US20220002722A1 - Nucleic acids for inhibiting expression of lpa in a cell - Google Patents

Nucleic acids for inhibiting expression of lpa in a cell Download PDF

Info

Publication number
US20220002722A1
US20220002722A1 US17/293,303 US201917293303A US2022002722A1 US 20220002722 A1 US20220002722 A1 US 20220002722A1 US 201917293303 A US201917293303 A US 201917293303A US 2022002722 A1 US2022002722 A1 US 2022002722A1
Authority
US
United States
Prior art keywords
strand
nucleic acid
nucleotides
modified
nucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/293,303
Other languages
English (en)
Inventor
David Anthony Rider
Lucas Bethge
Christian Frauendorf
Adrien Weingaertner
Judith Flauptmann
Sibylle DAMES
Steffen Schubert
Stephan Tenbaum
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Silence Therapeutics GmbH
Original Assignee
Silence Therapeutics GmbH
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 PCT/EP2018/081106 external-priority patent/WO2019092283A1/en
Application filed by Silence Therapeutics GmbH filed Critical Silence Therapeutics GmbH
Assigned to SILENCE THERAPEUTICS GMBH reassignment SILENCE THERAPEUTICS GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRAUENDORF, CHRISTIAN, HAUPTMANN, Judith, TENBAUM, Stephan, BETHGE, LUCAS, RIDER, David Anthony, WEINGAERTNER, Adrien, DAMES, Sibylle, SCHUBERT, STEFFEN
Publication of US20220002722A1 publication Critical patent/US20220002722A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • 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
    • C12N2310/3212'-O-R Modification
    • 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
    • C12N2310/3222'-R Modification
    • 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
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • 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
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • 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
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/532Closed or circular
    • 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

Definitions

  • the present invention relates to products and compositions and their uses.
  • the invention relates to nucleic acid products that interfere with the LPA gene expression or inhibit its expression.
  • Such therapeutic Lp(a) lowering therapy serves to prevent and reduce the risk of suffering stroke, atherosclerosis, thrombosis and cardiovascular diseases such as coronary heart disease and aortic stenosis or any other disorder, pathology or syndrome linked to elevated levels of Lp(a) particles.
  • dsRNA Double stranded RNAs (dsRNA) able to complementarily bind expressed mRNA have been shown to be able to block gene expression (Fire et al., 1998, Nature. 1998 Feb. 19; 391(6669):806-11 and Elbashir et al., 2001, Nature. 2001 May 24; 411(6836):494-88) by a mechanism that has been termed RNA interference (RNAi).
  • RNAi RNA interference
  • Short dsRNAs direct gene specific, post transcriptional silencing in many organisms, including vertebrates, and have become a useful tool for studying gene function.
  • RNAi is mediated by the RNA induced silencing complex (RISC), a sequence specific, multi component nuclease that degrades messenger RNAs homologous to the silencing trigger loaded into the RISC complex.
  • RISC RNA induced silencing complex
  • Interfering RNA such as siRNAs, antisense RNAs, and micro RNAs are oligonucleotides that prevent the formation of proteins by gene silencing i.e. inhibiting gene translation of the protein through degradation of mRNA molecules.
  • Gene silencing agents are becoming increasingly important for therapeutic applications in medicine.
  • Lp(a) particles are heterogeneous low-density lipoprotein particles expressed predominantly in the liver (Witztum and Ginsberg, J Lipid Res. 2016 March; 57(3):336-9). They are composed of Apolipoprotein(a) (Apo(a) or Lp(a) encoded by the LPA gene) linked to an LDL-like particle via the ApoB poly-peptide. Genetically defined high Lp(a) particle serum levels are unaffected by diet and exercise and are associated to increased risk to suffer from cardiovascular disease through the associated atherosclerotic potential (Alonso et al., Journal of the American College of Cardiology Vol. 63, No. 19, 2014).
  • Lp(a) particles In terms of diagnostics and preventive medicine the patient's serum level of Lp(a) particles is a highly prevalent, independent, genetic risk factor for coronary heart disease and aortic stenosis (Saeedi and Frohlich Clinical Diabetes and Endocrinology (2016) 2:7). There is no current approved specific Lp(a) particle reduction therapy beyond indirect standard general LDL-lowering measures. Accordingly, methods for effective treatment, prevention and reduction of risk of suffering from disorders such as and associated with stroke, atherosclerosis, thrombosis and cardiovascular diseases such as coronary heart disease, aortic stenosis and other yet unidentified associated disorders, pathologies or syndromes are currently needed. The present invention addresses this unmet medical need.
  • One aspect of the invention relates to a nucleic acid for inhibiting expression of LPA in a cell, comprising at least one duplex region that comprises at least a portion of a first strand and at least a portion of a second strand that is at least partially complementary to the first strand, wherein said first strand is at least partially complementary to at least a portion of a RNA transcribed from the LPA gene, wherein said first strand comprises a nucleotide sequence selected from the following sequences: SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41 or 43, wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are modified with a 2′ fluoro modification, and the nucleotides on the second strand which correspond to positions 11-13 of the first strand are modified with a 2′ fluoro modification.
  • the invention also provides a composition comprising the nucleic acid or conjugated nucleic acid of any aspect of the invention, and optionally a physiologically acceptable excipient.
  • One aspect relates to a nucleic acid that is capable of inhibiting expression of LPA for use as a medicament.
  • nucleic acid or conjugated nucleic acid for use in the treatment of a disease, disorder or syndrome and/or in the manufacture of a medicament for treating a disease, disorder, or syndrome.
  • the invention provides a method of treating or preventing a disease, disorder or syndrome comprising administration of a composition comprising a nucleic acid or conjugated nucleic acid according to any aspect of the invention to an individual in need of treatment.
  • the nucleic acid or conjugated nucleic acid may be administered to the subject subcutaneously, intravenously or using any other application routes such as oral, rectal or intraperitoneal.
  • a method of making the nucleic acid or conjugated nucleic acid according to the invention is also included.
  • the nucleic acid or the composition comprising the nucleic acid or conjugated nucleic acid of the invention may be used in the treatment of a disease, disorder or syndrome.
  • the treatment may be to prevent and reduce risk to suffer from stroke, atherosclerosis, thrombosis or cardiovascular diseases such as coronary heart disease or aortic stenosis and any other disease or pathology associated to elevated levels of Lp(a)-containing particles.
  • the present invention relates to a nucleic acid which is double stranded and directed to an expressed RNA transcript of LPA and compositions thereof.
  • These nucleic acids or conjugated nucleic acids can be used in the treatment and prevention of a variety of diseases, disorders and syndromes where reduced expression of LPA gene product is desirable.
  • One aspect relates to a nucleic acid for inhibiting expression of LPA in a cell, comprising at least one duplex region that comprises at least a portion of a first strand and at least a portion of a second strand that is at least partially complementary to the first strand, wherein said first strand is at least partially complementary to at least a portion of a RNA transcribed from the LPA gene, wherein said first strand comprises, or preferably consists of, a nucleotide sequence selected from the following sequences: SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are modified with a 2′ fluoro modification, and the nucleotides on the second strand which correspond to positions 11-13 of the first strand are modified with a 2′ fluoro modification.
  • One aspect relates to a nucleic acid for inhibiting expression of LPA in a cell, comprising at least one duplex region that comprises a first strand and a second strand that is at least partially complementary to the first strand, wherein said first strand is at least partially complementary to at least a portion of a RNA transcribed from the LPA gene, wherein said first strand comprises, or preferably consists of, a nucleotide sequence selected from the following sequences: SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are modified with a 2′ fluoro modification, and the nucleotides on the second strand which correspond to positions 11-13 of the first strand are modified with a 2′ fluoro modification.
  • nucleic acids are able to efficiently reduce the expression of LPA in a cell and are very stable.
  • the nucleic acid of the invention is preferably capable of inhibiting expression of LPA in a cell to a similar, such as the same, or a higher degree than the same nucleic acids with a different modification pattern in comparable conditions. More specifically, the nucleic acid of the invention is preferably capable of inhibiting expression of LPA in a cell by 80, 90, 100, 105, 110 or more percent as compared to the same nucleic acid with a different modification pattern in comparable conditions.
  • One aspect relates to a nucleic acid wherein all nucleotides of the nucleic acid are modified at the 2′ position of the sugar.
  • One aspect relates to a nucleic acid wherein the nucleic acid is modified preferably along the entire length of the first strand with alternating 2′ O-methyl modifications and 2′ fluoro modifications.
  • One aspect relates to a nucleic acid wherein the remaining modifications of the second strand are naturally occurring modifications, preferably 2′ O-methyl.
  • nucleotides on the second strand which correspond to positions 11-13 of the first strand are modified with a 2′ fluoro modification and all other nucleotides of the second strand are modified with a naturally occurring modification, which is preferably 2′ O-methyl.
  • the second strand may comprise a nucleotide sequence of SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 38, 40, 42, or 44.
  • the nucleic acid may: a) be blunt ended at both ends; b) have an overhang at one end and a blunt end at the other; or c) have an overhang at both ends.
  • the nucleic acid is preferably blunt ended at both ends.
  • nucleic acids among others have the advantage of being active in various species that are relevant for pre-clinical and clinical development and/or of having few relevant off-target effects as well as being stable in vivo and having a long duration of action. They also comprise comprises relatively few non-naturally occurring modified nucleotides but are nonetheless able to efficiently inhibit the target gene for long periods of time. The specific modification pattern with few non-naturally occurring modified nucleotides (2′F modified nucleotides) also makes them easier to synthesise.
  • the nucleic acid may comprise a first strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 9, and optionally a second strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 10; or a first strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 5, and optionally a second strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 6; or a first strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 1, and optionally a second strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 2; or a first strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO:3, and optionally a second strand that comprises or preferably consists of a nucleotide sequence of SEQ ID NO: 4; or a first
  • the nucleic acid wherein said first strand comprises, and preferably consists of, the nucleotide sequence of SEQ ID NO: 9 and optionally, wherein said second strand comprises, and preferably consists of, the nucleotide sequence of SEQ ID NO. 10.
  • the LPA gene comprises highly repetitive sequences. First strand nucleic acids with very similar sequences can therefore have perfect sequence complementarity to very different target regions of the mRNA.
  • nucleic acid for inhibiting expression of LPA in a cell
  • the nucleic acid comprises at least one duplex region that comprises: a first strand; and a second strand, wherein said second strand is at least partially complementary to the first strand
  • said first strand comprises a sequence of at least 15, preferably at least 16, more preferably at least 17, yet more preferably at least 18 and most preferably at least 19 nucleotides of any one of the reference sequences SEQ ID NO: 9, 5, 1, 3, 7, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, and wherein the number of single nucleotide mismatches and/or deletions and/or insertions in the first strand sequence relative to the portion of the reference sequence that is comprised in the first strand sequence is at most three, preferably at most two, more preferably at most one and most preferably zero.
  • sequence may however be modified by a number of modifications that do not change the identity of the nucleotide.
  • modifications of the backbone of the nucleic acid do not change the identity of the nucleotide because the base itself remains the same as in the reference sequence.
  • the first strand of the nucleic acid comprises a sequence of at least 18 nucleotides of any one of the reference sequences, preferably of any one of the reference sequences SEQ ID NO: 9 and 5, and wherein the number of single-nucleotide mismatches and/or deletions and/or insertions in the first strand sequence relative to the portion of the reference sequence that is comprised in the first strand sequence is at most one, and preferably zero.
  • the first strand of the nucleic acid comprises a sequence of at least 19 nucleotides of any of the reference sequences SEQ ID NO: 9 and 5.
  • a reference sequence comprising or consisting of unmodified nucleotides
  • this reference is not limited to the sequence with unmodified nucleotides.
  • the same reference also encompasses the same nucleotide sequence in which one, several, such as two, three, four, five, six, seven or more, including all, nucleotides are modified by modifications such as 2′-OMe, 2′-F, a ligand, a linker, a 3′ end or 5′ end modification or any other modification. It also refers to sequences in which two or more nucleotides are linked to each other by the natural phosphodiester linkage or by any other linkage such as a phosphorothioate or a phosphorodithioate linkage.
  • a double-stranded nucleic acid is a nucleic acid in which the first strand and the second strand hybridise to each other over at least part of their lengths and are therefore capable of forming a duplex region under physiological conditions, such as in PBS at 37° C. at a concentration of 1 ⁇ M of each strand.
  • the first and second strand are preferably able to hybridise to each other and therefore to form a duplex region over a region of at least 15 nucleotides, preferably 16, 17, 18 or 19 nucleotides.
  • This duplex region comprises nucleotide base parings between the two strands, preferably based on Watson-Crick base pairing and/or wobble base pairing (such as GU base pairing).
  • the double stranded nucleic acid is preferably a stable double stranded nucleic acid under physiological conditions and preferably has a melting temperature (Tm) of 45° C. or more, preferably 50° C. or more, and more preferably 55° C.
  • each first strand and the second strand are preferably capable of forming a duplex region (ie are complementary to each other) over i) at least a portion of their lengths, preferably over at least 15 nucleotides of both of their lengths, ii) over the entire length of the first strand, iii) over the entire length of the second strand and/or iv) over the entire length of both the first and the second strand.
  • Strands being complementary to each other over a certain length means that the strands are able to base pair to each other, either via Watson-Crick or wobble base pairing, over that length.
  • Each nucleotide of the length does not necessarily have to be able to base pair with its counterpart in the other strand over the entire given length as long as a stable double-stranded nucleotide under physiological conditions can be formed. This is however preferred.
  • a certain number of mismatches, deletions or insertions between the first (antisense) strand and the target sequence, or between the first strand and the second (sense) strand can be tolerated in the context of siRNA and even have the potential in certain cases to increase activity.
  • nucleic acid it is meant a nucleic acid comprising two strands comprising nucleotides, that is able to interfere with gene expression. Inhibition may be complete or partial and results in down regulation of gene expression in a targeted manner.
  • the nucleic acid comprises two separate polynucleotide strands; the first strand, which may also be a guide strand; and a second strand, which may also be a passenger strand.
  • the first strand and the second strand may be part of the same polynucleotide molecule that is self-complementary which ‘folds’ back to form a double stranded molecule.
  • the nucleic acid may be an siRNA molecule.
  • the nucleic acid may comprise ribonucleotides, modified ribonucleotides, deoxynucleotides, deoxyribonucleotides, or nucleotide analogues non-nucleotides that are able to mimic nucleotides such that they may ‘pair’ with the corresponding base on the target sequence or complementary strand.
  • the nucleic acid may further comprise a double-stranded nucleic acid portion or duplex region formed by all or a portion of the first strand (also known in the art as a guide strand) and all or a portion of the second strand (also known in the art as a passenger strand).
  • the duplex region is defined as beginning with the first base pair formed between the first strand and the second strand and ending with the last base pair formed between the first strand and the second strand, inclusive.
  • duplex region it is meant the region in two complementary or substantially complementary oligonucleotides that form base pairs with one another, either by Watson-Crick base pairing or any other manner that allows for a duplex between oligonucleotide strands that are complementary or substantially complementary.
  • an oligonucleotide strand having 21 nucleotide units can base pair with another oligonucleotide of 21 nucleotide units, yet only 19 nucleotides on each strand are complementary or substantially complementary, such that the “duplex region” consists of 19 base pairs.
  • the remaining base pairs may exist as 5′ and 3′ overhangs, or as single stranded regions.
  • complementarity refers to complementarity between the strands such that they are capable of annealing under biological conditions. Techniques to empirically determine if two strands are capable of annealing under biological conditions are well known in the art. Alternatively, two strands can be synthesised and added together under biological conditions to determine if they anneal to one another.
  • the portion of the first strand and second strand that form at least one duplex region may be fully complementary or be at least partially complementary to each other.
  • first and second strands must be able to hybridise under physiological conditions.
  • the complementarity between the first strand and second strand in the at least one duplex region may be perfect in that there are no nucleotide mismatches or additional/deleted nucleotides in either strand. Alternatively, the complementarity may not be perfect.
  • the complementarity may be from about 70% to about 100%. More specifically, the complementarity may be at least 70%, 75%, 80%, 85%, 90% or 95% and intermediate values.
  • a portion of as for example in “one duplex region that comprises at least a portion of a first strand” should be understood to mean that the duplex region comprises at least 10, preferably at least 12, more preferably at least 14, yet more preferably at least 16, even more preferably at least 18 and most preferably all of the nucleotides of a given reference strand sequence.
  • the portion of the reference sequence in the duplex region is at least 70%, preferably at least 80%, more preferably at least 90%, yet more preferably at least 95% and most preferably 100% identical to the corresponding portion of the reference sequence.
  • the number of single nucleotide mismatches relative to the portion of the reference sequence is at most three, preferably at most two, more preferably at most one and most preferably zero.
  • the first strand and the second strand may each comprise a region of complementarity which comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the sequences listed in Table 1.
  • a nucleic acid involves the formation of a duplex region between all or a portion of the first strand and a portion of a target nucleic acid.
  • the portion of the target nucleic acid that forms a duplex region with the first strand defined as beginning with the first base pair formed between the first strand and the target sequence and ending with the last base pair formed between the first strand and the target sequence, inclusive, is the target nucleic acid sequence or simply, target sequence.
  • the duplex region formed between the first strand and the second strand need not be the same as the duplex region formed between the first strand and the target sequence. That is, the second strand may have a sequence different from the target sequence; however, the first strand must be able to form a duplex structure with both the second strand and the target sequence, at least under physiological conditions.
  • the complementarity between the first strand and the target sequence may be perfect (no nucleotide mismatches or additional/deleted nucleotides in either nucleic acid).
  • the complementarity between the first strand and the target sequence may not be perfect.
  • the complementarity may be from about 70% to about 100%. More specifically, the complementarity may be at least 70%, 80%, 85%, 90% or 95% and intermediate values.
  • the identity between the first strand and the complementary sequence of the target sequence may range from about 75% to about 100%. More specifically, the complementarity may be at least 75%, 80%, 85%, 90% or 95% and intermediate values, provided a nucleic acid is capable of reducing or inhibiting the expression of LPA.
  • a nucleic acid having less than 100% complementarity between the first strand and the target sequence may be able to reduce the expression of LPA to the same level as a nucleic acid having perfect complementarity between the first strand and target sequence. Alternatively, it may be able to reduce expression of LPA to a level that is 15%-100% of the level of reduction achieved by the nucleic acid with perfect complementarity.
  • the nucleic acid comprises a first nucleic acid strand and a second nucleic acid strand, wherein the first strand is capable of hybridising under physiological conditions to a nucleic acid of sequence SEQ ID NO: 10, 6, 2, 4, 8, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, or 44;
  • the second strand is capable of hybridising under physiological conditions to the first strand to form a duplex region
  • nucleotides at positions 2 and 14 from the 5′ end of the first strand are modified with a 2′ fluoro modification
  • nucleotides on the second strand which correspond to positions 11-13 of the first strand are modified with a 2′ fluoro modification
  • Nucleic acids that are capable of hybridising under physiological conditions are nucleic acids that are capable of forming base pairs, preferably Watson-Crick or wobble base-pairs, between at least a portion of the opposed nucleotides in the strands so as to form at least a duplex region.
  • Such a double-stranded nucleic acid is preferably a stable double-stranded nucleic acid under physiological conditions (for example in PBS at 37° C. at a concentration of 1 ⁇ M of each strand), meaning that under such conditions, the two strands stay hybridised to each other.
  • the Tm of the double-stranded nucleotide is preferably 45° C. or more, preferably 50° C. or more and more preferably 55° C. or more.
  • nucleic acid for inhibiting expression of LPA, wherein the nucleic acid comprises a first sequence of at least 15, preferably at least 16, more preferably at least 17, yet more preferably at least 18 and most preferably all nucleotides differing by no more than 3 nucleotides, preferably no more than 2 nucleotides, more preferably no more than 1 nucleotide and most preferably not differing by any nucleotide from any of the sequences of Table 1, the first sequence being able to hybridise to a target gene transcript (such as an mRNA) under physiological conditions.
  • a target gene transcript such as an mRNA
  • the nucleic acid further comprises a second sequence of at least 15, preferably at least 16, more preferably at least 17, yet more preferably at least 18 and most preferably all nucleotides differing by no more than 3 nucleotides, preferably no more than 2 nucleotides, more preferably no more than 1 nucleotide and most preferably not differing by any nucleotide from any of the sequences of Table 1, the second sequence being able to hybridise to the first sequence under physiological conditions and preferably the nucleic acid being an siRNA that is capable of inhibiting LPA expression via the RNAi pathway.
  • the nucleic acids described herein may be capable of inhibiting the expression of LPA. Inhibition may be complete, i.e. 0% remaining expression compared of the expression level of LPA in the absence of the nucleic acid of the invention. Inhibition of LPA expression may be partial, i.e. it may be 15%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% or intermediate values of LPA expression in the absence of a nucleic acid of the invention. The level of inhibition may be measured by comparing a treated sample with an untreated sample or with a sample treated with a control such as for example a siRNA that does not target LPA.
  • Inhibition may be measured by measuring LPA mRNA and/or protein levels or levels of a biomarker or indicator that correlates with LPA presence or activity. It may be measured in cells that may have been treated in vitro with a nucleic acid described herein. Alternatively, or in addition, inhibition may be measured in cells, such as hepatocytes, or tissue, such as liver tissue, or an organ, such as the liver, or in a body fluid such as blood, serum, lymph or any other body part that has been taken from a subject previously treated with a nucleic acid disclosed herein.
  • Preferably inhibition of LPA expression is determined by comparing the LPA mRNA level measured in LPA-expressing cells after 24 or 48 hours in vitro treatment under ideal conditions (see the examples for appropriate concentrations and conditions) with a nucleic acid disclosed herein to the LPA mRNA level measured in the same cells that were untreated or mock treated or treated with a control nucleic acid.
  • the term “inhibit”, “down-regulate”, or “reduce” with respect to gene expression means the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits (e.g., mRNA), or activity of one or more proteins or protein subunits, is reduced below that observed in the absence of the nucleic acid or conjugated nucleic acid of the invention or in reference to an siRNA molecule with no known homology to human transcripts (herein termed non-silencing control).
  • control may be conjugated and modified in an analogous manner to the molecule of the invention and delivered into the target cell by the same route; for example the expression may be reduced to 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 15%, or to intermediate values, or less than that observed in the absence of the nucleic acid or conjugated nucleic acid or in the presence of a non-silencing control.
  • the nucleic acid may comprise a first strand and a second strand that are each from 19-25 nucleotides in length.
  • the first strand and the second strand may be of different lengths.
  • the first strand and/or the second strand may each be from 17-35, preferably 18-30, more preferably 19-25 and most preferably 19 nucleotides in length and at least one duplex region may be from 10-25 nucleotides, preferably 18-23 nucleotides in length.
  • the duplex may comprise two separate strands or it may comprise a single strand which comprises the first strand and the second strand.
  • the first strand may be 17-25 nucleotides in length, preferably it may be 18-24 nucleotides in length, it may be 18, 19, 20, 21, 22, 23 or 24 nucleotides in length. Most preferably, the first strand is 19 nucleotides in length.
  • the second strand may independently be 17-25 nucleotides in length, preferably it may be 18-24 nucleotides in length, it may be 18, 19, 20, 21, 22, 23 or 24 nucleotides in length. More preferably, the second strand is 18 or 19 nucleotides in length, and most preferably it is 18 nucleotides in length.
  • the nucleic acid may be 15-25 nucleotide pairs in length.
  • the nucleic acid may be 17-23 nucleotide pairs in length.
  • the nucleic acid may be 17-25 nucleotide pairs in length.
  • the nucleic acid may be 23-24 nucleotide pairs in length.
  • the nucleic acid may be 19-21 nucleotide pairs in length.
  • the nucleic acid may be 21-23 nucleotide pairs in length.
  • the nucleic acid may comprise a duplex region that consists of 19-25 nucleotide base pairs.
  • the duplex region may consist of 17, 18, 19, 20, 21, 22, 23, 24 or 25 base pairs, which may be contiguous.
  • the duplex region consists of 19 base pairs.
  • the nucleic acid mediates RNA interference.
  • the nucleic acid for inhibiting expression of LPA in a cell comprises at least one duplex region that comprises a first strand and a second strand that is at least partially complementary to the first strand, wherein said first strand comprises a sequence of at least 15, preferably at least 16, more preferably at least 17, yet more preferably at least 18 and most preferably at least 19 nucleotides with a sequence identity of at least 70%, preferably at least 80%, more preferably at least 90%, yet more preferably at least 95% and most preferably 100% of any of sequences SEQ ID NOs: 9, 5, 1, 3, 7, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43.
  • nucleic acid or conjugated nucleic acid as described may reduce the expression of LPA by at least 15% compared to the expression observed in the absence of the nucleic acid or conjugated nucleic acid. All preferred features of any of the previous aspects also apply to this aspect.
  • the expression of LPA may be reduced to at least the following given % or less than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 15% or less, and intermediate values, than that observed in the absence of the nucleic acid or conjugated nucleic acid or in the presence of a non-silencing control.
  • the nucleic acid may have an overhang at one end and a blunt end at the other.
  • the nucleic acid may have an overhang at both ends.
  • the nucleic acid may be blunt ended at both ends.
  • the nucleic acid may be blunt ended at the end with the 5′-end of the first strand and the 3′-end of the second strand or at the 3′-end of the first strand and the 5′-end of the second strand.
  • an “overhang” as used herein has its normal and customary meaning in the art, i.e. a single stranded portion of a nucleic acid that extends beyond the terminal nucleotide of a complementary strand in a double strand nucleic acid.
  • the term “blunt end” includes double stranded nucleic acid whereby both strands terminate at the same position, regardless of whether the terminal nucleotide(s) are base-paired.
  • the terminal nucleotide of a first strand and a second strand at a blunt end may be base paired.
  • the terminal nucleotide of a first strand and a second strand at a blunt end may not be paired.
  • the terminal two nucleotides of a first strand and a second strand at a blunt end may be base-paired.
  • the terminal two nucleotides of a first strand and a second strand at a blunt end may not be paired.
  • the nucleic acid may comprise an overhang at a 3′- or 5′-end.
  • the nucleic acid may have a 3′-overhang on the first strand.
  • the nucleic acid may have a 3′-overhang on the second strand.
  • the nucleic acid may have a 5′-overhang on the first strand.
  • the nucleic acid may have a 5′-overhang on the second strand.
  • the nucleic acid may have an overhang at both the 5′-end and 3′-end of the first strand.
  • the nucleic acid may have an overhang at both the 5′-end and 3′-end of the second strand.
  • the nucleic acid may have a 5′ overhang on the first strand and a 3′ overhang on the second strand.
  • the nucleic acid may have a 3′ overhang on the first strand and a 5′ overhang on the second strand.
  • the nucleic acid may have a 3′ overhang on the first strand and a 3′ overhang on the second strand.
  • the nucleic acid may have a 5′ overhang on the first strand and a 5′ overhang on the second strand.
  • An overhang at the 3′-end or 5′ end of the second strand or the first strand may be selected from consisting of 1, 2, 3, 4 and 5 nucleotides in length.
  • an overhang may consist of 1 or 2 nucleotides, which may or may not be modified.
  • the nucleic acid is an siRNA.
  • siRNAs are short interfering or short silencing RNAs that are able to inhibit the expression of a target gene through the RNA interference (RNAi) pathway. Inhibition occurs through targeted degradation of mRNA transcripts of the target gene after transcription.
  • RNAi RNA interference
  • the siRNA forms part of the RISC complex.
  • the RISC complex specifically targets the target RNA by sequence complementarity of the first (antisense) strand with the target sequence.
  • the nucleic acid mediates RNA interference (RNAi).
  • RNAi RNA interference
  • the nucleic acid, or at least the first strand of the nucleic acid is therefore preferably able to be incorporated into the RISC complex.
  • the nucleic acid, or at least the first strand of the nucleic acid is therefore able to guide the RISC complex to a specific target RNA with which the nucleic acid, or at least the first strand of the nucleic acid, is at least partially complementary.
  • the RISC complex then specifically cleaves this target RNA and as a result leads to inhibition of the expression of the gene from which the RNA stems.
  • Unmodified polynucleotides may be prone to degradation by cellular nucleases, and, as such, modifications/modified nucleotides may be included in the nucleic acid of the invention. Such modifications may help to stabilise the nucleic acid by making them more resistant against nucleases. This improved resistance allows nucleic acids to be active in mediating RNA interference for longer time periods and is especially desirable when the nucleic acids are to be used for treatment.
  • Modifications of the nucleic acid of the present invention generally provide a powerful tool in overcoming potential limitations including, but not limited to, in vitro and in vivo stability and bioavailability inherent to native RNA molecules.
  • the nucleic acid according to the invention may be modified by chemical modifications. Modified nucleic acid can also minimise the possibility of inducing interferon activity in humans. Modification can further enhance the functional delivery of a nucleic acid to a target cell.
  • the modified nucleic acid of the present invention may comprise one or more chemically modified ribonucleotides of either or both of the first strand or the second strand.
  • a ribonucleotide may comprise a chemical modification of the base, sugar or phosphate moieties.
  • the ribonucleic acid may be modified by substitution or insertion with analogues of nucleic acids or bases.
  • At least one nucleotide of the first and/or second strand of the nucleic acid is a modified nucleotide, preferably a non-naturally occurring nucleotide such as preferably a 2′-F modified nucleotide.
  • nucleic acid of the present invention may be modified.
  • the nucleic acid may comprise at least one modified nucleotide.
  • the modified nucleotide may be in the first strand.
  • the modified nucleotide may be in the second strand.
  • the modified nucleotide may be in the duplex region.
  • the modified nucleotide may be outside the duplex region, i.e., in a single stranded region.
  • the modified nucleotide may be on the first strand and may be outside the duplex region.
  • the modified nucleotide may be on the second strand and may be outside the duplex region.
  • the 3′-terminal nucleotide of the first strand may be a modified nucleotide.
  • the 3′-terminal nucleotide of the second strand may be a modified nucleotide.
  • the 5′-terminal nucleotide of the first strand may be a modified nucleotide.
  • the 5′-terminal nucleotide of the second strand may be a modified nucleotide.
  • a nucleic acid of the invention may have 1 modified nucleotide or a nucleic acid of the invention may have about 2-4 modified nucleotides, or a nucleic acid may have about 4-6 modified nucleotides, about 6-8 modified nucleotides, about 8-10 modified nucleotides, about 10-12 modified nucleotides, about 12-14 modified nucleotides, about 14-16 modified nucleotides about 16-18 modified nucleotides, about 18-20 modified nucleotides, about 20-22 modified nucleotides, about 22-24 modified nucleotides, 24-26 modified nucleotides or about 26-28 modified nucleotides or all of the nucleotides may be modified.
  • nucleic acid comprising said modified nucleotides retains at least 50% of its activity as compared to the same nucleic acid but without said modified nucleotides or vice versa.
  • the nucleic acid may retain 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% and intermediate values of its activity as compared to the same nucleic acid but without said modified nucleotides, or may have more than 100% of the activity of the same nucleic acid without said modified nucleotides.
  • the modified nucleotide may be a purine or a pyrimidine. At least half of the purines may be modified. At least half of the pyrimidines may be modified. All of the purines may be modified. All of the pyrimidines may be modified.
  • the modified nucleotides may be selected from the group consisting of a 3′-terminal deoxy-thymine (dT) nucleotide, a 2′-O-methyl modified nucleotide, a 2′ modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, a 2′-amino-modified nucleotide, a 2′-alkyl-modified nucleotide, a morpholino nucleotide, a phosphoramidate, a non-natural base comprising nucleotide, a nucleotide comprising a 5′-phosphorothioate group, a nucleotide comprising a 5′ phosphate or 5′ phosphate mimic and a terminal nucleotide linked to a cholesteryl derivative or a dodecanoic acid bisdecylamide group.
  • dT
  • the nucleic acid may comprise a nucleotide comprising a modified nucleotide, wherein the base is selected from 2-aminoadenosine, 2,6-diaminopurine, inosine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidine (e.g., 5-methylcytidine), 5-alkyluridine (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine), 6-azapyrimidine, 6-alkylpyrimidine (e.g.
  • Nucleic acids discussed herein include unmodified RNA as well as RNA which has been modified, e.g., to improve efficacy, and polymers of nucleoside surrogates.
  • Unmodified RNA refers to a molecule in which the components of the nucleic acid, namely sugars, bases, and phosphate moieties, are the same or essentially the same as that which occur in nature, for example as occur naturally in the human body.
  • Modified nucleotide as used herein refers to a nucleotide in which one or more of the components of the nucleotides, namely sugars, bases, and phosphate moieties, are different from those which occur in nature.
  • modified nucleotides While they are referred to as modified nucleotides they will of course, because of the modification, the term also includes molecules which are not nucleotides, for example a polynucleotide molecule in which the ribophosphate backbone is replaced with a non-ribophosphate construct that allows hybridisation between strands i.e. the modified nucleotides mimic the ribophosphate backbone.
  • a modification may only occur at a 3′ or 5′ terminal position, may only occur in a terminal regions, such as at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand.
  • a modification may occur in a double strand region, a single strand region, or in both.
  • a modification may occur only in the double strand region of a nucleic acid of the invention or may only occur in a single strand region of a nucleic acid of the invention.
  • a phosphorothioate modification at a non-linking O position may only occur at one or both termini, may only occur in a terminal region, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4 or 5 nucleotides of a strand, or may occur in duplex and/or in single strand regions, particularly at termini.
  • the 5′ end or 3′ ends may be phosphorylated.
  • Stability of a nucleic acid of the invention may be increased by including particular bases in overhangs, or to include modified nucleotides, in single strand overhangs, e.g., in a 5′ or 3′ overhang, or in both.
  • Purine nucleotides may be included in overhangs. All or some of the bases in a 3′ or 5′ overhang may be modified. Modifications can include the use of modifications at the 2′ OH group of the ribose sugar, the use of deoxyribonucleotides, instead of ribonucleotides, and modifications in the phosphate group, such as phosphorothioate modifications. Overhangs need not be homologous with the target sequence.
  • Nucleases can hydrolyse nucleic acid phosphodiester bonds. However, chemical modifications to nucleic acids can confer improved properties, and, can render oligoribonucleotides more stable to nucleases.
  • Modified nucleic acids can include one or more of:
  • modified phosphate groups include phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulphur.
  • One, each or both non-linking oxygens in the phosphate group can be independently any one of 5, Se, B, C, H, N, or OR (R is alkyl or aryl).
  • the phosphate linker can also be modified by replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylenephosphonates).
  • the replacement can occur at a terminal oxygen. Replacement of the non-linking oxygens with nitrogen is possible.
  • a modified nucleotide can include modification of the sugar groups.
  • the 2′ hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents.
  • Other substitutents of certain embodiments include 2
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified nucleotide may contain a sugar such as arabinose.
  • Modified nucleotides can also include “abasic” sugars, which lack a nucleobase at C-1′. These abasic sugars can further contain modifications at one or more of the constituent sugar atoms.
  • the 2′ modifications may be used in combination with one or more phosphate linker modifications (e.g., phosphorothioate).
  • phosphate linker modifications e.g., phosphorothioate
  • the phosphate groups can individually be replaced by non-phosphorus containing connectors.
  • moieties which can replace the phosphate group include siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • replacements may include the methylenecarbonylamino and methylenemethylimino groups.
  • the phosphate linker and ribose sugar may be replaced by nuclease resistant nucleotides.
  • PNA peptide nucleic acid
  • the 3′ and 5′ ends of an oligonucleotide can be modified. Such modifications can be at the 3′ end or the 5′ end or both ends of the molecule. They can include modification or replacement of an entire terminal phosphate or of one or more of the atoms of the phosphate group.
  • the 3′ and 5′ ends of an oligonucleotide can be conjugated to other functional molecular entities such as labelling moieties, e.g., fluorophores (e.g., pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes) or protecting groups (based e.g., on sulfur, silicon, boron or ester).
  • labelling moieties e.g., fluorophores (e.g., pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes) or protecting groups (based e.g., on sulfur, silicon, boron or ester).
  • the functional molecular entities can be attached to the sugar through a phosphate group and/or a linker.
  • the terminal atom of the linker can connect to or replace the linking atom of the phosphate group or the C-3′ or C-5′ O, N, S or C group of the sugar.
  • the linker can connect to or replace the terminal atom of a nucleotide surrogate (e.g., PNAs).
  • the 3′ end can be an —OH group.
  • terminal modifications include dyes, intercalating agents (e.g., acridines), cross-linkers (e.g., psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases, EDTA, lipophilic carriers (e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, O3-
  • Terminal modifications can be added for a number of reasons, including to modulate activity or to modulate resistance to degradation.
  • Terminal modifications useful for modulating activity include modification of the 5′ end with phosphate or phosphate analogues.
  • Nucleic acids of the invention, on the first or second strand, may be 5′ phosphorylated or include a phosphoryl analogue at the 5′ prime terminus.
  • 5′-phosphate modifications include those which are compatible with RISC mediated gene silencing.
  • Suitable modifications include: 5′-monophosphate ((HO) 2 (O)P—O-5′); 5′-diphosphate ((HO) 2 (O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO) 2 (O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-monothiophosphate (phosphorothioate; (HO) 2 (S)P—O-5′); 5′
  • Terminal modifications can also be useful for monitoring distribution, and in such cases the groups to be added may include fluorophores, e.g., fluorescein or an Alexa dye. Terminal modifications can also be useful for enhancing uptake, useful modifications for this include cholesterol. Terminal modifications can also be useful for cross-linking an RNA agent to another moiety.
  • fluorophores e.g., fluorescein or an Alexa dye.
  • Terminal modifications can also be useful for enhancing uptake, useful modifications for this include cholesterol. Terminal modifications can also be useful for cross-linking an RNA agent to another moiety.
  • Adenine, guanine, cytosine and uracil are the most common bases found in RNA. These bases can be modified or replaced to provide RNAs having improved properties.
  • nuclease resistant oligoribonucleotides can be prepared with these bases or with synthetic and natural nucleobases (e.g., inosine, thymine, xanthine, hypoxanthine, nubularine, isoguanisine, or tubercidine) and any one of the above modifications.
  • substituted or modified analogues of any of the above bases and “universal bases” can be employed.
  • Examples include 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thynine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines,
  • non-pairing nucleotide analogue means a nucleotide analogue which includes a non-base pairing moiety including but not limited to: 6 des amino adenosine (Nebularine), 4-Me-indole, 3-nitropyrrole, 5-nitroindole, Ds, Pa, N3-Me ribo U, N3-Me riboT, N3-Me dC, N3-Me-dT, N1-Me-dG, N1-Me-dA, N3-ethyl-dC, N3-Me dC.
  • the non-base pairing nucleotide analogue is a ribonucleotide. In other embodiments it is a deoxyribonucleotide.
  • terminal functional group includes without limitation a halogen, alcohol, amine, carboxylic, ester, amide, aldehyde, ketone, ether groups.
  • moieties may be linked to the 5′ terminus of the first strand or the second strand. These include abasic ribose moiety, abasic deoxyribose moiety, modifications abasic ribose and abasic deoxyribose moieties including 2′ 0 alkyl modifications; inverted abasic ribose and abasic deoxyribose moieties and modifications thereof, C6-imino-Pi; a mirror nucleotide including L-DNA and L-RNA; 5′OMe nucleotide; and nucleotide analogues including 4′,5′-methylene nucleotide; 1-( ⁇ -D-erythrofuranosyl)nucleotide; 4′-thio nucleotide, carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate, 3-aminopropy
  • the nucleic acid of the present invention may comprise an abasic nucleotide.
  • abasic refers to moieties lacking a base or having other chemical groups in place of a base at the 1′ position, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative.
  • the nucleic acid may comprise one or more nucleotides on the second and/or first strands that are modified. Alternating nucleotides may be modified, to form modified nucleotides.
  • Alternating as described herein means to occur one after another in a regular way. In other words, alternating means to occur in turn repeatedly. For example, if one nucleotide is modified, the next contiguous nucleotide is not modified and the following contiguous nucleotide is modified and so on. One nucleotide may be modified with a first modification, the next contiguous nucleotide may be modified with a second modification and the following contiguous nucleotide is modified with the first modification and so on, where the first and second modifications are different.
  • One or more of the odd numbered nucleotides of the first strand of the nucleic acid of the invention may be modified wherein the first strand is numbered 5′ to 3′, the 5′-most nucleotide being nucleotide number 1 of the first strand.
  • the term “odd numbered” as described herein means a number not divisible by two. Examples of odd numbers are 1, 3, 5, 7, 9, 11 and so on.
  • One or more of the even numbered nucleotides of the first strand of the nucleic acid of the invention may be modified, wherein the first strand is numbered 5′ to 3′.
  • the term “even numbered” as described herein means a number which is evenly divisible by two. Examples of even numbers are 2, 4, 6, 8, 10, 12, 14 and so on.
  • One or more of the odd numbered nucleotides of the second strand of the nucleic acid of the invention may be modified wherein the second strand is numbered 3′ to 5′, the 3′-most nucleotide being nucleotide number 1 of the second strand.
  • One or more of the even numbered nucleotides of the second strand of the nucleic acid of the invention may be modified, wherein the second strand is numbered 3′ to 5′.
  • One or more nucleotides on the first and/or second strand may be modified, to form modified nucleotides.
  • One or more of the odd numbered nucleotides of the first strand may be modified.
  • One or more of the even numbered nucleotides of the first strand may be modified by at least a second modification, wherein the at least second modification is different from the modification on the one or more odd nucleotides.
  • At least one of the one or more modified even numbered nucleotides may be adjacent to at least one of the one or more modified odd numbered nucleotides.
  • a plurality of odd numbered nucleotides in the first strand may be modified in the nucleic acid of the invention.
  • a plurality of even numbered nucleotides in the first strand may be modified by a second modification.
  • the first strand may comprise adjacent nucleotides that are modified by a common modification.
  • the first strand may also comprise adjacent nucleotides that are modified by a second different modification.
  • One or more of the odd numbered nucleotides of the second strand may be modified by a modification that is different to the modification of the odd numbered nucleotides on the first strand and/or one or more of the even numbered nucleotides of the second strand may be modified by the same modification of the odd numbered nucleotides of the first strand. At least one of the one or more modified even numbered nucleotides of the second strand may be adjacent to the one or more modified odd numbered nucleotides.
  • a plurality of odd numbered nucleotides of the second strand may be modified by a common modification and/or a plurality of even numbered nucleotides may be modified by the same modification that is present on the first stand odd numbered nucleotides.
  • a plurality of odd numbered nucleotides on the second strand may be modified by a second modification, wherein the second modification is different from the modification of the first strand odd numbered nucleotides.
  • the second strand may comprise adjacent nucleotides that are modified by a common modification, which may be a second modification that is different from the modification of the odd numbered nucleotides of the first strand.
  • each of the odd numbered nucleotides in the first strand and each of the even numbered nucleotides in the second strand may be modified with a common modification and, each of the even numbered nucleotides may be modified in the first strand with a second modification and each of the odd numbered nucleotides may be modified in the second strand with a second different modification.
  • the nucleic acid of the invention may have the modified nucleotides of the first strand shifted by at least one nucleotide relative to the unmodified or differently modified nucleotides of the second strand.
  • One or more or each of the odd numbered nucleotides may be modified in the first strand and one or more or each of the even numbered nucleotides may be modified in the second strand.
  • One or more or each of the alternating nucleotides on either or both strands may be modified by a second modification.
  • One or more or each of the even numbered nucleotides may be modified in the first strand and one or more or each of the even numbered nucleotides may be modified in the second strand.
  • One or more or each of the alternating nucleotides on either or both strands may be modified by a second modification.
  • One or more or each of the odd numbered nucleotides may be modified in the first strand and one or more of the odd numbered nucleotides may be modified in the second strand by a common modification.
  • One or more or each of the alternating nucleotides on either or both strands may be modified by a second modification.
  • One or more or each of the even numbered nucleotides may be modified in the first strand and one or more or each of the odd numbered nucleotides may be modified in the second strand by a common modification.
  • One or more or each of the alternating nucleotides on either or both strands may be modified by a second modification.
  • the nucleic acid of the invention may comprise single or double stranded constructs that comprise at least two regions of alternating modifications in one or both of the strands. These alternating regions can comprise up to about 12 nucleotides but preferably comprise from about 3 to about 10 nucleotides. The regions of alternating nucleotides may be located at the termini of one or both strands of the nucleic acid of the invention.
  • the nucleic acid may comprise from 4 to about 10 nucleotides of alternating nucleotides at each termini (3′ and 5′) and these regions may be separated by from about 5 to about 12 contiguous unmodified or differently or commonly modified nucleotides.
  • the odd numbered nucleotides of the first strand may be modified and the even numbered nucleotides may be modified with a second modification.
  • the second strand may comprise adjacent nucleotides that are modified with a common modification, which may be the same as the modification of the odd numbered nucleotides of the first strand.
  • One or more nucleotides of second strand may also be modified with the second modification.
  • One or more nucleotides with the second modification may be adjacent to each other and to nucleotides having a modification that is the same as the modification of the odd numbered nucleotides of the first strand.
  • the first strand may also comprise phosphorothioate linkages between the two nucleotides at the 3′ end and at the 5′ end.
  • the second strand may comprise a phosphorothioate linkage between the two nucleotides at 5′ end.
  • the second strand may also be conjugated to a ligand at the 5′ end.
  • the nucleic acid of the invention may comprise a first strand comprising adjacent nucleotides that are modified with a common modification.
  • One or more of such nucleotides may be adjacent to one or more nucleotides which may be modified with a second modification.
  • One or more nucleotides with the second modification may be adjacent.
  • the second strand may comprise adjacent nucleotides that are modified with a common modification, which may be the same as one of the modifications of one or more nucleotides of the first strand.
  • One or more nucleotides of second strand may also be modified with the second modification.
  • One or more nucleotides with the second modification may be adjacent.
  • the first strand may also comprise phosphorothioate linkages between the two nucleotides at the 5′ end and at the 3′ end.
  • the second strand may comprise a phosphorothioate linkage between the two nucleotides at the 3′ end.
  • the second strand may also be conjugated to a ligand at the 5′ end.
  • nucleotides numbered from 5′ to 3′ on the first strand and 3′ to 5′ on the second strand, 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 and 25 may be modified by a modification on the first strand.
  • the nucleotides numbered 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22 and 24 may be modified by a second modification on the first strand.
  • the nucleotides numbered 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 may be modified by a modification on the second strand.
  • the nucleotides numbered 2, 4, 8, 10, 12, 14, 16, 18, 20, 22 and 24 may be modified by a second modification on the second strand.
  • Nucleotides are numbered for the sake of the nucleic acid of the present invention from 5′ to 3′ on the first strand and 3′ to 5′ on the second strand
  • the nucleotides numbered 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22 and 24 may be modified by a modification on the first strand.
  • the nucleotides numbered 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 may be modified by a second modification on the first strand.
  • the nucleotides numbered 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23 may be modified by a modification on the second strand.
  • the nucleotides numbered 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22 and 24 may be modified by a second modification on the second strand.
  • first and/or the second strand are shorter than 25 nucleotides in length, such as 19 nucleotides in length, there are no nucleotides numbered 20, 21, 22, 23, 24 and 25 to be modified.
  • nucleotides numbered 20, 21, 22, 23, 24 and 25 there are no nucleotides numbered 20, 21, 22, 23, 24 and 25 to be modified. The skilled person understands the description above to apply to shorter strands, accordingly.
  • One or more modified nucleotides on the first strand may be paired with modified nucleotides on the second strand having a common modification.
  • One or more modified nucleotides on the first strand may be paired with modified nucleotides on the second strand having a different modification.
  • One or more modified nucleotides on the first strand may be paired with unmodified nucleotides on the second strand.
  • One or more modified nucleotides on the second strand may be paired with unmodified nucleotides on the first strand.
  • the alternating nucleotides can be aligned on the two strands such as, for example, all the modifications in the alternating regions of the second strand are paired with identical modifications in the first strand or alternatively the modifications can be offset by one nucleotide with the common modifications in the alternating regions of one strand pairing with dissimilar modifications (i.e. a second or further modification) in the other strand.
  • dissimilar modifications i.e. a second or further modification
  • the modifications on the first strand may be shifted by one nucleotide relative to the modified nucleotides on the second strand, such that common modified nucleotides are not paired with each other.
  • the modification and/or modifications may each and individually be selected from the group consisting of 3′-terminal deoxy-thymine, 2′-O-methyl, a 2′-deoxy-modification, a 2′-amino-modification, a 2′-alkyl-modification, a morpholino modification, a phosphoramidate modification, 5′-phosphorothioate group modification, a 5′ phosphate or 5′ phosphate mimic modification and a cholesteryl derivative or a dodecanoic acid bisdecylamide group modification and/or the modified nucleotide may be any one of a locked nucleotide, an abasic nucleotide or a non-natural base comprising nucleotide.
  • At least one modification may be 2′-O-methyl and/or at least one modification may be 2′-F. Further modifications as described herein may be present on the first and/or second strand.
  • “same or common modification” means the same modification to any nucleotide, be that A, G, C or U modified with a group such as a methyl group or a fluoro group. Is it not taken to mean the same addition on the same nucleotide.
  • 2′F-dU, 2′F-dA, 2′F-dC, 2′F-dG are all considered to be the same or common modification, as are 2′-OMe-rU, 2′-OMe-rA; 2′-OMe-rC; 2′-OMe-rG
  • a 2-′F modification is a different modification to a 2′-OMe modification.
  • the nucleic acid may comprise a modification and a second or further modification which are each and individually selected from the group comprising 2′-O-methyl modification and 2′-F modification.
  • the nucleic acid may comprise a modification that is 2′-O-methyl (2′-OMe) that may be a first modification, and a second modification that is 2′-F.
  • the nucleic acid of the invention may also include a phosphorothioate modification and/or a deoxy modification which may be present in or between the terminal 2 or 3 nucleotides of each or any end of each or both strands.
  • the invention provides as a further aspect, a nucleic acid for inhibiting expression of LPA in a cell, comprising a nucleotide sequence of SEQ ID NO: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, wherein the nucleotides of first strand are modified by a first modification on the odd numbered nucleotides, and modified by a second modification on the even numbered nucleotides, and nucleotides of the second strand are modified by a third modification on the even numbered nucleotides and modified by a fourth modification the odd numbered nucleotides, wherein at least the first modification is different to the second modification and the third modification is different to the fourth modification.
  • the third and first modifications may be the same or different
  • the second and fourth modifications may be the same or different.
  • the first and second modifications may be different to each other and the third and fourth modifications may be different to each other.
  • the second strand may comprise a nucleotide sequence of SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, or 44.
  • the nucleotides of the first strand may be modified by a first modification on the odd numbered nucleotides, and modified with a second modification on the even numbered nucleotides, and the second strand may be modified on the odd numbered nucleotides with the second modification and modified with the first modification on the even numbered nucleotides.
  • the first modification may be 2′OMe and the second modification may be 2′ F.
  • the first strand may comprise the nucleotide sequence of SEQ ID NO: 5 or SEQ ID NO: 9 and/or the second strand may comprise the nucleotide sequence of SEQ ID NO: 6, or SEQ ID NO:10.
  • the modifications may be those as set out in Table 1.
  • a nucleotide on the second strand that “corresponds to” a position on the first strand is suitably the nucleotide that base pairs with that nucleotide on the first strand.
  • the nucleotide on the second strand which corresponds to position 13 of the first strand is the nucleotide that forms a base pair with position 13 of the first strand.
  • the nucleotide on the second strand which corresponds to position 11 of the first strand is the nucleotide that forms a base pair with position 11 of the first strand.
  • the nucleotide on the second strand which corresponds to position 12 of the first strand is the nucleotide that forms a base pair with position 12 of the first strand.
  • This nomenclature may be applied to other positions of the second strand.
  • position 13 of the first strand would pair with position 7 of the second strand.
  • Position 11 of the first strand would pair with position 9 of the second strand.
  • This nomenclature may be applied to other positions of the second strand.
  • the nucleotide that corresponds to position 13 of the first strand is suitably position 13 of the second strand, counting from the 3′ of the second strand, starting from the first nucleotide of the double stranded region.
  • position 11 of the second strand is suitably the 11 th nucleotide from the 3′ of the second strand, starting from the first nucleotide of the double stranded region. This nomenclature may be applied to other positions of the second strand.
  • the nucleotide on the second strand that “corresponds to” a position on the first strand may not necessarily form a base pair if that position is the position in which there is a mismatch, but the principle of the nomenclature still applies.
  • Preferred is a first and second strand that are fully complementary over the duplex region (ignoring any overhang regions) and there are no mismatches within the double stranded region of the nucleic acid.
  • a nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotides on the second strand which corresponds to position 11 and 13 of the first strand are not modified with a 2′ O-methyl modification.
  • nucleotide on the second strand which corresponds to position 12 of the first strand is not modified with a 2′ O-methyl modification.
  • This limitation on the nucleic acid may be seen with any other limitation described herein.
  • nucleic acid as disclosed herein, wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotides on the second strand which corresponds to position 11-13 of the first strand are not modified with a 2′ O-methyl modification.
  • all even numbered nucleotide of the first strand are modified with a 2′ fluoro modification and all odd numbered nucleotides of the first strand are modified with a 2′ O-methyl modification.
  • nucleotides on the second strand other than those which correspond to position 11, or 13, or 11 and 13, or preferably 11-13 of the first strand are modified with a 2′ O-methyl modification.
  • One advantage of such a nucleic is that it comprises relatively few non-naturally occurring modified nucleotides but is nonetheless able to efficiently inhibit the target gene for long periods of time.
  • Such a nucleic acid is easier to synthesise than corresponding nucleic acids with more non-naturally occurring (2′F modified) nucleotides.
  • Suitable naturally occurring modifications include, as well as 2 O′ methyl, other 2′ sugar modifications, in particular a 2′ H modification resulting in a DNA nucleotide.
  • a nucleic acid as disclosed herein comprising no more than 20%, such as no more than 15% such as no more than 10%, of nucleotides which have 2′ modifications that are not 2′ O methyl modifications on the first and/or second strand, preferably as a percentage of the total nucleotides of both the first and second strands.
  • a nucleic acid as disclosed herein comprising no more than 20%, (such as no more than 15% or no more than 10%) of 2′ fluoro modifications on the first and/or second strand, preferably as a percentage of the total nucleotides of both strands.
  • nucleic acid as disclosed herein wherein all nucleotides are modified with a 2′ O-methyl modification except positions 2 and 14 from the 5′ end of the first strand and the nucleotides on the second strand which correspond to position 11, or 13, or 11 and 13, or preferably 11-13 of the first strand.
  • nucleotides that are not modified with 2′ O-methyl are modified with fluoro at the 2′ position.
  • nucleic acid as disclosed herein wherein all nucleotides of the nucleic acid are modified at the 2′ position of the sugar.
  • these nucleotides are modified with a 2′-fluoro modification where the modification is not a 2′ O-Methyl modification.
  • Nucleic acids of the invention may comprise one or more nucleotides modified at the 2′ position with a 2′ H, and therefore having a DNA nucleotide within the nucleic acid.
  • Nucleic acids of the invention may comprise DNA nucleotides at positions 2 and/or 14 of the first strand counting from the 5′ end of the first strand.
  • Nucleic acids may comprise DNA nucleotides on the second strand which correspond to position 11, or 13, or 11 and 13, or 11-13 of the first strand.
  • Nucleic acids of the invention may comprise one or more LNA nucleotides. Nucleic acids of the invention may comprise LNA nucleotides at positions 2 and/or 14 of the first strand counting from the 5′ end of the first strand. Nucleic acids may comprise LNA on the second strand which correspond to position 11, or 13, or 11 and 13, or 11-13 of the first strand.
  • the nucleic acid is modified on the first strand, preferably along the entire strand, with alternating 2′ O-methyl modifications and 2′ fluoro modifications, and positions 2 and 14 (starting from the 5′ end) are modified with 2′ fluoro.
  • the second strand is modified with 2′ fluoro modifications at nucleotides on the second strand which correspond to position 11, or 13, or 11 and 13, or preferably 11-13 of the first strand.
  • the second strand is modified with 2′ fluoro modifications at positions 11-13 counting from the 3′ end starting at the first position of the complementary (double stranded) region, and the remaining modifications are naturally occurring modifications, preferably 2′ O-methyl.
  • the nucleic acid preferably has a blunt end at least at the end that comprises the 5′ end of the first strand.
  • a nucleotide of the second strand that is in a position corresponding for example to an even-numbered nucleotide of the first strand is a nucleotide of the second strand that is base-paired to an even-numbered nucleotide of the first strand.
  • the nucleotide/nucleotides of the second strand in a position corresponding to nucleotide 11 or nucleotide 13 or nucleotides 11 and 13 or preferably nucleotides 11-13 of the first strand is/are modified by a fourth modification.
  • all the nucleotides of the second strand other than the nucleotide/nucleotides in a position corresponding to nucleotide 11 or nucleotide 13 or nucleotides 11 and 13 or preferably nucleotides 11-13 of the first strand is/are modified by a third modification.
  • nucleotides 2 and 14 or preferably all the even numbered nucleotides of the first strand are modified with a first modification.
  • the odd-numbered nucleotides of the first strand are modified with a second modification.
  • the fourth modification is preferably different from the second modification and preferably different from the third modification and the fourth modification is preferably the same as the first modification.
  • the first and the fourth modification are preferably a 2′-OMe modification and the second and third modification are preferably a 2′-F modification.
  • the nucleotides on the first strand are numbered consecutively starting with nucleotide number 1 at the 5′ end of the first strand.
  • all the even-numbered nucleotides of the first strand are modified by a first modification
  • all the odd-numbered nucleotides of the first strand are modified by a second modification
  • all the nucleotides of the second strand in positions corresponding to nucleotides 11-13 of the first strand are modified by a fourth modification
  • all the nucleotides of the second strand other than the nucleotides corresponding to nucleotides 11-13 of the first strand are modified by a third modification, wherein the first and fourth modification are 2′-F and the second and third modification are 2′-OMe.
  • the nucleotides on the first strand are numbered consecutively starting with nucleotide number 1 at the 5′ end of the first strand.
  • each of the nucleotides of the first strand and of the second strand is a modified nucleotide.
  • One aspect is a double-stranded nucleic acid for inhibiting expression of LPA, preferably in a cell, wherein the nucleic acid comprises a first strand and a second strand, wherein the first strand sequence comprises a sequence of at least 15 nucleotides differing by no more than 3 nucleotides from any one of the sequences SEQ ID NO: 9, 5, 1, 3, 7, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, preferably SEQ ID NO: 9, wherein all the even-numbered nucleotides of the first strand are modified by a first modification, all the odd-numbered nucleotides of the first strand are modified by a second modification, all the nucleotides of the second strand in positions corresponding to nucleotides 11-13 of the first strand are modified by a fourth modification, all the nucleotides of the second strand other than the nucleotides corresponding to nucleotides 11-13 of the first strand modified by a
  • nucleic acids which are siRNA molecules wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleic acid comprises one or more or all of:
  • nucleic acid as disclosed herein, wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand and the nucleotides at positions 7 and/or 9, or 7-9 from the 5′ end of the second strand are modified with a 2′ fluoro modification, and at least 90% of the remaining nucleotides are 2′-O methyl modified or comprise another naturally occurring 2′ modification.
  • blunt double stranded 19 base nucleic acid with no overhang, are:
  • a nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotide at position 7 from the 5′ end of the second strand is not modified with a 2′ O-methyl modification.
  • nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotide at position 9 from the 5′ end of the second strand is not modified with a 2′ O-methyl modification
  • a nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotides at position 7 and 9 from the 5′ end of the second strand are not modified with a 2′ O-methyl modification.
  • a nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotides at positions 7-9 from the 5′ end of the second strand are not modified with a 2′ O-methyl modification.
  • nucleic acid as disclosed herein wherein the nucleotides at positions 2 and 14 from the 5′ end of the first strand are not modified with a 2′ O-methyl modification, and the nucleotides at positions 7 and/or 9, or 7-9 from the 5′ end of the second strand are modified with a 2′ fluoro modification.
  • Suitable naturally occurring modifications include, as well as 2 O′ methyl, other 2′ sugar modifications, in particular a 2′ H modification resulting in a DNA nucleotide.
  • a nucleic acid as disclosed herein comprising no more than 20%, such as no more than 15% such as more than 10%, of nucleotides which have 2′ modifications that are not 2′ 0 methyl modifications on the first and/or second strand, preferably as a percentage of the total nucleotides of both the first and second strands.
  • a nucleic acid as disclosed herein comprising no more than 20%, (such as no more than 15% or no more than 10%) of 2′ fluoro modifications on the first and/or second strand, preferably as a percentage of the total nucleotides of both strands.
  • nucleic acid as disclosed herein wherein all nucleotides are modified with a 2′ O-methyl modification except positions 2 and 14 from the 5′ end of the first strand and the nucleotides at positions 7 and/or 9 from the 5′ end of the second strand.
  • nucleotides that are not modified with 2′ O-methyl are modified with fluoro at the 2′ position.
  • nucleic acid as disclosed herein wherein all nucleotides are modified with a 2′ O-methyl modification except positions 2 and 14 from the 5′ end of the first strand and the nucleotides at positions 7-9 from the 5′ end of the second strand.
  • nucleotides that are not modified with 2′ O-methyl are modified with fluoro at the 2′ position.
  • the second strand preferably does not have a 2′ O-methyl group at nucleotides 8 or 9 or 10 counting from the 5′ end of the duplex corresponding to positions 13, 12, and 11 of the first strand respectively.
  • the second strand preferably does not have a 2′ O-methyl group at nucleotides 9 or 10 or 11 counting from the 5′ end of the duplex corresponding to positions 13, 12, and 11 of the first strand respectively.
  • the nucleic acid of the present invention may include one or more phosphorothioate modifications on one or more of the ends of the first and/or the second strand.
  • each or either end of the first strand may comprise one or two or three phosphorothioate modified nucleotides.
  • each or either end of the second strand may comprise one or two or three phosphorothioate modified nucleotides.
  • the first strand may include at least one phosphorothioate (ps) linkage.
  • the first strand may further comprise a phosphorothioate linkage between the terminal two 3′ nucleotides or phosphorothioate linkages between the terminal three 3′ nucleotides.
  • the linkages between the other nucleotides in the first strand are phosphodiester linkages.
  • the first strand may include more than 1 phosphorothioate linkage.
  • the second strand may comprise a phosphorothioate linkage between the terminal two 3′ nucleotides or phosphorothioate linkages between the terminal three 3′ nucleotides.
  • the second strand may comprise a phosphorothioate linkage between the terminal two 5′ nucleotides or phosphorothioate linkages between the terminal three 5′ nucleotides.
  • the nucleic acid comprises one or more phosphorodithioate linkages, such as 1, 2, 3 or 4 phosphorodithioate linkages. Preferably there are up to 4 phosphorodithioate linkages, one each at the 5′ and 3′ ends of the first and second strands.
  • a phosphorodithioate linkage in the nucleic acid of the invention reduces the variation in the stereochemistry of a population of nucleic acid molecules compared to molecules comprising a phosphorothioate in that same position.
  • Phosphorothioate linkage indeed introduce a chiral centre and it is difficult to control which non-linking oxygen is substituted for sulphur.
  • the use of a phosphorodithioate ensures that no chiral centre exists in that linkage and thus reduces or eliminates any variation in the population of nucleic acid molecules, depending on the number of phosphorodithioate and phosphorothioate linkages used in the nucleic acid molecule.
  • the nucleic acid comprises a phosphorothioate linkage between each of the three terminal 3′ nucleotides and/or between each of the three terminal 5′ nucleotides on the first strand, and/or between each of the three terminal 3′ nucleotides and/or between each of the three terminal 5′ nucleotides of the second strand when there is no phosphorodithioate linkage present at that end.
  • No phosphorodithioate linkage being present at an end means that the linkage between the two terminal nucleotides, or preferably between the three terminal nucleotides of the nucleic acid end in question are linkages other than phosphorodithioate linkages.
  • the invention also provides a nucleic acid according to any aspect of the invention described herein, wherein the first RNA strand has a terminal 5′ (E)-vinylphosphonate nucleotide, and the terminal 5′ (E)-vinylphosphonate nucleotide is linked to the second nucleotide in the first strand by a phosphodiester linkage.
  • the first strand may include more than one phosphodiester linkage.
  • the first strand may comprise phosphodiester linkages between at least the terminal three 5′ nucleotides.
  • the first strand may comprise phosphodiester linkages between at least the terminal four 5′ nucleotides.
  • the first strand may comprise formula (IV):
  • ‘(vp)-’ is the 5′ (E)-vinylphosphonate
  • ‘N’ is a nucleotide
  • ‘po’ is a phosphodiester linkage
  • n is from 1 to (the total number of nucleotides in the first strand—2), preferably wherein n is from 1 to (the total number of nucleotides in the first strand—3), more preferably wherein n is from 1 to (the total number of nucleotides in the first strand—4).
  • the 5′-most nucleotide of the first strand is a nucleotide other than A or U, this nucleotide is replaced by A or U in the sequence.
  • this nucleotide is replaced by U, and more preferably by U with a 5′ vinylphosphonate, in the sequence.
  • a terminal 5′ (E)-vinylphosphonate nucleotide is a nucleotide wherein the natural phosphate group at the 5′-end has been replaced with a E-vinylphosphonate, in which the bridging 5′-oxygen atom of the terminal nucleotide of the 5′ phosphorylated strand is replaced with a methynyl (—CH ⁇ ) group:
  • 5′ (E) vinylphosphonate is a 5′ phosphate mimic.
  • a biological mimic is a molecule that is capable of carrying out the same function as and is structurally very similar to the original molecule that is being mimicked.
  • 5′ (E) vinylphosphonate mimics the function of a normal 5′ phosphate, e.g. enabling efficient RISC loading.
  • 5′ (E) vinylphosphonate is capable of stabilizing the 5′-end nucleotide by protecting it from dephosphorylation by enzymes such as phosphatases.
  • the terminal 5′ (E)-vinylphosphonate nucleotide is an RNA nucleotide.
  • nucleic acid in one aspect, the nucleic acid:
  • nucleic acid in one aspect, the nucleic acid:
  • the nucleic acid which is preferably an siRNA that inhibits expression of LPA, preferably via RNAi, comprises one or more or all of:
  • the nucleic acids of the invention may include one or more inverted nucleotides, for example inverted thymidine or inverted adenine (for example see Takei, et al., 2002. JBC 277 (26):23800-06).
  • the nucleic acid of the invention comprises one or more inverted ribonucleotides, preferably an inverted adenine, using a 5′-5′ linkage or a 3′-3′ linkage, preferably a 3′-3′ linkage at the 3′ end of the second strand.
  • the nucleic acid of the invention may comprise an inverted RNA nucleotide at one or several of the strand ends.
  • Such inverted nucleotides provide stability to the nucleic acid.
  • the nucleic acid comprises at least an inverted nucleotide at one or several of the 3′ end of at least one of the strands and/or at the 5′ end of the of the second strand. More preferably, the nucleic acid comprises an inverted nucleotide at the 3′ end of the second strand.
  • the nucleic acid comprises an inverted RNA nucleotide at the 3′ end of the second strand and this nucleotide is preferably an inverted A.
  • the inverted nucleotide is preferably present at an end of a strand not as an overhang but opposite a corresponding nucleotide in the other strand. A nucleic acid with such a modification is stable and easy to synthesise.
  • the nucleic acid of the invention may be conjugated to a ligand.
  • Efficient delivery of oligonucleotides, in particular double stranded nucleic acids of the invention, to cells in vivo is important and requires specific targeting and substantial protection from the extracellular environment, particularly serum proteins.
  • One method of achieving specific targeting is to conjugate a ligand to the nucleic acid.
  • the ligand helps in targeting the nucleic acid to the required target site.
  • conjugate appropriate ligands for the desired receptor molecules in order for the conjugated molecules to be taken up by the target cells by mechanisms such as different receptor-mediated endocytosis pathways or functionally analogous processes.
  • ASGP-R asialoglycoprotein receptor complex
  • ASGP-R asialoglycoprotein receptor complex
  • GalNAc N-Acetyl-D-Galactosylamine
  • the asialoglycoprotein receptor complex (ASGP-R), which recognizes specifically terminal ⁇ -galactosyl subunits of glycosylated proteins or other oligosaccharides (Weigel, P. H. et. al., Biochim. Biophys. Acta. 2002 Sep. 19; 1572(2-3):341-63) can be used for delivering a drug to the liver's hepatocytes expressing the receptor complex by covalent coupling of galactose or galactosamine to the drug substance (Ishibashi, S.; et. al., J Biol. Chem. 1994 Nov. 11; 269(45):27803-6). Furthermore the binding affinity can be significantly increased by the multi-valency effect, which is achieved by the repetition of the targeting moiety (Biessen E A, et al., J Med Chem. 1995 Apr. 28; 38(9):1538-46.).
  • the ASGP-R complex is a mediator for an active uptake of terminal ⁇ -galactosyl containing glycoproteins to the cell's endosomes.
  • the ASGPR is highly suitable for targeted delivery of drug candidates conjugated to such ligands like, e.g., nucleic acids into receptor-expressing cells (Akinc et al., Mol Ther. 2010 July; 18(7):1357-64).
  • the ligand can comprise a saccharide that is selected to have an affinity for at least one type of receptor on a target cell.
  • the receptor is on the surface of a mammalian liver cell, for example, the hepatic asialoglycoprotein receptor complex described before (ASGP-R).
  • the saccharide may be selected from N-acetyl galactosamine, mannose, galactose, glucose, glucosamine and fucose.
  • the saccharide may be N-acetyl galactosamine (GalNAc).
  • a ligand for use in the present invention may therefore comprise (i) one or more N-acetyl galactosamine (GalNAc) moieties and derivatives thereof, and (ii) a linker, wherein the linker conjugates the GalNAc moieties to a sequence as defined in any preceding aspects.
  • the linker may be a bivalent or trivalent or tetravalent branched structure.
  • the nucleotides may be modified as defined herein.
  • GalNAc refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose, commonly referred to in the literature as N-acetyl galactosamine.
  • Reference to “GalNAc” or “N-acetyl galactosamine” includes both the ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose and the ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose.
  • both the ⁇ -form: 2-(Acetylarnino)-2-deoxy- ⁇ -D-galactopyranose and ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose may be used interchangeably.
  • the compounds of the invention comprise the ⁇ -form, 2-(Acetylarnino)-2-deoxy- ⁇ -D-galactopyranose.
  • the ligand may therefore comprise GalNAc.
  • the ligand may comprise a compound of formula (I):
  • branching unit “A” branches into three in order to accommodate the three saccharide ligands.
  • the branching unit is covalently attached to the remaining tethered portions of the ligand and the nucleic acid.
  • the branching unit may comprise a branched aliphatic group comprising groups selected from alkyl, amide, disulphide, polyethylene glycol, ether, thioether and hydroxyamino groups.
  • the branching unit may comprise groups selected from alkyl and ether groups.
  • the branching unit A may have a structure selected from:
  • each A 1 independently represents O, S, C ⁇ O or NH
  • each n independently represents an integer from 1 to 20.
  • the branching unit may have a structure selected from:
  • each A 1 independently represents O, S, C ⁇ O or NH
  • each n independently represents an integer from 1 to 20.
  • the branching unit may have a structure selected from:
  • a 1 is O, S, C ⁇ O or NH
  • each n independently represents an integer from 1 to 20.
  • the branching unit may have the structure:
  • the branching unit may have the structure:
  • the branching unit may have the structure:
  • the branching unit consists of only a carbon atom.
  • the “X 3 ” portion is a bridging unit.
  • the bridging unit is linear and is covalently bound to the branching unit and the nucleic acid.
  • X 3 may be selected from —C 1 -C 20 alkylene-, —C 2 -C 20 alkenylene-, an alkylene ether of formula —(C 1 -C 20 alkylene)-O—(C 1-20 alkylene)-, —C(O)—C 1 -C 20 alkylene-, —C 0 -C 4 alkylene(Cy)C 0 -C 4 alkylene- wherein Cy represents a substituted or unsubstituted 5 or 6 membered cycloalkylene, arylene, heterocyclylene or heteroarylene ring, —C 1 -C 4 alkylene-NHC(O)—C 1 -C 4 alkylene-, —C 1 -C 4 alkylene-C(O)NH—C 1-4 alkylene-, —C 1-4 alkylene-SC(O)—C 1 -C 4 alkylene-, —C 1 -C 4 alkylene-C(O)S—
  • X 3 may be an alkylene ether of formula —(C 1 -C 20 alkylene)-O—(C 1 -C 20 alkylene)-.
  • X 3 may be an alkylene ether of formula —(C 1 -C 20 alkylene)-O—(C 4 -C 20 alkylene)-, wherein said (C 4 -C 20 alkylene) is linked to Z.
  • X 3 may be selected from the group consisting of —CH 2 —O—C 3 H 6 —, —CH 2 —O—C 4 H 8 —, —H 2 O—C 6 H 12 — and —H 2 —O—C 8 H 16 —, especially —H 2 —O—C 4 H 8 —, —CH 2 —O—C 6 H 12 — and —H 2 —O—C 8 H 16 —, wherein in each case the —CH 2 — group is linked to A.
  • the ligand may comprise a compound of formula (II):
  • Branching unit A may have the structure:
  • Branching unit A may have the structure:
  • X 3 may be C 1 -C 20 alkylene.
  • X 3 is selected from the group consisting of —C 3 H 6 —, —C 4 H 8 —, —C 6 H 12 — and —C 8 H 16 — especially —C 4 H 8 —, —C 6 H 12 — and —C 8 H 16 —.
  • the ligand may comprise a compound of formula (III):
  • the branching unit may comprise carbon.
  • the branching unit is carbon.
  • X 3 may be selected from the group consisting of —CH 2 —O—C 4 H 8 —, —CH 2 —O—C 5 H 10 —, —CH 2 —O—C 6 H 12 —, —CH 2 —O—C 7 H 14 —, and —CH 2 —O—C 8 H 16 —.
  • X 3 is selected from the group consisting of —CH 2 —O—C 4 H 8 —, —CH 2 —O—C 6 H 12 — and —CH 2 —O—C 8 H 16 .
  • P when P represents a modified phosphate group, P can be represented by:
  • Y 1 and Y 2 each independently represent ⁇ O, ⁇ S, —O ⁇ , —OH, —SH, —BH 3 , —OCH 2 CO 2 , —OCH 2 CO 2 R x , —OCH 2 C(S)OR x , and —OR x , wherein R x represents C 1 -C 6 alkyl and wherein indicates attachment to the remainder of the compound.
  • modified phosphate it is meant a phosphate group wherein one or more of the non-linking oxygens is replaced.
  • modified phosphate groups include phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulphur.
  • One, each or both non-linking oxygens in the phosphate group can be independently any one of S, Se, B, C, H, N, or OR (R is alkyl or aryl).
  • the phosphate can also be modified by replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylenephosphonates).
  • the replacement can occur at a terminal oxygen. Replacement of the non-linking oxygens with nitrogen is possible.
  • Y 1 may represent —OH and Y 2 may represent ⁇ O or ⁇ S; or
  • Y 1 may represent —O ⁇ and Y 2 may represent ⁇ O or ⁇ S;
  • Y 1 may represent ⁇ O and Y 2 may represent —CH 3 , —SH, —OR x , or —BH 3
  • Y 1 may represent ⁇ S and Y 2 may represent —CH 3 , OR x or —SH.
  • the modified phosphate group is a thiophosphate group.
  • Thiophosphate groups include bithiophosphate (i.e. where Y 1 represents ⁇ S and Y 2 represents —S ⁇ ) and monothiophosphate (i.e. where Y 1 represents —O ⁇ and Y 2 represents ⁇ S, or where Y 1 represents ⁇ O and Y 2 represents —S ⁇ ).
  • P is a monothiophosphate.
  • conjugates having thiophosphate groups in replacement of phosphate groups have improved potency and duration of action in vivo.
  • P may also be an ethylphosphate (i.e. where Y 1 represents ⁇ O and Y 2 represents OCH 2 CH 3 ).
  • the saccharide may be selected to have an affinity for at least one type of receptor on a target cell.
  • the receptor is on the surface of a mammalian liver cell, for example, the hepatic asialoglycoprotein receptor complex (ASGP-R).
  • the saccharide may be selected from N-acetyl with one or more of galactosamine, mannose, galactose, glucose, glucosamine and fructose.
  • a ligand to be used in the present invention may include N-acetyl galactosamine (GalNAc).
  • the compounds of the invention may have 3 ligands, which will each preferably include N-acetyl galactosamine.
  • GalNAc refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose, commonly referred to in the literature as N-acetyl galactosamine.
  • Reference to “GalNAc” or “N-acetyl galactosamine” includes both the ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose and the ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose.
  • both the ⁇ -form: 2-(Acetylarnino)-2-deoxy- ⁇ -D-galactopyranose and ⁇ -form: 2-(Acetylamino)-2-deoxy- ⁇ -D-galactopyranose may be used interchangeably.
  • the compounds of the invention comprise the ⁇ -form, 2-(Acetylarnino)-2-deoxy- ⁇ -D-galactopyranose.
  • X 1 may be (—CH 2 —CH 2 —O)(—CH 2 ) 2 —.
  • X 1 may be (—CH 2 —CH 2 —O) 2 (—CH 2 ) 2 —.
  • X 1 may be (—CH 2 —CH 2 —O) 3 (—CH 2 ) 2 —.
  • X 1 is (—CH 2 —CH 2 —O) 2 (—CH 2 ) 2 —.
  • X 1 represents C 3 -C 6 alkylene.
  • X 1 may be propylene.
  • X 1 may be butylene.
  • X 1 may be pentylene.
  • X 1 may be hexylene.
  • the alkyl is a linear alkylene. In particular, X 1 may be butylene.
  • X 2 represents an alkylene ether of formula —C 3 H 6 —O—CH 2 —i.e. C 3 alkoxy methylene, or —CH 2 CH 2 CH 2 OCH 2 —.
  • the invention provides a conjugated nucleic acid having one of the following structures:
  • Z is a nucleic acid as defined herein before and is preferably conjugated to the 5′ end of the second strand of the nucleic acid.
  • the conjugated nucleic acid has the following structure:
  • Z is a nucleic acid as defined herein before and is preferably conjugated to the 5′ end of the second strand.
  • a ligand of formula (I), (II) or (III) can be attached at the 3′-end of the first (antisense) strand and/or at any of the 3′- and/or 5′-end of the second (sense) strand.
  • the nucleic acid can comprise more than one ligand of formula (I), (II), or (III).
  • a single ligand of formula (I), (II) or (III) is preferred because a single such ligand is sufficient for efficient targeting of the nucleic acid to the target cells.
  • at least the last two, preferably at least the last three and more preferably at least the last four nucleotides at the end of the nucleic acid to which the ligand is attached are linked by a phosphodiester linkage.
  • the 5′-end of the first (antisense) strand is not attached to a ligand of formula (I), (II) or (III), since a ligand in this position can potentially interfere with the biological activity of the nucleic acid.
  • a nucleic acid with a single ligand of formula (I), (II) or (III) at the 5′-end of a strand is easier and therefore cheaper to synthesis than the same nucleic acid with the same ligand at the 3′-end.
  • a single ligand of any of formulae (I), (II) or (III) is covalently attached to (conjugated with) the 5′-end of the second strand of the nucleic acid.
  • the nucleic acid is conjugated to a ligand that comprises a lipid, and more preferably a ligand that comprises a cholesterol.
  • nucleic acid according to the present invention may be conjugated to a ligand of the following structure
  • a conjugate of the invention can comprise any nucleic acid as disclosed herein conjugated to any ligand or ligands as disclosed herein.
  • the present invention also relates to a conjugate for inhibiting expression of a LPA gene in a cell, said conjugate comprising a nucleic acid portion, comprising the nucleic acid of any aspect of the invention, and at least one ligand portion, said nucleic acid portion comprising at least one duplex region that comprises at least a portion of a first RNA strand and at least a portion of a second RNA strand that is at least partially complementary to the first strand, wherein said first strand is at least partially complementary to at least a portion of RNA transcribed from said LPA gene, said at least one ligand portion comprising a linker moiety, preferably a serinol-derived linker moiety, and a targeting ligand for in vivo targeting of cells and being conjugated exclusively to the 3′ and/or 5′ ends of one or both RNA strands, wherein the 5′ end of the first RNA strand is not conjugated, wherein:
  • the ligands may be monomeric or multimeric (e.g. dimeric, trimeric, etc.).
  • the ligands are monomeric, thus containing a single targeting ligand moiety, e.g. a single GalNAc moiety.
  • the ligands may be dimeric ligands wherein the ligand portions comprise two linker moieties, such as serinol-derived linker moieties or non-serinol linker moieties, each linked to a single targeting ligand moiety.
  • the ligands may be trimeric ligands wherein the ligand portions comprise three linker moieties, such as serinol-derived linker moieties or non-serinol linker moieties, each linked to a single targeting ligand moiety.
  • the two or three serinol-derived linker moieties may be linked in series e.g. as shown below:
  • n 1 or 2 and Y is S or O.
  • the ligands are monomeric.
  • the conjugated RNA strands are conjugated to a targeting ligand via a linker moiety including a further linker wherein the further linker is or comprises a saturated, unbranched or branched C 1-5 alkyl chain, wherein optionally one or more carbons (for example 1, 2 or 3 carbons, suitably 1 or 2, in particular 1) is/are replaced by a heteroatom selected from O, N, S(O) p , wherein p is 0, 1 or 2 (for example a CH 2 group is replaced with O, or with NH, or with S, or with SO 2 or a —CHs group at the terminus of the chain or on a branch is replaced with OH or with NH 2 ) wherein said chain is optionally substituted by one or more oxo groups (for example 1 to 3, such as 1 group).
  • oxo groups for example 1 to 3, such as 1 group.
  • the linker moiety is a serinol-derived linker moiety.
  • An O atom of said structure typically links to an RNA strand and the N atom typically links to the targeting ligand.
  • the further linker comprises a saturated, unbranched C 1-15 alkyl chain wherein one or more carbons (for example 1, 2 or 3 carbons, suitably 1 or 2, in particular 1) is/are replaced by an oxygen atom.
  • the further linker comprises a PEG-chain.
  • the further linker comprises a saturated, unbranched C 1-15 alkyl chain.
  • the further linker comprises a saturated, unbranched C 1-6 alkyl chain.
  • the further linker comprises a saturated, unbranched C 4 or C 6 alkyl chain, e.g. a C 4 alkyl chain.
  • the first RNA strand is a compound of formula (X):
  • the second RNA strand is a compound of formula (XI):
  • the first RNA strand is a compound of formula (XV)
  • the second RNA strand is a compound of formula (XVI):
  • the first RNA strand is a compound of formula (XII):
  • the second RNA strand is a compound of formula (XIII):
  • the GalNAc may be substituted for any other targeting ligand, such as those mentioned herein.
  • b is 0, c is 1 and d is 1; b is 1, c is 0 and d is 1; b is 1, c is 1 and d is 0; or b is 1, c is 1 and d is 1.
  • b is 0, c is 1 and d is 1; b is 1, c is 0 and d is 1; or b is 1, c is 1 and d is 1.
  • b is 0, c is 1 and d is 1.
  • Y is O. In another embodiment, Y is S.
  • R 1 is H or methyl. In one embodiment, R 1 is H. In another embodiment, R 1 is methyl.
  • n 0, 1, 2 or 3.
  • n is 0.
  • L is selected from the group consisting of:
  • L is:
  • Example F moieties include (CH 2 ) 1-6 e.g. (CH 2 ) 1-4 e.g. CH 2 , (CH 2 ) 4 , (CH 2 ) 5 or (CH 2 ) 6 , or CH 2 O(CH 2 ) 2-3 , e.g. CH 2 O(CH 2 )CH 3 .
  • L 2 is:
  • L 2 is:
  • L 2 is:
  • L 2 is:
  • n 0 and L 2 is:
  • L 2 is typically the same. Between moieties bracketed by b, c and d, L 2 may be the same or different. In an embodiment, L 2 in the moiety bracketed by c is the same as the L 2 in the moiety bracketed by d. In an embodiment, L 2 in the moiety bracketed by c is not the same as L 2 in the moiety bracketed by d. In an embodiment, the L 2 in the moieties bracketed by b, c and d is the same, for example when the linker moiety is a serinol-derived linker moiety.
  • Serinol derived linker moieties may be based on serinol in any stereochemistry i.e. derived from L-serine isomer, D-serine isomer, a racemic serine or other combination of isomers.
  • the serinol-GalNAc moiety (SerGN) has the following stereochemistry:
  • the targeted cells are hepatocytes.
  • One aspect is a nucleic acid for inhibiting expression of LPA in a cell, comprising at least one duplex region that comprises at least a portion of a first strand and at least a portion of a second strand that is at least partially complementary to the first strand, wherein said first strand is at least partially complementary to at least a portion of a RNA transcribed from the LPA gene, wherein said first strand comprises a nucleotide sequence selected from the following sequences: SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, or 43, wherein the nucleic acid is conjugated to a ligand.
  • the second strand may comprise a nucleotide sequence of SEQ ID NO: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, or 44.
  • the nucleotides of the first and/or second strand may be modified, as herein described.
  • the nucleic acid comprises SEQ ID NO:5 or SEQ ID NO:9 and SEQ ID NO:6 or SEQ ID NO: 10 conjugated to a ligand of formula (I) (as set out above), wherein the ligand is conjugated to the nucleic acid as described and wherein the first strand is modified with a 2′OMe modification on the odd numbered nucleotides, and modified with a 2′F on the even numbered nucleotides, and the second strand is modified with a 2′OMe on the even numbered nucleotides and modified with a 2′F on the odd numbered nucleotides.
  • formula (I) as set out above
  • nucleic acid wherein the first strand comprises, or preferably consists of SEQ ID NO: 165 and the second strand optionally comprises, or preferably consists of SEQ ID NO: 163.
  • This nucleic acid can be further conjugated to a ligand.
  • siRNA that consists of SEQ ID NO: 165 and SEQ ID NO: 164.
  • One aspect of the invention is conjugate 21.
  • the present invention also provides pharmaceutical compositions comprising the nucleic acid or conjugated nucleic acid of the invention.
  • the pharmaceutical compositions may be used as medicaments or as diagnostic agents, alone or in combination with other agents.
  • one or more nucleic acid conjugates of the invention can be combined with a delivery vehicle (e.g., liposomes) and/or excipients, such as carriers, diluents.
  • a delivery vehicle e.g., liposomes
  • excipients such as carriers, diluents.
  • Other agents such as preservatives and stabilizers can also be added.
  • the invention also includes a pharmaceutical composition
  • a pharmaceutical composition comprising one or more nucleic acids or conjugated nucleic acids according to the present invention in a physiologically/pharmaceutically acceptable excipient, such as a stabilizer, preservative, diluent, buffer, and the like.
  • the pharmaceutical composition may be a sterile injectable aqueous suspension or solution, or in a lyophilised form or adhered, absorbed or included to or into any other suitable galenic carrier substance such as pellets, tablets, capsules, nanoparticles, gels, tablets, beads or similar structures.
  • One aspect relates to a double-stranded nucleic acid that is capable of inhibiting expression LPA, preferably in a cell, for use as a medicament.
  • a further aspect of the invention relates to a nucleic acid or conjugated nucleic acid of the invention or the pharmaceutical composition comprising the nucleic acid or conjugated nucleic acid of the invention for use in the treatment of a disease, disorder or syndrome, preferably a disease, disorder or syndrome associated with elevated levels of Lp(a)-containing particles.
  • the treatment may be to prevent and/or reduce the risk to suffer from and/or treat stroke, atherosclerosis, thrombosis or cardiovascular diseases such as coronary heart disease or aortic stenosis and any other disease or pathology associated to elevated levels of Lp(a)-containing particles.
  • the treatment may be to prevent and/or reduce the risk of suffering from and/or treat an atherosclerotic cardiovascular disease, an atherosclerotic cerebrovascular disease, hyperlipidaemia, and dyslipidaemia, preferably wherein the disease is associated with elevated levels of Lp(a)-containing particles.
  • the treatment may be to prevent and/or reduce the risk of suffering from and/or treat calcific aortic stenosis, ischaemic stroke, coronary artery disease, peripheral arterial disease, abdominal aortic aneurysm, heart failure secondary to ischaemic cardiomyopathy, or familial hypercholesterolaemia, preferably wherein the disease is associated with elevated levels of Lp(a)-containing particles.
  • the treatment is to prevent and/or reduce the risk of suffering from and/or treat aortic stenosis, such as calcific aortic stenosis, or familial hypercholesterolaemia, preferably in each case when the disease or disorder is associated with elevated levels of Lp(a)-containing particles.
  • a desirable level of Lp(a)-containing particles in serum is generally described as a level of under 14 mg/dL.
  • An elevated level of Lp(a)-containing particles is a level of at least 14, preferably at least 20, more preferably at least 30, more preferably at least 40 and most preferably at least 50 mg/dL of Lp(a)-containing particles in the serum of a subject.
  • the invention includes a pharmaceutical composition
  • a pharmaceutical composition comprising one or more nucleic acids or conjugated nucleic acids according to the present invention in a physiologically/pharmaceutically acceptable excipient, such as a stabiliser, preservative, diluent, buffer and the like.
  • Lp(a)-containing particles and “Lp(a) particles” are used interchangeably throughout this disclosure.
  • compositions and medicaments of the present invention may be administered to a mammalian subject in a pharmaceutically effective dose.
  • the mammal may be selected from a human, a non-human primate, a simian or prosimian, a dog, a cat, a horse, cattle, a pig, a goat, a sheep, a mouse, a rat, a hamster, a hedgehog and a guinea pig, or other species of relevance.
  • the wording “LPA” or “LPA” as used herein denotes nucleic acid or protein in any of the above-mentioned species, if expressed therein naturally or artificially, but preferably this wording denotes human nucleic acids or proteins.
  • the nucleic acid or conjugated nucleic acid of the present invention can also be administered in combination with other therapeutic compounds, either administrated separately or simultaneously, e.g., as a combined unit dose.
  • the further therapeutic agent can be selected from the group comprising an oligonucleotide, a small molecule, a monoclonal antibody, a polyclonal antibody and a peptide.
  • a molecular conjugation to other biologically active molecular entities such as peptides, cellular or artificial ligands or small and large molecules is also possible.
  • a unit dose may contain between about 0.01 mg/kg and about 100 mg/kg body weight of nucleic acid or conjugated nucleic acid.
  • the dose can be from 10 mg/kg to 25 mg/kg body weight, or 1 mg/kg to 10 mg/kg body weight, or 0.05 mg/kg to 5 mg/kg body weight, or 0.1 mg/kg to 5 mg/kg body weight, or 0.1 mg/kg to 1 mg/kg body weight, or 0.1 mg/kg to 0.5 mg/kg body weight, or 0.5 mg/kg to 1 mg/kg body weight.
  • the dose can be from about 0.5 mg/kg to about 10 mg/kg body weight, or about 0.6 mg/kg to about 8 mg/kg body weight, or about 0.7 mg/kg to about 5 mg/kg body weight, or about 0.8 mg/kg to about 4 mg/kg body weight, or about 0.9 mg/kg to about 3.5 mg/kg body weight, or about 1 mg/kg to about 3 mg/kg body weight, or about 1 mg/kg body weight, or about 3 mg/kg body weight, wherein “about” is a deviation of up to 30%, preferably up to 20%, more preferably up to 10%, yet more preferably up to 5% and most preferably 0% of the indicated value. Dosage levels may also be calculated via other parameters such as, e.g., body surface area.
  • the nucleic acid described herein may be capable of inhibiting the expression of LPA.
  • the nucleic acid described herein may be capable of partially inhibiting the expression of LPA. Inhibition may be complete, i.e. 0% compared of the expression level of LPA in the absence of the nucleic acid of the invention. Inhibition of LPA expression may be partial, i.e. it may be 15%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% or intermediate values of LPA expression in the absence of a nucleic acid of the invention.
  • a nucleic acid or conjugated nucleic acid of the invention, or compositions including the same may be for use in a regimen comprising treatments once or twice weekly, every week, every two weeks, every three weeks, every four weeks, every five weeks, every six weeks, every seven weeks, or every eight weeks, or in regimens with varying dosing frequency such as combinations of the before-mentioned intervals.
  • the nucleic acid may be for use subcutaneously, intravenously or using any other application routes such as oral, rectal or intraperitoneal, preferably for use subcutaneously.
  • the LPA expression may be inhibited compared to untreated cells and/or subjects by a range from 15% up to 100% but at least about 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 100% or intermediate values.
  • the level of inhibition may allow treatment of a disease associated with LPA expression or overexpression, or may serve to further investigate the functions and physiological roles of the LPA gene product.
  • a further aspect of the invention relates to a nucleic acid or conjugated nucleic acid of the invention in the manufacture of a medicament for treating a disease, disorder or syndromes, such as those as listed above or additional pathologies associated with elevated levels of Lp(a), or additional therapeutic approaches where inhibition of LPA expression is desired.
  • nucleic acid composition comprising a nucleic acid or conjugated nucleic acid as described herein, to an individual in need of treatment (to improve such pathologies).
  • the nucleic acid composition may be administered in a regimen comprising treatments twice every week, once every week, every two weeks, every three weeks, every four weeks, every five weeks, every six weeks, every seven weeks, or every eight weeks or at administration intervals of more than once every eight weeks or in regimens with varying dosing frequency such as combinations of the before-mentioned intervals.
  • the nucleic acid or conjugated nucleic acid may be for use subcutaneously or intravenously or other application routes such as oral, rectal or intraperitoneal.
  • the nucleic acid or conjugated nucleic acid is injected subcutaneously.
  • the nucleic acid or conjugated nucleic acid of the present invention can be produced using routine methods in the art including chemical synthesis or expressing the nucleic acid either in vitro (e.g., run off transcription) or in vivo.
  • chemical synthesis or expressing the nucleic acid either in vitro (e.g., run off transcription) or in vivo.
  • solid phase chemical synthesis or using a nucleic acid-based expression vector including viral derivates or partially or completely synthetic expression systems can be used to produce the nucleic acid of the invention in vitro, within an intermediate host organism or cell type, within an intermediate or the final organism or within the desired target cell.
  • Methods for the production (synthesis or enzymatic transcription) of the nucleic acid described herein are known to persons skilled in the art
  • the present invention also relates to the unmodified sequences of all modified sequences disclosed herein.
  • FIG. 1 shows the results of a non-conjugated siRNA molecule screen for inhibition of LPA mRNA expression in human RT-4 cells.
  • FIGS. 2A and 2B show the dose response of non-conjugated LPA-targeting siRNA molecules on LPA mRNA expression in human RT-4 cells.
  • FIG. 3 shows the inhibition of LPA mRNA expression in human and cynomolgus primary hepatocytes by different doses of GalNAc-L1 LPA-1038 conjugated siRNA molecules delivered by receptor-mediated uptake.
  • FIG. 4 shows representative examples of the knockdown of LPA-mRNA by L6-conjugated GalNAc siRNAs indicated in primary human hepatocytes delivered by receptor-mediated uptake.
  • FIG. 5 shows the synthesis of A0268, which is a 3′ mono-GalNAc conjugated single stranded oligonucleotide and is the starting material in the synthesis of Conjugate 1 and Conjugate 3.
  • (ps) denotes phosphorothioate linkage.
  • FIG. 6 shows the synthesis of A0006 which is a 5′ tri-antennary GalNAc conjugated single stranded oligonucleotide used for the synthesis of Reference Conjugate 4.
  • ps denotes phosphorothioate linkage.
  • FIG. 7 illustrates the in vitro determination of TTR knockdown.
  • FIG. 7A shows the in vitro determination of TTR knockdown by Reference Conjugates (RC) 1 and 3 as well as the untreated control “UT”
  • FIG. 7B shows the in vitro determination of TTR knockdown by Reference Conjugates (RC) 2 and 3, as well as the untreated control “UT”
  • FIG. 7C shows the in vitro determination of TTR knockdown by Conjugates 1, 2 and 3, as well as by RC3 and untreated control “UT”.
  • Reference Conjugates 1 and 2 represent comparator conjugates.
  • Reference Conjugate 3 represents a non-targeting GalNAc siRNA and “untreated” (“UT”) represents untreated cells. Both RC3 and UT are negative controls. mRNA levels were normalised against PtenII.
  • FIG. 9 shows oligonucleotide synthesis of 3′ and 5′ GalNAc conjugated oligonucleotides precursors (such as compound X0385B-prec).
  • FIG. 10 shows equal dose response of knock down for LPA targeting siRNA with two single GalNAc units conjugated to the second strand as compared to a triantennary GalNAc unit at the 5′ second strand in primary cynomolgus hepatocytes.
  • FIG. 11 illustrates the in vitro determination of TTR knockdown.
  • FIG. 11A shows the in vitro determination of TTR knockdown by Conjugates 4, 5, 6 and 2 compared to “Luc” (Reference Conjugate 3) as well as the untreated control “UT”
  • FIG. 11B shows the in vitro determination of TTR knockdown by Conjugates 7 and 2, compared to “Luc” (Reference Conjugate 3) as well as the untreated control “UT”.
  • Luc or Reference Conjugate 3 represents a non-targeting GalNAc siRNA and “untreated” (“UT”) represents untreated cells. Both RC3 and UT are negative controls. mRNA level were normalised against PtenII.
  • FIG. 12 illustrates the in vitro determination of TTR knockdown.
  • FIG. 12A shows the in vitro determination of TTR knockdown by Conjugates 8, 9, 10, 11 and 2 compared to “Luc” (Reference Conjugate 3) as well as the untreated control “UT”
  • FIG. 12B shows the in vitro determination of TTR knockdown by Conjugates 12 and 2, compared to “Luc” (Reference Conjugate 3) as well as the untreated control “UT”.
  • Luc or Reference Conjugate 3 represents a non-targeting GalNAc siRNA and “untreated” (“UT”) represents untreated cells. Both RC3 and UT are negative controls. mRNA level were normalised against PtenII.
  • FIG. 13 illustrates the in vitro determination of LPA mRNA knockdown by Conjugate 19 compared to controls.
  • Ctr represents a non-targeting GalNAc siRNA and “untreated” (“UT”) represents untreated cells. Both Ctr and UT are negative controls.
  • mRNA level were normalised against ACTB.
  • FIG. 17 shows serum stability of Conjugates 4, 5, 6, 7 and 2, and untreated control (UT) at 37° C. over 3 days.
  • FIG. 18 shows serum stability of Conjugates 8, 9, 10, 11, 12 and 2, and untreated control (UT) at 37° C. over 3 days.
  • FIG. 19 shows the reduction in LPA mRNA in primary human hepatocytes by Conjugate 21.
  • FIG. 20 shows the reduction in LPA mRNA in primary cynonolgus hepatocytes by Conjugate 21.
  • FIG. 21 shows that Conjugate 21 does not affect the level of APOB gene expression.
  • FIG. 22 shows that Conjugate 21 does not affect the level of PLG gene expression.
  • FIG. 23 shows a time course of serum Lp(a) inhibition over 29 days in cynomolgus with different dosages of conjugate 21.
  • FIG. 24 shows a dose response curve of conjugate 21 showing reduction of serum Lp(a) at day 29 in cynomolgus.
  • modified conjugated siRNA molecules s or functional examples are shown re.
  • GalNAc-LPA-1038-L1 First strand (SEQ ID NO: 119, based on SEQ ID NO 5) OMeA-(ps)-FU-(ps)-OMeA-FA-OMeC-FU-OMeC-FU-OMeG-FU- OMeC-FC-OMeA-FU-OMeU-FA-OMeC-(ps)-FC-(ps)-OMeA 3′
  • Second strand (SEQ ID NO: 120, based on SEQ ID NO SEQ ID NO 6) 5′[ST23 (ps)]3 long trebler (ps)FU-OMeG-FG-OMeU- FA-OMeA-FU-OMeG-FG-OMeA-FC-OMeA-FG-OMeA-FG-OMeU- FU-(ps)-OMeA-(ps)-FU 3′ GalNAc-LPA-1038-L6 First strand (SEQ ID NO: 121, based on
  • GalNAc-LPA-1041-L1 First strand (SEQ ID NO: 123, based on SEQ ID NO 9) 5′ OMeA-(ps)-FU-(ps)-OMeA-FA-OMeC-FU-OMeC-FU-OMeG- FU-OMeC-FC-OMeA-FU-OMeU-FA-OMeC-(ps)-FC-(ps)-OMeG 3′
  • Second strand (SEQ ID NO: 124, based on SEQ ID NO 10) 5′[ST23 (ps)]3 long trebler (ps) FC-OMeG-FG-OMeU- FA-OMeA-FU-OMeG-FG-OMeA-FC-OMeA-FG-OMeA-FG-OMeU- FU-(ps)-OMeA-(ps)-FU 3′ GalNAc-LPA-1041-L6 First strand (SEQ ID NO: 125, based on SEQ
  • oligonucleotides were either obtained from commercial oligonucleotide manufacturers (Biospring, Frankfurt, Germany, or RiboBio, Guangzhou, Guangdong, PRO) or synthesized on an AKTA oligopilot synthesizer (in house) using standard phosphoramidite chemistry.
  • Commercially available solid support and 2′O-Methyl RNA phosphoramidites, 2′Fluoro DNA phosphoramidites (all standard protection) and commercially available long trebler phosphoramidite (Glen research) were used.
  • Synthesis was performed using 0.1 M solutions of the phosphoramidite in dry acetonitrile and benzylthiotetrazole (BTT) was used as activator (0.3M in acetonitrile). All other reagents were commercially available standard reagents.
  • Conjugation of the respective GalNac synthon (e.g., ST23, ST41 or ST43) was achieved by coupling of the respective phosphoramidite to the 5′end of the oligochain under standard phosphoramidite coupling conditions.
  • Phosphorothioates were introduced using standard commercially available thiolation reagents (EDITH, Link technologies).
  • the single strands were cleaved off the CPG by using methylamine (40% aqueous) and the resulting crude oligonucleotide was purified by Ion exchange chromatography (Resource Q, 6 mL, GE Healthcare) on a AKTA Pure HPLC System using a Sodium chloride gradient.
  • equimolar amounts of the respective single strands were dissolved in water and heated to 80° C. for 5 min. After gradual cooling to RT the resulting duplex was lyophilised.
  • siRNAs The sequences of the resulting nucleic acids (siRNAs) are set out in Table 1 below.
  • Liposomal transfection complexes were prepared in triplicate at a ratio of 1.5 ⁇ l RNAiMax (ThermoFisher)/80 pmol of the indicated siRNA molecules. The complex was diluted to the indicated concentrations of 2.5 nM and 25 nM, respectively (values represented pairwise as light and darker grey bars).
  • RT4 human urinary bladder transitional cell papilloma cells expressing endogenously LPA were seeded at a density of 125.000 cells per well in 24-well format on top of previously plated transfection complexes (reverse transfection) at the indicated concentration. 24 hours after transfection total RNA was isolated using the Spin Cell Mini Kit 250 (Stratec).
  • LPA mRNA levels were determined by qRT-PCR relative to PP/B mRNA expression in the respective samples as housekeeping transcript. Values were normalized to the amount of LPA mRNA detected in untreated cells (intraplate). A non-silencing siRNA compound was transfected as an additional control. Means and SD of normalized triplicate values are shown. Results are shown in FIG. 1 .
  • RT4 human urinary bladder transitional cell papilloma cells were reversely transfected as described above (Example 2) and treated at the indicated concentration (range 100 nM to 0.2 nM) with the different non-conjugated siRNA compounds (Table 1) as labeled.
  • 24 h post transfection total RNA was isolated using the Spin Cell Mini Kit 250 (Stratec).
  • LPA mRNA levels were determined by qRT-PCR relative to PP/B mRNA expression in the respective samples as housekeeping transcript. Values were normalized to the amount of LPA mRNA detected in untreated cells. The bars represent the remaining LPA mRNA expression for each data point. Results are shown in FIG. 2 .
  • hepatocytes Primary hepatocytes (ThermoFisher) were plated on collagen-coated 96-well plates at densities of 45,000 cells per well (cynomolgus) and 30,000 cells per well (human). GalNAc-L1-conjugated LPA-1038 was added immediately after plating at the indicated concentrations (nM). 24 hours after siRNA treatment total RNA was isolated using the InviTrap RNA cell HTS 96 well kit (Stratec). LPA mRNA levels were determined by qRT-PCR relative to Actin (cynomolgus) or APOB (human) mRNA levels in the respective samples as housekeeping transcript. Values were normalized to LPA expression in untreated cells. Means and SD of normalized triplicate values of remaining LPA mRNA levels are shown as black bars. Results shown in FIG. 3 .
  • hepatocytes Primary human hepatocytes (ThermoFisher) were plated on collagen-coated 96-well plates at 30,000 cells per well (96 well format). GalNAc-L6-conjugated siRNAs including a non-silencing control were added immediately after cell plating at the two indicated concentrations. 24 hours after siRNA treatment total RNA was isolated using the InviTrap RNA cell HTS 96 well kit (Stratec). LPA mRNA expression levels were determined by qRT-PCR relative to APOB mRNA as housekeeping transcript. Values were normalized to LPA mRNA expression in untreated cells and remaining LPA mRNA levels represented pairwise as bars (100 nM black bars, 20 nM grey bars). Means and SD of normalized triplicate values are shown in FIG. 4 .
  • Murine primary hepatocytes were seeded into collagen pre-coated 96 well plates (Thermo Fisher Scientific, #A1142803) at a cell density of 30,000 cells per well and treated with siRNA-conjugates at concentrations ranging from 10 nM to 0.0001 nM.
  • 24 h post treatment cells were lysed and RNA extracted with InviTrap® RNA Cell HTS 96 Kit/C24 ⁇ 96 preps (Stratec #7061300400) according to the manufactures protocol.
  • Transcripts levels of TTR and housekeeping mRNA (PtenII) were quantified by TaqMan analysis.
  • FIG. 7 also shows the target gene expression levels of Reference Conjugates 1 and 2 which act as comparator conjugates.
  • the conjugates of the invention decrease the target gene expression compared to Reference Conjugates 1 and 2.
  • mice C57BL/6 mice were treated s.c. with 1 mg/kg siRNA-conjugates at day 0. Serum samples were taken at day 7, 14, and 27 by orbital sinus bleeding and stored at ⁇ 20° C. until analysis. Serum TTR quantification was performed with a Mouse Prealbumin ELISA (ALPCO, 41-PALMS/lot 22, 2008003B) according to the manufacturers protocol (sample dilution 1:8000 or 1:800).
  • Conjugate 2 resulted in 3-fold lower target protein level in serum at day seven and 4-fold lower target protein level in serum at day 27 compared to Reference Conjugate 4 at equimolar dose in wild type mice. Furthermore, Conjugate 2 resulted in 85% reduction of target serum protein level at day 27 after single injection, compared to 36% reduction by equimolar amount of Reference Conjugate 4.
  • the siRNAs are modified with alternating 2′-OMe/2′-F and contain each two phosphorothioate (PS) internucleotide linkages at their 5′ and 3′ terminal two internucleotide linkages.
  • PS phosphorothioate
  • conjugate 19 one serinol-GalNAc unit each is attached via a PS-bond to the 5′ and 3′ of the second strand.
  • conjugate 20 the two terminal 5′ internucleotides of the second strand are phosphodiesters and a triantennary GalNAc linker is attached via a PS bond to this end.
  • Dose response of LPA knockdown in primary cynomolgus hepatocytes was assessed 24 h post treatment with 100, 20, 4, 0.8, 0.16, 0.032, and 0.006 nM siRNA.
  • the reference control is construct 2
  • the non-targeting control is named Cte.
  • the transcript ct-value for each treatment group was normalized to the transcript ct value for the house keeping gen ACTB ( ⁇ ct) and to untreated hepatocytes, named ut ( ⁇ ct).
  • siRNAs SEQ ID NO: name batch strand sequence 135 Conjugate X0373 X0373A mA (ps) fU 19 (ps) mA fA mC fU mC fU mG fU mC fC mA fU mU fA mC (ps) fC (ps) mG 136 X0373B Ser(GN) (ps) fC (ps) mG (ps) fG mU fA mA fU mG fG mA fC mA fG mA fG mU fU (ps) mA (ps) fU (ps) Ser(GN) 135 Ref.
  • RNA-Lysis Buffer S (Stratec) was added. Following 15 mi incubation at room temperature plates were storage at ⁇ 80° C. until RNA isolation according to the manufacturers protocol.
  • RNA for each treatment group were mixed with 10 ⁇ l PCR mastermix (TAKYON low Rox) containing 600 nM mTTR-primer, 400 nM ApoB-primer and 200 nM of each probe as well as 0.5 units Euroscript II RT polymerase with 0.2 units RNAse inhibitor.
  • TaqMan analysis was performed in 384-well plate with a 10 min RT step at 48° C., 3 mi initial denaturation at 95° C. and 40 cycles of 95° C. for 10 sec and 60° C. for 1 mi.
  • the primers contain two of BHQ1, FAM and YY, one at each end of the sequence.
  • TMPRSS6 & ApoB MultiPlex TaqMan analysis 10 ⁇ l isolated RNA for each treatment group were mixed with 10 ⁇ l PCR mastermix (TAKYON low Rox) containing 800 nM TMPRSS6 primer, 100 nM ApoB primer and 200 nM of either probe as well as 0.5 units Euroscript II RT polymerase with 0.2 units RNAse inhibitor. TaqMan analysis was performed in 384-well plate with a 10 min RT step at 48° C., 3 min initial denaturation at 95° C. and 40 cycles of 95° C. for 10 sec and 60° C. for 1 mi.
  • siRNA dissolved in PBS was administered sub cutaneous in the scapular region of c57BL/6 mice.
  • Liver samples were snap frozen in liquid nitrogen and stored at ⁇ 80° C. until extraction RNA with InviTrap Spin Tissue RNA Mini Kit (stratec) according to the manufacturers manual. Following, transcript level of Aldh2, Tmprss6 and Pten were quantified as described above.
  • RNAase stability in the endosomal/lysosomal compartment of hepatic cells in vitro siRNA was incubated for 0 h, 4 h, 24 h or 72 h in Sprague Dawley Rat Liver Tritosomes (Tebu-Bio, CatN.: R0610.LT, lot: 1610405, pH: 7.4, 2.827 Units/ml).
  • Tritosomes were mixed 1:10 with low pH buffer (1.5M acetic acid, 1.5M sodium acetate pH 4.75). 30 ⁇ l of this acidified Tritosomes.
  • siRNA (20 ⁇ M) were mixed with and incubated for the indicated times at 37° C.
  • RNA was isolated with the Clarity OTX Starter Kit-Cartriges (Phenomenex CatNo: KSO-8494) according to the manufactures protocol for biological fluids. Lyophilized RNA was reconstituted in 30 ⁇ l H 2 O, mixed with 4 ⁇ loading buffer and 5 ⁇ l were loaded to a 20% TBE-polyacrylamide gel electrophoresis (PAGE) for separation qualitative semi-quantitative analysis. PAGE was run at 120 V for 2 h and RNA visualized by Ethidum-bromide staining with subsequent digital imaging with a Biorad Imaging system.
  • Clarity OTX Starter Kit-Cartriges Phenomenex CatNo: KSO-8494
  • Example compounds were synthesised according to methods described below and methods known to the person skilled in the art. Assembly of the oligonucleotide chain and linker building blocks was performed by solid phase synthesis applying phosphoramidite methodology. GalNAc conjugation was achieved by peptide bond formation of a GalNAc-carboxylic acid building block to the prior assembled and purified oligonucleotide having the necessary number of amino modified linker building blocks attached.
  • Ancillary reagents were purchased from EMP Biotech. Synthesis was performed using a 0.1 M solution of the phosphoramidite in dry acetonitrile and benzyithiotetrazole (BTT) was used as activator (0.3M in acetonitrile). Coupling time was 15 min. A Cap/OX/Cap or Cap/Thio/Cap cycle was applied (Cap: Ac 2 O/NMI/Lutidine/Acetonitrile, Oxidizer: 0.1M I 2 in pyridine/H 2 O). Phosphorothioates were introduced using standard commercially available thiolation reagent (EDITH, Link technologies).
  • DMT cleavage was achieved by treatment with 3% dichloroacetic acid in toluene. Upon completion of the programmed synthesis cycles a diethylamine (DEA) wash was performed. All oligonucleotides were synthesized in DMT-off mode.
  • DEA diethylamine
  • the single strands were cleaved off the CPG by 40% aq. methylamine treatment.
  • the resulting crude oligonucleotide was purified by ion exchange chromatography (Resource Q, 6 mL, GE Healthcare) on a AKTA Pure HPLC System using a sodium chloride gradient. Product containing fractions were pooled, desalted on a size exclusion column (Zetadex, EMP Biotech) and lyophilised.
  • % FLP % full length product
  • 5′1 ⁇ NH2 means refers to the position (5′ end) and number (1 ⁇ NH2) of free serinol derived amino groups which are available for conjugation.
  • 1 ⁇ 3′NH2 on A0264 means there is free amino group which can be reacted with GalNAc synthon 9 at the 3′ end of the strand A0264.
  • 3′5′1 ⁇ NH2 means there is one serinol-derived free amino group which can be reacted with GalNAc linker 9 at the 3′ end and the 5′ end of the strand.
  • 3′5′1 ⁇ NH2 refers to the position (3′ and 5′ end) and number (1 ⁇ NH2 each) of free amino groups which are available for conjugation.
  • 3′5′1 ⁇ NH2 on A0561 means there are 2 free amino group (1 at the 3′ AND 1 at the 5′ end) which can be reacted with GalNAc synthon 9 at the 3′ end of the strand A0561.
  • Conjugation of the GalNac synthon (9) was achieved by coupling to the serinol-amino function of the respective oligonucleotide strand 11 using a peptide coupling reagent. Therefore, the respective amino-modified precursor molecule 11 was dissolved in H 2 O (500 OD/mL) and DMSO (DMSO/H 2 O, 2/1, v/v) was added, followed by DIPEA (2.5% of total volume). In a separate reaction vessel pre-activation of the GalN(Ac4)-C4-acid (9) was performed by reacting 2 eq. (per amino function in the amino-modified precursor oligonucleotide 11) of the carboxylic acid component with 2 eq.
  • Conjugation of the GalNac synthon (9) was achieved by coupling to the amino function of the respective oligonucleotide strand 14 using a peptide coupling reagent. Therefore, the respective amino-modified precursor molecule 14 was dissolved in H 2 O (500 OD/mL) and DMSO (DMSO/H 2 O, 2/1, v/v) was added, followed by DIPEA (2.5% of total volume). In a separate reaction vessel pre-activation of the GalN(Ac4)-C4-acid (9) was performed by reacting 2 eq. (per amino function in the amino-modified precursor oligonucleotide 14) of the carboxylic acid component with 2 eq.
  • Double strand formation was performed according to the methods described above.
  • the double strand purity is given in % double strand which is the percentage of the UV-area under the assigned product signal in the UV-trace of the IP-RP-HPLC analysis (Table 7).
  • f denotes 2′Fluoro 2′deoxyribonucleotide or 2′-fluoro ribonucleotide (the terms are interchangeable)
  • n 2′O Methyl ribonucleotide
  • Ser(GN) is a GalNAc-C4 building block attached to serinol derived linker moiety:
  • O— is the linkage between the oxygen atom and e.g. H, phosphordiester linkage or phosphorothioate linkage.
  • GN is:
  • Antisense strand-STS16001AL33 (SEQ ID NO: 127) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU (ps) Ser(GN) 3′ Sense strand-STS16001BL20 (SEQ ID NO: 128) 5′ Ser(GN) (ps) fA mA fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-STS16001BV1L42 (SEQ ID NO: 130) Ser(GN) (ps) fA (ps) mA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA (ps) Ser(GN)
  • Antisense strand-STS16001AL33 (SEQ ID NO: 127) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU (ps) Ser(GN) 3′ Sense strand-STS16001BV1L42 (SEQ ID NO: 130) 5′ Ser(GN) (ps) fA (ps) mA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA (ps) Ser(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-STS16001BV1L75 (SEQ ID NO: 142) 5′ Ser(GN) fA (ps) mA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA Ser(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-STS16001BV16L42 (SEQ ID NO: 143) 5′ Ser(GN) (ps) fA mA fC mA fG mU fG mU fU mC fU mC fU mA fU mA fU mA fA (ps) Ser(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-5T516001BV20L75 (SEQ ID NO: 144) 5′ Ser(GN)fA mA fC mA fG mU fG mU fU mC fU mU fU mA fU mA fU mA fA Ser(GN) 3′
  • Antisense strand- (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-STS16001BV1L94 (SEQ ID NO: 145) 5′ Ser(GN) (ps) Ser(GN) (ps) fA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA (ps) Ser(GN) (ps) Ser(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU 3′
  • Sense strand-STS16001V1BL96 (SEQ ID NO: 146) 5′ C6Am(GN) (ps) fA (ps) mA (ps) mA (ps) mA (ps) mA (ps) fC mA fG mU fU mC fU mU fU mC fU mA fU (ps) mA (ps) fA (ps) C7Am(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU 3′
  • Sense strand-STS16001V1BL97 (SEQ ID NO: 147) 5′ GlyC3Am(GN) (ps) fA (ps) mA (ps) mA (ps) fC mA fG mU fU mC fU mU fG mC fU mA fU (ps) mA (ps) fA (ps) GlyC3Am(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU 3′ Sense strand (SEQ ID NO: 148) 5′ PipAm(GN) (ps) fA (ps) mA (ps) mA (ps) fC mA fG mU fU mC fU mU fG mC fU mC fU mA fU (ps) mA (ps) fA (ps) PipAm(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU 3′
  • Sense strand-STS16001V1BL88 (SEQ ID NO: 149) 5′ C3Am(GN) (ps) fA (ps) mA (ps) mA (ps) mA (ps) fC mA fG mU fU mC fU mU fG mC fU mA fU (ps) mA (ps) fA (ps) C3Am(GN) 3′
  • Antisense strand-STS16001A (SEQ ID NO: 129) 5′ mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU 3′
  • Sense strand-STS16001V1BL87 (SEQ ID NO: 150) 5′ C6Am(GN) (ps) fA (ps) mA (ps) mA (ps) mA (ps) fC mA fG mU fU mC fU mU fG mC fU mA fU (ps) mA (ps) fA (ps) GlyC3Am(GN) 3′
  • Antisense strand (SEQ ID NO: 151) mU (ps) fC (ps) mU fU mC fU mU fA mA fA mC fU mG fA mG fU mU (ps) fU (ps) mC Sense strand (SEQ ID NO: 152) Ser(GN) (ps) fG (ps) mA (ps) fA mA fC mU fC mA fG mU fU mU fA mA fG mA fA (ps) mG (ps) fA (ps) Ser(GN)
  • Antisense strand (SEQ ID NO: 153) mA (ps) fU (ps) mG fU mA fG mC fC mG fA mG fG mA fU mC fU mU (ps) fC (ps) mU Sense strand (SEQ ID NO: 154) Ser(GN) (ps) fA (ps) mG (ps) fA mA fG mA fU mC fC mG fG mC fU mA fC (ps) mA (ps) fU (ps) Ser(GN)
  • Antisense strand (SEQ ID NO: 155) mA (ps) fA (ps) mC fC mA fG mA fA mG fA mA fG mC fA mG fG mU (ps) fG (ps) mA Sense strand (SEQ ID NO: 156) Ser(GN) (ps) fU (ps) mC (ps) fA mC fC mU fG mC fU mU fC mU fU mC fU mG fG (ps) mU (ps) mU (ps) fU (ps) Ser(GN)
  • Antisense strand (SEQ ID NO: 135) mA (ps) fU (ps) mA fA mC fU mC fU mG fU mC fC mA fU mU fA mC (ps) fC (ps) mG Sense strand (SEQ ID NO: 136) Ser(GN) (ps) fC (ps) mG (ps) fG mU fA mA fU mG fG mA fC mA fG mA fG mU fU (ps) mA (ps) fU (ps) Ser(GN)
  • Antisense strand - STS16001A (SEQ ID NO: 129) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand - STS16001BL20 (SEQ ID NO: 128) Ser(GN) (ps) fA mA fC mA fG mU fG mU fU mC fU mU fU mC fU mA fU (ps) mA (ps) fA
  • Antisense strand-STS16001AL33 (SEQ ID NO: 127) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU (ps) Ser(GN) Sense strand-STS16001BV1 (SEQ ID NO: 157) fA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mU fU mC fU mA fU (ps) mA (ps) fA
  • Antisense strand-STS18001A (A0130, SEQ ID NO: 132) mU (ps) fC (ps) mG fA mA fG mU fA mU fU mC fC mG fC mG fU mA(ps)fC(ps)mG Sense strand-STS18001BL4 (A0131, SEQ ID NO: 133) [(ST23) (ps)] 3 C4XLT (ps) fC mG fU mA fC mG fC mG fG mA fA mU fA mC fU mU fC (ps) mG (ps) fA
  • Antisense strand-STS16001AL33 (SEQ ID NO: 127) mU (ps) fU (ps) mA fU mA fG mA fG mC fA mA fG mA fA mC fU mG (ps) fU (ps) mU Sense strand-STS16001BL4 (SEQ ID NO: 134) 5′[(ST23) (ps) 3 C4XLT(ps) fA (ps) mA (ps) mA (ps) mA (ps) fC mA fG mU fG mU fU mC fU mC fU mA fU (ps) mA (ps) fA
  • Antisense strand (SEQ ID NO: 138) mC (ps) fU (ps) mU fA mC fU mC fU mC fG mC fC mC fA mA fG mC (ps) fG (ps) mA Sense strand (SEQ ID NO: 139) [(ST23) (ps)]3 (C6XLT) (ps) fU mC fG mC fU mU fG mG fG mC fG mA fG mA fG mU fA (ps) mA (ps) fG
  • Antisense strand (SEQ ID NO: 151) mU (ps) fC (ps) mU fU mC fU mU fA mA fA mC fU mG fA mG fU mU (ps) fU (ps) mC Sense strand (SEQ ID NO: 158) [ST23 (ps)]3 Itrb (ps) fG mA fA mA fC mU fC mA fG mU fU mU fA mA fG mA fA (ps) mG (ps) fA
  • Antisense strand (SEQ ID NO: 160) mU (ps) fA (ps) mC fC mA fG mA fA mG fA mA fG mC fA mG fG mU (ps) fG (ps) mA Sense strand (SEQ ID NO: 161) [ST23 (ps)]3 ST41 (ps)fU mC fA mC fC mU fG mC fU mU fC mU fU mC fU mG fG (ps) mU (ps) fA
  • Antisense strand (SEQ ID NO: 135) mA (ps) fU (ps) mA fA mC fU mC fU mG fU mC fC mA fU mU fA mC (ps) fC (ps) mG Sense strand (SEQ ID NO: 162) [ST23 (ps)]3 C6XLT (ps) fC mG fG mU fA mA fU mG fG mA fC mA fG mA fG mU fU (ps) mA (ps) fU
  • the in vitro data show that in the context of one or two serinol-derived linker moieties being provided at 5′ and 3′ ends of the sense strand in Conjugates 4-7, the number of phosphorothioate (PS) bonds between the terminal nucleotide and the linker, and/or between the terminal three nucleotides in the sense strand, can be varied whilst maintaining efficacy for decreasing target gene expression.
  • PS phosphorothioate
  • Conjugates 8, 9, 10 and 11 appear to be comparable to or better than Conjugate 2 which was previously shown to be the most effective conjugate at 0.01 nM.
  • Conjugate 19 was also shown to decrease target gene expression compared to the negative controls (see “UT” column and Ctr which is a non-targeting siRNA and also referred to as Reference Conjugate 5), as shown in FIG. 13 . This indicates that the first strand is binding to the target gene, thus lowering gene expression.
  • Conjugates 8-12 and 19 show that a number of linkers which are structurally diverse and which are conjugated at both termini of the sense strand are effective at decreasing target gene expression. Conjugates 8-12 and 19 decrease target gene expression more effectively than “Luc” which is Reference Conjugate 3 (for Conjugates 8-12), “Ctr” which is Reference Conjugate 5 (for Conjugate 19) and untreated control.
  • the conjugates of the invention are particularly effective at reducing target gene expression compared to the negative control (PBS) and Reference Conjugates 6 and 7 respectively.
  • the in vivo data show that a variety of example linkers which are conjugated at both termini of the second strand are effective at decreasing target gene expression in vivo.
  • the positioning of the linker improves in vivo potency conjugates, as compared to a triantennary GalNAc-linker control at the 5′ terminus of the second strand (Reference Conjugates 6, 7 and 8).
  • FIG. 17 shows the results from the serum stability studies in respect of Conjugates 2, 4, 5, 6 and 7.
  • FIG. 18 shows the serum stability of Conjugates 2, 8, 9, 10, 11 and 12.
  • All tested conjugates contain each one GalNAc linker unit at the 5′ end and another at the 3′ end of the second strand.
  • the siRNAs are modified with alternating 2′-OMe/2′-F and contain each two phosphorothioate (PS) internucleotide linkages at their 5′ and 3′ terminal two internucleotide linkages, unless stated differently.
  • PS phosphorothioate
  • Conjugate 4 the serinol-GalNAc units are attached via a phosphodiester bond.
  • Conjugate 5 the serinol-GalNAc units are conjugated via PS, whereas all internucleotide linkage in the second strand are phosphodiesters.
  • Conjugate 6 the second strand contains no PS.
  • Conjugate 7 two serinol-GalNAc units are attached to each second strand terminus and to each other via a PS-bonds at the respective ends.
  • Conjugate 8 a C6-amino-modifier at 5′ and a C7-amino-modifier at the 3′ end of the second strand were applied for ligand attachment.
  • Conjugate 9 Gly-C3-amino-modifiers, in Conjugate 10 piperidyl-amino-modifiers, in Conjugate 11 C3-amino-modifiers and in Conjugate 2 serinol-GalNAc units were used as linkers for conjugation to both ends of the second strand.
  • Conjugate 2 both terminal internucleotides as well as the nucleotide-serinol bonds are PS.
  • Conjugate 12 a C6-amino-modifier at the 5′ and a GlyC3-amino-modifier at the 3′ end of second strand were applied for ligand attachment.
  • “ut” indicates an untreated sample which the other samples were normalised to.
  • “Luc” indicates an siRNA targeting Luciferase (Reference Conjugate 3), which was used as non-targeting control and does not reduce target mRNA levels.
  • the data show that in context of a serinol-derived linker moiety being provided at 5′ and 3′ ends of the sense strand, the number of phosphorothioate (PS) bonds between the terminal nucleotide and the linker, and/or between the terminal three nucleotides in the sense strand, can be varied whilst maintaining stability in serum.
  • PS phosphorothioate
  • Human primary hepatocytes were seeded into collagen 1-coated 96-well plates (Life Technologies) at a density of 30,000 cells per well. Cynomolgus hepatocytes were seeded at a density of 45,000 cells per well.
  • Conjugate 21 and a control GalNAc-conjugated siRNA were serially diluted 5-fold at a concentration range of 0.006-100 nM and added immediately after plating in plating media. Plates were then incubated at 37° C. in a 5% CO 2 atmosphere for 24 hours. Subsequently, cells were lysed and RNA was isolated using the method described below.
  • Antisense strand-Conjugate 21 (SEQ ID NO: 165) 5′ mA (ps) fU (ps) mA fA mC fU mC fU mG fU mC fC mA fU mU fA mC (ps) fC (ps) mG 3′
  • Sense strand-STS16001BL20 (SEQ ID NO: 164) 5′ [ST23 (ps)]3 C6XLT (ps) mC mG mG mU mA mA fU fG mA mC mA mG mA mG mU mU (ps) mA (ps) mU 3′
  • RNA-solution Ten ⁇ l of RNA-solution was used for gene expression analysis by reverse transcription quantitative polymerase chain reaction (RT-qPCR) performed with amplicon sets/sequences for LPA, ACTB (Eurogentec Deutschland GmbH, Cologne, Germany), APOB and PLG (BioTez GmbH, Berlin, Germany).
  • RT-qPCR reverse transcription quantitative polymerase chain reaction
  • RT-qPCR reactions were carried out with an ABI StepOne Plus (Applied Biosystems, part of Thermo Fisher Scientific, Massachusetts, USA) using standard protocols for RT-PCR (48° C. 30 min, 95° C. 10 mi, 40 cycles at 95° C. 15 s followed by 60° C. 1 min).
  • LPA and PLG analyses were performed in singleplex assays (primers: 300 nM, probe: 100 nM).
  • APOB 200 nM
  • ACTB 300 nM
  • APOB 200 nM
  • ACTB 300 nM
  • LPA mRNA expression levels were normalised to ACTB and relative to cells treated with a non-targeting siRNA control measured by RT-qPCR. Data is presented as the mean ⁇ SD of a single experiment.
  • LPA mRNA expression levels were normalised to ACTB and relative to cells treated with a non-targeting siRNA control measured by RT-qPCR. Data is presented as the mean ⁇ SD of a single experiment.
  • FIG. 21 shows Knockdown of APOB mRNA by Conjugate 21 through receptor-mediated uptake into primary human hepatocytes 24 hours post siRNA treatment.
  • APOB mRNA expression levels were normalised to ACTB and relative to cells treated with a non-targeting siRNA control measured by RT-qPCR. Data is presented as the mean ⁇ SD of a single experiment.
  • FIG. 22 shows Knockdown of PLG mRNA by Conjugate 21 through receptor-mediated uptake into primary human hepatocytes 24 hours post siRNA treatment.
  • PLG mRNA expression levels were normalised to ACTB and relative to cells treated with a non-targeting siRNA control measured by RT-qPCR. Data is presented as the mean ⁇ SD of a single experiment.
  • Conjugate 21 achieves high levels of LPA mRNA inhibition without affecting the levels of APOB and PLG mRNAs.
  • conjugate 21 was formulated in 0.9% saline and each animal received a single dose of conjugate 21 at doses of 0.1, 0.3, 1.0 and 3.0 mg/kg.
  • a single sequence may have more than one name. In those cases, one of those names is given in the summary sequence table.
  • RNA sequence such as (ps) and [ST23 (ps)]3 ST41 (ps) etc, these are optional parts of the sequence, but are a preferred embodiment of that sequence.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cardiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
US17/293,303 2018-11-13 2019-11-13 Nucleic acids for inhibiting expression of lpa in a cell Pending US20220002722A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
PCT/EP2018/081106 WO2019092283A1 (en) 2017-11-13 2018-11-13 Nucleic acids for inhibiting expression of lpa in a cell
EPPCT/EP2018/081106 2018-11-13
EP19174466 2019-05-14
EP19174466.3 2019-05-14
PCT/EP2019/081158 WO2020099476A1 (en) 2018-11-13 2019-11-13 Nucleic acids for inhibiting expression of lpa in a cell

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/081158 A-371-Of-International WO2020099476A1 (en) 2018-11-13 2019-11-13 Nucleic acids for inhibiting expression of lpa in a cell

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/559,479 Continuation US11499153B2 (en) 2018-11-13 2021-12-22 Nucleic acids for inhibiting expression of LPA in a cell

Publications (1)

Publication Number Publication Date
US20220002722A1 true US20220002722A1 (en) 2022-01-06

Family

ID=66554180

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/293,303 Pending US20220002722A1 (en) 2018-11-13 2019-11-13 Nucleic acids for inhibiting expression of lpa in a cell
US17/559,479 Active US11499153B2 (en) 2018-11-13 2021-12-22 Nucleic acids for inhibiting expression of LPA in a cell

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/559,479 Active US11499153B2 (en) 2018-11-13 2021-12-22 Nucleic acids for inhibiting expression of LPA in a cell

Country Status (29)

Country Link
US (2) US20220002722A1 (ru)
EP (2) EP4219712A3 (ru)
JP (1) JP7245328B2 (ru)
KR (1) KR102553386B1 (ru)
CN (1) CN113227372A (ru)
AU (1) AU2019380628A1 (ru)
BR (1) BR112021009213A2 (ru)
CA (1) CA3119239A1 (ru)
CL (1) CL2021001225A1 (ru)
CO (1) CO2021007372A2 (ru)
CR (1) CR20210301A (ru)
DK (1) DK3880818T3 (ru)
EA (1) EA202190895A1 (ru)
ES (1) ES2932295T3 (ru)
FI (1) FI3880818T3 (ru)
HR (1) HRP20221358T1 (ru)
HU (1) HUE060703T2 (ru)
IL (1) IL283159A (ru)
LT (1) LT3880818T (ru)
MA (1) MA58208B1 (ru)
MX (1) MX2021005590A (ru)
PE (1) PE20211282A1 (ru)
PH (1) PH12021550801A1 (ru)
PL (1) PL3880818T3 (ru)
PT (1) PT3880818T (ru)
RS (1) RS63778B1 (ru)
SG (1) SG11202103882QA (ru)
SI (1) SI3880818T1 (ru)
WO (1) WO2020099476A1 (ru)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110520531A (zh) * 2017-04-05 2019-11-29 赛伦斯治疗有限责任公司 产品和组合物
EP4219716A3 (en) 2017-11-13 2024-03-27 Silence Therapeutics GmbH Nucleic acids for inhibiting expression of lpa in a cell
DK3880818T3 (da) 2018-11-13 2022-12-05 Silence Therapeutics Gmbh Nukleinsyrer til inhibering af ekspression af LPA i en celle
JP2023545502A (ja) 2020-10-16 2023-10-30 サノフイ リポタンパク質(a)を阻害するためのrna組成物および方法
WO2023041508A2 (en) * 2021-09-14 2023-03-23 Argonaute RNA Limited Treatment of cardiovascular disease
WO2023152194A1 (en) 2022-02-09 2023-08-17 Silence Therapeutics Gmbh Therapeutic inhibition of lpa expression
CN114703184A (zh) * 2022-03-11 2022-07-05 厦门甘宝利生物医药有限公司 Lpa抑制剂及其用途
WO2024013334A1 (en) * 2022-07-15 2024-01-18 Silence Therapeutics Gmbh Nucleic acids for inhibiting expression of agt in a cell
CN115851723B (zh) * 2022-10-24 2023-10-03 厦门甘宝利生物医药有限公司 一种抑制lpa基因表达的rna抑制剂及其应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170096665A1 (en) * 2015-10-01 2017-04-06 Arrowhead Pharmaceuticals, Inc. Compositions and Methods for Inhibiting Gene Expression of LPA

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL9201440A (nl) 1992-08-11 1994-03-01 Univ Leiden Triantennaire clusterglycosiden, hun bereiding en toepassing.
US7259150B2 (en) * 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US20060160759A1 (en) 2002-09-28 2006-07-20 Jianzhu Chen Influenza therapeutic
CN101278059A (zh) * 2005-07-28 2008-10-01 肿瘤疗法科学股份有限公司 诊断和治疗肾细胞癌的方法
US20110110886A1 (en) 2008-06-13 2011-05-12 Yale University Small molecule inhibitors of autotaxin and methods of use
WO2016149020A1 (en) * 2015-03-17 2016-09-22 Arrowhead Research Corporation Rna interference agents
EP3228326A1 (en) * 2016-04-05 2017-10-11 Silence Therapeutics GmbH Nucleic acid linked to a trivalent glycoconjugate
CN110520531A (zh) 2017-04-05 2019-11-29 赛伦斯治疗有限责任公司 产品和组合物
EP3735461A1 (en) * 2017-11-13 2020-11-11 Silence Therapeutics GmbH Nucleic acids for inhibiting expression of aldh2 in a cell
EP4219716A3 (en) * 2017-11-13 2024-03-27 Silence Therapeutics GmbH Nucleic acids for inhibiting expression of lpa in a cell
DK3880818T3 (da) 2018-11-13 2022-12-05 Silence Therapeutics Gmbh Nukleinsyrer til inhibering af ekspression af LPA i en celle

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170096665A1 (en) * 2015-10-01 2017-04-06 Arrowhead Pharmaceuticals, Inc. Compositions and Methods for Inhibiting Gene Expression of LPA

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Watts, Jonathan K., Glen F. Deleavey, and Masad J. Damha. "Chemically modified siRNA: tools and applications." Drug discovery today 13.19-20 (2008): 842-855. (Year: 2008) *

Also Published As

Publication number Publication date
KR102553386B1 (ko) 2023-07-07
PT3880818T (pt) 2022-12-06
CR20210301A (es) 2021-09-16
CN113227372A (zh) 2021-08-06
WO2020099476A1 (en) 2020-05-22
DK3880818T3 (da) 2022-12-05
EP3880818A1 (en) 2021-09-22
EA202190895A1 (ru) 2021-08-10
CO2021007372A2 (es) 2021-07-09
SG11202103882QA (en) 2021-05-28
ES2932295T3 (es) 2023-01-17
PL3880818T3 (pl) 2023-01-23
HRP20221358T1 (hr) 2022-12-23
MX2021005590A (es) 2023-01-24
BR112021009213A2 (pt) 2021-08-10
MA58208B1 (fr) 2022-12-30
KR20210095878A (ko) 2021-08-03
FI3880818T3 (fi) 2022-12-15
US20220170016A1 (en) 2022-06-02
EP4219712A2 (en) 2023-08-02
LT3880818T (lt) 2022-12-27
SI3880818T1 (sl) 2023-01-31
CL2021001225A1 (es) 2021-12-24
US11499153B2 (en) 2022-11-15
IL283159A (en) 2021-06-30
AU2019380628A1 (en) 2021-05-20
EP3880818B1 (en) 2022-09-28
JP2022507282A (ja) 2022-01-18
PH12021550801A1 (en) 2021-10-04
RS63778B1 (sr) 2022-12-30
CA3119239A1 (en) 2020-05-22
JP7245328B2 (ja) 2023-03-23
HUE060703T2 (hu) 2023-04-28
EP4219712A3 (en) 2023-11-22
PE20211282A1 (es) 2021-07-19

Similar Documents

Publication Publication Date Title
US20220290144A1 (en) Nucleic acids for inhibiting expression of lpa in a cell
US11499153B2 (en) Nucleic acids for inhibiting expression of LPA in a cell
US11560563B2 (en) SiRNAs with vinylphosphonate at the 5′ end of the antisense strand
US11987794B2 (en) Products and compositions
US20210155926A1 (en) siRNAs WITH AT LEAST TWO LIGANDS AT DIFFERENT ENDS
EP3735461A1 (en) Nucleic acids for inhibiting expression of aldh2 in a cell
US20230272385A1 (en) siRNAs WITH VINYLPHOSPHONATE AT THE 5' END OF THE ANTISENSE STRAND
EP3483271A2 (en) Products and compositions
AU2018247923B2 (en) Products and compositions
EP3483272A1 (en) Products and compositions
EA045986B1 (ru) Нуклеиновые кислоты для ингибирования экспрессии lpa в клетке

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

AS Assignment

Owner name: SILENCE THERAPEUTICS GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:RIDER, DAVID ANTHONY;BETHGE, LUCAS;FRAUENDORF, CHRISTIAN;AND OTHERS;SIGNING DATES FROM 20200304 TO 20200312;REEL/FRAME:057144/0200

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER