoproteins, polymers, or combinations thereof.
  • proteins may be antibodies for a target protein.
  • Typical polymers may be polyethylene glycol.
  • the oligomer of the invention may comprise both a polynucleotide region which typically consists of a contiguous sequence of nucleotides, and a further non-nucleotide region.
  • the compound may comprise non-nucleotide components, such as a conjugate component.
  • the oligomeric compound is linked to ligands/conjugates, which may be used, e.g. to increase the cellular uptake of oligomeric compounds.
  • ligands/conjugates which may be used, e.g. to increase the cellular uptake of oligomeric compounds.
  • WO2007/031091 provides suitable ligands and conjugates, which are hereby incorporated by reference.
  • the invention also provides for a conjugate comprising the compound according to the invention as herein described, and at least one non-nucleotide or non-polynucleotide moiety covalently attached to said compound. Therefore, in various embodiments where the compound of the invention consists of a specified nucleic acid or nucleotide sequence, as herein disclosed, the compound may also comprise at least one non-nucleotide or non- polynucleotide moiety (e.g. not comprising one or more nucleotides or nucleotide analogues) covalently attached to said compound.
  • Conjugation may enhance the activity, cellular distribution or cellular uptake of the oligomer of the invention.
  • moieties include, but are not limited to, antibodies, polypeptides, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g.
  • a phospholipids e.g., di-hexadecyl-rac-glycerol or triethylammonium 1 ,2- di-o-hexadecyl-rac-glycero-3-h-phosphonate,
  • the oligomers of the invention may also be conjugated to active drug substances, for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • active drug substances for example, aspirin, ibuprofen, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • the conjugated moiety is a sterol, such as cholesterol.
  • the conjugated moiety comprises or consists of a positively charged polymer, such as a positively charged peptides of, for example 1 -50, such as 2 - 20 such as 3 - 10 amino acid residues in length, and/or polyalkylene oxide such as
  • the positively charged polymer such as a polyalkylene oxide may be attached to the oligomer of the invention via a linker such as the releasable inker described in WO 2008/034123.
  • conjugate moieties may be used in the conjugates of the invention: 5'- OLIGOM E R -3' o
  • activated oligomer refers to an oligomer of the invention that is covalently linked (i.e., functionalized) to at least one functional moiety that permits covalent linkage of the oligomer to one or more conjugated moieties, i.e., moieties that are not themselves nucleic acids or monomers, to form the conjugates herein described.
  • a functional moiety will comprise a chemical group that is capable of covalently bonding to the oligomer via, e.g., a 3'-hydroxyl group or the exocyclic NH 2 group of the adenine base, a spacer that is preferably hydrophilic and a terminal group that is capable of binding to a conjugated moiety (e.g., an amino, sulfhydryl or hydroxyl group).
  • this terminal group is not protected, e.g., is an NH 2 group.
  • the terminal group is protected, for example, by any suitable protecting group such as those described in "Protective Groups in Organic Synthesis” by Theodora W Greene and Peter G M Wuts, 3rd edition (John Wiley & Sons, 1999).
  • suitable hydroxyl protecting groups include esters such as acetate ester, aralkyl groups such as benzyl, diphenylmethyl, or triphenylmethyl, and tetrahydropyranyl.
  • suitable amino protecting groups include benzyl, alpha-methylbenzyl, diphenylmethyl, triphenylmethyl, benzyloxycarbonyl, tert-butoxycarbonyl, and acyl groups such as trichloroacetyl or trifluoroacetyl.
  • the functional moiety is self-cleaving. In other embodiments, the functional moiety is biodegradable. See e.g., U.S. Patent No. 7,087,229, which is incorporated by reference herein in its entirety.
  • oligomers of the invention are functionalized at the 5' end in order to allow covalent attachment of the conjugated moiety to the 5 ' end of the oligomer.
  • oligomers of the invention can be functionalized at the 3' end.
  • oligomers of the invention can be functionalized along the backbone or on the heterocyclic base moiety.
  • oligomers of the invention can be functionalized at more than one position independently selected from the 5' end, the 3' end, the backbone and the base.
  • activated oligomers of the invention are synthesized by incorporating during the synthesis one or more monomers that is covalently attached to a functional moiety. In other embodiments, activated oligomers of the invention are synthesized with monomers that have not been functionalized, and the oligomer is functionalized upon completion of synthesis.
  • the oligomers are functionalized with a hindered ester containing an aminoalkyl linker, wherein the alkyl portion has the formula (CH2) W , wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the alkylamino group can be straight chain or branched chain, and wherein the functional group is attached to the oligomer via an ester group (-0-
  • the oligomers are functionalized with a hindered ester containing a (CH 2 ) w -sulfhydryl (SH) linker, wherein w is an integer ranging from 1 to 10, preferably about 6, wherein the alkyl portion of the alkylamino group can be straight chain or branched chain, and wherein the functional group attached to the oligomer via an ester group
  • sulfhydryl-activated oligonucleotides are conjugated with polymer moieties such as polyethylene glycol or peptides (via formation of a disulfide bond).
  • Activated oligomers containing hindered esters as described above can be synthesized by any method known in the art, and in particular by methods disclosed in International Pub. No. WO 2008/034122 and the examples therein, which is incorporated herein by reference in its entirety.
  • the oligomers of the invention are functionalized by introducing sulfhydryl, amino or hydroxyl groups into the oligomer by means of a functionalizing reagent substantially as described in U.S. Patent Nos. 4,962,029 and 4,914,210, i.e., a substantially linear reagent having a phosphoramidite at one end linked through a hydrophilic spacer chain to the opposing end which comprises a protected or unprotected sulfhydryl, amino or hydroxyl group.
  • a functionalizing reagent substantially as described in U.S. Patent Nos. 4,962,029 and 4,914,210, i.e., a substantially linear reagent having a phosphoramidite at one end linked through a hydrophilic spacer chain to the opposing end which comprises a protected or unprotected sulfhydryl, amino or hydroxyl group.
  • Such reagents primarily react with hydroxyl groups of the oligomer.
  • such activated oligomers have a functionalizing reagent coupled to a 5 '-hydroxyl group of the oligomer. In other embodiments, the activated oligomers have a functionalizing reagent coupled to a 3'- hydroxyl group. In still other embodiments, the activated oligomers of the invention have a functionalizing reagent coupled to a hydroxyl group on the backbone of the oligomer. In yet further embodiments, the oligomer of the invention is functionalized with more than one of the functionalizing reagents as described in U.S. Patent Nos. 4,962,029 and 4,914,210, incorporated herein by reference in their entirety. Methods of synthesizing such functionalizing reagents and incorporating them into monomers or oligomers are disclosed in U.S. Patent Nos. 4,962,029 and 4,914,210.
  • the 5'-terminus of a solid-phase bound oligomer is
  • a dienyl phosphoramidite derivative functionalized with a dienyl phosphoramidite derivative, followed by conjugation of the deprotected oligomer with, e.g., an amino acid or peptide via a Diels-Alder cycloaddition reaction.
  • the incorporation of monomers containing 2'-sugar modifications; such as a 2'-carbamate substituted sugar or a 2'-(0-pentyl-N-phthalimido)- deoxyribose sugar into the oligomer facilitates covalent attachment of conjugated moieties to the sugars of the oligomer.
  • an oligomer with an amino-containing linker at the 2'-position of one or more monomers is prepared using a reagent such as, for example, 5'-dimethoxytrityl-2'-0-(e-phthalimidylaminopentyl)-2'-deoxyadenosine-3' ⁇ N,N- diisopropyl-cyanoethoxy phosphoramidite. See, e.g., Manoharan, et al., Tetrahedron Letters, 1991 , 34, 7171.
  • the oligomers of the invention may have amine-containing functional moieties on the nucleobase, including on the N6 purine amino groups, on the exocyclic N2 of guanine, or on the N4 or 5 positions of cytosine.
  • such functionalization may be achieved by using a commercial reagent that is already functionalized in the oligomer synthesis.
  • heterobifunctional and homobifunctional linking moieties are available from the Pierce Co. (Rockford, 111.).
  • Other commercially available linking groups are 5'-Amino-Modifier C6 and 3'-Amino- Modifier reagents, both available from Glen Research Corporation (Sterling, Va.).
  • 5'-Amino- Modifier C6 is also available from ABI (Applied Biosystems Inc., Foster City, Calif.) as
  • Aminolink-2, and 3'-Amino-Modifier is also available from Clontech Laboratories Inc. (Palo Alto, Calif.).
  • the oligomer of the invention may be used in pharmaceutical formulations and compositions.
  • such compositions comprise a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
  • PCT7D 2006/000512 provides suitable and preferred
  • Suitable dosages, formulations, administration routes, compositions, dosage forms, combinations with other therapeutic agents, pro-drug formulations are also provided in PCT/D 2006/000512 - which are also hereby incorporated by reference.
  • the oligomers of the invention may be utilized as research reagents for, for example, diagnostics, therapeutics and prophylaxis.
  • oligomers may be used to specifically inhibit the synthesis of androgen receptor protein (typically by degrading or inhibiting the mRNA and thereby prevent protein formation) in cells and experimental animals thereby facilitating functional analysis of the target or an appraisal of its usefulness as a target for therapeutic intervention.
  • androgen receptor protein typically by degrading or inhibiting the mRNA and thereby prevent protein formation
  • the oligomers may be used to detect and quantitate androgen receptor expression in cell and tissues by Northern blotting, in-situ hybridisation or similar techniques.
  • an animal or a human, suspected of having a disease or disorder, which can be treated by modulating the expression of androgen receptor is treated by administering antisense compounds in accordance with this invention, suitably in an effective amount.
  • methods of treating a mammal, such as treating a human, suspected of having or being prone to a disease or condition, associated with expression and/or activity of androgen receptor by administering a therapeutically or prophylactically effective amount of one or more of the oligomers or compositions of the invention.
  • T e pharmaceutical composition according to the invention may be used for the treatment of conditions associated with abnormal levels of androgen receptor activity, such as cancer, such as prostate or breast cancer,
  • the pharmaceutical composition according to the invention may be used for the treatment of alopecia, benign prostatic hyperplasia, spinal and muscular atrophy and
  • treatment as referred to herein may be prophylactic.
  • PCT/DK2006/000512 Suitable dosages, formulations, administration routes, compositions, dosage forms, combinations with other therapeutic agents, pro-drug formulations are also provided in PCT/DK2006/000512 - which are hereby incorporated by reference., although it should be recognised that the aspects of PCT/DK2006/000512 which are only specifically applicable to the treatment of cancer may not be appropriate in the therapeutic/pharmaceutical compositions and methods of the present invention.
  • the invention also provides for a pharmaceutical composition
  • a pharmaceutical composition comprising a compound or a conjugate as herein described or a conjugate, and a pharmaceutically acceptable diluent, carrier or adjuvant.
  • PCT DK2006/0005 12 provides suitable and preferred pharmaceutically acceptable diluent, carrier and adjuvants - which are hereby incorporated by reference.
  • the oligomer, a conjugate or a pharmaceutical composition according to the invention is typically administered in an effective amount.
  • the invention also provides for the use of the compound or conjugate of the invention as described for the manufacture of a medicament for the treatment of a disorder as referred to herein, or for a method of the treatment of as a disorder as referred to herein.
  • the invention also provides for a method for treating a disorder as referred to herein said method comprising administering a compound according to the invention as herein described, and/or a conjugate according to the invention, and/or a pharmaceutical composition according to the invention to a patient in need thereof.
  • Pharmaceutical compositions comprising more than one active ingredient
  • the pharmaceutical composition according to the invention may further comprise other active ingredients, including those which are indicated as being useful for the treatment of cancer such as prostate cancer or breast cancer, particularly agents used in conventional antiandrogen therapy.
  • Nonsteroidal antiandrogens NSAAs
  • Diarylhydantoin type androgen receptor ligand binding inhibitors or pharmaceutically acceptable salts thereof including, for example, those disclosed in U.S. Patent No. 7,709,517 may be used.
  • Luteinizing hormone-releasing hormone analogs suppress testicular production of androgens to castrate levels.
  • the present invention provides for a combined androgen blockade therapy, characterised in that the therapy comprises administering an antisense oligomer composition according to the invention, and an NSAA and/or LHRH-A agent, which may be administered prior to, during or subsequent to the administering of an antisense oligomer composition of the invention.
  • the antisense oligomer composition and one or more androgen blockade agents may be administered so that their effects are concurrent, i.e., temporally overlapping in the subject.
  • the invention also provides a kit of parts wherein a first part includes an antisense oligomer, conjugate thereof and/or oligomer pharmaceutical composition according to the invention and a further part comprises a Nonsteroidal antiandrogen (such as a
  • diarylhydantoin type androgen receptor ligand binding inhibitor or pharmaceutically acceptable salt thereof and/or bicalutamide) and/or a Luteinizing hormone-releasing hormone analog is provided.
  • One embodiment provides a kit that includes an antisense oligomer,
  • kit of parts may be used in a method of treatment, as referred to herein, where the method comprises administering both the first part and the further part, either simultaneously or one after the other.
  • AR antisense oligomer embodiments of the present invention may, for example, be used in combination with the diarylhydantoin type AR ligand binding inhibitor embodiments described herein and/or bicalutamide and/or LHRH-As.
  • An oligomer of 10-50 nucleobases in length which comprises a contiguous nucleobase sequence of a total of 10-50 nucleobases, wherein said contiguous nucleobase sequence is at least 80% homologous to a corresponding region of a nucleic acid which encodes a mammalian androgen receptor.
  • oligomer according to embodiment 1 or 2 wherein the contiguous nucleobase sequence comprises no more than 3, such as no more than 2 mismatches to the corresponding region of a nucleic acid which encodes a mammalian androgen receptor.
  • sequence of the oligomer consists of the contiguous nucleobase sequence.
  • nucleic acid which encodes a mammalian androgen receptor is the human androgen receptor nucleotide sequence such as SEQ ID No 1 , or a naturally occurring allelic variant thereof.
  • the contiguous nucleobase sequence comprises a contiguous subsequence of at least 7, nucleobase residues which, when formed in a duplex with the complementary androgen receptor target RNA is capable of recruiting RNaseH.
  • the contiguous nucleobase sequence comprises of a contiguous subsequence of at least 8, at least 9 or at least 10 nucleobase residues which, when formed in a duplex with the complementary androgen receptor target RNA is capable of recruiting RNaseH.
  • oligomer according to any one of embodiments 9 or 10 wherein said contiguous subsequence is at least 9 or at least 10 nucleobases in length, such as at least 12 nucleobases or at least 14 nucleobases in length, such as 14, 15 or 16 nucleobases residues which, when formed in a duplex with the complementary androgen receptor target RNA is capable of recruiting RNaseH.
  • oligomer according to any one of embodiments 1 - 14, wherein said oligomer has a length of 14, 15 or 16 nucleobases.
  • oligomer according to any one of embodiments 1 - 16, wherein the oligomer or contiguous nucleobase sequence comprises, or is selected from a corresponding nucleobase sequence present in a nucleotide sequence selected from the group consisting of SEQ ID NO 2 - 22.
  • sequence comprises a total of 2, 3, 4, 5, 6, 7, 8, 9 or 10 affinity enhancing nucleotide analogues! such as 5 and 8 affinity enhancing nucleotide analogues.
  • the oligomer according to any one of embodiments 1 - 19 which comprises at least one affinity enhancing nucleotide analogue, wherein the remaining nucleobases are selected from the group consisting of DNA nucleotides and RNA nucleotides, preferably DNA nucleotides.
  • 21 The oligomer according to any one of embodiments 1 - 20, wherein the oligomer comprises of a sequence of nucleobases of formula, in 5' to 3' direction, A-B-C, and optionally of formula A-B-C-D, wherein:
  • A consists or comprises of at least one nucleotide analogue, such as 1 , 2, 3, 4, 5 or 6 nucleotide analogues, preferably 2-5 nucleotide analogues, preferably 2, 3 or 4 nucleotide analogues, most preferably 2, 3 or 4 consecutive nucleotide analogues and;
  • B consists or comprises at least five consecutive nucleobases which are capable of recruiting RNAseH (when formed in a duplex with a complementary RNA molecule, such as the AR mRNA target), such as DNA nucleobases, such as 5, 6,
  • C consists or comprises of at least one nucleotide analogue, such as 1 , 2, 3, 4, 5, or 6 nucleotide analogues, preferably 2-5 nucleotide analogues, such as 2, 3 or 4 nucleotide analogues, most preferably 2, 3 or 4 consecutive nucleotide analogues, and;
  • D when present, consists or comprises, preferably consists, of one or more DNA nucleotide, such as 1 -3 or 1 -2 DNA nucleotides.
  • region A consists or comprises of 2, 3 or 4 consecutive nucleotide analogues.
  • region B consists or comprises of 7, 8, 9 or 10 consecutive DNA nucleotides or equivalent nucleobases which are capable of recruiting RNAseH when formed in a duplex with a
  • complementary RNA such as the androgen receptor mRNA target.
  • oligomer according to any one of embodiments 21 - 23, wherein region C consists or comprises of 2, 3 or 4 consecutive nucleotide analogues.
  • nucleobases which are capable of recruiting RNAseH when formed in a duplex with a complementary RNA ,such as the androgen receptor mRNA target;
  • C Consists or comprises of 3 contiguous nucleotide analogues
  • D Consists where present, of one or two DNA nucleotides.
  • oligomer according to embodiment 26 wherein the contiguous nucleobase sequence consists of 10, 1 1 , 12, 13 or 14 nucleobases, and wherein;
  • LNA Locked Nucleic Acid
  • he oligomer according to any one of embodiments 1 - 30, which comprises 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 LNA units such as 2 and 8 nucleotide LNA units.
  • the LNAs are independently selected from oxy-LNA, thio-LNA, and amino-LNA, in either of the beta- D and alpha-L configurations or combinations thereof.
  • he oligomer according to embodiment 32 wherein the LNAs are all beta-D-oxy-LNA.
  • oligomer according to any one of embodiments 1 - 34, wherein at least one of the nucleobases present in the oligomer is a modified nucleobase selected from the group consisting of 5-methylcytosine, isocytosine, pseudoisocytosine, 5-bromouracil, 5- propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, and 2-chloro-6- aminopurine.
  • oligomer hybridises with a corresponding mammalian androgen receptor mRNA with a T m of at least 50°C.
  • oligomer according to embodiment 38 wherein the oligomer comprises at least one phosphorothioate internucleoside linkage.
  • phosphorothioate linkages adjacent to or between DNA or RNA units, or within region B are phosphorothioate linkages.
  • a conjugate comprising the oligomer according to any one of the embodiments 1 -43 and at least one non-nucleotide or non-polynucleotide moiety covalently attached to said compound.
  • a pharmaceutical composition comprising an oligomer as defined in any of
  • diarylhydantoin type androgen receptor ligand binding inhibitor is synonymous with the term “diarylthiohydantoin androgen receptor ligand binding inhibitor”. It relates to a structure wherein the -N-C(0)-N- portion of the hydantoin ring is replaced with -N-C(S)-N-.
  • diarylhydantoin type androgen receptor ligand binding inhibitor is a compound of the formula (I):
  • R 1 is selected from the group consisting of trifluoromethyl and iodo
  • R 2 and R3 are each independently selected from C
  • R 1 is trifluoromethyl.
  • R 2 and R3 are independently selected from C
  • R 2 and R3 are independently selected from Q-Cg alkyl groups; or R 2 and R3 together with the carbon to which they are linked form a C3-C 17 cycloalkyl group.
  • R 2 and R3 are independently selected from C
  • R2 and R3 are methyl; or R2 and R3 together with the carbon to which they are linked form a C4-C5 cycloalkyl group.
  • R 4 is a halogen selected from bromo, fluoro, chloro, iodo.
  • R is fluoro.
  • R5 is selected from the group consisting of methylcarbamoyl, methoxycarbonyl, acetamido and methanesulfonamino.
  • R5 is methylcarbamoyl.
  • the diarylhydantoin type androgen receptor ligand binding inhibitor is a compound of the formula (II):
  • the diarylhydantoin type androgen receptor ligand binding inhibitor is a compound of the formula (III):
  • diarylhydantoin type androgen receptor ligand binding inhibitor is a compound selected from the group consisting of:
  • alkyl denotes branched or unbranched hydrocarbon chains, preferably having about 1 to about 8 carbons, such as, methyl, ethyl, n-propyl, iso-propyl, n- butyl, sec-butyl, iso-butyl, tert-butyl, 2-methylpentyl pentyl, hexyl, isohexyl, heptyl, 4,4- dimethyl pentyl, octyl, 2,2,4-trimethylpentyl and the like.
  • cycloalkyl as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl.
  • the "cycloalkyl” is a saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 ring.
  • alkenyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbons in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4- pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3- nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8, 12-tetradecatrienyl, and the like.
  • alkynyl refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons in the normal chain, which include one or more triple bonds in the normal chain, such as 2-propynyl, 3-butynyl, 2-butynyl, 4-pentynyl, 3- pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4- decynyl, 3-undecynyl, 4-dodecynyl and the like.
  • arylalkyl refers to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2- phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • arylalkyl refers to alkyl, alkenyl, and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2- phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • halogen or "halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
  • haloalkyl refers to "alkyl” group which is substituted by one to three atoms selected from fluorine, chlorine, bromine, fluorine, and iodine.
  • aryl or “Ar” as employed herein alone or as part of another group refers to monocyclic and polycyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1 -naphthyl and 2-naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings).
  • heterocyclic represents an unsubstituted or substituted stable 5- to 10-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from N, O or S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinyl sulfoxide,
  • heterocyclic aromatic refers to a 5- or 7-membered aromatic ring which includes 1 , 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocycloalkyl ring (e.g. benzothiophenyl, indolyl), and includes possible N- oxides.
  • “Pharmaceutically acceptable” means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically acceptable salts” means salts of compounds of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)-benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid,
  • benzenesulfonic acid p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]oct-2-ene- l -carboxylic acid, glucoheptonic acid, 4,4'-methylenebis(3-hydroxy-2-ene- l -carboxylic acid),
  • 3-phenylpropionic acid trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid and the like.
  • Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases.
  • Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide.
  • Acceptable organic bases include
  • diarylhydantoin type androgen receptor ligand binding inhibitors are incorporated herein by reference and may be used in the present invention.
  • AR inhibitor compounds may be screened using, for example, hormone refractory prostate cancer cells for antagonistic and agonistic activities against AR. For example, see WO/2005/060661 , US05/05529 and US applications US2010/02 10665 and US20100172975, which are all, hereby incorporated by reference.
  • An in vitro assay. Potential AR inhibitor compounds may be subjected to tests using an artificial AR response reporter system in, for example, a hormone refractory prostate cancer cell line.
  • prostate cancer LNCaP cells may be engineered to stably express exogenous AR such that it is at a higher level of than endogenous AR.
  • the exogenous AR has similar properties to endogenous AR in that both are stabilized by a synthetic androgen R 1881 .
  • the AR-over expressed cells may also be engineered to stably incorporate an AR response reporter and the reporter activity of these cells shows features of hormone refractory prostate cancer. Said cells will respond to low concentrations of a synthetic androgen R 1881.
  • Engineered LNCaP cells can be maintained in Iscove's medium containing 10% fetal bovine serum (FBS). Two days prior to drug treatment, the cells are grown in Iscove's medium containing 10% charcoal-stripped FBS (CS-FBS) to deprive of androgens. The cells are split and grown in Iscove's medium containing 10% CS-FBS with 100 pM of R 1881 and increasing concentrations of test compounds. After two days of incubation, reporter activities were assayed.
  • FBS fetal bovine serum
  • bicalutamide will inhibit the AR response reporter and will not have agonistic activity in hormone sensitive prostate cancer cells.
  • the antagonistic activity of the diarylhydantoin type compounds mentioned above can be determined using such a reporter assay.
  • PSA prostate specific antigen
  • LNCaP- AR also abbreviated LN- AR.
  • LNCaP-AR cells are a line of lymph node carcinoma of prostate cells transduced with a plasmid that expresses androgen receptors.
  • LNCaP-AR cells can be maintained in Iscove's medium containing 10% FBS. Two days prior to drug treatment, the cells are grown in Iscove's medium containing 10% CS-FBS to deprive of androgens.
  • the cells are split and grown in Iscove's medium containing 10% CS-FBS with appropriate concentrations of R 1881 and the test compounds. After four days incubation, secreted PSA levels may be assayed using PSA ELISA kits (American Qualex, San Clemente. Calif.).
  • PSA ELISA kits American Qualex, San Clemente. Calif.
  • Other suitable assays to determine the effect of compounds on AR include in vivo assays - US2010/0210665 and US20100172975 describe such an assay; this assay is incorporated herein by reference.
  • Diarylhydantoin derivative androgen receptor ligand binding inhibitors that may be used in combination with AR antisense oligomers according to the invention include those disclosed in U.S. Patent No. 7,709,5 17 which is incorporated by reference in its entirety as if fully set forth herein.
  • diarylhydantoin androgen receptor ligand binding inhibitors for use according to the invention include, for example the following diarylthiohydantoin compounds:
  • the compounds of the invention are useful as pharmaceutical compositions prepared with a therapeutically effective amount of a compound of the invention, as defined herein, and a pharmaceutically acceptable carrier or diluent.
  • the diarylhydantoin compounds may, for example, be formulated as pharmaceutical compositions and administered to a subject in need of treatment, for example a mammal, such as a human patient, in a variety of forms adapted to the chosen route of administration, for example, orally, nasally, intraperitoneally, or parenterally, by intravenous, intramuscular, topical or subcutaneous routes, or by injection into tissue.
  • diarylhydantoin compounds used according to the invention may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier; or by inhalation or insufflation. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier
  • the diarylhydantoin compounds may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • the diarylhydantoin compounds may be combined with a fine inert powdered carrier and inhaled by the subject or insufflated. Such compositions and preparations should contain at least 0.1 % diarylhydantoin compounds. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of a given unit dosage form. The amount of diarylhydantoin compounds in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the diarylhydantoin compounds may be incorporated into sustained-release preparations and devices.
  • the diarylhydantoin compounds may be incorporated into time release capsules, time release tablets, and time release pills.
  • the diarylhydantoin compounds may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the diarylhydantoin compounds can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the diarylhydantoin compounds which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms may be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. It may be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions may be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the diarylhydantoin compounds in the required amount in the appropriate solvent with, for example, various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation include vacuum drying and freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • Useful dosages of the compounds of the diarylhydantoin compounds can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949, which is hereby incorporated by reference.
  • the concentration of the diarylhydantoin compounds in a liquid composition can be from about 0. 1 -25% by weight, or from about 0.5- 10% by weight.
  • concentration in a semi-solid or solid composition such as a gel or a powder can be about 0. 1 -5% by weight, or about 0.5-2.5% by weight.
  • diarylhydantoin compounds and antisense AR oligomers required for use in treatment may vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • Effective dosages and routes of administration of agents of the diarylhydantoin compounds may, for example, be conventional.
  • the exact amount (effective dose) of the agent may vary from subject to subject, depending on, for example, the species, age, weight and general or clinical condition of the subject, the severity or mechanism of any disorder being treated, the particular agent or vehicle used, the method and scheduling of administration, and the like.
  • a therapeutically effective dose can be determined empirically, by conventional procedures known to those of skill in the art. See, e.g.. The Pharmacological Basis of Therapeutics, Goodman and Gilman, eds., Macmillan Publishing Co., New York.
  • an effective dose can be estimated initially either in cell culture assays or in suitable animal models.
  • the animal model may also be used to determine the appropriate concentration ranges and routes of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • a therapeutic dose can also be selected by analogy to dosages for comparable therapeutic agents.
  • the particular mode of administration and the dosage regimen for the diarylhydantoin compounds may be selected by the attending clinician, taking into account the particulars of the case (e.g., the subject, the disease, the disease state involved, and whether the treatment is prophylactic). Treatment may involve daily or multi-daily doses of compound(s) over a period of a few days to months, or even years.
  • a suitable dose will be in the range of from about 0.001 to about 100 mg/kg, e.g., from about 0.01 to about 100 mg/kg of body weight per day, such as above about 0. 1 mg per kilogram, or in a range of from about 1 to about 10 mg per kilogram body weight of the recipient per day.
  • a suitable dose may be about 1 mg/kg, 10 mg/kg, or 50 mg kg of body weight per day.
  • diarylhydantoin compounds are conveniently administered in unit dosage form; for example, containing 0.05 to 10000 mg, 0.5 to 10000 mg, 5 to 1000 mg, or about 100 mg of active ingredient per unit dosage form.
  • the diarylhydantoin compounds may be administered to achieve peak plasma concentrations of, for example, from about 0.5 to about 75 ⁇ , about 1 to 50 ⁇ , about 2 to about 30 ⁇ , or about 5 to about 25 ⁇ .
  • Exemplary desirable plasma concentrations include at least or no more than 0.25, 0.5, 1 , 5, 10, 25, 50, 75, 100 or 200 ⁇ .
  • plasma levels may be from about 1 to 100 micromolar or from about 10 to about 25 micromolar. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the diarylhydantoin compounds, optionally in saline, or orally administered as a bolus containing about 1 - 100 mg of the diarylhydantoin compounds.
  • Desirable blood levels may be maintained by continuous infusion to provide about 0.00005-5 mg per kg body weight per hour, for example at least or no more than 0.00005, 0.0005, 0.005, 0.05, 0.5, or 5 mg/kg/hr.
  • such levels can be obtained by intermittent infusions containing about 0.0002- 20 mg per kg body weight, for example, at least or no more than 0.0002, 0.002, 0.02, 0.2, 2, 20, or 50 mg of the diarylhydantoin compounds per kg of body weight.
  • the diarylhydantoin compounds may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator.
  • one aspect of the invention is directed to a method of treating a mammal, such as a human, suffering from or susceptible to conditions associated with abnormal levels of androgen receptor activity, for example as a result of overexpression of or mutation of AR, that includes administering to the mammal and antisense oligomer targeted to AR, for example, one comprised solely of DNA rather than RNA and including one or more LNA DNA monomers, or a derivative thereof, such as those described herein, for example, in the form of a pharmaceutical composition, for example via intravenous administration, and a diarylhydantoin type androgen receptor ligand binding inhibitor or pharmaceutically acceptable salt thereof including, for-example, those described herein and those disclosed in U.S. Patent No. 7,709,517, for example by oral administration or by intravenous administration or bolus injection.
  • the disorder may, for example, be a cancer such as breast cancer or prostate cancer, for example, advanced prostate cancer, castration-resistant prostate cancer, LHRH-resistant prostate cancer, androgen-sensitive prostate cancer, and androgen-insensitive prostate cancer, benign prostatic hyperplasia, AR-activity dependent breast cancer, alopecia, spinal and muscular atrophy, Kennedy disease and polyglutamate disease.
  • the antisense oligomer may optionally be linked ' to a ligand or conjugate.
  • a conjugate is a sterol, such as cholesterol.
  • the LNA antisense oligomers disclosed herein advantageously do not require conjugation for in vivo therapeutic use in mammals.
  • the invention is furthermore directed to a method for treating abnormal levels of androgen receptor activity, said method comprising administering an antisense oligomer of the invention, or a conjugate of the oligomer or a pharmaceutical composition of the oligomer or conjugate to a patient, such as a human or non-human mammal, in need thereof and further comprising the administration of and a diarylhydantoin type androgen receptor ligand binding inhibitor or pharmaceutically acceptable salt thereof including, for example, those described herein and those disclosed in U.S. Patent No.
  • Said further administration may be such that the further chemotherapeutic agent is conjugated to the antisense oligomer, is present in the pharmaceutical composition, or is administered in a separate formulation.
  • the combination methods of treatment according to the invention may be employed, for example, for at least one week, such as one week, for at least two weeks, such as two weeks, for at least one month, such as one month, for at least two months, such as two, three, four, five or six months, or longer than six months, such as one year.
  • the administration of antisense AR oligomer and antiandrogen compound may at least sometimes occur on the same days or may not occur on the same days; what is important is that each of the agents is present at the same time in the mammal subject.
  • antisense AR oligomer may be administered to the subject biweekly, weekly, every two weeks, or monthly, while the antiandrogen compound, such as MDV3100, is administered daily or every two days.
  • antiandrogen compound such as MDV3100
  • the invention provides a method of regulating genes, and their respective mRNA and protein products, which are modulated by the androgen receptor (i.e. androgen receptor targets) such as genes, mRNA and/or proteins selected form the group consisting of: Protein kinase C delta (PRKCD), Glutathione S- transferase theta 2 (GSTT2), transient receptor potential cation channel subfamily V member 3 (TRPV3), Pyrroline-5-carboxylate reductase 1 (PYCR 1 ) or ornithine aminotransferase (OAT), said methods including the step of administering to a mammal subject such as a human, an antisense oligomer targeting AR and a diarylhydantoin type androgen receptor ligand binding inhibitor or pharmaceutically acceptable salt thereof including, for example, those described herein and those disclosed in U.S. Patent No. 7,709,5 17, in amounts effective together to regulate expression of one or more androgen receptor modulated genes,
  • LNA monomer building blocks and derivatives were prepared following published procedures and references cited therein - see WO07/031081 and the references cited therein.
  • Oligonucleotides were synthesized according to the method described in
  • Table 1 shows examples of antisense oligonucleotide sequences of the invention.
  • Tables 2 and 3 show examples of antisense oligonucleotides (oligos) of the invention.
  • oligonucleotides were designed to target different regions of human Androgen receptor mRNA(GenBank accession number NM_000044, SEQ ID NO: 1 ).
  • Antisense oligonucleotide sequences of the invention are oligo sequences designed to target human Androgen receptor mRNA.
  • the effect of antisense oligonucleotides on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels.
  • the target can be expressed endogenously or by transient or stable transfection of a nucleic acid encoding said target.
  • the expression level of target nucleic acid can be routinely determined using, for example, Northern blot analysis, Real-Time PCR, Ribonuclease protection assays.
  • the following cell types are provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen.
  • Cells were cultured in the appropriate medium as described below and maintained at 37°C at 95-98% humidity and 5% C0 2 . Cells were routinely passaged 2-3 times weekly.
  • the human lung cancer cell line A5439 was cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25 ⁇ g/ml).
  • MCF7 The human breast cancer cell line MCF7 was cultured in EagleMEM (Sigma) +
  • Example 5 In vitro model: Treatment with antisense oligonucleotides
  • the cell lines listed in example 4 were treated with oligonucleotide using the cationic liposome formulation LipofectAMINE 2000 (Gibco) as transfection vehicle.
  • Cells were seeded in 6-well cell culture plates (NUNC) and treated when 80-90% confluent. Oligo concentrations used ranged from 1 nM to 16 nM final concentration.
  • Formulation of oligo- lipid complexes were carried out essentially as described by the manufacturer using serum- free OptiMEM (Gibco) and a final lipid concentration of 5 g/mL LipofectAMINE 2000.
  • Cells were incubated at 37°C for 4 hours and treatment was stopped by removal of oligo- containing culture medium. Cells were washed and serum-containing media was added. After oligo treatment cells were allowed to recover for 20 hours before they were harvested for RNA analysis.
  • Example 6 In vitro model: Extraction of RNA and cDNA synthesis
  • RNA 0.5 ⁇ g total RNA was adjusted to ( 10.8 ⁇ ) with RNase free H2O and mixed with 2 ⁇ random decamers (50 ⁇ ) and 4 ⁇ dNTP mix (2.5 mM each dNTP) and heated to 70 °C for 3 min after which the samples were rapidly cooled on ice. After cooling the samples on ice, 2 ⁇ ⁇ ⁇ Buffer RT, 1 ⁇ MMLV Reverse Transcriptase ( 100 U/ ⁇ ) and 0.25 ⁇ RNase inhibitor ( 10 U/ ⁇ ) was added to each sample, followed by incubation at 42 °C for 60 min, heat inactivation of the enzyme at 95°C for 10 min and then the sample was cooled to 4 °C.
  • Example 7 In vitro model: Analysis of Oligonucleotide Inhibition of Androgen receptor Expression by Real-time PCR
  • Antisense modulation of Androgen receptor expression can be assayed in a variety of ways known in the art. For example, Androgen receptor mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. Real-time quantitative PCR is presently preferred. RNA analysis can be performed on total cellular RNA or mRNA.
  • PCR competitive polymerase chain reaction
  • RNA isolation and RNA analysis such as Northern blot analysis is routine in the art and is taught in, for example, Current Protocols in Molecular Biology, John Wiley and Sons.
  • PCR Real-time quantitative
  • PCR can be conveniently accomplished using the commercially available Multi-Color Real Time PCR Detection System, available from Applied Biosystem.
  • Real-time Quantitative PCR Analysis of Androgen receptor mRNA Levels can be conveniently accomplished using the commercially available Multi-Color Real Time PCR Detection System, available from Applied Biosystem.
  • the sample content of human Androgen receptor mRNA was quantified using the human Androgen receptor ABI Prism Pre-Developed TaqMan Assay Reagents (Applied Biosystems cat. no. Hs00171 172_m l ) according to the manufacturer's instructions.
  • Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA quantity was used as an endogenous control for normalizing any variance in sample preparation.
  • the sample content of human GAPDH mRNA was quantified using the human GAPDH ABI Prism Pre-Developed TaqMan Assay Reagent (Applied Biosystems cat. no. 4310884E) according to the manufacturer's instructions.
  • Real-time Quantitative PCR is a technique well known in the art and is taught in for example Heid et al. Real time quantitative PCR, Genome Research ( 1996), 6: 986-994.
  • Real time PCR The cDNA from the first strand synthesis performed as described in example 6 was diluted 2-20 times, and analyzed by real time quantitative PCR using Taqman 7500 FAST or 7900 FAST from Applied Biosystems. The primers and probe were mixed with 2 x Taqman Fast Universal PCR master mix (2x) (Applied Biosystems Cat.# 4364103) and added to 4 ⁇ cDNA to a final volume of 10 ⁇ . Each sample was analysed in duplicate. Assaying 2 fold dilutions of a cDNA that had been prepared on material purified from a cell line expressing the RNA of interest generated standard curves for the assays. Sterile H 2 O was used instead of cDNA for the no template control.
  • PCR program 95° C for 30 seconds, followed by 40 cycles of 95° C, 3 seconds, 60° C, 20-30 seconds.
  • Relative quantities of target mRNA sequence were determined from the calculated Threshold cycle using the Applied Biosystems Fast System SDS Software Version 1.3.1.21. or SDS Software Version 2.3.
  • Example 8 In vitro analysis: Antisense Inhibition of Human Androgen receptor mRNA Expression by oligonucleotide compounds
  • Oligonucleotides presented in Table 3 were evaluated for their potential to knockdown of the androgen receptor mRNA at concentrations of 1 , 4 and 16 nM (see Figures 1 and 2).
  • Table 3 Antisense Inhibition of Human Androgen receptor mRNA expression by oligonucleotides.
  • Table 3 The data in Table 3 are presented as percentage down-regulation relative to mock transfected cells at 16 nM. Lower case letters represent DNA units, bold upper case letters represent ⁇ -D-oxy-LNA units. All LNA C are 5'methyl C. Subscript "s" represents phosphorothioate linkage. shown in Table 3, oligonucleotides of SEQ ID NOs: 48, 51 , 54, 58, 63, 69, 73 and 77 at nM demonstrated greater than 90% inhibition of Androgen receptor mRNA expression in A549 and MCF7 cells in these experiments and are therefore preferred.
  • oligonucleotides based on the illustrated antisense oligo sequences, for example varying the length (shorter or longer) and/or nucleobase content (e.g. the type and/or proportion of analogue units), which also provide good inhibition of Androgen receptor expression.
  • Example 9 In vivo analysis: Antisense Inhibition of mouse Androgen receptor mRNA liver
  • Nude mice were dosed i.v. q3dx4 with 100 mg/kg oligonucleotide (group size of 5 mice).
  • the antisense oligonucleotides (SEQ ID:48, SEQ ID:51 , SEQ ID:58, SEQ ID:63, SEQ ID:77) were dissolved in 0.9% phosphate buffered saline. Animals were sacrificed 24h after last dosing and liver tissue was sampled and stored in RNA later until RNA extraction and
  • oligonucleotides of SEQ ID NOs: 58 and 77 at 100 mg/kg demonstrated greater than 90% inhibition of Androgen receptor mRNA expression in mouse liver cells in these experiments and are therefore preferred.
  • Example 10 In vitro analysis: Antisense Inhibition of Human Androgen receptor mRNA Measurement of proliferating viable cells (MTS assay)
  • LNCaP prostate cancer and A549 lung cancer cells were seeded to a density of 150000 cells per well in a 6-well plate the day prior to transfection.
  • A549 cells were cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin
  • LNCaP cells were cultured in RPMI 1640 Medium (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25 g/ml). The next day medium was removed followed by addition of 1.2 ml OptiMEM containing 5 g/ml Lipofectamine2000 (Invitrogen). Cells were incubated for 7 min before adding 0.3 ml oligonucleotides diluted in OptiMEM. The final oligonucleotide concentrations were 4 and 16 nM.
  • Viable cells were measured at 490 nm in a Powerwave (Biotek Instruments). The OD490 nm were plotted against time/h. (See Figure 13 and Figure 14). As shown in figure 13 and figure 14, oligonucleotides of SEQ ID NOs: 58 and 77 inhibits growth of both LNCaP prostate and A549 lung cancer cells, and are therefore preferred.
  • Example 11 In vitro analysis: Caspase 3/7 activity by Antisense Inhibition of Human Androgen receptor mRNA
  • LNCaP prostate cancer and A549 lung cancer cells were seeded to a density of 150000 cells per well in a 6-well plate the day prior to transfection.
  • A549 cells were cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25 ⁇ g/ml) whereas LNCaP cells were cultured in RPMI 1640 Medium (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25 ⁇ 1). The next day medium was removed followed by addition of 1.2 ml OptiMEM containing 5 ⁇ Lipofectamine2000 (Invitrogen).
  • oligonucleotides were incubated for 7 min before adding 0.3 ml oligonucleotides diluted in OptiMEM. The final oligonucleotide concentrations were 4 and 16 nM. After 4 hours of treatment, media was removed and cells were trypsinized and seeded to a density of 5000 cells per well in white 96 well plate (Nunc) in 100 ⁇ media. Caspase 3/7 activity was measured at the times indicated by adding 100 ⁇ Caspase-Glo 3/7 assay (promega). Caspase 3/7 activity was measured using a luminometer. The caspase 3/7 activities were measured at three different time points 14h, 48h and 72h (See Figure 15 and Figure 16).
  • oligonucleotides of SEQ ID NOs: 58 and 77 induce caspase 3/7 activity in both LNCaP prostate and A549 lung cancer cells and are therefore preferred.
  • Example 12 In vitro analysis: Antisense Inhibition of Human Androgen receptor mRNA Expression by oligonucleotide compounds in prostate cancer cells LNCaP and the lung cancer cell line A549.
  • Oligonucleotides were evaluated for their potential to knockdown of the androgen receptor mRNA at concentrations of 0.5, 1 , 2, 4, 8 and 16 nM (see Figure 1 1 ).
  • LNCaP prostate cancer and A549 lung cancer cells were seeded to a density of 150000 cells per well in a 6-well plate the day prior to transfection.
  • A549 cells were cultured in DMEM (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25 g/ml) whereas LNCaP cells were cultured in RPMI 1640 Medium (Sigma) + 10% fetal bovine serum (FBS) + 2 mM Glutamax I + gentamicin (25pg/ml). The next day medium was removed followed by addition of 1.2 ml OptiMEM containing 5 g/ml Lipofectamine2000 (Invitrogen). Cells were incubated for 7 min before adding 0.3 ml oligonucleotides diluted in OptiMEM.
  • oligonucleotide concentrations were 0.5, 1 , 2, 4, 8 and 16 nM.
  • Cells were washed and serum- containing media was added. After oligo treatment cells were allowed to recover for 20 hours before they were harvested for RNA analysis.
  • the procedure for RNA isolation, cDNA synthesis and qPCR were as described in example 5, 6 and 7.
  • oligonucleotides of SEQ ID NOs: 58 and 77 were potent in knocking down AR mRNA expression in both the lung cancer cell line A549 and in the androgen receptor-dependent 22RV 1 prostate cancer cell line.
  • mice Six to seven week old male athymic nu/nu mice (Harlan Sprague Dawley) weighing an average of 27.3+2.4g were used in the study. Ten million cells of 22RV 1 (androgen- independent prostate cancer line) were suspended in PBS (Gibco#14190) and Matrigel (BD#356234) with a ratio of 1 : 1 were injected subcutaneously into each mouse. When tumors reach an average volume of 150-200 mm 3 , the mice were divided into nine experimental groups. Two hundred ⁇ of oligo was injected intravenously when the average tumor size reached 152.66+27.97 mm 3 . Oligos were given every 3 days for a total of 5 times.
  • mice were sacrificed and blood collected in EDTA laced tubes and spun for 5 min. 50 ⁇ of the supernatants were then subjected to PSA assay using the ELISA kit from ALPCO
  • Oligos were gi ven every 3 days for a total of 5 times.
  • the control vehicles were given the same regime as the oligos.
  • Example 14 Preparation of conjugates of oligomers with polyethylene glycol
  • the oligomers having sequences shown as SEQ ID NO: 48 or SEQ ID NO: 63 are functionalized on the 5' terminus by attaching an aminoalkyl group, such as hexan- 1 -amine blocked with a blocking group such as Fmoc to the 5' phosphate groups of the oligomers using routine phosphoramidite chemistry, oxidizing the resultant compounds, deprotecting them and purifying them to achieve the functionalized oligomers, respectively, having the formulas (IA) an aminoalkyl group, such as hexan- 1 -amine blocked with a blocking group such as Fmoc to the 5' phosphate groups of the oligomers using routine phosphoramidite chemistry, oxidizing the resultant compounds, deprotecting them and purifying them to achieve the functionalized oligomers, respectively, having the formulas (IA) an aminoalkyl group, such as hexan- 1 -amine blocked with a blocking group such as Fmoc to the 5' phosphate groups
  • each of the oligomers of SEQ ID NOs: 48 and 63 is attached to a PEG polymer having average molecular weight of 12,000 via a releasable linker.
  • Activated oligomers that can be used in this process to respectively make the conjugates shown in formulas (IVA), (IVB) and (IVC) have the chemical structures shown in formulas (VA), (VB) and (VC):
  • a mismatch oligomer control (“MM-oligo") having the sequence 5'-A s C s C s ⁇ s a s / s i s c s a s c s i s fs/ s A s G s C-3' (SEQ ID NO: 107) was used as indicated (mismatched bases indicated in italics; this sequence does not have 100% match with any mRNA in the mouse or human database). Mice were purposely dosed with a low dose of SEQ ID NO:58 (40 mg/kg, q3dx4, i.v.), which showed minimum or no significant tumor growth inhibition (TGI) by itself in this experiment.
  • TGI tumor growth inhibition
  • FIG. 21 A shows effect of the combination of SEQ ID NO:58 and MDV3100 in a colony formation growth assay using the AR-positive, androgen-resistant human cell line, C4-2b-AR.
  • SEQ ID NO:58 or MDV3100 alone showed dose-dependent growth inhibition. However, when combined, growth of colonies was inhibited more effectively than either agent alone.
  • FIG. 2 I B a similar result was obtained when SEQ ID NO:58 was combined with bicalutamide, but only at a higher concentration.
  • C4-2b cells were plated at 400 cells/well in a 24-well plate.
  • FIG. 21 A shows results for SEQ ID NO:58 (indicated EZN-4176) and MDV3100.
  • FIG. 21 B shows results for SEQ ID NO:58 (indicated EZN-4176) and bicalutamide.
  • MDV3100 has demonstrated antitumor effects in castration-resistant prostate cancer (CRPC) patients since it decreased serum PSA by 50% or more in 56% of patients, induced responses in soft tissue in 22% of patients, stabilized bone disease in 56% of patients, and reduced circulating tumor cell counts in 49% of patients (Scher et al., Lancet 375, 1437 (April 24, 2010). Nevertheless, a certain population of patients failed initially to respond to MDV3100 and the median time to progression was approximately 47 weeks. Not only can resistance to MDV3100 occur, but MDV3 100, which competes with androgen for the ligand- binding domain of the AR, may be less effective on AR variants devoid of the ligand-binding domain. Such variants have already been documented in prostate cancer patients.
  • CRPC castration-resistant prostate cancer
  • combination treatment with an antisense oligomer targeting AR such as SEQ ID NO:58
  • diarylhydantoin type AR binding inhibitor such as MDV3100
  • Example 16 In vitro analysis: antisense inhibition of human androgen receptor mRNA expression and prostate specific antigen (PSA) mRNA by oligonucleotide compounds in the prostate cancer cell line C4-2b.
  • PSA prostate specific antigen
  • a method for treating an androgen receptor dependent hyperproliferative disorder in a mammal comprising the steps of:
  • hyperproliferative disorder is selected from the group consisting of prostate cancer, benign prostatic hyperplasia and breast cancer.
  • a kit comprising:
  • composition comprising an antisense oligomer selected from the group consisting of:
  • composition comprising a compound selected from the group consisting of:
  • a method for treating an androgen receptor dependent hyperproliferative disorder in a mammal comprising the steps of:
  • hyperproliferative disorder is prostate cancer.
  • An oligomer of between 10 - 30 nucleotides in length which comprises a contiguous nucleotide sequence of a total of between 10 - 30 nucleotides, wherein said contiguous nucleotide sequence is at least 80% homologous to a region corresponding to a mammalian androgen receptor gene or the reverse complement of an mRNA, such as SEQ ID NO: 1 or naturally occurring variant thereof, wherein said oligomer comprises at least one LNA unit.
  • the contiguous nucleotide sequence is at least 80% homologous to a region corresponding to i) SEQ ID NO 94 or SEQ ID NO 58, ii) SEQ ID NO 105 or SEQ ID NO 77 or iii) a sequence selected from the group consisting of SEQ ID NO: 2 - 22 and 86 - 106. 19.
  • the oligomer according to paragraph 17 or 18, wherein the contiguous nucleotide sequence comprises no mismatches or no more than one or two mismatches with the reverse complement of the corresponding region of 1.
  • the nucleotide sequence of the oligomer consists of the contiguous nucleotide sequence.
  • nucleotide analogues are sugar modified nucleotides, such as sugar modified nucleotides selected from the group consisting of: Locked Nucleic Acid (LNA) units; 2'-0-alkyl-RNA units, 2'-OMe-RNA units, 2'-amino- DNA units, and 2'-fluoro-DNA units.
  • LNA Locked Nucleic Acid
  • a pharmaceutical composition comprising the oligomer according to any one of paragraphs 17 - 27, or the conjugate according to paragraph 28, and a pharmaceutically acceptable diluent, carrier, salt or adjuvant.
  • a method of treating a disease or a medical disorder as disclosed herein, such as a hyperproliferative disorder, such as cancer comprising administering an effective amount of an oligomer according to any one of the paragraphs 17-27, or a conjugate according to paragraph 28, or a pharmaceutical composition according to paragraph 29, to a patient suffering from, or likely to suffer from a disease or a medical disorder as disclosed herein, such as a hyperproliferative disorder, such as cancer.
  • a method for the inhibition of Androgen Receptor in a cell which is expressing the androgen receptor comprising administering an oligomer according to any one of the paragraphs 17-27, or a conjugate according to paragraph 28 to said cell so as to inhibit androgen receptor in said cell.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne l'utilisation combinée d'oligomères antisens ciblant l'ARNm de récepteurs androgènes et d'inhibiteurs de liaison aux récepteurs androgènes qui réduisent l'activité des récepteurs androgènes pour le traitement de troubles médicaux liés aux récepteurs androgènes, tels que les cancers, en particulier les cancers de la prostate et les cancers du sein.
PCT/US2011/060352 2010-11-12 2011-11-11 Compositions et procédés pour le traitement de troubles dépendant des récepteurs androgènes y compris les cancers WO2012065051A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP11839515.1A EP2646553A4 (fr) 2010-11-12 2011-11-11 Compositions et procédés pour le traitement de troubles dépendant des récepteurs androgènes y compris les cancers
AU2011326034A AU2011326034B2 (en) 2010-11-12 2011-11-11 Compositions and methods for treating androgen receptor dependent disorders including cancers

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US12/945,551 2010-11-12
US12/945,551 US8450290B2 (en) 2007-11-26 2010-11-12 Methods for treating androgen receptor dependent disorders including cancers
GB1101969.2 2011-02-04
GBGB1101969.2A GB201101969D0 (en) 2011-02-04 2011-02-04 Methods

Publications (1)

Publication Number Publication Date
WO2012065051A1 true WO2012065051A1 (fr) 2012-05-18

Family

ID=46051313

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/060352 WO2012065051A1 (fr) 2010-11-12 2011-11-11 Compositions et procédés pour le traitement de troubles dépendant des récepteurs androgènes y compris les cancers

Country Status (3)

Country Link
EP (1) EP2646553A4 (fr)
AU (1) AU2011326034B2 (fr)
WO (1) WO2012065051A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014059364A1 (fr) * 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Procédés de traitement de la maladie de kennedy
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
EP3327144A1 (fr) 2013-02-25 2018-05-30 Novartis AG Nouvelle mutation du récepteur d'androgène
US20220193110A1 (en) * 2020-12-17 2022-06-23 Washington University Nxtar-derived oligonucleotides and uses thereof

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4914210A (en) 1987-10-02 1990-04-03 Cetus Corporation Oligonucleotide functionalizing reagents
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US4962029A (en) 1987-10-02 1990-10-09 Cetus Corporation Covalent oligonucleotide-horseradish peroxidase conjugate
EP0692972A1 (fr) 1993-04-02 1996-01-24 Anticancer, Inc. Procede permettant l'apport aux follicules pileux de compositions profitables
WO1997011170A1 (fr) 1995-09-20 1997-03-27 Worcester Foundation For Biomedical Research Chimiotherapie par oligonucleotides antisens de l'hypertrophie ou du cancer de la prostate
WO2001083740A2 (fr) 2000-05-04 2001-11-08 Avi Biopharma, Inc. Composition anti-sens a region d'epissage et methode associee
EP1222309A1 (fr) 1999-09-30 2002-07-17 Isis Pharmaceuticals, Inc. Rnase h humaine et compositions nucleotidiques correspondantes
US6733776B1 (en) 1992-04-02 2004-05-11 Anticancer, Inc. Method for promoting hair growth
WO2004046160A2 (fr) 2002-11-18 2004-06-03 Santaris Pharma A/S Conception antisens
WO2005027833A2 (fr) 2003-09-12 2005-03-31 Avi Biopharma, Inc. Compose et methode de traitement du cancer de la prostate androgeno-independant
WO2005060661A2 (fr) 2003-12-19 2005-07-07 The Regents Of The University Of California Procedes et materiaux pour evaluer des therapies du cancer de la prostate
US20050164970A1 (en) 2003-12-22 2005-07-28 University Of Kansas Medical Center Method for treating prostate cancer using siRNA duplex for androgen receptor
US7067256B2 (en) 1996-05-08 2006-06-27 Board Of Regents, The University Of Texas System Ribozyme mediated inactivation of the androgen receptor
US7087229B2 (en) 2003-05-30 2006-08-08 Enzon Pharmaceuticals, Inc. Releasable polymeric conjugates based on aliphatic biodegradable linkers
WO2007031091A2 (fr) 2005-09-15 2007-03-22 Santaris Pharma A/S Composes antagonistes d'arn de modulation de l'expression de p21 ras
WO2008034123A2 (fr) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Conjugués polymères contenant des fragments chargés positivement
WO2008034122A2 (fr) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Segments de liaison biodégradables à base d'ester encombré pour distribution d'oligonuclotides
US20080090888A2 (en) * 2004-02-24 2008-04-17 Michael Jung Methods and materials for assessing prostate cancer therapies and compounds
WO2008053314A2 (fr) 2006-10-30 2008-05-08 Nokia Corporation Procédé, appareil et système fournissant une mobilité commandée par un opérateur pour un équipement utilisateur
WO2009055053A2 (fr) 2007-10-26 2009-04-30 The Regents Of The University Of California Composés de diarylhydantoïne
WO2009068033A2 (fr) 2007-11-26 2009-06-04 Santaris Pharma A/S Antagonistes basés sur les lna ciblant le récepteur de l'androgène
US20100048676A1 (en) * 2008-08-04 2010-02-25 University Of Rochester Non-androgen dependent roles for androgen receptor in liver cancer
US7709517B2 (en) 2005-05-13 2010-05-04 The Regents Of The University Of California Diarylhydantoin compounds
US7772433B2 (en) * 2002-02-28 2010-08-10 University Of Tennessee Research Foundation SARMS and method of use thereof

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4962029A (en) 1987-10-02 1990-10-09 Cetus Corporation Covalent oligonucleotide-horseradish peroxidase conjugate
US4914210A (en) 1987-10-02 1990-04-03 Cetus Corporation Oligonucleotide functionalizing reagents
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US6733776B1 (en) 1992-04-02 2004-05-11 Anticancer, Inc. Method for promoting hair growth
EP0692972A1 (fr) 1993-04-02 1996-01-24 Anticancer, Inc. Procede permettant l'apport aux follicules pileux de compositions profitables
WO1997011170A1 (fr) 1995-09-20 1997-03-27 Worcester Foundation For Biomedical Research Chimiotherapie par oligonucleotides antisens de l'hypertrophie ou du cancer de la prostate
US7067256B2 (en) 1996-05-08 2006-06-27 Board Of Regents, The University Of Texas System Ribozyme mediated inactivation of the androgen receptor
EP1222309A1 (fr) 1999-09-30 2002-07-17 Isis Pharmaceuticals, Inc. Rnase h humaine et compositions nucleotidiques correspondantes
WO2001083740A2 (fr) 2000-05-04 2001-11-08 Avi Biopharma, Inc. Composition anti-sens a region d'epissage et methode associee
US7772433B2 (en) * 2002-02-28 2010-08-10 University Of Tennessee Research Foundation SARMS and method of use thereof
WO2004046160A2 (fr) 2002-11-18 2004-06-03 Santaris Pharma A/S Conception antisens
US7087229B2 (en) 2003-05-30 2006-08-08 Enzon Pharmaceuticals, Inc. Releasable polymeric conjugates based on aliphatic biodegradable linkers
WO2005027833A2 (fr) 2003-09-12 2005-03-31 Avi Biopharma, Inc. Compose et methode de traitement du cancer de la prostate androgeno-independant
WO2005060661A2 (fr) 2003-12-19 2005-07-07 The Regents Of The University Of California Procedes et materiaux pour evaluer des therapies du cancer de la prostate
US20050164970A1 (en) 2003-12-22 2005-07-28 University Of Kansas Medical Center Method for treating prostate cancer using siRNA duplex for androgen receptor
US20080090888A2 (en) * 2004-02-24 2008-04-17 Michael Jung Methods and materials for assessing prostate cancer therapies and compounds
US7709517B2 (en) 2005-05-13 2010-05-04 The Regents Of The University Of California Diarylhydantoin compounds
US20100210665A1 (en) 2005-05-13 2010-08-19 The Regents Of The University Of California Diarylhydantoin compounds
US20100172975A1 (en) 2005-05-13 2010-07-08 The Regents Of The University Of California Treatment of hyperproliferative disorders with diarylhydantoin compounds
WO2007031091A2 (fr) 2005-09-15 2007-03-22 Santaris Pharma A/S Composes antagonistes d'arn de modulation de l'expression de p21 ras
WO2008034122A2 (fr) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Segments de liaison biodégradables à base d'ester encombré pour distribution d'oligonuclotides
WO2008034123A2 (fr) 2006-09-15 2008-03-20 Enzon Pharmaceuticals, Inc. Conjugués polymères contenant des fragments chargés positivement
WO2008053314A2 (fr) 2006-10-30 2008-05-08 Nokia Corporation Procédé, appareil et système fournissant une mobilité commandée par un opérateur pour un équipement utilisateur
WO2009055053A2 (fr) 2007-10-26 2009-04-30 The Regents Of The University Of California Composés de diarylhydantoïne
WO2009068033A2 (fr) 2007-11-26 2009-06-04 Santaris Pharma A/S Antagonistes basés sur les lna ciblant le récepteur de l'androgène
US7737125B2 (en) * 2007-11-26 2010-06-15 Enzon Pharamaceuticals, Inc. LNA antagonists targeting the androgen receptor
US20100234451A1 (en) * 2007-11-26 2010-09-16 Enzon Pharmaceuticals, Inc. Lna antagonists targeting the androgen receptor
US20100048676A1 (en) * 2008-08-04 2010-02-25 University Of Rochester Non-androgen dependent roles for androgen receptor in liver cancer

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
CHRISTENSEN, NUCL. ACIDS. RES., vol. 30, 2002, pages 4918 - 4925
FREIER; ALTMANN, NUCL. ACID RES., vol. 25, 1997, pages 4429 - 4443
HOIMES ET AL.: "Redefining hormone resistance in prostate cancer", THERAPEUTIC ADVANCES IN MEDICAL ONCOLOGY, vol. 2, no. 2, 1 March 2010 (2010-03-01), pages 107 - 123, XP055114294 *
KNUDSEN. ET AL.: "Starving the addiction: new opportunities for durable suppression of AR signaling in prostate cancer", CLIN CANCER RES., vol. 15, no. 15, 2009, pages 4792 - 4798, XP055114296 *
MANOHARAN ET AL., TETRAHEDRON LETTERS, vol. 34, 1991, pages 7171
MONKS ET AL., PNAS, 2 November 2007 (2007-11-02)
MOONEY ET AL., NAR, vol. 31, no. 8, 15 December 2002 (2002-12-15)
SCHER ET AL.: "Antitumour activity of MDV3100 in castration-resistant prostate cancer: a Phase 1-2 study", LANCET, vol. 375, no. 9724, 24 April 2010 (2010-04-24), pages 1437 - 1446, XP027182350 *
See also references of EP2646553A4 *
TAPLIN: "Drug insight: role of the androgen receptor in the development and progression of prostate cancer", NAT CLIN PRACT ONCOL., vol. 4, no. 4, 2007, pages 236 - 244, XP055114300 *
THEODORA W GREENE; PETER G M WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
TRAN ET AL.: "Development of a Second-Generation Antiandrogen for Treatment of Advanced Prostate Cancer", SCIENCE, vol. 324, no. 5928, 2009, pages 787 - 790, XP002696684 *
UHLMANN, CURR. OPINION IN DRUG DEVELOPMENT, vol. 3, no. 2, 2000, pages 293 - 213

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014059364A1 (fr) * 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Procédés de traitement de la maladie de kennedy
US9175291B2 (en) 2012-10-11 2015-11-03 Isis Pharmaceuticals Inc. Modulation of androgen receptor expression
US9523094B2 (en) 2012-10-11 2016-12-20 Ionis Pharmaceuticals, Inc. Methods of treating kennedy's disease
EP4052709A1 (fr) * 2012-10-11 2022-09-07 Ionis Pharmaceuticals, Inc. Procédés de traitement de la maladie de kennedy
EP3327144A1 (fr) 2013-02-25 2018-05-30 Novartis AG Nouvelle mutation du récepteur d'androgène
US10011874B2 (en) 2013-02-25 2018-07-03 Novartis Ag Androgen receptor mutation
EP3696276A1 (fr) 2013-02-25 2020-08-19 Novartis AG Nouvelle mutation du récepteur d'androgène
US20220193110A1 (en) * 2020-12-17 2022-06-23 Washington University Nxtar-derived oligonucleotides and uses thereof

Also Published As

Publication number Publication date
EP2646553A4 (fr) 2015-01-07
EP2646553A1 (fr) 2013-10-09
AU2011326034A1 (en) 2013-05-02
AU2011326034B2 (en) 2016-02-04

Similar Documents

Publication Publication Date Title
EP2225377B1 (fr) Antagonistes basés sur les lna ciblant le récepteur de l'androgène
US8450290B2 (en) Methods for treating androgen receptor dependent disorders including cancers
EP2152879B1 (fr) Composés antagonistes de l'arn pour la modulation de la béta caténine
EP2225376B1 (fr) Composés d'antagonistes d'arn utilisés pour moduler l'expression de pik3ca
JP2010526797A (ja) Her3のモジュレーションのためのrnaアンタゴニスト化合物
JP2011527901A (ja) Gli2を標的化するrnaアンタゴニスト
AU2011326034B2 (en) Compositions and methods for treating androgen receptor dependent disorders including cancers
EP2238249A2 (fr) Composés d'antagonistes d'arn utiles pour moduler mcl-1
WO2011054811A1 (fr) Traitement combiné par des antagonistes d'arn ciblant hsp-27
US8440809B2 (en) RNA antagonists targeting Hsp27

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11839515

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2011326034

Country of ref document: AU

Date of ref document: 20111111

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE