WO2016170348A2 - Sarna compositions and methods of use - Google Patents

Sarna compositions and methods of use Download PDF

Info

Publication number
WO2016170348A2
WO2016170348A2 PCT/GB2016/051116 GB2016051116W WO2016170348A2 WO 2016170348 A2 WO2016170348 A2 WO 2016170348A2 GB 2016051116 W GB2016051116 W GB 2016051116W WO 2016170348 A2 WO2016170348 A2 WO 2016170348A2
Authority
WO
WIPO (PCT)
Prior art keywords
sarna
syndrome
lipid
disease
strand
Prior art date
Application number
PCT/GB2016/051116
Other languages
French (fr)
Other versions
WO2016170348A8 (en
WO2016170348A3 (en
Inventor
Pål SÆTROM
Endre Bakken STOVNER
Original Assignee
Mina Therapeutics Limited
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
Application filed by Mina Therapeutics Limited filed Critical Mina Therapeutics Limited
Priority to JP2017555233A priority Critical patent/JP2018512876A/en
Priority to US15/568,046 priority patent/US20180305689A1/en
Priority to EP16735927.2A priority patent/EP3286318A2/en
Publication of WO2016170348A2 publication Critical patent/WO2016170348A2/en
Publication of WO2016170348A3 publication Critical patent/WO2016170348A3/en
Publication of WO2016170348A8 publication Critical patent/WO2016170348A8/en
Priority to US17/319,744 priority patent/US20210363525A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • 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
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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

Definitions

  • the invention relates to oligonucleotides, specifically saRNA, compositions for modulating target gene expression and to the methods of using the compositions in diagnostic and therapeutic applications.
  • short RNAs can regulate transcription by destructing transcripts that are sense or antisense to a given niRNA and which are presumed to be non-coding transcripts. Destruction of the non-coding transcripts which are sense, or identical to, the given mRNA results in transcriptional repression of that mRNA, whereas destruction of the non-coding transcripts which are antisense to the given mRNA results in transcriptional activation and/or increased expression of the mRNA or the level of protein encoded by the mRNA. By targeting such non-coding transcripts, short RNAs can therefore be used to up- regulate specific genes at either the nucleic acid or protein.
  • RNAs have also been discovered to increase gene expression by targeting ncRNAs that overlap gene promoters (Janowski et al., Nature Chemical Biology, vol.3: 166-173 (2007), the contents of which are incorporated herein by reference in their entirety).
  • RNA which leads to up-regulation of the expression of a target gene by any mechanism is termed a short activating RNA or small activating RNA (saRNA).
  • saRNA small activating RNA
  • US Patent 8,288,354 to Wahlestedt discloses a method of modulating expression of a target gene comprising targeting a nucleic acid molecule to a naturally-occurring anti-sense transcript (NAT) of a sense strand of the targeted gene in a target cell, wherein the nucleic acid molecule targeting the NAT is complementary to the NAT.
  • the NAT may be a coding RNA transcript or a non-coding RNA transcript lacking any extensive open reading frame.
  • WO 2012/065143 to Krieg et al. teaches a method of activating expression of a target gene comprising blocking the binding of a long non-coding RNA (lnc-RNA) to Polycomb repressive complex 2 (PRC2) protein by a single-stranded oligonucleotide, thereby preventing the lnc-RNA from suppressing the target gene.
  • lnc-RNA long non-coding RNA
  • PRC2 Polycomb repressive complex 2
  • compositions and methods for the targeted modulation of genes via activation with saRNA which do not require the a priori identification of a NAT or rely on interactions at the polycomb complex for prophylactic diagnostic and/or therapeutic purposes.
  • FIG. 1 is a schematic illustrating the relationships among the nucleic acid moieties involved in the function of an saRNA of the invention.
  • compositions, methods and kits for the design, preparation, manufacture, formulation and/or use of short (or small) activating RNA (saRNA) that modulates target gene expression and/or function for therapeutic purposes, including diagnosing and prognosis.
  • saRNA short activating RNA
  • saRNA may refer to a single saRNA or saRNA in the plural (saRNAs).
  • One aspect of the invention provides an saRNA that targets an antisense RNA transcript of a target gene.
  • the antisense RNA transcript of the target gene is referred to thereafter as the target antisense RNA transcript.
  • the target antisense RNA transcript is transcribed from the coding strand of the target gene.
  • Another aspect of the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an saRNA that targets an antisense RNA transcript of a target gene and at least one pharmaceutically acceptable excipient, wherein the expression of the target gene is up-regulated.
  • Another aspect of the invention provides a method of modulating the expression of a target gene comprising administering an saRNA that targets an antisense RNA transcript of the target gene
  • Another aspect of the invention provides treating or preventing a disease comprising administering an saRNA that targets an antisense RNA transcript of a target gene, wherein the target gene is associated with the disease.
  • compositions, methods and kits for modulating target gene expression and/or function for therapeutic purposes comprise at least one saRNA that upregulates the expression of a target gene.
  • One aspect of the present invention provides a method to design and synthesize saRNA.
  • small activating RNA means a single-stranded or double-stranded RNA that upregulates or has a positive effect on the expression of a specific gene.
  • the saRNA may be single-stranded of 14 to 30 nucleotides.
  • the saRNA may also be double-stranded, each strand comprising 14 to 30 nucleotides.
  • the gene is called the target gene of the saRNA.
  • the target gene is a double-stranded DNA comprising a coding strand and a template strand.
  • an saRNA that upregulates the expression of the A3GALT2 gene is called an "A3 GALT2 -saRNA" and the A3GALT2 gene is the target gene of the A3GALT2 -saRNA.
  • a target gene may be any gene of interest.
  • a target gene has a promoter region on the template strand.
  • upregulation or “activation” of a gene is meant an increase in the level of expression of a gene, or levels of the polypeptide(s) encoded by a gene or the activity thereof, or levels of the RNA transcript(s) transcribed from the template strand of a gene above that observed in the absence of the saRNA of the present invention.
  • the saRNA of the present invention may have a direct upregulating effect on the expression of the target gene.
  • the saRNAs of the present invention may have an indirect upregulating effect on the RNA transcript(s) transcribed from the template strand of the target gene and/or the polypeptide(s) encoded by the target gene or mRNA.
  • the RNA transcript transcribed from the target gene is referred to thereafter as the target transcript.
  • the target transcript may be an mRNA of the target gene.
  • the target transcript may exist in the mitochondria.
  • the saRNAs of the present invention may have a downstream effect on a biological process or activity. In such embodiments, an saRNA targeting a first transcript may have an effect (either upregulating or downregulating) on a second, non-target transcript.
  • the saRNA of the present invention may show efficacy in proliferating cells.
  • proliferating means cells which are growing and/or reproducing rapidly.
  • Target antisense RNA transcript of a target gene Target antisense RNA transcript of a target gene
  • the saRNAs of the present invention is designed to be complementary to a target antisense RNA transcript of a target gene, and it may exert its effect on the target gene expression and/or function by down-regulating the target antisense RNA transcript.
  • the target antisense RNA transcript is transcribed from the coding strand of the target gene and may exist in the nucleus of a cell.
  • antisense when used to describe a target antisense RNA transcript in the context of the present invention means that the sequence is complementary to a sequence on the coding strand of a gene. [0025] It is to be understood that thymidine of the DNA is replaced by uridine in RNA and that this difference does not alter the understanding of the terms "antisense” or
  • the target antisense RNA transcript may be transcribed from a locus on the coding strand between up to 100, 80, 60, 40, 20 or 10 kb upstream of a location corresponding to the target gene's transcription start site (TSS) and up to 100, 80, 60, 40, 20 or 10 kb downstream of a location corresponding to the target gene's transcription stop site.
  • TSS target gene's transcription start site
  • the target antisense RNA transcript is transcribed from a locus on the coding strand located within +/- 1 kb of the target gene's transcription start site.
  • the target antisense RNA transcript is transcribed from a locus on the coding strand located within +/- 500 nt, +/- 250 nt, +/- 100 nt, +/- 10 nt, +/- 5 nt or +/- 1 nt of the target gene's transcription start site.
  • the target antisense RNA transcript is transcribed from a locus on the coding strand located +/- 2000 nucleotides of the target gene's transcription start site.
  • the locus on the coding strand is no more than 1000 nucleotides upstream or downstream from a location corresponding to the target gene's transcription start site.
  • the locus on the coding strand is no more than 500 nucleotides upstream or downstream from a location corresponding to the target gene's transcription start site.
  • transcription start site means a nucleotide on the template strand of a gene corresponding to or marking the location of the start of
  • the TSS may be located within the promoter region on the template strand of the gene.
  • transcription stop site means a region, which can be one or more nucleotides, on the template strand of a gene, which has at least one feature such as, but not limited to, a region which encodes at least one stop codon of the target transcript, a region encoding a sequence preceding the 3 'UTR of the target transcript, a region where the RNA polymerase releases the gene, a region encoding a splice site or an area before a splice site and a region on the template strand where transcription of the target transcript terminates.
  • the phrase "is transcribed from a particular locus" in the context of the target antisense RNA transcript of the invention means the transcription of the target antisense RNA transcript starts at the particular locus.
  • the target antisense RNA transcript is complementary to the coding strand of the genomic sequence of the target gene, and any reference herein to "genomic sequence” is shorthand for "coding strand of the genomic sequence”.
  • the "coding strand" of a gene has the same base sequence as the mRNA produced, except T is replayed by U in the mRNA.
  • the "template strand” of a gene is therefore complementary and antiparallel to the mRNA produced.
  • the target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence located between 100, 80, 60, 40, 20 or 10 kb upstream of the target gene's transcription start site and 100, 80, 60, 40, 20 or 10 kb downstream of the target gene's transcription stop site.
  • the target antisense RNA transcript comprises a sequence which is complementary to a genomic sequence located between 1 kb upstream of the target gene's transcription start site and 1 kb downstream of the target gene's transcription stop site.
  • the target antisense RNA transcript comprises a sequence which is complementary to a genomic sequence located between 500, 250, 100, 10, 5 or 1 nucleotide upstream of the target gene's transcription start site and ending 500, 250, 100, 10, 5 or 1 nucleotide downstream of the target gene's transcription stop site.
  • the target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence which includes the coding region of the target gene.
  • the target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence that aligns with the target gene's promoter region on the template strand.
  • Genes may possess a plurality of promoter regions, in which case the target antisense RNA transcript may align with one, two or more of the promoter regions.
  • An online database of annotated gene loci may be used to identify the promoter regions of genes.
  • the terms 'align' and 'alignment' when used in the context of a pair of nucleotide sequences mean the pair of nucleotide sequences are complementary to each other or have sequence identity with each other.
  • the region of alignment between the target antisense RNA transcript and the promoter region of the target gene may be partial and may be as short as a single nucleotide in length, although it may be at least 15 or at least 20 nucleotides in length, or at least 25 nucleotides in length, or at least 30, 35, 40, 45 or 50 nucleotides in length, or at least 55, 60, 65,
  • target antisense RNA transcript and the target gene's promoter region are identical in length and they align (i.e. they align over their entire lengths).
  • the target antisense RNA transcript is shorter than the target gene's promoter region and aligns over its entire length with the target gene's promoter region (i.e. it aligns over its entire length to a sequence within the target gene's promoter region).
  • the target antisense RNA transcript is longer than the target gene's promoter region and the target gene's promoter region is aligned fully by it (i.e. the target gene's promoter region is aligns over its entire length to a sequence within the target antisense RNA transcript).
  • the target antisense RNA transcript and the target gene's promoter region are of the same or different lengths and the region of alignment is shorter than both the length of the target antisense RNA transcript and the length of the target gene's promoter region.
  • the target antisense RNA transcript is at least 1 kb, or at least 2, 3, 4, 5, 6, 7, 8, 9 or 10, e.g., 20, 25, 30, 35 or 40 kb long.
  • the target antisense RNA transcript comprises a sequence which is at least 75%, or at least 85%, or at least 90%, or at least 95% complementary along its full length to a sequence on the coding strand of the target gene.
  • the present invention provides saRNAs targeting the target antisense RNA transcript and may effectively and specifically down-regulate such target antisense RNA transcripts. This can be achieved by saRNA having a high degree of complementarity to a region within the target antisense RNA transcript.
  • the saRNA will have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the region within the target antisense RNA transcript to be targeted.
  • the target antisense RNA transcript has sequence identity with a region of the template strand of the target gene
  • the target antisense RNA transcript will be in part identical to a region within the template strand of the target gene allowing reference to be made either to the template strand of the gene or to a target antisense RNA transcript.
  • the location at which the saRNA hybridizes or binds to the target antisense RNA transcript (and hence the same location on the template strand) is referred to as the "targeted sequence" or "target site”.
  • the antisense strand of the saRNA may be at least 80%, 90%, 95%, 98%, 99% or 100% identical with the reverse complement of the targeted sequence.
  • the reverse complement of the antisense strand of the saRNA has a high degree of sequence identity with the targeted sequence.
  • the targeted sequence may have the same length, i.e., the same number of nucleotides, as the saRNA and/or the reverse complement of the saRNA,
  • the targeted sequence comprises at least 14 and less than
  • the targeted sequence has 19, 20, 21 , 22, or 23 nucleotides.
  • the location of the targeted sequence is situated within a promoter area of the template strand.
  • the targeted sequence is located within a TSS
  • TSS core or “TSS core sequence” as used herein, refers to a region between 2000 nucleotides upstream and 2000 nucleotides downstream of the TSS (transcription start site). Therefore, the TSS core comprises 4001 nucleotides and the TSS is located at position 2001 from the 5' end of the TSS core sequence.
  • the targeted sequence is located between 1000 nucleotides upstream and 1000 nucleotides downstream of the TSS.
  • the targeted sequence is located between 500 nucleotides upstream and 500 nucleotides downstream of the TSS.
  • the targeted sequence is located between 250 nucleotides upstream and 250 nucleotides downstream of the TSS. [0059] In some embodiments, the targeted sequence is located between 100 nucleotides upstream and 100 nucleotides downstream of the TSS.
  • the targeted sequence is located between 10 nucleotides upstream and 10 nucleotides downstream of the TSS.
  • the targeted sequence is located between 5 nucleotides upstream and 5 nucleotides downstream of the TSS.
  • the targeted sequence is located between 1 nucleotide upstream and 1 nucleotide downstream of the TSS.
  • the targeted sequence is located upstream of the TSS in the TSS core.
  • the targeted sequence may be less than 2000, less than 1000, less than 500, less than 250, less than 100, less than 10 or less than 5 nucleotides upstream of the TSS.
  • the targeted sequence is located downstream of the TSS in the TSS core.
  • the targeted sequence may be less than 2000, less than 1000, less than 500, less than 250, less than 100, less than 10 or less than 5 nucleotides downstream of the TSS,
  • the targeted sequence is located +/- 50 nucleotides surrounding the TSS of the TSS core. In some embodiments, the targeted sequence substantially overlaps the TSS of the TSS core. In some embodiments, the targeted sequence overlaps begins or ends at the TSS of the TSS core. In some embodiments, the targeted sequence overlaps the TSS ofthe TSS core by 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18 or 19 nucleotides in either the upstream or downstream direction.
  • the location of the targeted sequence on the template strand is defined by the location of the 5 ' end of the targeted sequence.
  • the 5 ' end of the targeted sequence may be at any position of the TSS core and the targeted sequence may start at any position selected from position 1 to position 4001 of the TSS core.
  • the targeted sequence when the 5' most end of the targeted sequence from position 1 to position 2000 of the TSS core, the targeted sequence is considered upstream of the TSS and when the 5 ' most end of the targeted sequence is from position 2002 to 4001 , the targeted sequence is considered downstream of the TSS.
  • the targeted sequence is considered to be a TSS centric sequence and is neither upstream nor downstream of the TSS.
  • the targeted sequence when the 5' end of the targeted sequence is at position 1600 of the TSS core, i.e., it is the 1600 th nucleotide of the TSS core, the targeted sequence starts at position 1600 of the TSS core and is considered to be upstream of the TSS,
  • the TSS core is a sequence for the target gene as described in Table 1 and 2.
  • the TSS core is a sequence such as, but not limited to, SEQ
  • the saRNA of the present invention may have two strands that form a duplex, one strand being a guide strand.
  • the saRNA duplex is also called a double- stranded saRNA.
  • a double-stranded saRNA or saRNA duplex, as used herein, is an saRNA that includes more than one, and preferably, two, strands in which interstrand hybridization can form a region of duplex structure.
  • the two strands of a double-stranded saRNA are referred to as an antisense strand or a guide strand, and a sense strand or a passenger strand.
  • the antisense strand of an saRNA duplex used interchangeably with antisense strand saRNA or antisense saRNA, has a high degree of complementarity to a region within the target antisense RNA transcript.
  • the antisense strand may have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the region within the target antisense RNA transcript or targeted sequence. Therefore, the antisense strand has a high degree of complementary to the targeted sequence on the template strand.
  • the sense strand of the saRNA duplex used interchangeably with sense strand saRNA or sense saRNA, has a high degree of sequence identity with the targeted sequence on the template strand.
  • the targeted sequence is located within the promoter area of the template strand. In some embodiments, the targeted sequence is located within the TSS core of the template stand.
  • the location of the antisense strand and/or sense strand of the saRNA duplex, relative to the targeted sequence is defined by making reference to the TSS core sequence. For example, when the targeted sequence is downstream of the TSS, the antisense saRNA and the sense saRNA start downstream of the TSS. In another example, when the targeted sequence starts at position 200 of the TSS core, the antisense saRNA and the sense saRNA start upstream of the TSS.
  • FIG. 1 The relationships among the saRNAs, a target gene, a coding strand of the target gene, a template strand of the target gene, a target antisense RNA transcript, a target transcript, a targeted sequence/target site, and the TSS are shown in FIG. 1 ,
  • a "strand” in the context of the present invention means a contiguous sequence of nucleotides, including non-natu rally occurring or modified nucleotides. Two or more strands may be, or each form a part of, separate molecules, or they may be connected covalently, e.g., by a linker such as a polyethyleneglyeol linker. At least one strand of an saRNA may comprise a region that is complementary to a target antisense RNA. Such a strand is called an antisense or guide strand of the saRNA duplex. A second strand of an saRNA that comprises a region complementary to the antisense strand of the saRNA is called a sense or passenger strand.
  • An saRNA duplex may also be formed from a single molecule that is at least partly self-complementary forming a hairpin structure, including a duplex region.
  • the term "strand" refers to one of the regions of the saRNA that is complementary to another internal region of the saRNA.
  • the guide strand of the saRNA will have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the sequence within the target antisense RNA transcript.
  • the passenger strand of an saRNA may comprise at least one nucleotide that is not complementary to the corresponding nucleotide on the guide strand, called a mismatch with the guide strand.
  • the mismatch with the guide strand may encourage preferential loading of the guide strand (Wu et al, PLoS ONE, vol.6 (12):e28580 (201 1), the contents of which are incorporated herein by reference in their entirety).
  • the at least one mismatch with the guide strand may be at 3 ' end of the passenger strand.
  • the 3 ' end of the passenger strand may comprise 1-5 mismatches with the guide strand.
  • the 3 ' end of the passenger strand may comprise 2-3 mismatches with the guide strand.
  • the 3 ' end of the passenger strand may comprise 6-10 mismatches with the guide strand.
  • an saRNA duplex may show efficacy in proliferating cells.
  • An saRNA duplex may have siRNA-like complementarity to a region of a target antisense RNA transcript; that is, 100% complementarity between nucleotides 2-6 from the 5' end of the guide strand in the saRNA duplex and a region of the target antisense RNA transcript.
  • Other nucleotides of the saRNA may, in addition, have at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to a region of the target antisense RNA transcript.
  • nucleotides 7 counted from the 5' end until the 3' end of the saRNA may have least 80%, 90%, 95%, 98%o, 99% or 100% complementarity to a region of the target antisense RNA transcript.
  • small interfering RNA or "siRNA” in the context mean a double- stranded RNA typically 20-25 nucleotides long involved in the RNA interference (RNAi) pathway and interfering with or inhibiting the expression of a specific gene.
  • the gene is the target gene of the siRNA.
  • siRNA that interferes the expression of A3GALT2 gene is called “A3GALT2-siRNA” and the A3GALT2 gene is the target gene.
  • An siRNA is usually about 21 nucleotides long, with 3' overhangs (e.g., 2 nucleotides) at each end of the two strands.
  • siRNA inhibits target gene expression by binding to and promoting the cleavage of one or more RNA transcripts of the target gene at specific sequences.
  • RNAi the RNA transcripts are in RNA, so cleavage of mRNA results in the down-regulation of gene expression.
  • saRNA of the present invention may modulate the target gene expression by cleavage of the target antisense RNA transcript.
  • a double-stranded saRNA may include one or more single-stranded nucleotide overhangs.
  • the term "overhang” or “tail” in the context of double-stranded saRNA and siRNA refers to at least one unpaired nucleotide that protrudes from the duplex structure of saRNA or siRNA. For example, when a 3 '-end of one strand of an saRNA extends beyond the 5 '-end of the other strand, or vice versa, there is a nucleotide overhang.
  • An saRNA may comprise an overhang of at least one nucleotide; alternatively the overhang may comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more.
  • a nucleotide overhang may comprise of consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside.
  • the overhang(s) may be on the sense strand, the antisense strand or any combination thereof.
  • the nucleotide(s) of an overhang can be present on the 5 ' end, 3 ' end or both ends of either an antisense or sense strand of an saRNA.
  • oligonucleotides are designed to form, upon hybridization, one or more single-stranded overhangs, and such overhangs shall not be regarded as mismatches with regard to the determination of complementarity.
  • an saRNA comprising one oligonucleotide 19 nucleotides in length and another oligonucleotide 21 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 19 nucleotides that is fully complementary to the shorter oligonucleotide, can yet be referred to as "fully complementary" for the purposes described herein.
  • the antisense strand of a double-stranded saRNA has a 1-10 nucleotide overhang at the 3' end and/or the 5' end. In one embodiment, the antisense strand of a double-stranded saRNA has 1-4 nucleotide overhang at its 3' end, or 1-2 nucleotide overhang at its 3' end. In one embodiment, the sense strand of a double-stranded saRNA has a 1 -10 nucleotide overhang at the 3' end and/or the 5' end.
  • the sense strand of a double-stranded saRNA has 1-4 nucleotide overhang at its 3' end, or 1-2 nucleotide overhang at its 3' end. In one embodiment, both the sense strand and the antisense strand of a double- stranded saRNA have 3' overhangs.
  • the 3' overhangs may comprise one or more uracils, e.g., the sequences UU or UUU.
  • one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate, wherein the internucleoside linkage is
  • the overhang comprises one or more deoxyribonucleoside, e.g., the sequence dTdT or dTdTdT. In one embodiments, the overhang comprises the sequence dT*dT, wherein * is a thiophosphate internucleoside linkage.
  • the saRNA of the present invention may alternatively be defined by reference to the target gene.
  • the target antisense RNA transcript is complementary to a genomic region on the coding strand of the target gene, and the saRNA of the present invention is in turn complementary to a region of the target antisense RNA transcript, so the saRNA of the present invention may be defined as having sequence identity to a region on the coding strand of the target gene.
  • the saRNA of the present invention may have a high percent identity, e.g. at least 80%, 90%, 95%, 98% or 99%, or 100% identity, to a genomic sequence on the target gene.
  • the genomic sequence may be up to 2000, 1000, 500, 250, or 100 nucleotides upstream or downstream of the target gene's transcription start site. It may align with the target gene's promoter region.
  • the saRNA may have sequence identity to a sequence that aligns with the promoter region of the target gene,
  • the existence of the target antisense RNA transcript does not need to be determined to design the saRNA of the present invention.
  • the design of the saRNA does not require the identification of the target antisense RNA transcript.
  • the nucleotide sequence of the TSS core i.e., the sequence in the region 2000 nucleotides upstream of the target gene's transcription start site to 2000 nucleotides downstream of the target gene's transcription start may be obtained by the genomic sequence of the coding strand of the target gene, by sequencing or by searching in a database.
  • Targeted sequence within the TSS core starting at any position from position 1 to position 4001 of the TSS core on the template strand can be selected and can then be used to design saRNA sequences.
  • the saRNA has a high degree of sequence identity with the reverse complement of the targeted sequence.
  • Off-target hit number refers to the number of other sites in the whole genome that are identical to the saRNA's targeted sequence on the template strand of the target gene.
  • the term “Omm hit number” refers to the number of known protein coding transcript other than the target transcript of the saRNA, the complement of which the saRNA may hybridize with or bind to with 0 mismatch. In another word, "Omm hit number” counts the number of known protein coding transcript, other than the target transcript of the saRNA that comprises a region completely identical with the saRNA sequence.
  • 1mm hit number refers to the number of known protein coding transcript other than the target transcript of the saRNA, the complement of which the saRNA may hybridize with or bind to with 1 mismatch.
  • “lmm hit number” counts the number of known protein coding transcript, other than the target transcript of the saRNA that comprises a region identical with the saRNA sequence with only 1 mismatch.
  • only saRNA sequences that have no off-target hit, no Omm hit and no 1mm hit are selected. For those saRNA sequences disclosed in the present application, each has no off- target hit, no Omm hit and no 1mm hit,
  • Determination of existence means either searching databases of ESTs and/or antisense UNA transcripts around the locus of the target gene to identify a suitable target antisense RNA transcript, or using RT PCR or any other known technique to confirm the physical presence of a target antisense RNA transcript in a cell.
  • the saRNA of the present invention may be single or, double-stranded. Double-stranded molecules comprise a first strand and a second strand. If double-stranded, each strand of the duplex may be at least 14, or at least 18, e.g. 19, 20, 21 or 22 nucleotides in length. The duplex may be hybridized over a length of at least 12, or at least 15, or at least 17, or at least 19 nucleotides. Each strand may be exactly 19 nucleotides in length. Preferably, the length of the saRNA is less than 30 nucleotides since oligonucleotide duplex exceeding this length may have an increased risk of inducing the interferon response. In one embodiment, the length of the saRNA is 19 to 25 nucleotides. The strands forming the saRNA duplex may be of equal or unequal lengths.
  • the saRNAs of the present invention comprise a sequence of at least 14 nucleotides and less than 30 nucleotides which has at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to the targeted sequence.
  • the sequence which has at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to the targeted sequence is at least 15, 16, 17, 18 or 19 nucleotides in length, or 18-22 or 19 to 21 , or exactly 19.
  • the saRNA of the present invention may include a short 3' or 5' sequence which is not complementary to the target antisense RNA transcript. In one embodiment, such a sequence is at 3 ' end of the strand.
  • the sequence may be 1 -5 nucleotides in length, or 2 or 3.
  • the sequence may comprises uracil, so it may be a 3' stretch of 2 or 3 uracils.
  • the sequence may comprise one or more deoxyribonucleoside, such as dT. In one embodiment, one or more of the nucleotides in the sequence is replaced with a nucleoside thiophosphate, wherein the
  • internucleoside linkage is thiophosphate.
  • the sequence comprises the sequence dT*dT, wherein * is a thiophosphate internucleoside linkage.
  • This non-complementary sequence may be referred to as "tail". If a 3' tail is present, the strand may be longer, e.g., 19 nucleotides plus a 3' tail, which may be UU or UUU. Such a 3 ' tail shall not be regarded as mismatches with regard to determine complementarity between the saRNA and the target antisense RNA transcript.
  • the saRNA of the present invention may consist of (i) a sequence having at least 80% complementarity to a region of the target antisense RNA transcript; and (ii) a 3' tail of 1 -5 nucleotides, which may comprise or consist of uracil residues.
  • the saRNA will thus typically have complementarity to a region of the target antisense RNA transcript over its whole length, except for the 3' tail, if present.
  • Any of the saRNA sequences disclosed in the present application may optionally include such a 3' tail.
  • any of the saRNA sequences disclosed in the saRNA Tables and Sequence Listing may optionally include such a 3' tail.
  • the saRNA of the present invention may further comprise Dicer or Drosha substrate sequences.
  • the saRNA of the present invention may contain a flanking sequence.
  • the flanking sequence may be inserted in the 3 ' end or 5 ' end of the saRNA of the present invention.
  • the flanking sequence is the sequence of a miRNA, rendering the saRNA to have miRNA configuration and may be processed with Drosha and Dicer.
  • the saRNA of the present invention has two strands and is cloned into a microRNA precursor, e.g., miR-30 backbone flanking sequence.
  • the saRNA of the present invention may comprise a restriction enzyme substrate or recognition sequence.
  • the restriction enzyme recognition sequence may be at the 3 ' end or 5 5 end of the saRNA of the present invention.
  • restriction enzymes include Notl and Ascl.
  • the saRNA of the present invention consists of two strands stably base-paired together.
  • the passenger strand may comprise at least one nucleotide that is not complementary to the corresponding nucleotide on the guide strand, called a mismatch with the guide strand.
  • the at least one mismatch with the guide strand may be at 3 ' end of the passenger strand.
  • the 3 ' end of the passenger strand may comprise 1-5 mismatches with the guide strand.
  • the 3 ' end of the passenger strand may comprise 2-3 mismatches with the guide strand.
  • the 3 ' end of the passenger strand may comprise 6-10 mismatches with the guide strand.
  • the double-stranded saRNA may comprise a number of unpaired nucleotides at the 3' end of each strand forming 3' overhangs.
  • the number of unpaired nucleotides forming the 3' overhang of each strand may be in the range of 1 to 5 nucleotides, or I to 3 nucleotides, or 2 nucleotides.
  • the 3' overhang may be formed on the 3' tail mentioned above, so the 3' tail may be the 3' overhang of a double-stranded saRNA,
  • the saRNA of the present invention may be single-stranded and consists of
  • saRNA of the present invention may have complementarity to a region of the target antisense RNA transcript over its whole length, except for the 3' tail, if present.
  • the saRNA of the present invention may also be defined as having "identity" to the coding strand of the target gene.
  • the saRNA of the present invention may be double-stranded and consists of a first strand comprising (i) a first sequence having at least 80% complementarity to a region of the target antisense RNA transcript and (ii) a 3' overhang of 1 -5 nucleotides; and a second strand comprising (i) a second sequence that forms a duplex with the first sequence and (ii) a 3' overhang of 1-5 nucleotides.
  • the genomic sequence of the target gene may be used to design saRNA of the target gene.
  • the sequence of a target antisense RNA transcript may be determined from the sequence of the target gene for designing saRNA of the target gene.
  • One aspect of the present invention provides an saRNA that modulates the expression of a target gene. Also provided is an saRNA that modulates the level of a target transcript.
  • the target transcript is a coding transcript, e.g., mRNA.
  • Another aspect of the present invention provides an saRNA that modulates the level of a protein encoded by the coding target transcript.
  • the expression of target gene is increased by at least 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80% in the presence of the saRNA of the present invention compared to the expression of target gene in the absence of the saRNA of the present invention.
  • the expression of target gene is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, in the presence of the saRNA of the present invention compared to the expression of target gene in the absence of the saRNA of the present invention.
  • the modulation of the expression of target gene may be reflected or determined by the change of mRNA levels encoding the target gene.
  • Non-limiting examples of target genes which can be modulated by the saRNA of the present invention may be coding genes, including the target genes described in Table 1 such
  • Table 2 below describes non-coding target genes and their TSS locations.
  • the saRNAs of the present invention may be single-stranded and comprise 14-
  • sequence of a single-stranded saRNA may have at least 60%, 70%>, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs; 4048-315235, 318727- 584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208- 2585259.
  • the single-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532- 1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259.
  • the saRNA is a single-stranded saRNA which comprises an antisense sequence such as, but not limited to any of the antisense sequences described in the sequence listing referenced at the beginning of this application.
  • the saRNA is a single-stranded saRNA which comprises an antisense sequence such as, but not limited to any of the sense sequences described in the sequence listing referenced at the beginning of this application.
  • the single stranded saRNAs of the present invention may be modified or unmodified.
  • the single-stranded saRNA may have a 3' tail.
  • the saRNAs may be double-stranded.
  • the two strands form a duplex, also known as an saRNA duplex, and each strand comprises 14-30 nucleotides.
  • the first strand of a double-stranded saRNA may have at least 60%, 70%, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062- 913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259.
  • the first strand of the double-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259.
  • the second strand of a double- stranded saRNA may have at least 60%, 70%o, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259.
  • the second strand of the double-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931 , 1564373- 1879188, and 1889208-2585259.
  • the double-stranded saRNA may have a 3' overhang on each strand.
  • the saRNA of the present invention is an saRNA duplex.
  • the saRNA duplex may be a pair of sense and antisense sequences such as, but not limited to, any of the sense sequence and corresponding antisense sequences described in the sequence listing referenced at the beginning of this application.
  • the saRNA of the present invention may be the pair of the sense sequence and antisense sequence described in the sequence listing referenced at the beginning of this application,
  • the double-stranded saRNA of the present invention may be modified or unmodified.
  • Bifunction or dual-functional oligonucleotides e.g., saRNA may be designed to up-regulate the expression of a first gene and down-regulate the expression of at least one second gene.
  • One strand of the dual-functional oligonucleotide activates the expression of the first gene and the other strand inhibits the expression of the second gene.
  • Each strand might further comprise a Dicer substrate sequence.
  • modification refers to structural and/or chemical modifications with respect to A, G, U or C ribonucleotides.
  • Nucleotides in the saRNAs of the present invention may comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynueleotides.
  • the saRNA of the present invention may include any useful modification, such as to the sugar, the nucleobase, or the intemucleoside linkage (e.g. to a linking phosphate / to a phosphodiester linkage / to the phosphodiester backbone).
  • One or more atoms of a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro).
  • modifications e.g., one or more modifications are present in each of the sugar and the intemucleoside linkage.
  • Modifications according to the present invention may be modifications of ribonucleic acids (RNAs) to deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof
  • RNAs ribonucleic acids
  • DNAs deoxyribonucleic acids
  • TAAs threose nucleic acids
  • GNAs glycol nucleic acids
  • PNAs peptide nucleic acids
  • LNAs locked nucleic acids
  • the saRNAs of the present invention may comprise at least one modification described herein,
  • the saRNA is an saRNA duplex and the sense strand and antisense sequence may independently comprise at least one modification.
  • the sense sequence may comprises a modification and the antisense strand may be unmodified.
  • the antisense sequence may comprises a modification and the sense strand may be unmodified.
  • the sense sequence may comprises more than one modification and the antisense strand may comprise one modification.
  • the antisense sequence may comprises more than one modification and the sense strand may comprise one modification.
  • the saRNA of the present invention can include a combination of modifications to the sugar, the nucleobase, and/or the intemueleoside linkage. These combinations can include any one or more modifications described herein or in International Application Publication
  • WO2013/052523 filed October 3, 2012, in particular Formulas (Ia)-(Ia-5), (Ib)-(If), (Ila)-(IIp), (IIb-1 ), (IIb-2), (IIc-l )-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the contents of which are incorporated herein by reference in their entirety.
  • the saRNA of the present invention may or may not be uniformly modified along the entire length of the molecule.
  • one or more or all types of nucleotide e.g., purine or pyrimidine, or any one or more or all of A, G, U, C
  • nucleotides X in an saRNA of the invention are modified, wherein X may be any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
  • nucleotide modifications may exist at various positions in an saRNA.
  • nucleotide analogs or other modification(s) may be located at any position(s) of an saRNA such that the function of saRNA is not substantially decreased.
  • the saRNA of the present invention may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e.
  • any one or more of A, G, U or C) or any intervening percentage e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 90% to 100%, and from 95% to 100%).
  • any intervening percentage e.g.,
  • the saRNA of the present invention may be modified to be a spherical nucleic acid (SNA) or a circular nucleic acid.
  • the terminals of the saRNA of the present invention may be linked by chemical reagents or enzymes, producing spherical saRNA that has no free ends.
  • Spherical saRNA is expected to be more stable than its linear counterpart and to be resistant to digestion with RNase R exonuclease.
  • Spherical saRNA may former comprise other structural and/or chemical modifications with respect to A, G, U or C
  • the saRNA of the present invention may comprise inverted deoxy abasic modifications on the passenger strand.
  • the at least one inverted deoxy abasic modification may be on 5' end, or 3' end, or both ends of the passenger strand.
  • the inverted deoxy basic modification may encourage preferential loading of the guide strand,
  • the saRNA of the present invention may be modified with any modifications of an oligonucleotide or polynucleotide disclosed in pages 136 to 247 of PCT Publication
  • Conjugation may result in increased stability and/or half-life and may be particularly useful in targeting the saRNA of the present invention to specific sites in the cell, tissue or organism.
  • the saRNA of the present invention can be designed to be conjugated to other polynucleotides, 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 (e.g.
  • alkylating agents phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEGJi, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g.
  • biotin e.g., aspirin, vitamin E, folic acid
  • transport/absorption facilitators e.g., aspirin, vitamin E, folic acid
  • synthetic ribonucleases proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell, hormones and hormone receptors, non-peptidic species, such as lipids, lectins,
  • nucleic acid molecules carbohydrates, vitamins, cofactors, or a drag.
  • Suitable conjugates for nucleic acid molecules are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
  • saRNA of the present invention may be administered with, or further include one or more of RNAi agents, small interfering RNAs (siRNAs), small hairpin RNAs (shRNAs), long non-coding RNAs (IncRNAs), enhancer RNAs, enhancer-derived RNAs or enhancer-driven RNAs (eRNAs), mieroRNAs (miRNAs), miRNA binding sites, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, ap tamers or vectors, and the like to achieve different functions.
  • RNAi agents small interfering RNAs
  • shRNAs small hairpin RNAs
  • IncRNAs long non-coding RNAs
  • eRNAs enhancer RNAs
  • eRNAs enhancer-derived RNAs or enhancer-driven RNAs
  • miRNAs mieroRNAs
  • miRNA binding sites antisense RNAs, ribozymes
  • RNAi agents small interfering RNAs (siRNAs), small hairpin RNAs (shRNAs), long non- coding RNAs (IncRNA), mieroRNAs (miRNAs), miRNA binding sites, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers or vectors may comprise at least one modification or substitution.
  • the modification is selected from a chemical substitution of the nucleic acid at a sugar position, a chemical substitution at a phosphate position and a chemical substitution at a base position.
  • the chemical modification is selected from incorporation of a modified nucleotide; 3' capping; conjugation to a high molecular weight, non-immunogenic compound; conjugation to a lipophilic compound; and incorporation of phosphorothioate into the phosphate backbone.
  • the high molecular weight, non-immunogenic compound is polyalkylene glycol, or polyethylene glycol (PEG).
  • saRNA comprising at least one modification may show efficacy in proliferating cells.
  • saRNA of the present invention may be attached to a transgene so it can be co-expressed from an RNA polymerase II promoter.
  • saRNA of the present invention is attached to green fluorescent protein gene (GFP).
  • saRNA of the present invention may be attached to a DNA or RNA aptamer, thereby producing saRNA-aptamer conjugate
  • Aptamers are oligonucleotides or peptides with high selectivity, affinity and stability. They assume specific and stable three- dimensional shapes, thereby providing highly specific, tight binding to target molecules.
  • An aptamer may be a nucleic acid species that has been engineered through repeated rounds ⁇ vitro selection or equivalently, SELEX (systematic evolution of ligands by exponential enrichment) to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms.
  • Nucleic acid aptamers have specific binding affinity to molecules through interactions other than classic Watson-Crick base pairing. Nucleic acid aptamers, like peptides generated by phage display or monoclonal antibodies (mAhs), are capable of specifically binding to selected targets and, through binding, block their targets' ability to function. In some cases, aptamers may also be peptide aptamers. For any specific molecular target, nucleic acid aptamers can be identified from combinatorial libraries of nucleic acids, e.g. by SELEX. Peptide aptamers may be identified using a yeast two hybrid system. A skilled person is therefore able to design suitable aptamers for delivering the saRNAs or cells of the present invention to target cells such as liver cells. DNA aptamers, RNA aptamers and peptide aptamers are contemplated. Administration of saRNA of the present invention to the liver using liver-specific aptamers is preferred.
  • nucleic acid aptamer As used herein, a typical nucleic acid aptamer is approximately 10-15 kDa in size (20-45 nucleotides), binds its target with at least nanomolar affinity, and discriminates against closely related targets.
  • Nucleic acid aptamers may be ribonucleic acid, deoxyribonucleic acid, or mixed ribonucleic acid and deoxyribonucleic acid. Aptamers may be single-stranded ribonucleic acid, deoxyribonucleic acid or mixed ribonucleic acid and deoxyribonucleic acid. Aptamers may comprise at least one chemical modification.
  • a suitable nucleotide length for an aptamer ranges from about 15 to about 100 nucleotides (nt), and in various other embodiments, 15-30 nt, 20-25 nt, 30-100 nt, 30-60 nt, 25- 70 nt, 25-60 nt, 40-60 nt, 25-40 nt, 30-40 nt, any of 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39 or 40 nt or 40-70 nt in length.
  • the sequence can be designed with sufficient flexibility such that it can accommodate interactions of a tamers with two targets at the distances described herein. Aptamers may be further modified to provide protection from nuclease and other enzymatic activities.
  • the aptamcr sequence can be modified by any suitable methods known in the art.
  • the saRNA-aptamer conjugate may be formed using any known method for linking two moieties, such as direct chemical bond formation, linkage via a linker such as streptavidin and so on.
  • saRNA of the present invention may be attached to an antibody.
  • Methods of generating antibodies against a target cell surface receptor are well known.
  • the saRNAs of the invention may be attached to such antibodies with known methods, for example using RNA carrier proteins.
  • the resulting complex may then be administered to a subject and taken up by the target cells via receptor-mediated endocytosis.
  • saRNA of the present invention may be conjugated with lipid moieties such as a cholesterol moiety (Letsinger et al, Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), eholie acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4: 1053-1060), a thioether, e.g., beryl-5-tritylthiol (Manoharan et al, Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manoharan et al, Biorg. Med. Chem.
  • lipid moieties such as a cholesterol moiety (Letsinger et al, Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), eholie acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4: 1053-1060), a
  • Acids Res., 1990, 18:3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al, Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manohara et ah, Tetrahedron Lett., 1995, 36:3651 -3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229- 237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et ah, J. Pharmacol. Exp. Ther., 1996, 277:923-937), the content of each of which is herein incorporated by reference in its entirety.
  • the saRNA of the present invention is conjugated with a ligand.
  • the ligand may be any ligand disclosed in US
  • the conjugate has a formula of Ligand-[linker] 0 ptionai-[tether] op tionai-oligonucleotide agent.
  • the oligonucleotide agent may comprise a subunit having formulae (I) disclosed by US 20130184328 to Manoharan et al., the contents of which are incorporated herein by reference in their entirety.
  • the ligand may be any ligand disclosed in US 20130317081 to Akinc et al., the contents of which are incorporated herein by reference in their entirety, such as a lipid, a protein, a hormone, or a carbohydrate ligand of Formula 1 1 -X XV I .
  • the ligand may be coupled with the saRNA with a bivalent or traivalent branched linker in Formula XXXI-XXXV disclosed in Akinc.
  • the saRNA of the present invention may be provided in combination with other active ingredients known to have an effect in the particular method being considered.
  • the other active ingredients may be administered simultaneously, separately, or sequentially with the saRNA of the present invention.
  • saRNA of the present invention is administered with saRNA modulating a different target gene.
  • Non-limiting examples include saRNA that modulates albumin, insulin or FTNF4A genes. Modulating any gene may be achieved using a single saRNA or a combination of two or more different saRNAs.
  • Non-limiting examples of saRNA that can be administered with saRNA of the present invention include saRNA modulating albumin or BNF4A disclosed in International Publication WO 2012/175958 filed June 20, 2012, saRNA modulating insulin disclosed in International Publications WO 2012/046084 and WO 2012/046085 both filed Oct. 10, 2011, saRNA modulating human progesterone receptor, human major vault protein (IrMVP), E-cadherin gene, p53 gene, or PTEN gene disclosed in US Pat. No. 7,709,456 filed November 13, 2006 and US Pat. Publication US 2010/0273863 filed April 23, 2010, saRNAs targeting p21 gene disclosed in International Publication WO 2006/113246 filed April 1 1, 2006, any nucleic acid disclosed in
  • WO2012/065143 filed November 12, 2011 that upregulates the expression of genes in Table 8 of WO 2012/065143 or increases the expression of a tumor suppressor, any oligonucleotide that activates target genes in Table 4 of W ( ) 2013 / 173 35 filed May 16, 2013, any oligonucleotide that activates target genes in Table 4 of WO2013/173637 filed May 16, 2013, any
  • the saRNA is conjugated with a carbohydrate ligand, such as any carbohydrate ligand disclosed in US Pat No. 8106022 and 8828956 to Manoharan et al. (Alnylam Pharmaceuticals), the contents of which are incorporated herein by reference in their entirety.
  • the carbohydrate ligand may be monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide.
  • carbohydrate- conjugated RNA agents may target the parenchymal cells of the liver.
  • the saRNA is conjugated with more than one carbohydrate ligand, preferably two or three.
  • the saRNA is conjugated with one or more galactose moiety. In another embodiment, the saRNA is conjugated at least one (e.g., two or three or more) lactose molecules (lactose is a glucose coupled to a galactose). In another embodiment, the saRNA is conjugated with at least one (e.g., two or three or more) N-Acetyl-Galaetosamine (GalNAc), N-Ac- Glucosamine (GluNAc), or mannose (e.g., mannose-6-phosphate). In one embodiment, the saRNA is conjugated with at least one mannose ligand, and the conjugated saRNA targets macrophages.
  • lactose molecules lactose is a glucose coupled to a galactose
  • the saRNA is conjugated with at least one (e.g., two or three or more) N-Acetyl-Galaetosamine (GalNA
  • saRNA of the present invention is administered with a small interfering RNA or siRNA that inhibits the expression of a gene.
  • saRNA of the present invention is administered with one or more drugs for therapeutic purposes.
  • compositions comprising a small activating RNA (saRNA) that upregulates a target gene, and at least one pharmaceutically acceptable carrier.
  • saRNA small activating RNA
  • compositions may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable excipient includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired.
  • excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21 st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD,
  • any conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other eomponent(s) of the pharmaceutical composition.
  • compositions are administered to humans, human patients or subjects.
  • active ingredient generally refers to saRNA to be delivered as described herein.
  • compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the
  • compositions include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
  • the efficacy of the formulated saRNA described herein may be determined in proliferating cells
  • Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
  • compositions in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1 % and 100%, e.g., between .5 and 50%, between 1 -30%, between 5-80%, at least 80% (w/w) active ingredient.
  • the formulations described herein may contain at least one saRNA.
  • the formulations may contain 1 , 2, 3, 4 or 5 saRNAs with different sequences.
  • the formulation contains at least three saRNAs with different sequences.
  • the formulation contains at least five saRNAs with different sequences.
  • the saRNA of the present invention can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the saRNA); (4) alter the biodistribution (e.g., target the saRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo.
  • excipients of the present invention can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core-shell nanoparticles, peptides, proteins, cells transfected with saRNA (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics and combinations thereof.
  • the formulations of the invention can include one or more excipients, each in an amount that together increases the stability of the saRNA and/or increases cell transfection by the saRNA.
  • the saRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles.
  • Pharmaceutically acceptable carriers, excipients, and delivery agents for nucleic acids that may be used in the formulation with the saRNA of the present invention are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
  • the saRNA of the present invention comprises two single
  • composition further comprises a salt buffer composed of 50mM Tris-HCl, pH 8.0, lOOmM NaCl and 5mM EDTA.
  • the saRNA of the present invention may be delivered with dendrimers.
  • Dendrimers are highly branched macromolecules.
  • the saRNA of the present invention is complexed with structurally flexible poly(amidoamine) (PAMAM) dendrimers for targeted in vivo delivery.
  • PAMAM structurally flexible poly(amidoamine)
  • the complex is called sa R A-d cndri mers .
  • Dendrimers have a high degree of molecular uniformity, narrow molecular weight distribution, specific size and shape characteristics, and a highly-functional izcd terminal surface.
  • the manufacturing process is a series of repetitive steps starting with a central initiator core. Each subsequent growth step represents a new generation of polymers with a larger molecular diameter and molecular weight, and more reactive surface sites than the preceding generation.
  • PAMAM dendrimers are efficient nucleotide delivery systems that bear primary amine groups on their surface and also a tertiary amine group inside of the structure.
  • the primary amine group participates in nucleotide binding and promotes their cellular uptake, while the buried tertiary amino groups act as a proton sponge in endosomes and enhance the release of nucleic acid into the cytoplasm.
  • These dendrimers protect the saRNA carried by them from ribonuclease degradation and achieves substantial release of saRNA over an extended period of time via endocytosis for efficient gene targeting.
  • PAMAM ' dendrimers may comprise a triethanolamine (TEA) core, a diaminobutane (DAB) core, a cystamine core, a diaminohexane (HEX) core, a
  • PAMAM dendrimers comprise a TEA core or a DAB core.
  • saRNA complexes, micelles, liposomes or particles can be prepared containing these lipidoids and therefore, can result in an effective delivery of the saRNA following the injection of a lipidoid formulation via localized and/or systemic routes of administration.
  • Lipidoid complexes of saRNA can be administered by various means including, but not limited to, intravenous, intramuscular, or subcutaneous routes,
  • nucleic acids may be affected by many parameters, including, but not limited to, the formulation composition, nature of particle PEGylation, degree of loading, oligonucleotide to lipid ratio, and biophysical parameters such as, but not limited to, particle size (Akinc et al., Mol Ther. 2009 17:872-879; the contents of which are herein incorporated by reference in its entirety).
  • particle size Akinc et al., Mol Ther. 2009 17:872-879; the contents of which are herein incorporated by reference in its entirety.
  • poly(ethylene glycol) (PEG) lipids may result in significant effects on in vivo efficacy.
  • Formulations with the different lipidoids including, but not limited to penta[3-(l - laurylaminopropionyl)]-triethylenetetramine hydrochloride (TETA-5LAP; aka 98N12-5, see Murugaiah et al, Analytical Biochemistry, 401 :61 (2010); the contents of which are herein incorporated by reference in its entirety), C 12-200 (including derivatives and variants), and MD1 , can be tested for in vivo activity.
  • TETA-5LAP penta[3-(l - laurylaminopropionyl)]-triethylenetetramine hydrochloride
  • C 12-200 including derivatives and variants
  • MD1 can be tested for in vivo activity.
  • CI 2-200 The lipidoid referred to herein as "CI 2-200" is disclosed by Love et al., Proc
  • the lipidoid formulations can include particles comprising either 3 or 4 or more components in addition to the saRNA.
  • formulations with certain lipidoids include, but are not limited to, 98N12-5 and may contain 42% lipidoid, 48% cholesterol and 10% PEG (C14 alkyl chain length).
  • formulations with certain lipidoids include, but are not limited to, C12-200 and may contain 50% lipidoid, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and 1.5% PEG-DMG.
  • an saRNA formulated with a lipidoid for systemic intravenous administration can target the liver.
  • a final optimized intravenous formulation using saRNA and comprising a lipid molar composition of 42% 98N12-5, 48% cholesterol, and 10% PHG-lipid with a final weight ratio of about 7,5 to 1 total lipid to saRNA and a C14 alkyl chain length on the PEG lipid, with a mean particle size of roughly 50-60 nm, can result in the distribution of the formulation to be greater than 90%o to the liver, (see, Akinc et al, Mol Ther. 2009 17:872-879; the contents of which are herein incorporated by reference in its entirety).
  • an intravenous formulation using a CI 2-200 may have a molar ratio of 50/10/38.5/1.5 of C12- 200/disteroylphosphatidyl choline/cholesterol/PEG-DMG, with a weight ratio of 7 to 1 total lipid to nucleic acid and a mean particle size of 80 nm may be effective to deliver saRNA (see, Love et al., Proc Natl Acad Sci U S A. 2010 107: 1864-1869, the contents of which are herein incorporated by reference in its entirety).
  • an MD1 lipidoid-containing formulation may be used to effectively deliver saRNA to hepatocytes in vivo.
  • the characteristics of optimized lipidoid formulations for intramuscular or subcutaneous routes may vary significantly depending on the target cell type and the ability of formulations to diffuse through the extracellular matrix into the blood stream. While a particle size of less than 150 nm may be desired for effective hepatocyte delivery due to the size of the endothelial fenestrae (see, Akinc et al., Mol Ther.
  • lipidoid- formulated saRNA to deliver the formulation to other cells types including, but not limited to, endothelial cells, myeloid cells, and muscle cells may not be similarly size-limited.
  • lipidoid formulations to deliver siRNA in vivo to other non-hep atocyte cells such as myeloid cells and endothelium has been reported (see Akinc et al, Nat Biotechnol. 2008 26:561-569; Leuschner et al, Nat Biotechnol. 2011 29:1005-1010; Cho et al. Adv. Fund. Mater. 2009 19:31 12-3118; 8 th International Judah Folkman Conference, Cambridge, MA October 8-9, 2010; the contents of each of which is herein incorporated by reference in its entirety).
  • Effective delivery to myeloid cells, such as monocytes lipidoid formulations may have a similar component molar ratio.
  • lipidoids and other components including, but not limited to, disteroylphosphatidyl choline, cholesterol and PEG-DMG, may be used to optimize the formulation of saRNA for delivery to different cell types including, but not limited to, hepatoeytes, myeloid cells, muscle cells, etc.
  • the component molar ratio may include, but is not limited to, 50% C12-200, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and %1.5 PEG-DMG (see Leuschner et al, Nat Biotechnol 2011 29: 1005-1010; the contents of which are herein incorporated by reference in its entirety).
  • lipidoid formulations for the localized delivery of nucleic acids to cells (such as, but not limited to, adipose cells and muscle cells) via either subcutaneous or intramuscular delivery, may not require all of the formulation components desired for systemic delivery, and as such may comprise only the lipidoid and saRNA,
  • Liposomes Liposomes, Lipoplexes, and Lipid Nanoparticles
  • the saRNA of the invention can be formulated using one or more liposomes, lipoplexes, or lipid nanoparticles.
  • pharmaceutical compositions of saRNA include liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of nutrients and pharmaceutical formulations.
  • Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter.
  • MLV multilamellar vesicle
  • SUV small unicellular vesicle
  • LUV large unilamellar vesicle
  • Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis.
  • Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical formulations.
  • liposomes may depend on the physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped and the liposomal ingredients, the nature of the medium in which the lipid vesicles are dispersed, the effective concentration of the entrapped substance and its potential toxicity, any additional processes involved during the application and/or delivery of the vesicles, the optimization size,
  • compositions described herein may include, without limitation, liposomes such as those formed from 1 ,2-dioleyloxy-JV,iV- dimethylaminopropane (DODMA) liposomes, DiLa2 liposomes from Marina Biotech (Bothell, WA), l ,2-dilinoleyloxy-3-dimethylaminopropane (DLin-DMA), 2 ,2-dilinoleyl-4-(2- dimethylaminoethyl)-[l ,3]-dioxolane (DLin-KC2-DMA), and MC3 (US20100324120; the contents of which are herein incorporated by reference in its entirety) and liposomes which may deliver small molecule drugs such as, but not limited to, DOX1
  • compositions described herein may include, without limitation, liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271 -281 ; Zhang et al. Gene Therapy. 1999 6: 1438-1447; Jeffs et al. Pharm Res. 2005 22:362-372; Morrissey et al, Nat BiotechnoL 2005 2: 1002-1007; Zimmermann et al., Nature.
  • liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy.
  • the liposome formulations may be composed of 3 to 4 lipid components in addition to the saRNA.
  • a liposome can contain, but is not limited to, 55% cholesterol, 20% disteroylphosphatidyl choline (DSPC), 10% PEG-S-DSG, and 15% 1 ,2-dioleyloxy-N.N- dimcthylaminopropane (DODMA), as described by Jeffs et al.
  • DSPC disteroylphosphatidyl choline
  • PEG-S-DSG 10%
  • DODMA 1,2-dioleyloxy-N.N- dimcthylaminopropane
  • certain liposome formulations may contain, but are not limited to, 48% cholesterol, 20% DSPC, 2% PEG-c-DMA, and 30% cationic lipid, where the cationic lipid can be 1 ,2-distearloxy-JV,N- dimethylaminopropane (DSDMA), DODMA, DLin-DMA, or 1 ,2-dilinolcnv loxy-3 - dimethylaminopropane (DLenDMA), as described by Heyes et al
  • the nucleic acid-lipid particle may comprise a cationic lipid comprising from about 50 mol % to about 85 mol % of the total lipid present in the particle; a non-cationic lipid comprising from about 13 mol % to about 49.5 mol % of the total lipid present in the particle; and a conjugated lipid that inhibits aggregation of particles comprising from about 0.5 mol % to about 2 mol % of the total
  • the nucleic acid-lipid particle may be any nucleic acid-lipid particle disclosed in US2006008910 to Maelaehlan et al., the contents of which are incorporated herein by reference in their entirety.
  • the nucleic acid-lipid particle may comprise a cationic lipid of Formula I, a non-eationic lipid, and a conjugated lipid that inhibits aggregation of particles.
  • the saRNA of the present invention may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers.
  • the liposome may contain a sugar-modified lipid disclosed in US5595756 to Bally et al., the contents of which are incorporated herein by reference in their entirety.
  • the lipid may be a ganglioside and cerebroside in an amount of about 10 mol percent
  • the saRNA of the present invention may be formulated in a liposome comprising a cationic lipid.
  • the liposome may have a molar ratio of nitrogen atoms in the cationic lipid to the phosphates in the saRNA (N:P ratio) of between 1 :1 and 20:1 as described in International Publication No. WO2013006825, the contents of which are herein incorporated by reference in its entirety.
  • the liposome may have a N;P ratio of greater than 20:1 or less than 1 : 1.
  • the saRNA of the present invention may be formulated in a lipid-polycation complex.
  • the formation of the lipid-polycation complex may be accomplished by methods known in the art and/or as described in U.S. Pub. No. 20120178702, the contents of which are herein incorporated by reference in its entirety.
  • the polycation may include a cationic peptide or a polypeptide such as, but not limited to, polylysine, poly ornithine and/or polyarginine and the cationic peptides described in International Pub. No. WO2012013326; herein incorporated by reference in its entirety.
  • the saRNA may be formulated in a lipid-polycation complex which may further include a neutral lipid such as, but not limited to, cholesterol or dioleoyl phosphatidylethanolamine (DOPE), [00170]
  • DOPE dioleoyl phosphatidylethanolamine
  • the liposome formulation may be influenced by, but not limited to, the selection of the catiomc lipid component, the degree of cationic lipid saturation, the nature of the
  • the liposome formulation was composed of 57.1 % catiomc lipid, 7.1% d i almitoylphosphatidylcholine, 34.3 % cholesterol, and 1.4% PEG-c-DMA, [00171]
  • the ratio of PEG in the lipid nanoparticle (LNP) formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified from C 14 to CI 8 to alter the pharmacokinetics and/or biodistribution of the LNP formulations.
  • LNP formulations may contain 1 -5% of the lipid molar ratio of PEG-c-DOMG as compared to the cationic lipid, DSPC and cholesterol
  • the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG-DSG ( 1 ,2-Distearoyl-sn-g ycerol, methoxypolyethylene glycol) or PEG-DPG ( 1 ,2-Dipalmitoyl-sn -glycerol, methoxypolyethylene glycol).
  • the cationic lipid may be selected from any lipid known in the art such as, but not limited to, DLin-MC3-DMA, D Lin-DM A , C 1 2- 200 and DLin-KC2 -DMA,
  • the saRNA of the present invention may be formulated in a lipid nanoparticle such as the lipid nanopartieles described in International Publication No.
  • the cationic lipid which may be used in formulations of the present invention may be selected from, but not limited to, a cationic lipid described in
  • the cationic lipid may be selected from, but not limited to, formula A described in International Publication Nos. WO2012040184, WO201 1 153120, WO201 1 149733, WO201 1090965, WO201 1043913, WO201 1022460, WO2012061259, WO2012054365 and WO2012044638; the contents of each of which is herein incorporated by reference in their entirety.
  • the cationic lipid may be selected from, but not limited to, formula CLI-CLXXIX of International Publication No, WO2008103276, formula CLI-CLXXIX of US Patent No. 7,893,302, formula CLI-CLXXXII of US Patent No. 7,404,969 and formula I- VI of US Patent Publication No. US201000361 15; the contents of each of which are herein incorporated by reference in their entirety.
  • the cationic lipid may be a multivalent cationic lipid such as the cationic lipid disclosed in US Patent No. 7223887 to Gaucheron et al, the contents of which are incorporated herein by reference in their entirety.
  • the cationic lipid may have a positively-charged head group including two quaternary amine groups and a hydrophobic portion including four hydrocarbon chains as described in US Patent No, 7223887 to Gaucheron et al., the contents of which are incorporated herein by reference in their entirety.
  • the cationic lipid may be biodegradable as the biodegradable lipids disclosed in US20130195920 to Maier et al., the contents of which are incorporated herein by reference in their entirety.
  • the cationic lipid may have one or more biodegradable groups located in a lipidic moiety of the cationic lipid as described in formula I- IV in US 20130195920 to Maier et al, the contents of which are incorporated herein by reference in their entirety.
  • the cationic lipid may be selected from (2QZ,23Z)-
  • N,N-dimethylnonacosa-20,23-dien- 10-amine ( 17Z,20Z)-N,N-dimemylhexacosa- 17,20-dien-9- amine, ( 1 Z, 19Z)-N5N-dimethy lpentaeosa-1 6, 19-dien-8-amine, ( 13Z, 16Z)-N,N-dimethyldoeosa- 13,16-dien-5-arnine, ( 12Z.15Z)-N,N-dimethylhenieosa-l 2, 15-dien-4-amine, ( 14Z, 17Z)-N,N- dimethyltricosa-14, 17-dien-6-amine, ( 15Z, 18Z)-N,N-dimethyltetraeosa- 15,18-dien-7 -amine, ( 18Z,2 lZ)-N,N-dimethylheptacosa- 18,21 -dien-10-amine, ( 15ZJ 8
  • the lipid may be a eleavable lipid such as those described in
  • the nanopartieles described herein may comprise at least one cationic polymer described herein and or known in the art.
  • the cationic lipid may be synthesized by methods known in the art and/or as described in International Publication Nos. WO2012040184, WO201 1 153120, WO201 1 149733, WO201 1090965, WO201 1043913, WO201 1022460, WO2012061259, WO2012054365, WO2012044638, WO2010080724 and WO201021865; the contents of each of which is herein incorporated by reference in their entirety.
  • the LNP formulations of the saRNA may contain PEG-c-
  • the LNP formulations of the saRNA may contain PEG-c-DOMG at 1.5% lipid molar ratio.
  • the pharmaceutical compositions of the saRNA may include at least one of the PEGylated lipids described in International Publication No. 2012099755, the contents of which is herein incorporated by reference in its entirety.
  • the LNP formulation may contain PEG-DMG 2000 (l ⁇ - dimyristoyl-sn-glycero-3-phophoethanolamine-N-[methoxy(polyethylene glycol)-2000).
  • the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art and at least one other component.
  • the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art, DSPC and cholesterol.
  • the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol.
  • the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol in a molar ratio of 2:40: 10:48 (see e.g., Geall et al, Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety).
  • the saRNA described herein may be formulated in a nanoparticle to be delivered by a parenteral route as described in U.S. Pub. No. 20120207845; the contents of which is herein incorporated by reference in its entirety.
  • the cationic lipid may also be the cationic lipids disclosed in US20130156845 to Manoharan et al. and US 20130129785 to Manoharan et al, WO 2012047656 to Wasan et al., WO 2010144740 to Chen et al, WO 2013086322 to Ansell et al, or WO 2012016184 to Manoharan et al, the contents of each of which are incorporated herein by reference in their entirety,
  • the saRNA of the present invention may be formulated with a plurality of cationic lipids, such as a first and a second cationic lipid as described in
  • the first cationic lipid can be selected on the basis of a first property and the second cationic lipid can be selected on the basis of a second property, where the properties may be determined as outlined in US20130017223, the contents of which are herein incorporated by reference in its entirety.
  • the first and second properties are
  • the saRNA may be formulated with a lipid particle comprising one or more cationic lipids and one or more second lipids, and one or more nucleic acids, wherein the lipid particle comprises a solid core, as described in US Patent Publication No. US20120276209 to Cullis et al., the contents of which are incorporated herein by reference in their entirety.
  • the saRNA of the present invention may be complexed with a cationic amphiphile in an oil-in-water (o/w) emulsion such as described in EP2298358 to Satishchandran et al., the contents of which are incorporated herein by reference in their entirety.
  • the cationic amphiphile may be a cationic lipid, modified or unmodified spermine, bupivacaine, or benzalkonium chloride and the oil may be a vegetable or an animal oil.
  • nucleic acid-cationic amphiphile complex is in the oil phase of the oil-in-water emulsion (see e.g., the complex described in European Publication No. EP2298358 to Satishchandran et al, the contents of which are herein incorporated by reference in its entirety).
  • the saRNA of the present invention may be formulated with a composition comprising a mixture of cationic compounds and neutral lipids.
  • the cationic compounds may be formula (I) disclosed in WO 1999010390 to Ansell et al., the contents of which are disclosed herein by reference in their entirety
  • the neutral lipid may be selected from the group consisting of diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide and sphingomyelin.
  • the lipid formulation may comprise a eationie lipid of formula A, a neutral lipid, a sterol and a PEG or PEG-modified lipid disclosed in US 20120101 148 to Akinc et al, the contents of which are incorporated herein by reference in their entirety.
  • the LNP formulation may be formulated by the methods described in International Publication Nos. WO2011127255 or WO2008103276, each of which are herein incorporated by reference in their entirety.
  • the saRNA of the present invention may be encapsulated in any of the lipid nanoparticle (LNP) formulations described in WO2011 127255 and/or WO2008103276; the contents of each of which are herein incorporated by reference in their entirety.
  • the LNP formulations described herein may comprise a polycationic composition.
  • the polycationic composition may be selected from formula 1-60 of US Patent Publication No. US20050222064; the contents of which is herein incorporated by reference in its entirety.
  • the LNP formulations comprising a polycationic composition may be used for the delivery of the saRNA described herein in vivo and/or in vitro,
  • the LNP formulations described herein may additionally comprise a permeability enhancer molecule.
  • a permeability enhancer molecule are described in US Patent Publication No. US20050222064; the contents of which is herein incorporated by reference in its entirety.
  • the pharmaceutical compositions may be formulated in liposomes such as, but not limited to, DiLa2 liposomes (Marina Biotech, Bothell. WA), SMARTICLES®/NOV340 (Marina Biotech, Bothell, WA), neutral DOPC (1,2-dioleoyl-sn- glycero-3 -phosphoeholine) based liposomes (e.g., siRNA delivery for ovarian cancer (Landen et al. Cancer Biology & Therapy 2006 5(12)1708-1713); the contents of which is herein incorporated by reference in its entirety) and hyaluronan-coated liposomes (Quiet Therapeutics, Israel).
  • DiLa2 liposomes Marina Biotech, Bothell. WA
  • SMARTICLES®/NOV340 Marina Biotech, Bothell, WA
  • neutral DOPC 1,2-dioleoyl-sn- glycero-3 -phosphoeholine
  • the pharmaceutical compositions may be formulated with any amphoteric liposome disclosed in WO 2008/043575 to Panzner and US 8580297 to Essler et al. (Marina Biotech), the contents of which are incorporated herein by reference in their entirety.
  • the amphoteric liposome may comprise a mixture of lipids including a eationie amphiphile, an anionic amphiphile and optional one or more neutral amphiphiles.
  • the amphoteric liposome may comprise amphoteric compounds based on amphiphilic molecules, the head groups of which being substituted with one or more amphoteric groups.
  • the pharmaceutical compositions may be formulated with any amphoteric liposome disclosed in WO 2008/043575 to Panzner and US 8580297 to Essler et al. (Marina Biotech), the contents of which are incorporated herein by reference in their entirety.
  • the amphoteric liposome may comprise a mixture of lipids including a eation
  • compositions may be formulated with an amphoteric lipid comprising one or more amphoteric groups having an isoelectric point between 4 and 9, as disclosed in US
  • the pharmaceutical composition may be formulated with liposomes comprising a sterol derivative as disclosed in US 7312206 to Panzner et al.
  • the pharmaceutical composition may be formulated with amphoteric liposomes comprising at least one amphipathic cationic lipid, at least one amphipathic anionic lipid, and at least one neutral lipid, or liposomes comprise at least one amphipathic lipid with both a positive and a negative charge, and at least one neutral lipid, wherein the liposomes are stable at pH 4.2 and pH 7.5, as disclosed in US Pat. No. 7780983 to Panzner et al. (Novosom), the contents of which are incorporated herein by reference in their entirety.
  • the pharmaceutical composition may be formulated with liposomes comprising a serum-stable mixture of lipids taught in US 201 10076322 to Panzner et al, the contents of which are incorporated herein by reference in their entirety, capable of encapsulating the saRNA of the present invention.
  • the lipid mixture comprises phosphatidylcholine and
  • the lipid mixture may also include pH sensitive anionic and cationic amphiphiles, such that the mixture is amphoteric, being negatively charged or neutral at pH 7.4 and positively charged at pH 4.
  • the drug/lipid ratio may be adjusted to target the liposomes to particular organs or other sites in the body.
  • liposomes loaded with the saRNA of the present invention as cargo are prepared by the method disclosed in US 20120021042 to Panzner et al, the contents of which are incorporated herein by reference in their entirety.
  • the method comprises steps of admixing an aqueous solution of a polyanionic active agent and an alcoholic solution of one or more amphiphiles and buffering said admixture to an acidic pH, wherein the one or more amphiphiles are susceptible of forming amphoteric liposomes at the acidic pH, thereby to form amphoteric liposomes in suspension encapsulating the active agent.
  • the nanoparticle formulations may be a carbohydrate nanoparticle comprising a carbohydrate carrier and a nucleic acid molecule (e.g., saR A),
  • the carbohydrate carrier may include, but is not limited to, an anhydride-modified phytoglyeogen or glycogen-type material, phtoglycogen octenyl succinate, phytoglyeogen beta-dextrin, anhydride- modified phytoglyeogen beta-dextrin. (See e.g., International Publication No. WO20 12 1 091 2 1 ; the contents of which is herein incorporated by reference in its entirety).
  • Lipid nanoparticle formulations may be improved by replacing the cationic lipid with a biodegradable cationic lipid which is known as a rapidly eliminated lipid nanoparticle (reLNP).
  • lonizable cationic lipids such as, but not limited to, DLinDMA, DLin-KC2-DMA, and DLin-MC3-DMA, have been shown to accumulate in plasma and tissues over time and may be a potential source of toxicity.
  • the rapid metabolism of the rapidly eliminated lipids can improve the toierabiiity and therapeutic index of the lipid nanoparticles by an order of magnitude from a 1 mg/kg dose to a 10 mg/kg dose in rat.
  • ester linkage can improve the degradation and metabolism profile of the cationic component, while still maintaining the activity of the reLNP formulation.
  • the ester linkage can be internally located within the lipid chain or it may be terminally located at the terminal end of the lipid chain.
  • the internal ester linkage may replace any carbon in the lipid chain.
  • the saRNA may be formulated as a lipoplex, such as, without limitation, the ATUPLEXTM system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom),
  • STEMFECTTM from STEMGENT® (Cambridge, MA), and polyethylenimine (PEI) or protamine-based targeted and non-targeted delivery of nucleic acids
  • STEMGENT® Cambridge, MA
  • PEI polyethylenimine
  • protamine-based targeted and non-targeted delivery of nucleic acids Aleku et al. Cancer Res. 2008 68:9788-9798; Striimberg et al. Int J Clin Pharmacol Ther 2012 50:76-78; Santel et al, Gene Ther 2006 13:1222-1234; Santel et al, Gene Ther 2006 13 :1360-1370; Gutbier et al, Pulm Pharmacol. Ther. 2010 23:334-344; Kaut ' mann et al. Mierovasc Res 2010 80:286-293Weide et al. J Immunother.
  • such formulations may also be constructed or compositions altered such that they passively or actively are directed to different cell types in vivo, including but not limited to hepatocytes, immune cells, tumor cells, endothelial cells, antigen presenting cells, and leukocytes (Akinc et al, Mol Ther. 2010 18:1357-1364; Song et al., Nat Biotechnol. 2005 23:709-717; Judge et al., J Clin Invest. 2009 1 19:661 -673; Kaufmann et al., Microvasc Res
  • passive targeting of formulations to liver cells includes the DLin-DMA, DLin-KC2-DMA and DLin- MC3-DMA-based lipid nanoparticle formulations which have been shown to bind to
  • Formulations can also be selectively targeted through expression of different ligands on their surface as exemplified by, but not limited by, folate, transferrin, N- aeetylgalactosamine (GalNAc), and antibody targeted approaches (Kolhatkar et al., Curt Drug Discov Technol. 201 1 8: 197-206; Musacchio and Torchilin, Front Biosci.
  • the saRNA is formulated as a solid lipid nanoparticle.
  • a solid lipid nanoparticle may be spherical with an average diameter between 10 to 1000 urn. SLN possess a solid lipid core matrix that can solubilize lipophilic molecules and may be stabilized with surfactants and/or emulsifiers.
  • the lipid nanoparticle may be a self-assembly lipid-polymer nanoparticle (see Zhang et al, ACS Nano, 2008, 2 (8), pp 1696-1702; the contents of which are herein incorporated by reference in its entirety).
  • the saRNA of the present invention can be formulated for controlled release and/or targeted delivery.
  • controlled release refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome.
  • the saRNA may be encapsulated into a delivery agent described herein and/or known in the art for controlled release and/or targeted delivery.
  • encapsulate means to enclose, surround or encase.
  • encapsulation may be substantial, complete or partial
  • substantially encapsulated means that at least greater than 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.9 or greater than 99.999% of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent.
  • Partially encapsulated means that less than 10, 10, 20, 30, 40 50 or less of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent.
  • encapsulation may be determined by measuring the escape or the activity of the pharmaceutical composition or compound of the invention using fluorescence and or electron micrograph.
  • At least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the pharmaceutical composition or compound of the invention are encapsulated in the delivery agent.
  • the saRNA may be encapsulated into a lipid
  • nanoparticle or a rapidly eliminated lipid nanoparticle and the lipid nanoparticles or a rapidly eliminated lipid nanoparticle may then be encapsulated into a polymer, hydrogel and/or surgical sealant described herein and/or known in the art.
  • the polymer, hydrogel or surgical sealant may be PLGA, ethylene vinyl acetate (EVAc), poloxamer,
  • GELSUE® Nanotherapeutics, Inc. Alachua, FL
  • HYLENEX® Hydrozyme Therapeutics, San Diego CA
  • surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA)
  • TISSELL® (Baxter International, Inc., Decrfield. IL)
  • PEG-based sealants PEG-based sealants
  • COSEAL® Baxter International, Inc., Decrfield, IL
  • the lipid nanoparticle may be encapsulated into any polymer known in the art which may form a gel when injected into a subject.
  • the lipid nanopartiele may be encapsulated into a polymer matrix which may be biodegradable.
  • the saRNA formulation for controlled release and/or targeted delivery may also include at least one controlled release coating.
  • Controlled release coatings include, but are not limited to, OPADRY®, polyvinylpyrrolidone/ vinyl acetate copolymer, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, EUDRAGIT RL®, EUDRAGIT RS® and cellulose derivatives such as ethylcellulose aqueous dispersions (AQUACOAT® and SURELEASE®).
  • the controlled release and/or targeted delivery formulation may comprise at least one degradable polyester which may contain polycationic side chains.
  • Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-laetide-co-L- lysine), poly(4-hydroxy-L-proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the saRNA of the present invention may be formulated with a targeting lipid with a targeting moiety such as the targeting moieties disclosed in
  • 20130202652 to Manoharan et al. may selected in order to favor the lipid being localized with a desired organ, tissue, cell, cell type or subtype, or organelle.
  • Non-limiting targeting moieties that are contemplated in the present invention include transferrin, anisamide, an RGD peptide, prostate specific membrane antigen (PSMA), fueose, an antibody, or an aptamer.
  • the saRNA of the present invention may be encapsulated in a therapeutic nanopartiele.
  • Therapeutic nanoparticles may be formulated by methods described herein and known in the art such as, but not limited to, International Pub Nos. WO2010005740, WO2010030763, WO2010005721 , WO2010005723, WO2012054923, US Pub. Nos.
  • therapeutic polymer nanoparticles may be identified by the methods described in US Pub No. US20120140790, the contents of which are herein incorporated by reference in its entirety.
  • sustained release refers to a pharmaceutical composition or compound that conforms to a release rate over a specific period of time.
  • the period of time may include, but is not limited to, hours, days, weeks, months and years.
  • the sustained release nanoparticle may comprise a polymer and a therapeutic agent such as, but not limited to, the saRNA of the present invention (see International Pub No. 2010075072 and US Pub No. US20100216804, US201 10217377 and US20120201859, the contents of each of which are herein incorporated by reference in their entirety).
  • the therapeutic nanoparticles may be formulated to be target specific.
  • the therapeutic nanoparticles may include a corticosteroid (see International Pub. No. WO201 1084518; the contents of which are herein incorporated by reference in its entirety), hi one embodiment, the therapeutic nanoparticles may be formulated to be cancer specific.
  • the therapeutic nanoparticles may be formulated in nanoparticles described in International Pub No. WO2008121949, WO2010005726,
  • the nanoparticles of the present invention may comprise a polymeric matrix.
  • the nanoparticle may comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides,
  • polyhydroxyacids polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes,
  • polyphosphazenes polyacrylates, polymethacrylates, polycyanoacrylates, polyureas,
  • polystyrenes polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co- L-lysine), poly(4-h yd rox y- . -p ro line ester) or combinations thereof.
  • the therapeutic nanoparticle comprises a diblock copolymer.
  • the diblock copolymer may include PEG in combination with a polymer such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L -lysine), poly(4- hydroxy-L-proline ester) or combinations thereof,
  • a polymer such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetal
  • the therapeutic nanopartiele comprises a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No, 8,236,330, each of which is herein incorporated by reference in their entirety).
  • the therapeutic nanopartiele is a stealth nanopartiele comprising a dibloek copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968 and International Publication No.
  • the therapeutic nanopartiele may comprise a multiblock copolymer such as, but not limited to the multiblock copolymers described in U.S. Pat. No. 8,263,665 and 8,287,910; the contents of each of which are herein incorporated by reference in its entirety.
  • the block copolymers described herein may be included in a polyion complex comprising a non-polymeric micelle and the block copolymer.
  • a polyion complex comprising a non-polymeric micelle and the block copolymer.
  • the therapeutic nanopartiele may comprise at least one acrylic polymer.
  • Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methaerylate copolymers, ethoxyethyl methacrylates, cyanoethyl methaerylate, amino alkyl methaerylate copolymer, polyiaerylic acid), poly(methacrylic acid), polyeyanoacrylates and combinations thereof.
  • the therapeutic nanoparticles may comprise at least one amine-containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; the contents of which are herein incorporated by reference in its entirety) and combinations thereof.
  • amine-containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; the contents of which are herein incorporated by reference in its entirety) and combinations thereof.
  • the therapeutic nanoparticles may comprise at least one degradable polyester which may contain polycationic side chains.
  • Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the therapeutic nanoparticle may include a conjugation of at least one targeting ligand.
  • the targeting ligand may be any ligand known in the art such as, but not limited to, a monoclonal antibody. ( Kirpotin et al, Cancer Res. 2006 66:6732-6740; the contents of which are herein incorporated by reference in its entirety).
  • the therapeutic nanoparticle may he formulated in an aqueous solution which may be used to target cancer (see International Pub No. WO201 1084513 and US Pub No. US201 10294717, the contents of each of which is herein incorporated by reference in their entirety).
  • the saRNA may be encapsulated in, linked to and/or associated with synthetic nanocarriers.
  • Synthetic nanocarriers include, but are not limited to, those described in International Pub. Nos. WO20 10005740. WO2010030763, WO201213501 , WO2012 149252. WO2 1 2 149255, WO20 12 149259. WO201 2149265, WO2012 149268.
  • the synthetic nanocarriers may be formulated using methods known in the art and/or described herein. As a non-limiting example, the synthetic nanocarriers may be formulated by the methods described in International Pub Nos. WO2010005740,
  • the synthetic nanocarrier formulations may be lyophilizcd by methods described in International Pub. No. WO201 1072218 and US Pat No. 8,21 1 ,473; the contents of each of which are herein incorporated by reference in their entirety.
  • the synthetic nanocarriers may contain reactive groups to release the saRNA described herein (see International Pub. No. WO20120952552 and US Pub No. US20120171229, the contents of each of which are herein incorporated by reference in their entirety).
  • the synthetic nanocarriers may be formulated for targeted release.
  • the synthetic nanocarrier may be formulated to release the saRNA at a specified pH and/or after a desired time interval.
  • the synthetic nanoparticle may be formulated to release the saRNA after 24 hours and/or at a pH of 4.5 (see International Pub, Nos. WO2010138193 and WO2010138194 and US Pub Nos. US20110020388 and US20110027217, the contents of each of which is herein incorporated by reference in their entireties).
  • the synthetic nanocarriers may be formulated for controlled and/or sustained release of the saRNA described herein.
  • the synthetic nanocarriers for sustained release may be formulated by methods known in the art, described herein and or as described in International Pub No. WO2010138192 and US Pub No. 20100303850, the contents each of which is herein incorporated by reference in their entirety.
  • the nanoparticle may be optimized for oral administration.
  • the nanoparticle may comprise at least one cationic biopolymer such as, but not limited to, chitosan or a derivative thereof.
  • the nanoparticle may be formulated by the methods described in U.S. Pub. No. 20120282343; the contents of which are herein incorporated by reference in its entirety.
  • the saRNA of the present invention may be formulated in a modular composition such as described in US 8575123 to Manoharan et ah, the contents of which are herein incorporated by reference in their entirety.
  • the modular composition may comprise a nucleic acid, e.g., the saRNA of the present invention, at least one endosomolytic component, and at least one targeting ligand.
  • the modular composition may have a formula such as any formula described in US 8575123 to Manoharan et ah, the contents of which are herein incorporated by reference in their entirety.
  • the saRNA of the present invention may be encapsulated in the lipid formulation to form a stable nucleic acid-lipid particle (SNALP) such as described in US8546554 to de Fougerolles et al., the contents of which are incorporated here by reference in their entirety.
  • SNALP stable nucleic acid-lipid particle
  • the lipid may be cationic or non-cationic.
  • the lipid to nucleic acid ratio (mass/mass ratio) (e.g., lipid to saRNA ratio) will be in the range of from about 1 :1 to about 50:1 , from about 1:1 to about 25:1, from about 3: 1 to about 15:1 , from about 4:1 to about 10:1, from about 5:1 to about 9:1 , or about 6: 1 to about 9:1, or 5: 1, 6:1, 7:1, 8:1, 9:1, 10:1 , or 11 : 1.
  • the SNALP includes 40% 2,2-Dilinoleyl-4- dimethylaminoethyl-[l ,3]-dioxolane (Lipid A), 10% dioleoylphosphatidylcholine (DSPC), 40% cholesterol, 10% polyethyleneglycol (PEG)-C -DOMG (mole percent) with a particle size of 63.0 ⁇ 2() nm and a 0.027 nucleic acid/lipid ratio.
  • the saRNA of the present invention may be formulated with a nucleic acid-lipid particle comprising an endosomal membrane destabilizer as disclosed in US 7189705 to Lam et al., the contents of which are incorporated herein by reference in their entirety.
  • the endosomal membrane destabilizer may be a Ca 2 ⁇ ion
  • the saRNA of the present invention may be formulated with formulated lipid particles (FLiPs) disclosed in US 8148344 to Akinc et al., the contents of which are herein incorporated by reference in their entirety.
  • FLiPs formulated lipid particles
  • Akinc et al. teach that FLiPs may comprise at least one of a single or double-stranded oligonucleotide, where the oligonucleotide has been conjugated to a lipophile and at least one of an emulsion or liposome to which the conjugated oligonucleotide has been aggregated, admixed or associated.
  • FLiPs formulated lipid particles
  • the saRNA of the present invention may be delivered to a cell using a composition comprising an expression vector in a lipid formulation as described in US 6086913 to Tam et al., the contents of which are incorporated herein by reference in their entirety.
  • the composition disclosed by Tam is serum-stable and comprises an expression vector comprising first and second inverted repeated sequences from an adeno associated virus (AAV), a rep gene from AAV, and a nucleic acid fragment.
  • AAV adeno associated virus
  • the expression vector in Tam is complexed with lipids.
  • the saRNA of the present invention may be formulated with a lipid formulation disclosed in US 20120270921 to de Fougerolles et al, the contents of which are incorporated herein by reference in their entirety.
  • the lipid formulation may include a cationic lipid having the formula A described in US 20120270921, the contents of which are herein incorporated by reference in its entirety.
  • the compositions of exemplary nucleic acid-lipid particles disclosed in Table A of US 20120270921, the contents of which are incorporated herein by reference in their entirety may be used with the saRNA of the present invention.
  • the saRNA of the present invention may be fully encapsulated in a lipid particle disclosed in US 20120276207 to Maurer et al, the contents of which are incorporated herein by reference in their entirety.
  • the particles may comprise a lipid composition comprising preformed lipid vesicles, a charged therapeutic agent, and a destabilizing agent to form a mixture of preformed vesicles and therapeutic agent in a
  • destabilizing solvent wherein the destabilizing solvent is effective to destabilize the membrane of the preformed lipid vesicles without disrupting the vesicles.
  • the saRNA of the present invention may be formulated with a conjugated lipid.
  • the conjugated lipid may have a formula such as described in US 20120264810 to Lin et ai, the contents of which are incorporated herein by reference in their entirety.
  • the conjugate lipid may form a lipid particle which further comprises a eationic lipid, a neutral lipid, and a lipid capable of reducing aggregation.
  • the saRNA of the present invention may be formulated in a neutral liposomal formulation such as disclosed in US 20120244207 to Fitzgerald et al., the contents of which are incorporated herein by reference in their entirety.
  • neutral liposomal formulation refers to a liposomal formulation with a near neutral or neutral surface charge at a physiological pH.
  • Physiological pH can be, e.g., about 7.0 to about 7.5, or, e.g., about 7.5, or, e.g., 7.0, 7.1 , 7.2, 7.3 , 7.4, or 7.5 , or, e.g., 7.3, or, e.g., 7.4.
  • a neutral liposomal formulation is an ionizable lipid nanoparticle (iLNP).
  • iLNP ionizable lipid nanoparticle
  • a neutral liposomal formulation can include an ionizable eationic lipid, e.g., DLin-KC2 -DMA.
  • the saRNA of the present invention may be formulated with a charged lipid or an amino lipid.
  • charged lipid is meant to include those lipids having one or two fatty acyl or fatty alkyl chains and a quaternary amino head group.
  • the quaternary amine carries a permanent positive charge.
  • the head group can optionally include an ionizable group, such as a primary, secondary, or tertiary amine that may be protonated at physiological pH.
  • the presence of the quaternary amine can alter the pKa of the ionizable group relative to the pKa of the group in a structurally similar compound that lacks the quaternary amine (e.g., the quaternary amine is replaced by a tertiary amine) In some
  • a charged lipid is referred to as an "amino lipid.”
  • the amino lipid may be any amino lipid described in US201 10256175 to Hope et al., the contents of which are incorporated herein by reference in their entirety.
  • the amino lipids may have the structure disclosed in Tables 3-7 of Hope, such as structure (II), DLin-K-C2 -DMA, DLin-K2-DMA, DLin-K6-DMA, etc..
  • the resulting pharmaceutical preparations may be lyophilized according to Hope.
  • the amino lipids may be any amino lipid described in US 201101 17125 to Hope et al, the contents of which are incorporated herein by reference in their entirety, such as a lipid of structure (I), DLin-K-DMA, DLin-C-DAP, DLin-DAC, D Lin -MA, DLin-S-DMA, etc.
  • the amino lipid may have the structure (I), (II), (III), or (IV), or 4-(R)-DUn-K-DMA (VI), 4-(S)-DUn-K-DMA (V) as described in WO2009132131 to Manoharan et al, the contents of which are incorporated herein by reference in their entirety.
  • the charged lipid used in any of the formulations described herein may be any charged lipid described in EP2509636 to Manoharan et al., the contents of which are incorporated herein by reference in their entirety,
  • the saRNA of the present invention may be formulated with an association complex containing lipids, liposomes, or lipoplexes.
  • the association complex comprises one or more compounds each having a structure defined by formula (I), a PEG-lipid having a structure defined by formula (XV), a steroid and a nucleic acid disclosed in US8034376 to Manoharan et al, the contents of which are incorporated herein by reference in their entirety.
  • the saRNA may be formulated with any combination of formula (I), a PEG-lipid having a structure defined by formula (XV), a steroid and a nucleic acid disclosed in US8034376 to Manoharan et al, the contents of which are incorporated herein by reference in their entirety.
  • the saRNA may be formulated with any combination of formula (I), a PEG-lipid having a structure defined by formula (XV), a steroid and a nucleic acid disclosed in US8034376 to Man
  • the saRNA of the present invention may be formulated with reverse head group lipids.
  • the saRNA may be formulated with a zwitterionic lipid comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the head group, such as a lipid having structure (A) or structure (I) described in WO2011056682 to Leung et al, the contents of which are incorporated herein by reference in their entirety.
  • the saRNA of the present invention may be formulated in a lipid bilayer carrier.
  • the saRNA may be combined with a lipid- detergent mixture comprising a lipid mixture of an aggregation-preventing agent in an amount of about 5 mol% to about 20 mol%, a cationic lipid in an amount of about 0.5 mol% to about 50 mol%, and a fusogenic lipid and a detergent, to provide a nucleic acid-lipid-detergent mixture; and then dialyzing the nucleic acid-lipid-detergent mixture against a buffered salt solution to remove the detergent and to encapsulate the nucleic acid in a lipid bilayer carrier and provide a lipid bilayer-nucleic acid composition, wherein the buffered salt solution has an ionic strength sufficient to encapsulate of from about 40 % to about 80 % of the nucleic acid, described in WO 1999018933 to Cullis
  • the saRNA of the present invention may be formulated in a nucleic acid-lipid particle capable of selectively targeting the saRNA to a heart, liver, or tumor tissue site.
  • the nucleic acid-lipid particle may comprise (a) a nucleic acid; (b) 1.0 mole % to 45 mole % of a cationic lipid; (c) 0,0 mole % to 90 mole % of another lipid; (d) 1 ,0 mole % to 10 mole % of a bilayer stabilizing component; (e) 0,0 mole % to 60 mole % cholesterol; and (f) 0,0 mole % to 10 mole % of cationic polymer lipid as described in
  • EP1328254 to Cullis et al the contents of which are incorporated herein by reference in their entirety.
  • Cullis teaches that varying the amount of each of the cationic lipid, bilayer stabilizing component, another lipid, cholesterol, and cationic polymer lipid can impart tissue selectivity for heart, liver, or tumor tissue site, thereby identifying a nucleic acid-lipid particle capable of selectively targeting a nucleic acid to the heart, liver, or tumor tissue site.
  • the saRNA of the invention can be formulated using natural and/or synthetic polymers.
  • Non-limiting examples of polymers which may be used for delivery include, but are not limited to, DYNAMIC POLYCONJUGATE® (Arrowhead Research Corp., Pasadena, CA) formulations from M1RUS® Bio (Madison, WI) and Roche Madison (Madison, WI),
  • PHASERXTM polymer formulations such as, without limitation, SMARTT POLYMER
  • TECHNOLOGYTM PHASERX®, Seattle, WA
  • DMRI/DOPE poloxamer
  • VAXFECTIN® adjuvant from Vical (San Diego, CA)
  • chitosan cyelodextrin from Calando Pharmaceuticals
  • PLGA poly(lactic-co-glycolie acid)
  • RON DEL i M RNAi Oligonucleotide Nanoparticle Delivery
  • pH responsive co-block polymers such as, but not limited to, PHASERX® (Seattle, WA).
  • a non-limiting example of chitosan formulation includes a core of positively charged chitosan and an outer portion of negatively charged substrate (U.S. Pub. No.
  • Chitosan includes, but is not limited to N-trimethyl chitosan, mono-N-carboxymethyl chitosan (MCC), N-palmitoyl chitosan (NPCS), EDTA-chitosan, low molecular weight chitosan, chitosan derivatives, or combinations thereof.
  • MCC mono-N-carboxymethyl chitosan
  • NPCS N-palmitoyl chitosan
  • EDTA-chitosan low molecular weight chitosan
  • chitosan derivatives or combinations thereof.
  • the polymers used in the present invention have undergone processing to reduce and/or inhibit the attachment of unwanted substances such as, but not limited to, bacteria, to the surface of the polymer.
  • the polymer may be processed by methods known and/or described in the art and/or described in International Pub. No. WO2012150467, herein incorporated by reference in its entirety .
  • a non-limiting example of PLGA formulations include, hut are not limited to,
  • PLGA injectable depots e.g., ELIGARD® which is formed by dissolving PLGA in 66% N- methyl-2-pyrrolidone (N MP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).
  • N MP N- methyl-2-pyrrolidone
  • the first of these delivery approaches uses dynamic polyconjugates and has been shown in vivo in mice to effectively deliver siRNA and silence endogenous target mRNA in hepatoeytes (Rozema et ah, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; herein incorporated by reference in its entirety).
  • This particular approach is a multicomponent polymer system whose key features include a membrane-active polymer to which nucleic acid, in this case siRNA, is covalently coupled via a disulfide bond and where both PEG (for charge masking) and N- acetylgalactosamine (for hepatocyte targeting) groups are linked via pH-sensitive bonds
  • Another polymer approach involves using transferrin-targeted cyclodextrin-containing polycation nanoparticles. These nanoparticles have demonstrated targeted silencing of the EWS-FLI1 gene product in transferrin receptor-expressing Ewing's sarcoma tumor cells (Hu-Lieskovan et al., Cancer Res.2005 65: 8984-8982; herein incorporated by reference in its entirety) and siRNA formulated in these nanoparticles was well tolerated in non-human primates ⁇ Heidel et al., Proe Natl Acad Sci USA 2007 104:5715-21; herein incorporated by reference in its entirety). Both of these delivery strategies incorporate rational approaches using both targeted delivery and endosomal escape mechanisms.
  • the polymer formulation can permit the sustained or delayed release of saRNA
  • the altered release profile for the saRNA can result in, for example, translation of an encoded protein over an extended period of time.
  • Biodegradable polymers have been previously used to protect nucleic acids from
  • the pharmaceutical compositions may be sustained release formulations.
  • the sustained release formulations may be for subcutaneous delivery.
  • Sustained release formulations may include, but are not limited to, PLGA microspheres, ethylene vinyl acetate (EVAc), poloxamer, GELSFTE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter Intemational, Inc Deer fie Id, IL), PEG-based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
  • saRNA may be formulated in PLGA microspheres by preparing the PLGA microspheres with tunable release rates (e.g., days and weeks) and encapsulating the saRNA in the PLGA microspheres while maintaining the integrity of the saRNA during the encapsulation process.
  • EVAc are non-biodegradeable, biocompatible polymers which are used extensively in pre-clinical sustained release implant applications (e.g., extended release products Ocusert a pilocarpine ophthalmic insert for glaucoma or progestasert a sustained release progesterone intrauterine device; transdermal delivery systems Testoderm, Duragesic and Selegiline; catheters).
  • Poloxamer F-407 NF is a hydrophilic, non-ionic surfactant tribloek copolymer of polyoxyethylene-polyoxypropylene-polyoxyethylene having a low viscosity at temperatures less than 5°C and forms a solid gel at temperatures greater than 15°C.
  • PEG-based surgical sealants comprise two synthetic PEG components mixed in a delivery device which can be prepared in one minute, seals in 3 minutes and is reabsorbed within 30 days.
  • GELSITE® and natural polymers are capable of in-situ gelation at the site of administration. They have been shown to interact with protein and peptide therapeutic candidates through ionic ineraction to provide a stabilizing effect.
  • Polymer formulations can also be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N- acetylgalactosamine (GalNAc) (Benoit et al., Biomacromolecules. 201 1 12:2708-2714; Rozema et al, Proc Natl Acad Sci U S A. 2007 104; 12982-12887; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464: 1067-1070; each of which is herein incorporated by reference in its entirety).
  • GalNAc N- acetylgalactosamine
  • the saRNA of the invention may be formulated with or in a polymeric compound.
  • the polymer may include at least one polymer such as, but not limited to, polyethen.es, polyethylene glycol (PEG), poly(l-lysine)(PLL), PEG grafted to PEL, cationic lipopolymer, biodegradable cationic lipopolymer, polyethyleneimine (PE1), cross-linked branched polyialkylene imines), a polyaminc derivative, a modified poloxamer, a biodegradable polymer, elastic biodegradable polymer, biodegradable block copolymer, biodegradable random copolymer, biodegradable polyester copolymer, biodegradable polyester block copolymer, biodegradable polyester block random copolymer, multiblock copolymers, linear biodegradable copolymer, poly[a-(4-aminobutyl)-L-glycolic acid) (PAGA),
  • polypropylramerates polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, polyiethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), acrylic polymers, amine-containing polymers, dextran polymers, dextran polymer derivatives or combinations thereof .
  • the saR NA of the invention may be formulated with the polymeric compound of PEG grafted with PLL as described in U.S. Pat. No, 6,177,274; herein incorporated by reference in its entirety.
  • the formulation may be used for transfecting cells in vitro or for in vivo delivery of the saRNA,
  • the saRNA may be suspended in a solution or medium with a cationic polymer, in a dry pharmaceutical composition or in a solution that is capable of being dried as described in U.S. Pub. Nos, 20090042829 and 20090042825; each of which are herein incorporated by reference in their entireties.
  • the saRNA of the invention may be formulated with a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No.
  • the saRNA of the invention may be formulated with a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety),
  • a polyamine derivative may be used to deliver nucleic acids or to treat and/or prevent a disease or to be included in an implantable or injectable device (U.S. Pub. No.
  • a pharmaceutical composition may include the saRNA and the polyamine derivative described in U.S. Pub. No. 20100260817 (the contents of which are incorporated herein by reference in its entirety).
  • the saRNA of the present invention may be delivered using a polyaminde polymer such as, but not limited to, a polymer comprising a 1 ,3 -dipolar addition polymer prepared by combining a carbohydrate diazide monomer with a dilkyne unite comprising oligoamines (U.S. Pat. No. 8,236,280; herein incorporated by reference in its entirety).
  • the saRNA of the present invention may be formulated with at least one polymer and/or derivatives thereof described in International Publication Nos.
  • the saRNA of the present invention may be formulated with a polymer of formula Z as described in WO201 1 1 15862, herein incorporated by reference in its entirety.
  • the saRNA may be formulated with a polymer of formula Z, Z' or Z " as described in International Pub. Nos. WO201 2082574 or WO20 1 2068 1 87 and U.S. Pub. No. 2012028342, the contents of each of which are herein incorporated by reference in their entireties.
  • the polymers formulated with the saRNA of the present invention may be
  • the saRNA of the invention may be formulated with at least one acrylic polymer.
  • Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl
  • methacrylates cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
  • Formulations of saRNA of the invention may include at least one amine- containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers or combinations thereof.
  • the saRNA of the invention may he formulated in a pharmaceutical compound including a poly(alkylene imine), a biodegradable cationic lipopolymer, a
  • biodegradable block copolymer a biodegradable polymer, or a biodegradable random
  • biodegradable cationic lipopolymer may be made by methods known in the art and/or described in U.S. Pat. No. 6,696,038, U.S. App. Nos. 20030073619 and 20040142474 each of which is herein incorporated by reference in their entireties.
  • the poly(alkylene imine) may be made using methods known in the art and/or as described in U.S. Pub. No. 20100004315, herein
  • biodegradable polymer, biodegradable block copolymer, the biodegradable random copolymer, biodegradable polyester block copolymer, biodegradable polyester polymer, or biodegradable polyester random copolymer may be made using methods known in the art and/or as described in U.S. Pat. Nos. 6,517,869 and 6,267,987, the contents of which are each incorporated herein by reference in their entirety.
  • the linear biodegradable copolymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,652,886.
  • the PAGA polymer may be made using methods known in the art and/or as described in U.S. Pat. No.
  • the PAGA polymer may be copolymerized to form a copolymer or block copolymer with polymers such as but not limited to, poly-L-lysine, polyargine, polyornithine, histones, avidin, protamines, polylactides and poly(lactide-co-glycolides).
  • the biodegradable cross-linked cationic multi-block copolymers may be made my methods known in the art and/or as described in U.S. Pat. No. 8,057,821 or U.S. Pub. No. 2012009145 each of which are herein incorporated by reference in their entireties.
  • the multi-block copolymers may be synthesized using linear polyethyleneimine (LPEI) blocks which have distinct patterns as compared to branched polyethyleneimines.
  • LPEI linear polyethyleneimine
  • the composition or pharmaceutical composition may be made by the methods known in the art, described herein, or as described in U.S. Pub. No.
  • the saRNA of the invention may be formulated with at least one degradable polyester which may contain polycationic side chains.
  • Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), p o 1 y (4 -h y dro x y- L -proline ester), and combinations thereof.
  • the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
  • the saRNA of the invention may be formulated with at least one crosslinkable polyester.
  • Crosslinkable polyesters include those known in the art and described in US Pub. No. 20120269761, herein incorporated by reference in its entirety.
  • the polymers described herein may be conjugated to a lipid- terminating PEG.
  • PLGA may be conjugated to a lipid-terminating PEG forming PLGA-DSPE-PEG.
  • PEG conjugates for use with the present invention are described in International Publication No. WO2008103276, herein incorporated by reference in its entirety.
  • the polymers may be conjugated using a ligand conjugate such as, but not limited to, the conjugates described in U.S. Pat. No. 8,273,363, herein incorporated by reference in its entirety.
  • the saRNA described herein may be conjugated with another compound.
  • conjugates are described in US Patent Nos.
  • saRNA of the present invention may be conjugated with conjugates of formula 1 -122 as described in US Patent Nos, 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties.
  • the saRNA described herein may be conjugated with a metal such as, but not limited to, gold. (See e.g., Giljohann et al Joum. Amer. Chem. Soc. 2009 131(6): 2072-2073; herein incorporated by reference in its entirety).
  • a metal such as, but not limited to, gold.
  • the saRNA described herein may be conjugated and/or encapsulated in gold- nanopartieles.
  • a gene delivery composition may include a nucleotide sequence and a poloxamer.
  • the saRNA of the present invention may be used in a gene delivery composition with the poloxamer described in U.S. Pub. No. 20100004313.
  • the polymer formulation of the present invention may be stabilized by contacting the polymer formulation, which may include a cationic carrier, with a cationie lipopolymer which may be covalently linked to cholesterol and polyethylene glycol groups.
  • the polymer formulation may be contacted with a cationic lipopolymer using the methods described in U.S. Pub. No. 20090042829 herein incorporated by reference in its entirety.
  • the cationic carrier may include, but is not limited to, polyethylenimine, poly(trimethylenimine), poly(tetramethylenimine), polypropylenimine, aminoglyeoside- polyamine, dideoxy-diamino -b-cyclodextrin, spermine, spermidine, poly(2-dimethylamino)ethyl methacrylate, poly(lysine), poly(histidine), poly(arginine), cationized gelatin, dendrimers, ehitosan, 1 ,2-Dioleoyl-3-Trimethylammomum-Pf opane(DOTAP), N- [ 1 -(2,3 - dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), 1 -[2-(oleoyloxy)ethyl]-2- oleyl-3-(2-hydroxyethyl)imidazolinium
  • the saRNA of the invention may be formulated in a polyplex of one or more polymers (U.S. Pub. No. 20120237565 and 20120270927; each of which is herein incorporated by reference in its entirety).
  • the polyplex comprises two or more cationic polymers.
  • the cationic polymer may comprise a poly(ethylene imine) (PEI) such as linear PEL
  • PEI poly(ethylene imine)
  • the saRNA of the invention can also be formulated as a nanoparticle using a combination of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphate.
  • Components may be combined in a core-shell, hybrid, and/or layer-by-layer architecture, to allow for fine-tuning of the nanoparticle so to delivery of the saRNA may be enhanced (Wang et al, Nat Mater. 2006 5 :791 -796; Fuller et al., Biomaterials. 2008 29:1526- 1532; DeKokcr et al, Adv Drug Deliv Rev. 201 1 63:748-761 ; Endres et al, Biomaterials. 201 1 32:7721 -7731 ; Su et al, Mol Pharm. 201 1 Jun 6;8(3):774-87; herein incorporated by reference in its entirety).
  • the nanoparticle may comprise a plurality of polymers such as, but not limited to hydrophilic -hydrophobic polymers (e.g., PEG-PLGA), hydrophobic polymers (e.g., PEG) and/or hydrophilic polymers (International Pub. No, WO201 20225 129; herein incorporated by reference in its entirety).
  • polymers such as, but not limited to hydrophilic -hydrophobic polymers (e.g., PEG-PLGA), hydrophobic polymers (e.g., PEG) and/or hydrophilic polymers (International Pub. No, WO201 20225 129; herein incorporated by reference in its entirety).
  • Biodegradable calcium phosphate nanoparticles in combination with lipids and/or polymers may be used to deliver saRNA in vivo.
  • a lipid coated calcium phosphate nanoparticle which may also contain a targeting ligand such as anisamide, may be used to deliver the saRNA of the present invention.
  • a targeting ligand such as anisamide
  • a lipid coated calcium phosphate nanoparticle was used (Li et al, J Contr Rel. 2010 142: 416-421 ; Li et al, J Contr Rel. 2012 158:108-1 14; Yang et al, Mol Ther, 2012 20:609-615; herein incorporated by reference in its entirety).
  • This delivery system combines both a targeted nanoparticle and a component to enhance the endosomal escape, calcium phosphate, in order to improve delivery of the siRNA.
  • calcium phosphate with a PEG-polyanion block copolymer may be used to delivery saRNA (Kazikawa et al, J Contr Rel. 2004 97:345-356; Kazikawa et al., J Contr Rel. 2006 1 1 1 :368-370; herein incorporated by reference in its entirety).
  • a PEG-charge-conversional polymer (Pitella et al.,
  • Biomaterials. 201 1 32:3106-31 14 may be used to form a nanoparticle to deliver the saRNA of the present invention.
  • the PEG-charge-conversional polymer may improve upon the PEG- polyanion block copolymers by being cleaved into a polycation at acidic pH. thus enhancing endosomal escape.
  • the use of core-shell nanoparticles has additionally focused on a high- throughput approach to synthesize cationic cross-linked nanogel cores and various shells (Siegwart et al, Proc Natl Acad Sci U S A. 201 1 108:12996-13001).
  • the complexation, delivery, and internalization of the polymeric nanoparticles can be precisely controlled by altering the chemical composition in both the core and shell components of the nanoparticle.
  • the core-shell nanoparticles may efficiently deliver saRNA to mouse hepatocytes after they covalently attach cholesterol to the nanoparticle,
  • a hollow lipid core comprising a middle PLGA layer and an outer neutral lipid layer containing PEG may be used to delivery of the saRNA of the present invention.
  • a luciferase-expressing tumor it was determined that the l ipid-polymer-l ipid hybrid nanoparticle significantly suppressed luciferase expression, as compared to a conventional lipoplex (Shi et al, Angew Chem Int Ed. 201 1 50:7027-7031 ; herein incorporated by reference in its entirety).
  • the lipid nanoparticles may comprise a core of the saRNA disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the modified nucleic acids in the core.
  • Core-shell nanoparticles for use with the saRNA of the present invention may be formed by the methods described in U.S. Pat. No. 8,313,777 herein incorporated by reference in its entirety.
  • the core-shell nanoparticles may comprise a core of the saRNA disclosed herein and a polymer shell.
  • the polymer shell may be any of the polymers described herein and are known in the art.
  • the polymer shell may be used to protect the saRNA in the core.
  • the core-shell nanoparticle may be used to treat an eye disease or disorder (See e.g. US Publication No. 20120321719, herein incorporated by reference in its entirety).
  • the polymer used with the formulations described herein may be a modified polymer (such as, but not limited to, a modified polyacetal) as described in International Publication No. WO201 1 120053, herein incorporated by reference in its entirety. Delivery
  • the present disclosure encompasses the delivery of saRNA for any of therapeutic, prophylactic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.
  • the saRNA of the present invention may be delivered to a cell naked.
  • naked refers to delivering saRNA free from agents which promote transfection.
  • the saRNA delivered to the cell may contain no modifications.
  • the naked saRNA may be delivered to the cell using routes of administration known in the art and described herein.
  • the saRNA of the present invention may be formulated, using the methods described herein.
  • the formulations may contain saRNA which may be modified and/or unmodified.
  • the formulations may further include, but are not limited to, cell penetration agents, a pharmaceutically acceptable carrier, a delivery agent, a bioerodible or biocompatible polymer, a solvent, and a sustained-release delivery depot.
  • the formulated saRNA may be delivered to the cell using routes of administration known in the art and described herein.
  • compositions may also be formulated for direct delivery to an organ or tissue in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with the compositions, and the like.
  • the saRNA of the present invention may also be cloned into a retroviral replicating vector (RRV) and transduced to cells.
  • RRV retroviral replicating vector
  • the saRNA of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavemous injection, ( into the base of the penis), intravaginal administration, intrauterine, extra-amniotic administration,
  • a pharmaceutical composition described herein can be formulated into a dosage form described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous).
  • injectable e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous.
  • Liquid dosage forms, injectable preparations, pulmonary forms, and solid dosage forms described in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety may be used as dosage forms for the saRNA of the present invention.
  • One aspect of the present invention provides methods of using saRNA of the present invention and pharmaceutical compositions comprising the saRNA and at least one pharmaceutically acceptable carrier.
  • the saRNA of the present invention modulates the expression of its target gene.
  • a method of regulating the expression of a target gene in vitro and/or in vivo comprising administering the saRNA of the present invention.
  • the expression of the target gene is increased by at least 5, 10, 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80% in the presence of the saRNA of the present invention compared to the expression of the target gene in the absence of the saRNA of the present invention.
  • the expression of the target gene is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, in the presence of the saRNA of the present invention compared to the expression of the target gene in the absence of the saRNA of the present invention.
  • the increase in gene expression of the saRNA descried herein is shown in proliferating cells.
  • the target gene may be any gene of the human genome.
  • the target gene may be any of the coding target genes listed in Table 1 or any of the non-coding genes listed in Table 2,
  • the saRNA described herein may be used as a spacer in a
  • CRISPR clustered regularly interspaced palindromic repeats
  • CRI PR Cas9 CRI PR Cas9 system
  • the CRISPR system comprising saRNA described herein may be used to cleave and edit a target gene.
  • the saRNA described herein may be used to treat blood disorders such as disorders affecting red blood cells (e.g., anemia, Thalassemia, sickle cell anemia and polycythemia vera), disorder affecting white blood cells (e.g., lymphoma, leukemia, multiple myeloma and myelodysplasitc syndrome), disorders affecting platelets (e.g., thrombocytopenia, idiopathic thrombocytopenic purpura, hcparin-induced thrombocytopenia, thrombotic thrombocytopenic purpura and essential thrombocytosis), disorders affecting blood plasma (e.g., sepsis, hemophilia, von Willebrand disease, hypercoagulable state, deep venous thrombosis and disseminated intravascular coagulation (DIC)).
  • red blood cells e.g., anemia, Thalassemia, sickle cell anemia and polycythemia vera
  • white blood cells e.
  • the saRNA described herein may be used to treat albumin disorders such as analbuminaemia, bisalbuminemia, familial dysalbuminemic hyperthyroxinemia and hypoalbuminemia.
  • the saRNA described herein may be used to treat coagulopathies such as haemophilia, activated protein C resistance, amyloid purpura, antiphospholipid syndrome, antithrombin III deficiency, Bernard-Soulier syndrome, bleeding diathesis, cavernous sinus thrombosis, coagulopathy, congenital afibrinogenemia, congenital amegakaryocytic thrombocytopenia, deep vein thrombosis, disseminated intravascular coagulation, dyiibrinogcncmia, essential thrombocythaemia, Evans syndrome, factor I deficiency, factor V leiden, factor XIII deficiency, Fechtners syndrome, giant platelet disorder, Glanzmann' s thrombasthenia, gray platelet syndrome, Harris platelet syndrome, heparain- induced thrombocytopenia, I lermansky-Pudlak Syndrome, hyperprothrombin
  • coagulopathies such as
  • hypoprothrombinemia idiopathic thrombocytopenic purpura, Kasabach-Merritt syndrome, May- Hegglin anomaly, neonatal alloimmune thrombocytopenia, Onyalai, phlebothrombosis, platelet storage pool deficiency, platelet transfusion refractoriness, Prett Test, primary fibrinogenolysis, protein C deficiency, protein S deficiency, prothrombin G2021 OA, pscudo gray platelet syndrome, purpura fulminans, Quebec platelet disorder, Scott syndrome, sticky platelet syndrome, superficial vein thrombosis, thrombocytopenia, thrombocytopenic purpura, thrombocytosis, thrombophilia, thrombotic microangiopathy, thrombotic thrombocytopenic purpura, Upshaw-Sehulman syndrome and Von Willebrand disease.
  • the saRNA described herein may be used to treat blood disease and disorders such as anemia, leukemia, lymphoma, multiple myeloma, sepsis and sickle-cell disease, atypical hemolytic uremic syndrome, chomeia, cytopenia, elliptocyte, fibrinogenolysis, hematolic disease, Hemolytic -uremic syndrome, hepatic veno-occlusive disease, hypervisocity syndrome, leukemoid reaction, leukocytosis, leukopenia, light chain deposition diease, macroglobulinemia, omithinaemia, pancytopenia, panmyelosis,
  • blood disease and disorders such as anemia, leukemia, lymphoma, multiple myeloma, sepsis and sickle-cell disease, atypical hemolytic uremic syndrome, chomeia, cytopenia, elliptocyte, fibrinogenolysis, hematolic disease, Hemo
  • pseudothrombocytopenia Shwachman-D ⁇ amond syndrome and tumor lysis syndrome.
  • the saRNA described herein may be used to treat hemorrhagic and hematological disorders of the fetus and newborn such as ABO, anti-Kell, anti- Rhc, anti-RhE, kernicterus, neonatal jaundice, placental insufficiency, polycythemia and Rh disease.
  • the saRNA described herein may be used to treat hematologic neoplasms such as hematologic malignant neoplasms (e.g., leukemia, lymphoma, multiple myeloma) and myeloid neoplasia (e.g., myeloid leukemia).
  • hematologic malignant neoplasms e.g., leukemia, lymphoma, multiple myeloma
  • myeloid neoplasia e.g., myeloid leukemia
  • the saRNA described herein may be used to treat histoeytosis related conditions such as Erdheim-Chester disease, Hemophagocytic
  • lymphohistiocytosis Langerhans cell histiocytosis, malignant histiocytosis, Non-Langerhans cell histiocytosis, Non-X histiocytosis, and X-type histiocytosis.
  • the saRNA described herein may be used to treat lymphocytic disorders such as lymphocytic immune system disorders (e.g., autoimmune lymphoproliferative syndrome, hypergammaglobulinemia, hypogammaglobulinemia, lymphorproiiferative disorders, paraproteinemia, persistent polyclonal B-ceil lymphocytosis and RASopathy) lymphocytic leukemia (e.g., acute lymphocytic leukemia and chronic lymphocytic leukemia) and lymphoma.
  • lymphocytic immune system disorders e.g., autoimmune lymphoproliferative syndrome, hypergammaglobulinemia, hypogammaglobulinemia, lymphorproiiferative disorders, paraproteinemia, persistent polyclonal B-ceil lymphocytosis and RASopathy
  • lymphocytic leukemia e.g., acute lymphocytic leukemia and chronic lymphocytic leukemia
  • the saRNA described herein may be used to treat monocyte and granulocyte disorders such as Agranulocytosis, Autoimmune neutropenia, Bandemia, Basopenia, Basophilia, Eosinopenia, Eosinophilia, Febrile neutropenia, Granulocytosis, Hypereosinophiiia, Hypereosinophilic syndrome, Hypersegmented neutrophil, Monocytopenia, Monocytosis, Morning pseudoneutropenia, Neutropenia and Neutrophilia.
  • monocyte and granulocyte disorders such as Agranulocytosis, Autoimmune neutropenia, Bandemia, Basopenia, Basophilia, Eosinopenia, Eosinophilia, Febrile neutropenia, Granulocytosis, Hypereosinophiiia, Hypereosinophilic syndrome, Hypersegmented neutrophil, Monocytopenia, Monocytosis, Morning pseudoneutropenia, Neutropenia
  • the saRNA described herein may be used to treat red blood cell disorders such as anemia, African iron overload, Aldolase A deficiency, Atransferrinemia, Bahima disease, Congenital dyserythropoietic anemia, Erythroid dysplasia, Haemochromatosis type 3, Hemoglobin Lepore syndrome, Hemoglobin variants, Hemoglobinemia,
  • red blood cell disorders such as anemia, African iron overload, Aldolase A deficiency, Atransferrinemia, Bahima disease, Congenital dyserythropoietic anemia, Erythroid dysplasia, Haemochromatosis type 3, Hemoglobin Lepore syndrome, Hemoglobin variants, Hemoglobinemia,
  • Hemosiderinuria Hereditary pyropoikilocytosis, HFE hereditary haemochromatosis, Iron deficiency, Latent iron deficiency, McLeod syndrome, Methemoglobinemia, Microeytosis, Myomatous erythrocytosis syndrome, Poikilocytosis, Polychromasia, Polycythemia, Porphyria, Reticulocytopenia, Rh deficiency syndrome, Sick cell syndrome, Spherocytosis,
  • the saRNA described herein may be used to treat breast disorders such as breast neoplasia (e.g., breast cancer), breast cyst, breast disease, breast engorgement, breast hematoma, breast lump, Duct ectasia of breast, Hypertrophy of breast, Inverted nipple, Fissure of the nipple, Mammary myofibroblastoma, Mastitis, Mastodynia, Mondor's disease, Nonpuerperal mastitis, Pseudoangiomatous stromal hyperplasia, Ptosis (breasts), Subareolar abscess, Tuberous breasts, and Zuska's disease
  • the saRNA described herein may be used to treat cardiovascular diseases such as cardiogenetic disorders, congenital disorders of the circulatory system (e.g., congenital heart disease, congenital vascular defects), pericardium disease (e.g., mesothelial hyperplasia), heart disease and vascular disease.
  • cardiovascular diseases such as cardiogenetic disorders, congenital disorders of the circulatory system (e.g., congenital heart disease, congenital vascular defects), pericardium disease (e.g., mesothelial hyperplasia), heart disease and vascular disease.
  • the saRNA described herein may be used to treat cardiogenetic disorders such as Alagille syndrome, Brugada syndrome, dilated cardiomyopathy, Familial atrial fibrillation.
  • cardiogenetic disorders such as Alagille syndrome, Brugada syndrome, dilated cardiomyopathy, Familial atrial fibrillation.
  • Hypertrophic cardiomyopathy Long QT syndrome, McLeod syndrome, Sakati-Nyhan-Tisdale syndrome, Short QT syndrome, Smith Martin Dodd syndrome and Timothy syndrome.
  • the saRNA described herein may be used to treat heart disease such as cardiac dysrhythmia, cardiomegaly, cardiomyopathy, cardiopulmonary resuscitation, chronic rheumatic heart disease, congenital heart disease, heart neoplasia, ischemic heart disease, pericardial disorder, valvular heart disease, Acute decompensated heart failure, Aneurysm of heart, Arteriosclerotic heart disease, Athletic heart syndrome, Atrioventricular fistula, Autoimmune heart disease, Brown atrophy of the heart, Cardiac amyloidosis, Cardiac asthma, Cardiac contractility modulation, Cardiac syndrome X, Cardiogenic shock,
  • heart disease such as cardiac dysrhythmia, cardiomegaly, cardiomyopathy, cardiopulmonary resuscitation, chronic rheumatic heart disease, congenital heart disease, heart neoplasia, ischemic heart disease, pericardial disorder, valvular heart disease, Acute decompensated heart failure
  • Cardiophobia Cardiorenal syndrome, Cardiotoxicity, Carditis, Coronary artery aneurysm, Coronary artery anomaly, Coronary artery disease, Coronary artery ectasia, Coronary occlusion, Coronary thrombosis, Coronary vasospasm, Coeur en sabot, Coxsackievirus-induced
  • cardiomyopathy Diastolic heart failure, Dressler's syndrome, Duroziez's disease, Eisenmenger's syndrome, Embryoeardia, Endocardial fibroelastosis, Heart failure, High-output cardiac failure, Hyperdynamic precordium.
  • Idiopathic giant-cell myocarditis Interventricular dyssynchrony, Intraventricular dyssynchrony, Isolated atrial amyloidosis, Keshan disease, Kounis syndrome, Mydicar, Myocardial bridge, Myocardial disarray, Myocardial rupture, Myocardial scarring, Myocardial stunning, Myocarditis, Nonbacterial thrombotic endocarditis, Ostial disease, Phosphorus and non-atherosclerotic heart disease, Postpericardiotomy syndrome, Right axis deviation, Roemheld syndrome, Shone's syndrome, Traumatic cardiac arrest, Ventricular aneurysm, Viral cardiomyopathy, and Waon therapy.
  • the saRNA described herein may be used to treat vascular disease such as cerebrovascular disease, congenital vascular defect, disease of the arteries, arterioles and capillaries, disease of the aorta, disease of veins, lymphatic vessels and lymph nodes, hypertension, ischemia, vascular neoplasia, Aggressive angiomyxoma, Anemic infarct, Aneurysm, Angiodysplasia, Angiopathy, Annuloaortic ectasia, Aortitis, Aortoiliac occlusive disease, Arterial stiffness, Arteriolosclerosis, Arteriosclerosis, Atheroma, Atherosclerosis, Brain ischemia, Thromboangiitis obliterans, Capillaritis, Carotid artery stenosis, Carotid Sonus, Cholesterol embolism, Chronic cerebrospinal ve
  • vascular disease such as cerebrovascular disease, congenital vascular defect,
  • arteriolosclerosis Hyperplastic arteriolosclerosis, Hypotension, Inferior vena cava syndrome, Intermittent claudication, Intradural pseudoaneurysm, Jugular vein ectasia, Kawasaki disease, Loeys Dictz syndrome, Lupus vasculitis, Macrovascular disease, Metanephric dysplastic hematoma of the sacral region, Monckeberg's arteriosclerosis, Obliterating endarteritis,
  • Orthostatic hypertension Orthostatic hypotension, Orthostatic intolerance, Paget Schroettcr disease, Paradoxical embolism, Peripheral vasculopathy, Phlebitis, Phlegmasia alba dolens, Phlegmasia cerulea dolens, Portacaval anastomosis, Portal vein thrombosis, Postural orthostatic tachycardia syndrome, Pscudothrombophlcbitis. Pulmonic regurgitation, Pylephlebitis,
  • Rasmussen's aneurysm Sack Barabas syndrome, Stewart-Treves syndrome, Superior mesenteric artery syndrome, Susac's syndrome, Takayasu's arteritis, Thoracic aortic aneurysm, Thrombophlebitis, Varicocele, Vascular anomaly, Vascular disease, Vascular malformation, Vaso-ocelusive crisis, Vegetative-vascular dystonia, Venous ulcer, Vertebral artery dissection, and Vertebrobasilar insufficiency,
  • the saRNA described herein may be used to treat cutaneous conditions such as acneiform eruptions, Autoinflammatory syndromes, , Chronic blistering cutaneous conditions, Conditions of the mucous membranes, Conditions of the skin appendages , Conditions of the subcutaneous fat , Connective tissue diseases (e.g., cutaneous lupus erythematosus and systemic connective tissue disorders), Cutaneous congenital anomalies (e.g., genodermatoses), Dermal and subcutaneous growths , Dermatitis (e.g., atopic dermatitis, contact dermatitis, eczema, pustular dermatitis, seborrheic dermatitis), Disturbances of human pigmentation , Drug eruptions, Endocrine-related cutaneous conditions , Eosinophilic cutaneous conditions , Epidermal nevi, Epidermal neoplasms, Epidermal cysts, Erythemas, Integumentary ne
  • cutaneous conditions such as acne
  • Neurocutaneous conditions Noninfectious immunodeficiency-related cutaneous conditions, Papulosquamous disorders (e.g., Lichenoid eruptions and psoriasis), Papulosquamous hyperkeratotic cutaneous conditions (e.g., palmoplantar keratodermas), Pregnancy-related cutaneous conditions, Pruritic skin conditions, Reactive neutrophilic cutaneous conditions, Recalcitrant palmoplantar eruptions , Skin conditions resulting from errors in metabolism , Skin conditions resulting from physical factors (e.g., bums, immersion foot syndrome, ionizing radiation-induced cutaneous conditions), Urticaria and angioedema, Vascular-related cutaneous conditions, atrophia maculosa varioliformis cutis, and dermatopathic lymphadenopathy.
  • Papulosquamous disorders e.g., Lichenoid eruptions and psoriasis
  • Papulosquamous hyperkeratotic cutaneous conditions e.g
  • the saRNA described herein may be used to treat digestive diseases such as accessory digestive gland disorders (e.g., diseases of the liver, gallbladder, biliary tract and pancreas, Alagille syndrome, biliary fever, Dub in -John son syndrome and Gilbert's syndrome), gastrointestinal cancer, tongue disorders, mesothelial hyperplasia, colitis, diseases of the appendix, diseases of the intestine, disease of the oesophagus, stomach and duodenum, steatorrhea-related diseases, peritoneum disorders, , Alvarez' syndrome, Heyde's syndrome, Reynolds syndrome, Rocmhcld syndrome, Abdominal epilepsy, Aerophagia, Ameboma, Anorectal abscess, Anismus, Anorectal disorder, Bezoar, Bile acid malabsorption, Blind loop syndrome, Bowel infarction, Bowel obstruction, Callous ulcer, Cameron lesions, Campylobacterio
  • digestive diseases such as accessory digestive
  • Dientamoebiasis Distal intestinal obstruction syndrome, Duodenal atresia, Dysphagia, Enteric neuropathy, Enterolith, Enteropathy, Eosinophilic gastroenteritis, Epiploic appendagitis, Fibrosing colonopathy, Functional constipation, Functional gastrointestinal disorder, Fundie gland polyposis, Gallstone ileus, Gastrointestinal bleeding, Gastrojejunocolic fistula, Globus pharyngis. Hematochezia, High-altitude flatus expulsion, Hirschsprung's disease,
  • Hyperehlorhydria Ileosigmoid knot, Inflammatory bowel disease, Intestinal metaplasia, Intestinal neuronal dysplasia, Intestinal spirochetosis, Intestinal varices, Kcriorrhea. Lower gastrointestinal bleeding, Malabsorption, Megacolon, Megarectum, Melanosis coli,
  • Microsporidiosis Microvillous inclusion disease, Mucositis, Necrotizing enterocolitis,
  • Pancolitis Perianal hematoma, Peutz-Jeghers syndrome, Pharyngitis, Pneumatosis intestinalis, Pouchitis, Proctocolitis, Protein losing enteropathy, Pseudodiarrhea, Radiation enteropathy, Radiation proctitis, Rectal bleeding, Rectal hemorrhage, Sandifer syndrome, Splenic flexure syndrome, Steatorrhea, Stercoral perforation, Stercoral ulcer, Stress ulcer, Superior mesenteric artery syndrome, Tropical sprue and Whipple's disease.
  • the saRNA described herein may be used to treat diseases of the ear and mastoid process such as Autoimmune inner ear disease, Balance disorder, Bilateral vestibulopathy, Cauliflower ear, Cryptotia, Ear disease, Earache, Enlarged vestibular aqueduct, Hcmotympanum. Hyperacusis, Macrotia, Mastoidectomy, Mastoiditis, Microtia, Misophonia, Mondini dysplasia, Otalgia, Otomycosis, Sopite syndrome, Superior canal dehiscence, Surfer's ear, Tinnitus, Usher syndrome, and Vestibular hyperacusis.
  • diseases of the ear and mastoid process such as Autoimmune inner ear disease, Balance disorder, Bilateral vestibulopathy, Cauliflower ear, Cryptotia, Ear disease, Earache, Enlarged vestibular aqueduct, Hcmotympanum. Hyperacusis,
  • the saRNA described herein may be used to treat endocrine disease such as adrenal gland disorders, congenital disorders of the endocrine system, disorders of the endocrine pancreas (e.g., diabetes), endocrine gonad disorders, endocrine neoplasia (e.g., thyroid cancer), endocrine-related cutaneous conditions, hypothalamus disorders, parathyroid disorders, pituitary disorders, thyroid disease (e.g., thyroid cancer and thyroid tumor), Acute infectious thyroiditis, Autoimmune thyroiditis, Colloid nodule, Congenital hypothyroidism, Cretinism, Dc Quervain's thyroiditis, Euthyroid, Euthyroid sick syndrome, Familial
  • endocrine disease such as adrenal gland disorders, congenital disorders of the endocrine system, disorders of the endocrine pancreas (e.g., diabetes), endocrine gonad disorders, endocrine neoplasia (e.g
  • ophthalmopathy Hashimoto's thyroiditis, Hashitoxicosis, Hyperthyroidism, Hyperthyroxmemia, Hypothyroidism, Maternal hypothyroidism, Myxedema coma, Myxedematous psychosis, Ord's thyroiditis, Palpation thyroiditis, Polar T3 syndrome, Postpartum thyroiditis, Radiation-induced thyroiditis, Radioactive iodine uptake test, Riedel's thyroiditis, Subacute lymphocytic thyroiditis, Subacute thyroiditis, Thyroid disease, Thyroid dyshormonogenesis, Thyroid hormone resistance, Thyroid neoplasm, Thyroid nodule, Thyroid storm, Thyroidectomy, Thyroiditis, Thyroidologist, Thyrotoxic periodic paralysis, Thyrotoxicosis factitia, Toxic multinodular goitre, Toxic nodular goiter, Van Wyk and G rum bach syndrome, Winter-over syndrome, Congenital hyperinsulinism
  • Hyperproinsulinemia Hypoglycemia, Idiopathic hypoglycemia, Impaired fasting glucose, Impaired glucose tolerance, Insulitis, Ketotic hypoglycemia, Nesidioblastosis,
  • the saRNA described herein may be used to treat metabolic diseases such as, acid-base disturbances, albinism, amyloidosis, electrolyte disturbances, gout, inborn errors of metabolism (e.g., amino acid metabolism disorders, cholesterol and steroid metabolism disorders, eieosanoid metabolism disorder, fatty-aeid metabolism disorders, glycoprotein metabolism disorders, glycoprotein metabolism disorders, heme metabolism disorders, lipid metabolism disorders, lysosomal storage diseases, phospholipid metabolism disorders, proteoglycan metabolism disorders, TCA and ETC metabolism disorders),
  • metabolic diseases such as, acid-base disturbances, albinism, amyloidosis, electrolyte disturbances, gout, inborn errors of metabolism (e.g., amino acid metabolism disorders, cholesterol and steroid metabolism disorders, eieosanoid metabolism disorder, fatty-aeid metabolism disorders, glycoprotein metabolism disorders, glycoprotein metabolism disorders, heme metabolism disorders, lipid metabolism disorders, lysosomal storage diseases
  • lipid disorders e.g., lipid metabolism disorders and lipid storage disorders
  • obesity e.g., lipid metabolism disorders and lipid storage disorders
  • lipid disorders e.g., lipid metabolism disorders and lipid storage disorders
  • obesity e.g., lipid metabolism disorders and lipid storage disorders
  • Abnormal basal metabolic rate Danon disease
  • Exercise-associated hyponatremia High anion gap metabolic acidosis
  • MDP syndrome Methys syndrome
  • Metabolic myopathy Pansteatitis
  • Refeeding syndrome Winchester syndrome
  • X-linked hypophosphatemia X-linked hypophosphatemia
  • Deficiency Deficiency, Metachromatic leukodystrophy, Multiple sulfatase deficiency, Neuronal ceroid lipofuscinosis, Niemann-Pick disease (e.g., Niemann-Pick disease, SMPD 1 -associated or Niemann Pick disease, type C), Pelizaeus-Merzbacher disease, Sandhoff disease, Schindlcr disease, Sphingolipidoses, Sulfatidosis, Tay-Sachs disease, and Wolman disease.
  • Niemann-Pick disease e.g., Niemann-Pick disease, SMPD 1 -associated or Niemann Pick disease, type C
  • Pelizaeus-Merzbacher disease e.g., Niemann-Pick disease, SMPD 1 -associated or Niemann Pick disease, type C
  • Pelizaeus-Merzbacher disease e.g., Sandhoff disease, Schindlcr disease
  • Sphingolipidoses e.g., Sulfatidosis
  • the saRNA described herein may be used to treat lysosomal storage diseases such as Activator Deficiency/GM2 Gangliosidosis, Alpha-mannosidosis, Aspartylglucosaminuria, Cholesteryl ester storage disease, Chronic Hexosaminidase A
  • Galactosialidosis e.g., Gaucher Disease Type I, Gaucher Disease Type II, Gaucher Disease Type III
  • GM1 gangliosidosis e.g Infantile, Late infantile/Juvenile
  • the saRNA described herein may be used to treat nutritional diseases such as nutritional deficiencies, hyperalimentation and malnutrition,
  • the saRNA described herein may be used to treat diseases of the eye and adnexa such as disorders of the choroid and retina, congenital disorders of the eyes, disorders of the conjunctiva, disorders of the eyelid, lacrimal system and orbit, glaucoma, disorders of the iris and ciliary body, disorders of the lens, disorders of the ocular muscles, binocular movement, accommodation and refraction, ocular neoplasia, disorders of the optic nerve and visual pathways, periorbital conditions, disorders of the sclera and cornea, disorders of the vitreous body and globe, Actinic conjunctivitis, Acute hemorrhagic conjunctivitis, Acute posterior multifocal placoid pigment epitheliopathy, Acute zonal occult outer retinopathy, Amaurosis, Amaurosis fugax, Angioid streaks, Anisocoria, Anterior segment mesenchymal
  • Keratomalacia Khodadoust line, Krukenberg's spindle, Late congenital syphilitic oculopathy, Lattice degeneration, Lecithin cholesterol aeyltransferase deficiency, Leukoeoria, Limbal nodule, Loa loa fdariasis, Macropsia, Macular hole, Macular hypoplasia, Macular telangiectasia,
  • Sympathetic ophthalmia Synchysis spurtillans, Teleopsia, Tenonitis, Terrien's marginal degeneration, Terson syndrome, Toxic anterior segment syndrome, Trachoma, Trochleitis, Uveoparotitis, Vernal keratoconjunctivitis, Vision loss, Visual impairment, Visual looming syndrome, Viircorctinopathy.
  • Vitreous touch syndrome Vossius ring, Wagner's disease, Weill- Marchesani syndrome, White dot syndromes, cataract, persistent fetal vasculature, and X-l inked congenital stationary night blindness.
  • the saRNA described herein may be used to treat foot diseases such as immersion foot syndrome, trench foot, Achilles bursitis, Ainhum, Athlete's foot, Blue toe syndrome, Brachymetatarsia, Bunion, Burning feet syndrome, Calcaneal spur, Callus, Chilblains, Equinovalgus, Haglund's deformity, Haglund's syndrome, Hallux varus, Janeway lesion, Metatarsophalangeal joint sprain, Morton's neuroma, Morton's toe, Palmoplantar keratoderma, Plantar calcaneal bursitis, Plantar fasciitis, Plantar wart, Ship foot, Syndesmosis procedure, Tailor's bunion, Tarsal coalition, Tarsal tunnel syndrome and Toe walking,
  • foot diseases such as immersion foot syndrome, trench foot, Achilles bursitis, Ainhum, Athlete's foot, Blue toe syndrome, Brachymetatarsia, Bunion, Burn
  • the saRNA described herein may be used to treat genitourinary system disease such as gynecologic disorders, male genital disorders, urological conditions, bulbar urethral necrosis, Frasier syndrome and reproductive system disease.
  • genitourinary system disease such as gynecologic disorders, male genital disorders, urological conditions, bulbar urethral necrosis, Frasier syndrome and reproductive system disease.
  • the saRNA described herein may be used to treat gynecologic disorders such as congential disorders of the female genital organs, gynecologic neoplasia (e.g., gynecological cancer), infertility, inflammatory diseases of the female pelvic organ, noninflammatory disorders of the female genital tract (e.g., menstrual disorders) and sexually transmitted diseases and infections, Asherman's syndrome, Breakthrough bleeding, Cervical motion tenderness, Cryptomenorrhea, Dermoid cyst, Endosalpingiosis, Esthiomene, Female genital disease, Fitz-Hugh-Curtis syndrome, Gynecologic hemorrhage, Hematocolpos, Hyperestrogenism, Hypergonadism, Hypogonadism, Labial fusion, Lipschutz ulcer, Meigs' syndrome, Metrophlebitis, Mixed gonadal dysgenesis, Ovarian apoplexy
  • the saRNA described herein may be used to treat gynecological cancer such as Adenomyoma, Chorioblastoma, Chorioepithelioma,
  • Dysgerminoma Embryonal carcinoma, Endometrial intraepithelial neoplasia, Gestational choriocarcinoma, Gonadoblastoma, Hereditary leiomyomatosis and renal cell cancer syndrome, Mixed Mullcrian tumor, Mucinous tumor, Ovarian fibroma, Ovarian serous cystadenoma, Peutz- Jeghers syndrome, Polyembryoma, Thecoma, Trophoblastic neoplasm, Uterine fibroid, Vaginal intraepithelial neoplasia, and Vulvar intraepithelial neoplasia,
  • the saRNA described herein may be used to treat noninflammatory disorders of the female genital tract such as menstrual disorders, Adenomyosis, Asherman's syndrome, Atrophic vaginitis, Atypical polypoid adenomyoma, Cervical disorders
  • noninflammatory disorders of the female genital tract such as menstrual disorders, Adenomyosis, Asherman's syndrome, Atrophic vaginitis, Atypical polypoid adenomyoma, Cervical
  • Ovarian torsion Paraovarian cyst, Pelvic congestion syndrome, Persistent genital arousal disorder, Rectocele, Rectovaginal fistula, Retroverted uterus, Salpingitis isthmica nodosa, Sigmoidocele, Stenosis of uterine cervix, Theca lutein cyst, Urethrocele, Uterine prolapse, Vaginal bleeding, Vaginismus, Van Wyk and Grumbaeh syndrome and Vesicovaginal fistula.
  • the saRNA described herein may be used to treat male genital disorders such as congenital disorders of the male genital organs, epididymis disorders, male genital neoplasia (e.g., prostate cancer), penis disorders, prostate disorders, sexually transmitted diseases and infections and testicle disorders.
  • male genital disorders such as congenital disorders of the male genital organs, epididymis disorders, male genital neoplasia (e.g., prostate cancer), penis disorders, prostate disorders, sexually transmitted diseases and infections and testicle disorders.
  • the saRNA described herein may be used to treat male genital neoplasia such as Bowen's disease, Penile cancer, Choriocarcinoma, Embryoma,
  • Embryonal carcinoma Endodermal sinus tumor, Extramammary Paget's disease, Germ cell tumor, Germinoma, Gonadoblastoma, Granulosa cell tumor, Gynandroblastoma, High-grade prostatic intraepithelial neoplasia, Intratubular germ cell neoplasia, Leydig cell tumour, Mule spinners' cancer, Prostate cancer, Reinke crystals, Seminoma, Sertoli cell tumour, Sertoli-Leydig cell tumour, Sex cord-gonadal stromal tumour, Spermatocyte seminoma and Teratoma.
  • the saRNA described herein may be used to treat urological conditions such as Bladder stone, Emphysematous cystitis, Enuresis, Eosinophilic cystitis, Foumier gangrene, Hydrocele, Kidney stone, Loin pain hematuria syndrome, Lower urinary tract symptoms, Malakoplakia, Micturition syncope, Nephrogenic adenoma, Obstructive uropathy, Ovarian vein syndrome, Paruresis, Pelvic myoneuropathy, Peutz-Jeghers syndrome, Pneumaturia, Pseudodyssynergia, Purohit-Blaivas Staging System, Renal colic, Renal stone formation in space, Retroperitoneal fibrosis, Sedoanalgesia, Transurethral resection of the prostate syndrome, Urinary tract infection, Urinary tract obstruction, Urinoma, Urolithiasis, Uropathy, Vesieouretic reflux, and Zellweg
  • the saRNA described herein may be used to treat hair disease such as Alopecia areata, Alopecia totalis, Alopecia universalis, Androgenic alopecia, Black piedra, Bubble hair deformity, Hair disease, Hair follicle nevus, Hair loss, Hypertrichosis, Hypotrichosis, Kinking hair, Loose anagen syndrome, Monilethrix, Pili annulati, Pili torti, Plica neuropathica, Telogen effluvium, Tinea capitis and Trichotillomania.
  • hair disease such as Alopecia areata, Alopecia totalis, Alopecia universalis, Androgenic alopecia, Black piedra, Bubble hair deformity, Hair disease, Hair follicle nevus, Hair loss, Hypertrichosis, Hypotrichosis, Kinking hair, Loose anagen syndrome, Monilethrix, Pili annulati, Pili torti, Plica neuropathica, Telogen effluvium, Tinea capit
  • the saRNA described herein may be used to treat immune system disorder such as asthma, autoimmune diseases, chronic fatigue syndrome, human MHC mediate diseases, hypersensitivity, immunodeficiency lymphocytic immune system disorders, Aagenaes syndrome, Animal allergy, Arthus reaction, Asplenia, Autoimmune heart disease, Autosplenectomy, Caspase-8 deficiency state, Castleman's disease, Chronic fatigue syndrome, Cryofibrinogenemia, Cutaneous small-vessel vasculitis, Cytokine release syndrome, Cytokine storm, Dermic Morgan fold, Ectopic thymus, Extracutaneous mastocytoma, Familial
  • immune system disorder such as asthma, autoimmune diseases, chronic fatigue syndrome, human MHC mediate diseases, hypersensitivity, immunodeficiency lymphocytic immune system disorders, Aagenaes syndrome, Animal allergy, Arthus reaction, Asplenia, Autoimmune heart disease, Autosplenectomy, Caspase-8 deficiency state, Castleman'
  • Lymphocytopenia Mastocytosis, Mortimer's disease, Oral allergy syndrome, Paraneoplastic syndrome, RAS -associated autoimmune leukoproliferative disorder, Seabather's eruption, Systemic inflammatory response syndrome, T cell deficiency, Thymus hyperplasia, Transplant rejection and XMEN disease.
  • the saRNA described herein may be used to treat asthma such as Acute severe asthma, Aspirin-induced asthma, Asthmagen, Baker's asthma, Brittle asthma, Bronchial thermoplasty, Bronchospasm, Cough-variant asthma, Dynamic hyperinflation, Exercise-induced bronchoconstriction, Occupational asthma, and Reactive airway disease.
  • asthma such as Acute severe asthma, Aspirin-induced asthma, Asthmagen, Baker's asthma, Brittle asthma, Bronchial thermoplasty, Bronchospasm, Cough-variant asthma, Dynamic hyperinflation, Exercise-induced bronchoconstriction, Occupational asthma, and Reactive airway disease.
  • the saRNA described herein may be used to treat mouth diseases such as chronic sclerosing sialadenitis and sialadenitis.
  • the saRNA described herein may be used to treat musculoskeletal disorders such as chrondropathies, congenital disorders of the muscoskeletal system (e.g., arthrogryposis and phocemelia), crystal deposition diseases, joint disorders (e.g., gout), muscular disorders, myoneural junction and neuromuscular diseases (e.g., motor neuron disease and muscular dystrophy), osteopathies (e.g., osteitis and osteonecrosis), skeletal disorders (e.g., osseous and chrondromatous neoplasia), soi tissue disorders, systemic connective tissue disorders, Achard syndrome, Acropachy, Ankylosing hyperostosis, Arterial tortuosity syndrome, Attenuated patella alta, Baker's cyst, BlackBerry thumb, Bone cyst, Bone disease, Cervical spinal stenosis, Cervical spine disorder, Chondroealeinosis, Condylar
  • Pseudarthrosis Radial tunnel syndrome, Radicular pain, List of repetitive strain injury software, Repetitive strain injury, Saber shin, Sacroiliac joint dysfunction, Seal finger, Sever's disease, Snapping hip syndrome, Spinal decompression, Spinal enthesopathy, Spondyloarthropathy, Sternal cleft, Subungual exostosis, Synovial sarcoma, Tailor's bunion, Temporomandibular joint dysfunction, Tension myositis syndrome, Tietze syndrome, Traumatic spondylopathy, Ulnar nerve entrapment, Ulnar tunnel syndrome, Unicameral bone cyst, Winged scapula, Yoga foot drop and Zenker's paralysis.
  • the saRNA described herein may be used to treat neurological disorders such as epilepsy, headaches, sleep disorders, stroke, central nervous system disorders, peripheral nervous system disorders, Tay-Sachs disease, dyslexia, Tourettes syndrome, Acute flaccid myelitis, Adrenoleukodystrophy, Aicardi syndrome, Alexander disease, Amblyaudia. Asperger syndrome, User:ParanoidLemmings sandbox.
  • neurological disorders such as epilepsy, headaches, sleep disorders, stroke, central nervous system disorders, peripheral nervous system disorders, Tay-Sachs disease, dyslexia, Tourettes syndrome, Acute flaccid myelitis, Adrenoleukodystrophy, Aicardi syndrome, Alexander disease, Amblyaudia. Asperger syndrome, User:ParanoidLemmings sandbox.
  • the saRNA described herein may be used to treat central nervous system disorders such as brain disorders, demyelmating diseases of CNS and spinal cord disorders, Anterior horn disease, Cavernous sinus thrombosis, Central nervous system
  • the saRNA described herein may be used to treat brain disorders such as agnosia, aphasias, brain tumor, hypothalamus disorders, leukodystrophies, neurological brain disorder, pituitary disorder, Acquired brain injury, Acute cerebellar ataxia of childhood, Aqueductal stenosis, Basal ganglia disease, Brain abscess, Cerebellopontine angle syndrome, Cerebral amyloid angiopathy, Cerebral hypoxia, Cerebral softening, Cerebral vasospasm, Cerebritis, Cerebrospinal fluid leak, Childhood acquired brain injury, Cortical blindness, Cortical visual impairment, Encephalopathy, Ethylmalonie encephalopathy,
  • the saRNA described herein may be used to treat demyelinating diseases of CNS such as multiple sclerosis, CNS demyelinating autoimmune diseases, Adrenoleukodystrophy, Alexander disease, Alpers' disease, Balo concentric sclerosis, CAMFAK syndrome, Canavan disease, Central pontine myelinolysis, Experimental autoimmune encephalomyelitis, Hereditary CNS demyelinating disease, Krabbe disease,
  • demyelinating diseases of CNS such as multiple sclerosis, CNS demyelinating autoimmune diseases, Adrenoleukodystrophy, Alexander disease, Alpers' disease, Balo concentric sclerosis, CAMFAK syndrome, Canavan disease, Central pontine myelinolysis, Experimental autoimmune encephalomyelitis, Hereditary CNS demyelinating disease, Krabbe disease,
  • the saRNA described herein may be used to treat peripheral nervous system disorders such as cranial nerve disorders, nerve disorder, nerve root disorder, plexus disorders, PNS neoplasia, Accessory nerve disorder, Anesthesia dolorosa, Anti-MAG peripheral neuropathy, Autonomic dysreflexia, Axillary nerve dysfunction, Axillary nerve palsy, Charcot-Marie-Tooth disease, Chemotherapy-induced peripheral neuropathy, Chronic solvent- induced encephalopathy, CMV polyradieulomyelopathy, Congenital insensitivity to pain with anhidrosis, Denervation, Diabetic neuropathy, Dysautonomia, Facial nerve paralysis, Familial dysautonomia, Hereditary sensory and autonomic neuropathy, Hereditary sensory and autonomic neuropathy type I, Horner's syndrome.
  • peripheral nervous system disorders such as cranial nerve disorders, nerve disorder, nerve root disorder, plexus disorders, PNS neoplasia, Accessory nerve disorder, Anesthesia dolor
  • Nerve compression syndrome Paine compression syndrome, Nerve injury, Orthostatic hypotension, Orthostatic intolerance, Paroxysmal sympathetic hyperactivity, Peripheral neuropathy, Piriformis syndrome, Plexopathy, Polyneuropathy, Postural orthostatic tachycardia syndrome, Primary autonomic failure, Pronator teres syndrome, Proximal diabetic neuropathy, Pudendal nerve entrapment, Pure autonomic failure, Radial neuropathy, Radiculopathy, Sciatica, Thoracic outlet syndrome, Ulnar neuropathy, Vegetative-vascular dystonia, Villaret's syndrome, and Wallenberg's Syndrome.
  • the saRNA described herein may be used to treat orthopedic problems such as Articular cartilage damage, Bankart lesion, Blount's disease, Calcific tendinitis, Dactylitis, Diastematomyelia, Failed back syndrome, Flat feet, Hill-Sachs lesion, Knee pain, Larsen syndrome, Myelopathy, Perthes Lesion, Pigeon toe, Shoulder problem and Spondylitis.
  • the saRNA described herein may be used to treat respiratory diseases such as congenital disorders of the respiratory system, diseases of pleura (e.g., mesothelioma, pleura neoplasia), lower respiratory tract diseases (e.g., asthma, acute lower respiratory diseases, bronchus disorders, chronic lower respiratory disease, lung disorders), pulmonary heart disease and disease of pulmonary circulation, pulmonary lesion, pulmonary tumor, respiratory and cardiovascular disorders specific to the perinatal period, respiratory system neoplasia and upper respiratory track disease.
  • respiratory diseases such as congenital disorders of the respiratory system, diseases of pleura (e.g., mesothelioma, pleura neoplasia), lower respiratory tract diseases (e.g., asthma, acute lower respiratory diseases, bronchus disorders, chronic lower respiratory disease, lung disorders), pulmonary heart disease and disease of pulmonary circulation, pulmonary lesion, pulmonary tumor, respiratory and cardiovascular disorders specific to the perinatal period, respiratory system neoplasia and
  • the saRNA described herein may be used to treat lung disorders such as tuberculosis, Alcoholic lung disease, Alpha 1 -antitrypsin deficiency, Alveolar capillary dysplasia, Alveolar lung disease, Antisynthetase syndrome, Asbestosis, Aspergilloma, Atypical pulmonary carcinoid tumor, Baritosis, Brown induration, Cavitation (biology),
  • lung disorders such as tuberculosis, Alcoholic lung disease, Alpha 1 -antitrypsin deficiency, Alveolar capillary dysplasia, Alveolar lung disease, Antisynthetase syndrome, Asbestosis, Aspergilloma, Atypical pulmonary carcinoid tumor, Baritosis, Brown induration, Cavitation (biology),
  • Pulmonary aspiration Pulmonary capillary hemangiomatosis, Pulmonary contusion, Pulmonary cyst, Pulmonary fibrosis, Pulmonary hematoma, Pulmonary hemorrhage, Pulmonary hyperplasia, Pulmonary laceration, Pulmonary toxicity, Pulmonary-renal syndrome, Respiratory bronchiolitis interstitial lung disease, Restrictive lung disease, Rheumatoid lung disease, Sarcoidosis,
  • the saRNA described herein may be used to treat sexual disorders such as erectile dysfunction, persistent genital arousal disorder, sexual masochism disorder, sexual sadism disorder and spermatorrhea.
  • the saRNA described herein may be used to treat voice disorders such as Bogart-Bacall syndrome, chroditis, laryngitis, muteness, Reinke's edema, spasmodic dysphonia, vocal cord paresis, vocal fold cyst and vocal fold nodule.
  • voice disorders such as Bogart-Bacall syndrome, chroditis, laryngitis, muteness, Reinke's edema, spasmodic dysphonia, vocal cord paresis, vocal fold cyst and vocal fold nodule.
  • the saRNA described herein may be used to treat immune system disorders such as asthma, autoimmune diseases, chronic fatigue syndrome, human MHC mediate disease, hypersensitivity, immunodeficiency and lymphocytic immune system disorders.
  • the saRNA described herein may be used to treat autoimmune diseases such as Acute disseminated encephalomyelitis, Alopecia areata, Alopecia universalis, Ankylosing spondylitis, Antiphospholipid syndrome, Antisynthetase syndrome, Aplastic anemia, Apolipoprotein H, Arthritis mutilans, Atrophic gastritis, Autoimmune enteropathy, Autoimmune gastrointestinal dysmotility, Autoimmune hemolytic anemia, Autoimmune hypophysitis, Autoimmune inner ear disease, Autoimmune Oophoritis,
  • autoimmune diseases such as Acute disseminated encephalomyelitis, Alopecia areata, Alopecia universalis, Ankylosing spondylitis, Antiphospholipid syndrome, Antisynthetase syndrome, Aplastic anemia, Apolipoprotein H, Arthritis mutilans, Atrophic gastritis, Autoimmune enteropathy, Auto
  • Autoimmune pancreatitis Autoimmune polyendocrine syndrome, Autoimmune polyendocrine syndrome type 1, Autoimmune polyendocrine syndrome type 2, Autoimmune polyendocrine syndrome type 3, Autoimmune/inflammatory syndrome induced by adjuvants, Balo concentric sclerosis, Behcet's disease, BENTA disease, Bickerstaff s encephalitis, Bullous pemphigoid, Chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, Cicatricial pemphigoid, CNS demyelinating autoimmune diseases, Coeliae disease, Cogan syndrome, Cold agglutinin disease, CREST syndrome, Crohn's disease, Cutaneous manifestations of sarcoidosis, Dermatitis, Dermatomyositis, Diabetes Health, Diabetes mellitus type 1 , Drug-induced lupus erythematosus, Eosinophilic fasciitis, Erythema nodosum, Gestation
  • the saRNA described herein may be used to treat lymphoma such as Adult T-cell leukemia/ lymphoma, Aggressive lymphoma, AIDS-rclated lymphoma, ALK+ large B-cell lymphoma, Anaplastic large-cell lymphoma, Angioimmunoblastic T-cell lymphoma, B-eell CLL/lymphoma, B-cell lymphoma, Blastic NK cell lymphoma, Burkitt's lymphoma, CD30+ cutaneous T-cell lymphoma, Cutaneous B-cell lymphoma, Cutaneous lymphoma, Cutaneous T cell lymphoma, Diffuse large B-cell lymphoma, Enteropathy-associated T-cell lymphoma, Extranodal NK/T-cell lymphoma, Follicular large-cell lymphoma, Follicular lymphoma, Gastric lymphoma, Germinal center B
  • lymphoma such as
  • the saRNA described herein may be used to treat cancers such as Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML),
  • Adrenocortical Carcinoma e.g., Kaposi Sarcoma and AIDS-Related Lymphoma
  • Anal Cancer e.g., Kaposi Sarcoma and AIDS-Related Lymphoma
  • Anal Cancer e.g., Kaposi Sarcoma and AIDS-Related Lymphoma
  • Anal Cancer e.g., Kaposi Sarcoma and AIDS-Related Lymphoma
  • Anal Cancer e.g., Kaposi Sarcoma and AIDS-Related Lymphoma
  • Anal Cancer e.g., Anal Cancer
  • Appendix Cancer e.g., Astrocytomas, Childhood, Atypical
  • Tumor Teratoid/Rhabdoid Tumor, Childhood, Central Nervous System, Basal Cell Carcinoma , Extrahepatic Bile Duct Cancer , Bladder Cancer, Bone Cancer (e.g., Ewing Sarcoma Family of Tumors, Osteosarcoma and Malignant Fibrous Histiocytoma), Brain Stem Glioma, Childhood, Brain Tumor (e.g., Astrocytoma, Brain Stem Glioma, Central Nervous System Atypical
  • Teratoid/Rhabdoid Tumor Central Nervous System Embryonal Tumors, Central Nervous System Germ Cell Tumors, Craniopharyngioma, Ependymoma), Breast Cancer , Bronchial Tumors, Burkitt Lymphoma , Carcinoid Tumor (e.g., childhood, Gastrointestinal), Carcinoma of Unknown Primary, Cardiac (Heart) Tumors, Childhood, Central Nervous System (e.g., Atypical Teratoid/Rhabdoid Tumor, Embryonal Tumors, Germ Cell Tumor and Lymphoma), Cervical Cancer, Chordoma, Childhood, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Myeloproliferative Neoplasms, Colon Cancer, Colorectal Cancer, Craniopharyngioma, Cutaneous T-Cell Lymphoma , Duct, Bile, Extrahepatic, Du
  • Ependymoma Esophageal Cancer, Esthesi ⁇ neuroblastoma, Childhood, Ewing Sarcoma, Extracranial Germ Cell Tumor, Childhood, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer (e.g., Intraocular Melanoma and Retinoblastoma), Fallopian Tube Cancer, Fibrous Histiocytoma of Bone, Malignant, and Osteosarcoma, Gallbladder Cancer, Gastric (Stomach) Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Stromal Tumors (GIST), Germ Cell Tumor (e.g., Central Nervous System, Childhood, Extracranial, Childhood, Extragonadal, Ovarian, Testicular), Gestational Trophoblastic Disease, Glioma , Hairy Cell Leukemia, Head and Neck Cancer, Heart Cancer, Hepatocellular (Liver) Cancer, Histiocytosis, Langerhans
  • Transitional Cell Cancer of the Renal Pelvis and Ureter Transitional Cell Cancer of the Renal Pelvis and Renal Pelvis, Urethral Cancer, Endometrial Uterine Cancer , Uterine Sarcoma, Vaginal Cancer, Vulvar Cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.
  • Additional disease and disorders which may be treated using the saRNA of the present invention including, but are not limited to, Pel-Ebstein fever, plasma cell dyscrasia, plasmacytoma, smouldering myeloma, Waldenstrom's macroglobulinemia, Chromosome 5q deletion syndrome, Myelofibrosis, Refractory anemia, Refractory cytopenia, Sideroblastic anemia, Bing-Neel syndrome, Actinic granuloma, Annular elastolytic giant-cell granuloma, Benign cephalic histiocytosis, Birbeck granules, Congenital self-healing reticulohistiocytosis, Darier-Roussy disease, Darier-Roussy saroid, Eruptive histiocytoma, Generalized eruptive histiocytoma, Generalized franuloma annulare, Giant cell elastophago
  • kits for conveniently and/or effectively carrying out methods of the present invention.
  • kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
  • kits for regulate the expression of genes in vitro or in vivo comprising saRNA of the present invention or a combination of saRNA of the present invention, saRNA modulating other genes, siRNAs, miRNAs or other oligonucleotide molecules.
  • the kit may fiirther comprise packaging and instructions and/or a delivery agent to form a formulation composition.
  • the delivery agent may comprise a saline, a buffered solution, a lipidoid, a dendrimer or any delivery agent disclosed herein.
  • Non-limiting examples of genes are described herein in Table 1 .
  • kits comprising saRNA described herein may be used with proliferating cells to show efficacy.
  • the buffer solution may include sodium chloride, calcium chloride, phosphate and/or EDTA.
  • the buffer solution may include, but is not limited to, saline, saline with 2mM calcium, 5% sucrose, 5% sucrose with 2mM calcium, 5% Mannitol, 5% Mannitol with 2mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2mM calcium and mannose (See U.S. Pub, No. 20120258046; herein incorporated by reference in its entirety).
  • the buffer solutions may be precipitated or it may be lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of saRNA in the buffer solution over a period of time and/or under a variety of conditions.
  • the present invention provides for devices which may incorporate saRNA of the present invention. These devices contain in a stable formulation available to be immediately delivered to a subject in need thereof, such as a human patient,
  • Non-limiting examples of the devices include a pump, a catheter, a needle, a transdermal patch, a pressurized olfactory delivery device, iontophoresis devices, multi-layered microfluidic devices.
  • the devices may be employed to deliver saRNA of the present invention according to single, multi- or split-dosing regiments.
  • the devices may be employed to deliver saRNA of the present invention across biological tissue, intradermal, subcutaneously, or intramuscularly. More examples of devices suitable for delivering oligonucleotides are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
  • Administered in combination means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. In some embodiments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently close together such that a combinatorial (e.g., a synergistic) effect is achieved.
  • a combinatorial e.g., a synergistic
  • amino acid As used herein, the terms "amino acid” and “amino acids” refer to all naturally occurring L-alpha-amino acids.
  • the amino acids are identified by either the onc- letter or three-letter designations as follows: aspartic acid (Asp:D), isoleucine (Ile:I), threonine (ThnT), leucine (Leu:L), serine (Ser:S), tyrosine (Tyr:Y), glutamic acid (Glu:E), phenylalanine (Phe:F), proline (Pro:P), histidine (His:H), glycine (Gly:G), lysine (Lys:K), alanine (Ala: A), arginine (Arg:R), cysteine (Cys:C), tryptophan (Trp:W), valine (Val:V), glutamine (Gln:Q) methionine (Met:M), asparagines (Asp:D
  • animal refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans at any stage of development. In some embodiments, “animal” refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
  • mammal e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig.
  • animals include, but are not limited to, mammals,
  • linking agent when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions.
  • An “association” need not be strictly through direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity sufficiently stable such that the "associated" entities remain physically associated.
  • bifunctional refers to any substance, molecule or moiety which is capable of or maintains at least two functions.
  • the functions may affect the same outcome or a different outcome.
  • the structure that produces the function may be the same or different.
  • bifunctional saRNA of the present invention may comprise a cytotoxic peptide (a first function) while those nucleosides which comprise the saRNA are, in and of themselves, cytotoxic (second function),
  • Biocompatible As used herein, the term “biocompatible” means compatible with living cells, tissues, organs or systems posing little to no risk of injury, toxicity or rejection by the immune system.
  • Biodegradable As used herein, the term “biodegradable” means capable of being broken down into innocuous products by the action of living things.
  • biologically active refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active.
  • the saRNA of the present invention may be considered biologically active if even a portion of the saRNA is biologically active or mimics an activity considered biologically relevant.
  • cancer in an individual refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Often, cancer cells will be in the form of a tumor, but such cells may exist alone within an individual, or may circulate in the blood stream as independent cells, such as leukemic cells.
  • Cell growth is principally associated with growth in cell numbers, which occurs by means of cell reproduction (i.e. proliferation) when the rate of the latter is greater than the rate of cell death (e.g. by apoptosis or necrosis), to produce an increase in the size of a population of cells, although a small component of that growth may in certain circumstances be due also to an increase in cell size or cytoplasmic volume of individual cells.
  • An agent that inhibits cell growth can thus do so by either inhibiting proliferation or stimulating cell death, or both, such that the equilibrium between these two opposing processes is altered.
  • Celt type refers to a cell from a given source (e.g., a tissue, organ) or a cell in a given state of differentiation, or a cell associated with a given pathology or genetic makeup.
  • Chromosome As used herein, the term “chromosome” refers to an organized structure of DNA and protein found in cells.
  • nucleic acids refers to hybridization or base pairing between nucleotides or nucleic acids, such as, for example, between the two strands of a double-stranded DNA molecule or between an oligonucleotide probe and a target are complementary.
  • Condition refers to the status of any cell, organ, organ system or organism. Conditions may refect a disease state or simply the physiologic presentation or situation of an entity. Conditions may be characterized as phenotypic conditions such as the macroscopic presentation of a disease or genotypic conditions such as the underlying gene or protein expression profiles associated with the condition. Conditions may be benign or malignant.
  • Controlled Release refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome
  • Cytostatic refers to inhibiting, reducing, suppressing the growth, division, or multiplication of a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
  • Cytotoxic refers to killing or causing injurious, toxic, or deadly effect on a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
  • Delivery refers to the act or manner of delivering a compound, substance, entity, moiety, cargo or payload.
  • Delivery Agent refers to any substance which facilitates, at least in part, the in vivo delivery of an saRNA of the present invention to targeted cells.
  • Delivery agent refers to any substance which facilitates, at least in part, the in vivo delivery of an saRNA of the present invention to targeted cells.
  • Destabilized As used herein, the term “destable,” “destabilize,” or “destabilizing region” means a region or molecule that is less stable than a starting, wild-type or native form of the same region or molecule.
  • Detectable label refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity that is readily detected by methods known in the art including radiography, fluorescence, chemiluminescence, enzymatic activity, absorb ancc and the like. Detectable labels include radioisotopes, fluorophores, chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin, streptavidin and haptens, quantum dots, and the like. Detectable labels may be located at any position in the oligonucleotides disclosed herein. They may be within the nucleotides or located at the 5' or 3' terminus.
  • Encapsulate As used herein, the term “encapsulate” means to enclose, surround or encase.
  • Engineered As used herein, embodiments of the invention are “engineered” when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule,
  • Equivalent subject may be e.g. a subject of similar age, sex and health such as liver health or cancer stage, or the same subject prior to treatment according to the invention.
  • the equivalent subject is "untreated” in that he does not receive treatment with an saRNA according to the invention. However, he may receive a conventional anti-cancer treatment, provided that the subject who is treated with the saRNA of the invention receives the same or equivalent conventional anti-cancer treatment.
  • Exosome is a vesicle secreted by mammalian cells.
  • expression refers to one or more of the following events: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3 ' end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post-translational modification of a polypeptide or protein.
  • Feature refers to a characteristic, a property, or a distinctive element.
  • Formulation includes at least one saRNA of the present invention and a delivery agent.
  • fragment refers to a portion.
  • fragments of proteins may comprise polypeptides obtained by digesting full-length protein isolated from cultured cells.
  • Fragments of oligonucleotides may comprise nucleotides, or regions of nucleotides.
  • a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
  • Gene refers to a nucleic acid sequence that comprises control and most often coding sequences necessary for producing a polypeptide or precursor. Genes, however, may not be translated and instead code for regulatory or structural RNA molecules.
  • a gene may be derived in whole or in part from any source known to the art, including a plant, a fungus, an animal, a bacterial genome or episome, eukaryotic, nuclear or plasmid DNA, cDNA, viral DNA, or chemically synthesized DNA.
  • a gene may contain one or more modifications in either the coding or the untranslated regions that could affect the biological activity or the chemical structure of the expression product, the rate of expression, or the manner of expression control. Such modifications include, but are not limited to, mutations, insertions, deletions, and substitutions of one or more nucleotides.
  • the gene may constitute an uninterrupted coding sequence or it may include one or more introns, bound by the appropriate splice junctions.
  • Gene expression refers to the process by which a nucleic acid sequence undergoes successful transcription and in most instances translation to produce a protein or peptide.
  • measurements may be of the nucleic acid product of transcription, e.g., RNA or mRNA or of the amino acid product of translation, e.g., polypeptides or peptides. Methods of measuring the amount or levels of RNA, mRNA, polypeptides and peptides are well known in the art.
  • Genome The term “genome” is intended to include the entire DNA complement of an organism, including the nuclear DNA component, chromosomal or extrachromosomal DNA, as well as the cytoplasmic domain (e.g., mitochondrial DNA).
  • homology As used herein, the term “homology” refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some
  • polymeric molecules are considered to be "homologous” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar.
  • the term “homologous” necessarily refers to a comparison between at least two sequences (polynucleotide or polypeptide sequences).
  • two polynucleotide sequences are considered to be homologous if the polypeptides they encode are at least about 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids.
  • homologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids.
  • homology is determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids.
  • two protein sequences are considered to be homologous if the proteins are at least about 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least about 20 amino acids.
  • hyperproliferative cell may refer to any cell that is proliferating at a rate that is abnormally high in comparison to the proliferating rate of an equivalent healthy cell (which may be referred to as a "control").
  • An "equivalent healthy” cell is the normal, healthy counterpart of a cell. Thus, it is a cell of the same type, e.g. from the same organ, which performs the same functions(s) as the comparator cell. For example, proliferation of a hyperproliferative hepatocyte should be assessed by reference to a healthy hepatocyte, whereas proliferation of a hyperproliferative prostate cell should be assessed by reference to a healthy prostate cell.
  • an "abnormally high" rate of proliferation it is meant that the rate of proliferation of the hyperproliferative cells is increased by at least 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80%, as compared to the proliferative rate of equivalent, healthy (non-hyperproliferative) cells.
  • the "abnormally high” rate of proliferation may also refer to a rate that is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, compared to the proliferative rate of equivalent, healthy cells.
  • Hyperproliferative disorder may be any disorder which involves hyperproliferative cells as defined above.
  • hyperproliferative disorders include neoplastic disorders such as cancer, psoriatic arthritis, rheumatoid arthritis, gastric hyperproliferative disorders such as inflammatory bowel disease, skin disorders including psoriasis, Reiter's syndrome, pityriasis rubra pilaris, and
  • hyperproliferative cell The presence of hyperproliferative cells within an animal may be identifiable using scans such as X- rays, MRI or CT scans.
  • the hyperproliferative cell may also be identified, or the proliferation of cells may be assayed, through the culturing of a sample in vitro using cell proliferation assays, such as MTT, XTT. MTS or WST-1 assays.
  • Cell proliferation in vitro can also be determined using flow cytometry,
  • Identity refers to the overall relatedness between polymeric molecules, e.g., between oligonucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g.
  • gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes), hi certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%», at least 50%», at least 60%, at least 70%», at least 80%», at least 90%, at least 95%>, or 100%» of the length of the reference sequence.
  • the nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M , ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W.. cd..
  • the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CAB IDS, 1989, 4: 1 1 - 17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H , and Lipman, D., SIAM J Applied Math., 48:1073 (1988); incorporated herein by reference.
  • Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et at, Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altsehul, S. F. et al., J. Molec. Biol, 215, 403 (1990)).
  • Inhibit expression of a gene means to cause a reduction in the amount of an expression product of the gene.
  • the expression product can be an RNA transcribed from the gene (e.g., an mRNA) or a polypeptide translated from an mRNA transcribed from the gene.
  • a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom.
  • the level of expression may be determined using standard techniques for measuring mRNA or protein.
  • vitro' 7 refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (eg., animal, plant, or microbe).
  • in vivo refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof).
  • Isolated refers to a substance or entity that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated.
  • isolated agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure.
  • a substance is "pure” if it is substantially free of other components.
  • substantially isolated By “substantially isolated” is meant that the compound is substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • Label refers to a substance or a compound which is incorporated into an object so that the substance, compound or object may be detectable.
  • Linker refers to a group of atoms, e.g., 10-1 ,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine.
  • the linker can be attached to a modified nucleoside or nucleotide on the nucleobase or sugar moiety at a first end, and to a payload, e.g., a detectable or therapeutic agent, at a second end.
  • the linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence.
  • the linker can be used for any useful purpose, such as to form saRNA conjugates, as well as to administer a payload, as described herein.
  • linker examples include, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, thioether, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein.
  • linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or tetraethylene glycol), and dextran polymers and derivatives thereof.
  • a selectively cleavable bond include an aniido bond can be cleaved for example by the use of tris(2 -carboxy ethy l)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond can be cleaved for example by acidic or basic hydrolysis,
  • Metastasis means the process by which cancer spreads from the place at which it first arose as a primary tumor to distant locations in the body. Metastasis also refers to cancers resulting from the spread of the primary tumor. For example, someone with breast cancer may show metastases in their lymph system, liver, bones or lungs.
  • Modified refers to a changed state or structure of a molecule of the invention. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the saRNAs of the present invention are modified by the introduction of non-natural nucleosides and/or nucleotides.
  • Naturally occurring As used herein, “naturally occurring” means existing in nature without artificial aid.
  • nucleic acid refers to a molecule comprised of one or more nucleotides, i.e., ribonucleotides, deoxyribonucleotides, or both.
  • the term includes monomers and polymers of ribonucleotides and deoxyribonucleotides, with the ribonucleotides and/or deoxyribonucleotides being bound together, in the case of the polymers, via 5' to 3' linkages.
  • the ribonucleotide and deoxyribonucleotide polymers may be single or double-stranded.
  • linkages may include any of the linkages known in the art including, for example, nucleic acids comprising 5' to 3' linkages.
  • the nucleotides may be naturally occurring or may be synthetically produced analogs that are capable of forming base-pair relationships with naturally occurring base pairs.
  • Examples of non-naturally occurring bases that are capable of forming base-pairing relationships include, but are not limited to, aza and deaza pyrimidine analogs, aza and deaza purine analogs, and other heterocyclic base analogs, wherein one or more of the carbon and nitrogen atoms of the pyrimidine rings have been substituted by heteroatoms, e.g., oxygen, sulfur, selenium, phosphorus, and the like.
  • patient refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained professional for a particular disease or condition.
  • Peptide As used herein, "peptide” is less than or equal to 50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
  • compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • compositions refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
  • Excipients may include, for example; antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration.
  • excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmeliose, crosslmked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose,
  • BHT butylated hydroxytoluene
  • calcium carbonate calcium phosphate (dibasic)
  • calcium stearate calcium stearate
  • croscarmeliose crosslmked polyvinyl pyrrolidone
  • citric acid crospovidone
  • cysteine ethylcellulose
  • gelatin hydroxypropyl cellulose
  • compositions described herein also includes pharmaceutically acceptable salts of the compounds described herein. As used herein,
  • “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid).
  • suitable organic acid examples include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsuifate, ethanesuifonate, fiimarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride,
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium,
  • the pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 17 th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth (eds.), Wiley- VCH, 2008, and Berge et al, Journal of Pharmaceutical Science, 66, 1 - 19 (1977), each of which is incorporated herein by reference in its entirety.
  • solvate means a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered.
  • solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-.
  • N-methylpyrrolidinone NMP
  • dimethyl sulfoxide DMSO
  • DMF dimethylformamide
  • DMAC ⁇ , ⁇ '-dimethylacetamide
  • DMEU l ,3-dimethyl-2-imidazolidmone
  • DMPU l ,3-dimethyl-3,4,5,6-tetrahydro-2-(lH)-pyrimidinone
  • ACN acetonitrile
  • propylene glycol ethyl acetate
  • benzyl alcohol 2-pyrrol idone. benzyl benzoate, and the like.
  • the solvent the solvate is referred to as a "hydrate.”
  • Pharmacologic effect is a measurable biologic phenomenon in an organism or system which occurs after the organism or system has been contacted with or exposed to an exogenous agent. Pharmacologic effects may result in therapeutically effective outcomes such as the treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset of disease, disorder, condition or infection. Measurement of such biologic phenomena may be quantitative, qualitative or relative to another biologic phenomenon. Quantitative measurements may be statistically significant. Qualitative measurements may be by degree or kind and may be at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more different. They may be observable as present or absent, better or worse, greater or less.
  • Exogenous agents when referring to pharmacologic effects are those agents which are, in whole or in part, foreign to the organism or system. For example, modifications to a wild type biomolecule, whether structural or chemical, would produce an exogenous agent. Likewise, incorporation or combination of a wild type molecule into or with a compound, molecule or substance not found naturally in the organism or system would also produce an exogenous agent.
  • the saRNA of the present invention comprises exogenous agents.
  • pharmacologic effects include, but are not limited to, alteration in cell count such as an increase or decrease in neutrophils, reticulocytes, granulocytes, erythrocytes (red blood cells), megakaryocytes, platelets, monocytes, connective tissue macrophages, epidermal langerhans cells, osteoclasts, dendritic cells, microglial cells, neutrophils, eosinophils, basophils, mast cells, helper T cells, suppressor T cells, cytotoxic T cells, natural killer T cells, B cells, natural killer cells, or reticulocytes.
  • Pharmacologic effects also include alterations in blood chemistry, pH, hemoglobin, hematocrit, changes in levels of enzymes such as, but not limited to, liver enzymes AST and ALT, changes in lipid profiles, electrolytes, metabolic markers, hormones or other marker or profile known to those of skill in the art,
  • Physicochemical As used herein, "physicoehemieal" means of or relating to a physical and/or chemical property.
  • the term "preventing” refers to partially or completely delaying onset of an infection, disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.
  • Prodrug The present disclosure also includes prodrugs of the compounds described herein.
  • prodrugs refer to any substance, molecule or entity which is in a form predicate for that substance, molecule or entity to act as a therapeutic upon chemical or physical alteration. Prodrugs may by covalently bonded or sequestered in some way and which release or are converted into the active drug moiety prior to, upon or after administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively.
  • Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol, 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
  • Prognosing means a statement or claim that a particular biologic event will, or is very likely to, occur in the future.
  • Progression As used herein, the term “progression” or “cancer progression” means the advancement or worsening of or toward a disease or condition.
  • Proliferate As used herein, the term “proliferate” means to grow, expand or increase or cause to grow, expand or increase rapidly, “Proliferative” means having the ability to proliferate. " Anti-pro liferative” means having properties counter to or inapposite to proliferative properties.
  • Protein means a polymer of amino acid residues linked together by peptide bonds.
  • a protein may be naturally occurring, recombinant, or synthetic, or any combination of these.
  • a protein may also comprise a fragment of a naturally occurring protein or peptide, A protein may be a single molecule or may be a multi-molecular complex.
  • the term protein may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analogue of a corresponding naturally occurring amino acid.
  • Protein expression refers to the process by which a nucleic acid sequence undergoes translation such that detectable levels of the amino acid sequence or protein are expressed.
  • Purified means to make substantially pure or clear from unwanted components, material defilement, admixture or imperfection.
  • regression As used herein, the term “regression” or “degree of regression” refers to the reversal, either phenotypically or genotypicaily, of a cancer progression. Slowing or stopping cancer progression may be considered regression.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Plant Pathology (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Transplantation (AREA)
  • Cardiology (AREA)
  • Endocrinology (AREA)
  • Dermatology (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Heart & Thoracic Surgery (AREA)

Abstract

The invention relates to oligonucleotides, e.g., saRNAs useful in upregulating the expression of a target gene and therapeutic compositions comprising such oligonucleotides. Methods of using the oligonucleotides and the therapeutic compositions are also provided.

Description

SARNA COMPOSITIONS AND METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to US Prov. Application No. 62/150,892 filed
Apr. 22, 2015, US Prov. Application No. 62/150,893 filed Apr. 22, 2015, and US Prov.
Application No. 62/150,897 filed Apr. 22, 2015 , US Prov. Application No. 62/150,895 filed Apr. 22, 2015, US Prov. Application No. 62/150,900 filed Apr. 22, 2015, US Prov. Application No. 62/150,904 filed Apr. 22, 2015, and US Prov. Application No. 62/150,908 filed Apr. 22, 2015, the contents of each of which are incorporated herein by reference in their entirety.
REFERENCE TO SEQUENCE LISTING
[0002] The instant application contains a "lengthy" Sequence Listing which has been filed electronically as a sequence listing text file and is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[0003] The invention relates to oligonucleotides, specifically saRNA, compositions for modulating target gene expression and to the methods of using the compositions in diagnostic and therapeutic applications.
BACKGROUND OF THE INVENTION
[0004] Recently it has been discovered that short RNAs can regulate transcription by destructing transcripts that are sense or antisense to a given niRNA and which are presumed to be non-coding transcripts. Destruction of the non-coding transcripts which are sense, or identical to, the given mRNA results in transcriptional repression of that mRNA, whereas destruction of the non-coding transcripts which are antisense to the given mRNA results in transcriptional activation and/or increased expression of the mRNA or the level of protein encoded by the mRNA. By targeting such non-coding transcripts, short RNAs can therefore be used to up- regulate specific genes at either the nucleic acid or protein. Small duplex RNAs have also been discovered to increase gene expression by targeting ncRNAs that overlap gene promoters (Janowski et al., Nature Chemical Biology, vol.3: 166-173 (2007), the contents of which are incorporated herein by reference in their entirety).
[ 0005] Any short RNA which leads to up-regulation of the expression of a target gene by any mechanism is termed a short activating RNA or small activating RNA (saRNA). Known methods of up-regulating a target gene by use of saRNAs can involve the detection of an RNA transcript which is antisense to the target gene of interest and designing short RNA molecules which down-regulate the identified transcript. For instance, US Patent 8,288,354 to Wahlestedt, the contents of which are incorporated herein by reference in their entirety, discloses a method of modulating expression of a target gene comprising targeting a nucleic acid molecule to a naturally-occurring anti-sense transcript (NAT) of a sense strand of the targeted gene in a target cell, wherein the nucleic acid molecule targeting the NAT is complementary to the NAT. The NAT may be a coding RNA transcript or a non-coding RNA transcript lacking any extensive open reading frame. In another example, WO 2012/065143 to Krieg et al., the contents of which are incorporated herein by reference in their entirety, teaches a method of activating expression of a target gene comprising blocking the binding of a long non-coding RNA (lnc-RNA) to Polycomb repressive complex 2 (PRC2) protein by a single-stranded oligonucleotide, thereby preventing the lnc-RNA from suppressing the target gene.
[0006] There remains, however, a need for compositions and methods for the targeted modulation of genes via activation with saRNA which do not require the a priori identification of a NAT or rely on interactions at the polycomb complex for prophylactic diagnostic and/or therapeutic purposes.
BRIEF DESCRI PTION OF THE DRAWINGS
[ 0007] The foregoing and other objects, features and advantages will be apparent from the following description of particular embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of various embodiments of the invention.
[0008] FIG. 1 is a schematic illustrating the relationships among the nucleic acid moieties involved in the function of an saRNA of the invention.
SUMMARY OF THE INVENTION
[ 0009] The present invention provides compositions, methods and kits for the design, preparation, manufacture, formulation and/or use of short (or small) activating RNA (saRNA) that modulates target gene expression and/or function for therapeutic purposes, including diagnosing and prognosis. [0010] It is understood by those of skill in the art that the term saRNA may refer to a single saRNA or saRNA in the plural (saRNAs).
[0011] One aspect of the invention provides an saRNA that targets an antisense RNA transcript of a target gene. The antisense RNA transcript of the target gene is referred to thereafter as the target antisense RNA transcript. The target antisense RNA transcript is transcribed from the coding strand of the target gene.
[0012] Another aspect of the invention provides a pharmaceutical composition comprising an saRNA that targets an antisense RNA transcript of a target gene and at least one pharmaceutically acceptable excipient, wherein the expression of the target gene is up-regulated.
[0013] Another aspect of the invention provides a method of modulating the expression of a target gene comprising administering an saRNA that targets an antisense RNA transcript of the target gene
[0014] Another aspect of the invention provides treating or preventing a disease comprising administering an saRNA that targets an antisense RNA transcript of a target gene, wherein the target gene is associated with the disease.
[0015] The details of various embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and the drawings, and from the claims.
DETAILED DESCRIPTION
[0016] The present invention provides compositions, methods and kits for modulating target gene expression and/or function for therapeutic purposes. These compositions, methods and kits comprise at least one saRNA that upregulates the expression of a target gene.
L Design and Synthesis of saRNA
[0017] One aspect of the present invention provides a method to design and synthesize saRNA.
[0018] The terms "small activating RNA", "short activating RNA", or "saRNA" in the context of the present invention means a single-stranded or double-stranded RNA that upregulates or has a positive effect on the expression of a specific gene. The saRNA may be single-stranded of 14 to 30 nucleotides. The saRNA may also be double-stranded, each strand comprising 14 to 30 nucleotides. The gene is called the target gene of the saRNA. As used herein, the target gene is a double-stranded DNA comprising a coding strand and a template strand. For example, an saRNA that upregulates the expression of the A3GALT2 gene is called an "A3 GALT2 -saRNA" and the A3GALT2 gene is the target gene of the A3GALT2 -saRNA. A target gene may be any gene of interest. In some embodiments, a target gene has a promoter region on the template strand.
[ 0019] By "upregulation" or "activation" of a gene is meant an increase in the level of expression of a gene, or levels of the polypeptide(s) encoded by a gene or the activity thereof, or levels of the RNA transcript(s) transcribed from the template strand of a gene above that observed in the absence of the saRNA of the present invention. The saRNA of the present invention may have a direct upregulating effect on the expression of the target gene.
[0020] The saRNAs of the present invention may have an indirect upregulating effect on the RNA transcript(s) transcribed from the template strand of the target gene and/or the polypeptide(s) encoded by the target gene or mRNA. The RNA transcript transcribed from the target gene is referred to thereafter as the target transcript. The target transcript may be an mRNA of the target gene. The target transcript may exist in the mitochondria. The saRNAs of the present invention may have a downstream effect on a biological process or activity. In such embodiments, an saRNA targeting a first transcript may have an effect (either upregulating or downregulating) on a second, non-target transcript.
[0021] In one embodiment, the saRNA of the present invention may show efficacy in proliferating cells. As used herein with respect to cells, "proliferating" means cells which are growing and/or reproducing rapidly.
Target antisense RNA transcript of a target gene
[0022] In one embodiment, the saRNAs of the present invention is designed to be complementary to a target antisense RNA transcript of a target gene, and it may exert its effect on the target gene expression and/or function by down-regulating the target antisense RNA transcript. The target antisense RNA transcript is transcribed from the coding strand of the target gene and may exist in the nucleus of a cell.
[ 0023 ] The term "complementary to5' in the context means being able to hybridize with the target antisense RNA transcript under stringent conditions.
[0024] The term "antisense" when used to describe a target antisense RNA transcript in the context of the present invention means that the sequence is complementary to a sequence on the coding strand of a gene. [0025] It is to be understood that thymidine of the DNA is replaced by uridine in RNA and that this difference does not alter the understanding of the terms "antisense" or
"complementarity" .
| (M)26| The target antisense RNA transcript may be transcribed from a locus on the coding strand between up to 100, 80, 60, 40, 20 or 10 kb upstream of a location corresponding to the target gene's transcription start site (TSS) and up to 100, 80, 60, 40, 20 or 10 kb downstream of a location corresponding to the target gene's transcription stop site.
[0027] In one embodiment, the target antisense RNA transcript is transcribed from a locus on the coding strand located within +/- 1 kb of the target gene's transcription start site.
[0028] In another embodiment, the target antisense RNA transcript is transcribed from a locus on the coding strand located within +/- 500 nt, +/- 250 nt, +/- 100 nt, +/- 10 nt, +/- 5 nt or +/- 1 nt of the target gene's transcription start site.
[0029] In another embodiment, the target antisense RNA transcript is transcribed from a locus on the coding strand located +/- 2000 nucleotides of the target gene's transcription start site.
[0030] In another embodiment, the locus on the coding strand is no more than 1000 nucleotides upstream or downstream from a location corresponding to the target gene's transcription start site.
[0031] In another embodiment, the locus on the coding strand is no more than 500 nucleotides upstream or downstream from a location corresponding to the target gene's transcription start site.
[0032] The term "transcription start site" (TSS) as used herein means a nucleotide on the template strand of a gene corresponding to or marking the location of the start of
transcription. The TSS may be located within the promoter region on the template strand of the gene.
[0033] The term "transcription stop site" as used herein means a region, which can be one or more nucleotides, on the template strand of a gene, which has at least one feature such as, but not limited to, a region which encodes at least one stop codon of the target transcript, a region encoding a sequence preceding the 3 'UTR of the target transcript, a region where the RNA polymerase releases the gene, a region encoding a splice site or an area before a splice site and a region on the template strand where transcription of the target transcript terminates. [0034] The phrase "is transcribed from a particular locus" in the context of the target antisense RNA transcript of the invention means the transcription of the target antisense RNA transcript starts at the particular locus.
[0035] The target antisense RNA transcript is complementary to the coding strand of the genomic sequence of the target gene, and any reference herein to "genomic sequence" is shorthand for "coding strand of the genomic sequence".
[0036] The "coding strand" of a gene has the same base sequence as the mRNA produced, except T is replayed by U in the mRNA. The "template strand" of a gene is therefore complementary and antiparallel to the mRNA produced.
[0037] Thus, the target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence located between 100, 80, 60, 40, 20 or 10 kb upstream of the target gene's transcription start site and 100, 80, 60, 40, 20 or 10 kb downstream of the target gene's transcription stop site.
[0038] In one embodiment, the target antisense RNA transcript comprises a sequence which is complementary to a genomic sequence located between 1 kb upstream of the target gene's transcription start site and 1 kb downstream of the target gene's transcription stop site.
[0039] In another embodiment, the target antisense RNA transcript comprises a sequence which is complementary to a genomic sequence located between 500, 250, 100, 10, 5 or 1 nucleotide upstream of the target gene's transcription start site and ending 500, 250, 100, 10, 5 or 1 nucleotide downstream of the target gene's transcription stop site.
[0040] The target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence which includes the coding region of the target gene. The target antisense RNA transcript may comprise a sequence which is complementary to a genomic sequence that aligns with the target gene's promoter region on the template strand. Genes may possess a plurality of promoter regions, in which case the target antisense RNA transcript may align with one, two or more of the promoter regions. An online database of annotated gene loci may be used to identify the promoter regions of genes. The terms 'align' and 'alignment' when used in the context of a pair of nucleotide sequences mean the pair of nucleotide sequences are complementary to each other or have sequence identity with each other.
[0041] The region of alignment between the target antisense RNA transcript and the promoter region of the target gene may be partial and may be as short as a single nucleotide in length, although it may be at least 15 or at least 20 nucleotides in length, or at least 25 nucleotides in length, or at least 30, 35, 40, 45 or 50 nucleotides in length, or at least 55, 60, 65,
70 or 75 nucleotides in length, or at least 100 nucleotides in length. Each of the following specific arrangements is intended to fall within the scope of the term "alignment":
[0042] a) The target antisense RNA transcript and the target gene's promoter region are identical in length and they align (i.e. they align over their entire lengths).
[0043] b) The target antisense RNA transcript is shorter than the target gene's promoter region and aligns over its entire length with the target gene's promoter region (i.e. it aligns over its entire length to a sequence within the target gene's promoter region).
[0044] c) The target antisense RNA transcript is longer than the target gene's promoter region and the target gene's promoter region is aligned fully by it (i.e. the target gene's promoter region is aligns over its entire length to a sequence within the target antisense RNA transcript).
[0045] d) The target antisense RNA transcript and the target gene's promoter region are of the same or different lengths and the region of alignment is shorter than both the length of the target antisense RNA transcript and the length of the target gene's promoter region.
[0046] The above definition of "align" and "alignment" applies mutatis mutandis to the description of other overlapping, e.g., aligned sequences throughout the description. Clearly, if a target antisense RNA transcript is described as aligning with a region of the target gene other than the promoter region then the sequence of the target antisense RNA transcript aligns with a sequence within the noted region rather than within the promoter region of the target gene.
[0047] In one embodiment, the target antisense RNA transcript is at least 1 kb, or at least 2, 3, 4, 5, 6, 7, 8, 9 or 10, e.g., 20, 25, 30, 35 or 40 kb long.
[0048] In one embodiment, the target antisense RNA transcript comprises a sequence which is at least 75%, or at least 85%, or at least 90%, or at least 95% complementary along its full length to a sequence on the coding strand of the target gene.
[0049] The present invention provides saRNAs targeting the target antisense RNA transcript and may effectively and specifically down-regulate such target antisense RNA transcripts. This can be achieved by saRNA having a high degree of complementarity to a region within the target antisense RNA transcript. The saRNA will have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the region within the target antisense RNA transcript to be targeted. [0050] Referring to Figure 1 , as the target antisense RNA transcript has sequence identity with a region of the template strand of the target gene, the target antisense RNA transcript will be in part identical to a region within the template strand of the target gene allowing reference to be made either to the template strand of the gene or to a target antisense RNA transcript. The location at which the saRNA hybridizes or binds to the target antisense RNA transcript (and hence the same location on the template strand) is referred to as the "targeted sequence" or "target site".
[0051] The antisense strand of the saRNA (whether single- or double-stranded) may be at least 80%, 90%, 95%, 98%, 99% or 100% identical with the reverse complement of the targeted sequence. Thus, the reverse complement of the antisense strand of the saRNA has a high degree of sequence identity with the targeted sequence. The targeted sequence may have the same length, i.e., the same number of nucleotides, as the saRNA and/or the reverse complement of the saRNA,
[0052] In some embodiments, the targeted sequence comprises at least 14 and less than
30 nucleotides.
[0053] In some embodiments, the targeted sequence has 19, 20, 21 , 22, or 23 nucleotides.
[0054] In some embodiments, the location of the targeted sequence is situated within a promoter area of the template strand.
[0055] In some embodiments, the targeted sequence is located within a TSS
(transcription start site) core of the template stand. A "TSS core" or "TSS core sequence" as used herein, refers to a region between 2000 nucleotides upstream and 2000 nucleotides downstream of the TSS (transcription start site). Therefore, the TSS core comprises 4001 nucleotides and the TSS is located at position 2001 from the 5' end of the TSS core sequence.
[0056] In some embodiments, the targeted sequence is located between 1000 nucleotides upstream and 1000 nucleotides downstream of the TSS.
[0057] In some embodiments, the targeted sequence is located between 500 nucleotides upstream and 500 nucleotides downstream of the TSS.
[0058] In some embodiments, the targeted sequence is located between 250 nucleotides upstream and 250 nucleotides downstream of the TSS. [0059] In some embodiments, the targeted sequence is located between 100 nucleotides upstream and 100 nucleotides downstream of the TSS.
[0060] In some embodiments, the targeted sequence is located between 10 nucleotides upstream and 10 nucleotides downstream of the TSS.
[ 0061] In some embodiments, the targeted sequence is located between 5 nucleotides upstream and 5 nucleotides downstream of the TSS.
[0062] In some embodiments, the targeted sequence is located between 1 nucleotide upstream and 1 nucleotide downstream of the TSS.
[0063] In some embodiments, the targeted sequence is located upstream of the TSS in the TSS core. The targeted sequence may be less than 2000, less than 1000, less than 500, less than 250, less than 100, less than 10 or less than 5 nucleotides upstream of the TSS.
[0064] In some embodiments, the targeted sequence is located downstream of the TSS in the TSS core. The targeted sequence may be less than 2000, less than 1000, less than 500, less than 250, less than 100, less than 10 or less than 5 nucleotides downstream of the TSS,
[0065] In some embodiments, the targeted sequence is located +/- 50 nucleotides surrounding the TSS of the TSS core. In some embodiments, the targeted sequence substantially overlaps the TSS of the TSS core. In some embodiments, the targeted sequence overlaps begins or ends at the TSS of the TSS core. In some embodiments, the targeted sequence overlaps the TSS ofthe TSS core by 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18 or 19 nucleotides in either the upstream or downstream direction.
[0066] The location of the targeted sequence on the template strand is defined by the location of the 5 ' end of the targeted sequence. The 5 ' end of the targeted sequence may be at any position of the TSS core and the targeted sequence may start at any position selected from position 1 to position 4001 of the TSS core. For reference herein, when the 5' most end of the targeted sequence from position 1 to position 2000 of the TSS core, the targeted sequence is considered upstream of the TSS and when the 5 ' most end of the targeted sequence is from position 2002 to 4001 , the targeted sequence is considered downstream of the TSS. When the 5' most end of the targeted sequence is at nucleotide 2001 , the targeted sequence is considered to be a TSS centric sequence and is neither upstream nor downstream of the TSS. [0067] For further reference, for example, when the 5' end of the targeted sequence is at position 1600 of the TSS core, i.e., it is the 1600th nucleotide of the TSS core, the targeted sequence starts at position 1600 of the TSS core and is considered to be upstream of the TSS,
[0068] In one embodiment, the TSS core is a sequence for the target gene as described in Table 1 and 2.
[0069] In one embodiment, the TSS core is a sequence such as, but not limited to, SEQ
ID NO: 1-4047, 315236-318726, 584785-589061, 913310-917531, 1241080-1245401, 1559932- 1564372 and 1879189-1889207.
[0070] In one embodiment, the saRNA of the present invention may have two strands that form a duplex, one strand being a guide strand. The saRNA duplex is also called a double- stranded saRNA. A double-stranded saRNA or saRNA duplex, as used herein, is an saRNA that includes more than one, and preferably, two, strands in which interstrand hybridization can form a region of duplex structure. The two strands of a double-stranded saRNA are referred to as an antisense strand or a guide strand, and a sense strand or a passenger strand.
[0071] The antisense strand of an saRNA duplex, used interchangeably with antisense strand saRNA or antisense saRNA, has a high degree of complementarity to a region within the target antisense RNA transcript. The antisense strand may have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the region within the target antisense RNA transcript or targeted sequence. Therefore, the antisense strand has a high degree of complementary to the targeted sequence on the template strand. The sense strand of the saRNA duplex, used interchangeably with sense strand saRNA or sense saRNA, has a high degree of sequence identity with the targeted sequence on the template strand. In some embodiments, the targeted sequence is located within the promoter area of the template strand. In some embodiments, the targeted sequence is located within the TSS core of the template stand.
[0072] The location of the antisense strand and/or sense strand of the saRNA duplex, relative to the targeted sequence is defined by making reference to the TSS core sequence. For example, when the targeted sequence is downstream of the TSS, the antisense saRNA and the sense saRNA start downstream of the TSS. In another example, when the targeted sequence starts at position 200 of the TSS core, the antisense saRNA and the sense saRNA start upstream of the TSS. [0073] The relationships among the saRNAs, a target gene, a coding strand of the target gene, a template strand of the target gene, a target antisense RNA transcript, a target transcript, a targeted sequence/target site, and the TSS are shown in FIG. 1 ,
[0074] A "strand" in the context of the present invention means a contiguous sequence of nucleotides, including non-natu rally occurring or modified nucleotides. Two or more strands may be, or each form a part of, separate molecules, or they may be connected covalently, e.g., by a linker such as a polyethyleneglyeol linker. At least one strand of an saRNA may comprise a region that is complementary to a target antisense RNA. Such a strand is called an antisense or guide strand of the saRNA duplex. A second strand of an saRNA that comprises a region complementary to the antisense strand of the saRNA is called a sense or passenger strand.
[0075] An saRNA duplex may also be formed from a single molecule that is at least partly self-complementary forming a hairpin structure, including a duplex region. In such case, the term "strand" refers to one of the regions of the saRNA that is complementary to another internal region of the saRNA. The guide strand of the saRNA will have no more than 5, or no more than 4 or 3, or no more than 2, or no more than 1 , or no mismatches with the sequence within the target antisense RNA transcript.
[0076] In some embodiments, the passenger strand of an saRNA may comprise at least one nucleotide that is not complementary to the corresponding nucleotide on the guide strand, called a mismatch with the guide strand. The mismatch with the guide strand may encourage preferential loading of the guide strand (Wu et al, PLoS ONE, vol.6 (12):e28580 (201 1), the contents of which are incorporated herein by reference in their entirety). In one embodiment, the at least one mismatch with the guide strand may be at 3 ' end of the passenger strand. In one embodiment, the 3 ' end of the passenger strand may comprise 1-5 mismatches with the guide strand. In one embodiment, the 3 ' end of the passenger strand may comprise 2-3 mismatches with the guide strand. In one embodiment, the 3 ' end of the passenger strand may comprise 6-10 mismatches with the guide strand.
[0077] In one embodiment, an saRNA duplex may show efficacy in proliferating cells.
[0078] An saRNA duplex may have siRNA-like complementarity to a region of a target antisense RNA transcript; that is, 100% complementarity between nucleotides 2-6 from the 5' end of the guide strand in the saRNA duplex and a region of the target antisense RNA transcript. Other nucleotides of the saRNA may, in addition, have at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to a region of the target antisense RNA transcript. For example, nucleotides 7 (counted from the 5' end) until the 3' end of the saRNA may have least 80%, 90%, 95%, 98%o, 99% or 100% complementarity to a region of the target antisense RNA transcript.
[0079] The terms "small interfering RNA" or "siRNA" in the context mean a double- stranded RNA typically 20-25 nucleotides long involved in the RNA interference (RNAi) pathway and interfering with or inhibiting the expression of a specific gene. The gene is the target gene of the siRNA. For example, siRNA that interferes the expression of A3GALT2 gene is called "A3GALT2-siRNA" and the A3GALT2 gene is the target gene. An siRNA is usually about 21 nucleotides long, with 3' overhangs (e.g., 2 nucleotides) at each end of the two strands.
[0080] An siRNA inhibits target gene expression by binding to and promoting the cleavage of one or more RNA transcripts of the target gene at specific sequences. Typically in RNAi the RNA transcripts are in RNA, so cleavage of mRNA results in the down-regulation of gene expression. In the present invention, not willing to be bound with any theory, one of the possible mechanisms is that saRNA of the present invention may modulate the target gene expression by cleavage of the target antisense RNA transcript.
[0081] A double-stranded saRNA may include one or more single-stranded nucleotide overhangs. The term "overhang" or "tail" in the context of double-stranded saRNA and siRNA refers to at least one unpaired nucleotide that protrudes from the duplex structure of saRNA or siRNA. For example, when a 3 '-end of one strand of an saRNA extends beyond the 5 '-end of the other strand, or vice versa, there is a nucleotide overhang. An saRNA may comprise an overhang of at least one nucleotide; alternatively the overhang may comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. A nucleotide overhang may comprise of consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) may be on the sense strand, the antisense strand or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5 ' end, 3 ' end or both ends of either an antisense or sense strand of an saRNA. Where two oligonucleotides are designed to form, upon hybridization, one or more single-stranded overhangs, and such overhangs shall not be regarded as mismatches with regard to the determination of complementarity. For example, an saRNA comprising one oligonucleotide 19 nucleotides in length and another oligonucleotide 21 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 19 nucleotides that is fully complementary to the shorter oligonucleotide, can yet be referred to as "fully complementary" for the purposes described herein.
[0082] In one embodiment, the antisense strand of a double-stranded saRNA has a 1-10 nucleotide overhang at the 3' end and/or the 5' end. In one embodiment, the antisense strand of a double-stranded saRNA has 1-4 nucleotide overhang at its 3' end, or 1-2 nucleotide overhang at its 3' end. In one embodiment, the sense strand of a double-stranded saRNA has a 1 -10 nucleotide overhang at the 3' end and/or the 5' end. In one embodiment, the sense strand of a double-stranded saRNA has 1-4 nucleotide overhang at its 3' end, or 1-2 nucleotide overhang at its 3' end. In one embodiment, both the sense strand and the antisense strand of a double- stranded saRNA have 3' overhangs. The 3' overhangs may comprise one or more uracils, e.g., the sequences UU or UUU. In one embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate, wherein the internucleoside linkage is
thiophosphate. In one embodiment, the overhang comprises one or more deoxyribonucleoside, e.g., the sequence dTdT or dTdTdT. In one embodiments, the overhang comprises the sequence dT*dT, wherein * is a thiophosphate internucleoside linkage.
[0083] The skilled person will appreciate that it is convenient to define the saRNA of the present invention by reference to the target antisense RNA transcript or the targeted sequence, regardless of the mechanism by which the saRNA modulates the target gene expression. However, the saRNA of the present invention may alternatively be defined by reference to the target gene. The target antisense RNA transcript is complementary to a genomic region on the coding strand of the target gene, and the saRNA of the present invention is in turn complementary to a region of the target antisense RNA transcript, so the saRNA of the present invention may be defined as having sequence identity to a region on the coding strand of the target gene. All of the features discussed herein with respect to the definition of the saRNA of the present invention by reference to the target antisense RNA transcript apply mutatis mutandis to the definition of the saRNA of the present invention by reference to the target gene so any discussion of complementarity to the target antisense RNA transcript should be understood to include identity to the genomic sequence of the target gene. Thus, the saRNA of the present invention may have a high percent identity, e.g. at least 80%, 90%, 95%, 98% or 99%, or 100% identity, to a genomic sequence on the target gene. The genomic sequence may be up to 2000, 1000, 500, 250, or 100 nucleotides upstream or downstream of the target gene's transcription start site. It may align with the target gene's promoter region. Thus, the saRNA may have sequence identity to a sequence that aligns with the promoter region of the target gene,
[0084] In one embodiment, the existence of the target antisense RNA transcript does not need to be determined to design the saRNA of the present invention. In another word, the design of the saRNA does not require the identification of the target antisense RNA transcript. For example, the nucleotide sequence of the TSS core, i.e., the sequence in the region 2000 nucleotides upstream of the target gene's transcription start site to 2000 nucleotides downstream of the target gene's transcription start may be obtained by the genomic sequence of the coding strand of the target gene, by sequencing or by searching in a database. Targeted sequence within the TSS core starting at any position from position 1 to position 4001 of the TSS core on the template strand can be selected and can then be used to design saRNA sequences. As discussed above, the saRNA has a high degree of sequence identity with the reverse complement of the targeted sequence.
[0085] The saRNA sequence's off-target hit number in the whole genome, 0 mismatch
(Omm) hit number, and 1 mismatch (1mm) hit number are then determined. The term "off-target hit number" refers to the number of other sites in the whole genome that are identical to the saRNA's targeted sequence on the template strand of the target gene. The term "Omm hit number" refers to the number of known protein coding transcript other than the target transcript of the saRNA, the complement of which the saRNA may hybridize with or bind to with 0 mismatch. In another word, "Omm hit number" counts the number of known protein coding transcript, other than the target transcript of the saRNA that comprises a region completely identical with the saRNA sequence. The term "1mm hit number" refers to the number of known protein coding transcript other than the target transcript of the saRNA, the complement of which the saRNA may hybridize with or bind to with 1 mismatch. In another word, "lmm hit number" counts the number of known protein coding transcript, other than the target transcript of the saRNA that comprises a region identical with the saRNA sequence with only 1 mismatch. In one embodiment, only saRNA sequences that have no off-target hit, no Omm hit and no 1mm hit are selected. For those saRNA sequences disclosed in the present application, each has no off- target hit, no Omm hit and no 1mm hit,
[0086] The method disclosed in US 2013/0164846 filed June 23, 2011 (saRNA algorithm), the contents of which are incorporated herein by reference in their entirety, may also be used to design saRNA. The design of saRNA is also disclosed in US Pat. No. 8,324, 181 and US Pat. No. 7,709,566 to Corey et al., US Pat. Pub. No. 2010/0210707 to Li et al., and Voutila et al, Mol Ther Nucleic Acids, vol 1 , e35 (2012), the contents of each of which are incorporated herein by reference in their entirety.
[0087] "Determination of existence" means either searching databases of ESTs and/or antisense UNA transcripts around the locus of the target gene to identify a suitable target antisense RNA transcript, or using RT PCR or any other known technique to confirm the physical presence of a target antisense RNA transcript in a cell.
[0088] In some embodiments, the saRNA of the present invention may be single or, double-stranded. Double-stranded molecules comprise a first strand and a second strand. If double-stranded, each strand of the duplex may be at least 14, or at least 18, e.g. 19, 20, 21 or 22 nucleotides in length. The duplex may be hybridized over a length of at least 12, or at least 15, or at least 17, or at least 19 nucleotides. Each strand may be exactly 19 nucleotides in length. Preferably, the length of the saRNA is less than 30 nucleotides since oligonucleotide duplex exceeding this length may have an increased risk of inducing the interferon response. In one embodiment, the length of the saRNA is 19 to 25 nucleotides. The strands forming the saRNA duplex may be of equal or unequal lengths.
[0089] In one embodiment, the saRNAs of the present invention comprise a sequence of at least 14 nucleotides and less than 30 nucleotides which has at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to the targeted sequence. In one embodiment, the sequence which has at least 80%, 90%, 95%, 98%, 99% or 100% complementarity to the targeted sequence is at least 15, 16, 17, 18 or 19 nucleotides in length, or 18-22 or 19 to 21 , or exactly 19.
[0090] The saRNA of the present invention may include a short 3' or 5' sequence which is not complementary to the target antisense RNA transcript. In one embodiment, such a sequence is at 3 ' end of the strand. The sequence may be 1 -5 nucleotides in length, or 2 or 3. The sequence may comprises uracil, so it may be a 3' stretch of 2 or 3 uracils. The sequence may comprise one or more deoxyribonucleoside, such as dT. In one embodiment, one or more of the nucleotides in the sequence is replaced with a nucleoside thiophosphate, wherein the
internucleoside linkage is thiophosphate. As a non-limiting example, the sequence comprises the sequence dT*dT, wherein * is a thiophosphate internucleoside linkage. This non-complementary sequence may be referred to as "tail". If a 3' tail is present, the strand may be longer, e.g., 19 nucleotides plus a 3' tail, which may be UU or UUU. Such a 3 ' tail shall not be regarded as mismatches with regard to determine complementarity between the saRNA and the target antisense RNA transcript.
[0091] Thus, the saRNA of the present invention may consist of (i) a sequence having at least 80% complementarity to a region of the target antisense RNA transcript; and (ii) a 3' tail of 1 -5 nucleotides, which may comprise or consist of uracil residues. The saRNA will thus typically have complementarity to a region of the target antisense RNA transcript over its whole length, except for the 3' tail, if present. Any of the saRNA sequences disclosed in the present application may optionally include such a 3' tail. Thus, any of the saRNA sequences disclosed in the saRNA Tables and Sequence Listing may optionally include such a 3' tail. The saRNA of the present invention may further comprise Dicer or Drosha substrate sequences.
[0092] The saRNA of the present invention may contain a flanking sequence. The flanking sequence may be inserted in the 3 ' end or 5 ' end of the saRNA of the present invention. In one embodiment, the flanking sequence is the sequence of a miRNA, rendering the saRNA to have miRNA configuration and may be processed with Drosha and Dicer. In a non-limiting example, the saRNA of the present invention has two strands and is cloned into a microRNA precursor, e.g., miR-30 backbone flanking sequence.
[0093] The saRNA of the present invention may comprise a restriction enzyme substrate or recognition sequence. The restriction enzyme recognition sequence may be at the 3 ' end or 55 end of the saRNA of the present invention. Non-limiting examples of restriction enzymes include Notl and Ascl.
[0094] In one embodiment, the saRNA of the present invention consists of two strands stably base-paired together. In some embodiments, the passenger strand may comprise at least one nucleotide that is not complementary to the corresponding nucleotide on the guide strand, called a mismatch with the guide strand. In one embodiment, the at least one mismatch with the guide strand may be at 3 ' end of the passenger strand. In one embodiment, the 3 ' end of the passenger strand may comprise 1-5 mismatches with the guide strand. In one embodiment, the 3 ' end of the passenger strand may comprise 2-3 mismatches with the guide strand. In one embodiment, the 3 ' end of the passenger strand may comprise 6-10 mismatches with the guide strand. [0095] In some embodiments, the double-stranded saRNA may comprise a number of unpaired nucleotides at the 3' end of each strand forming 3' overhangs. The number of unpaired nucleotides forming the 3' overhang of each strand may be in the range of 1 to 5 nucleotides, or I to 3 nucleotides, or 2 nucleotides. The 3' overhang may be formed on the 3' tail mentioned above, so the 3' tail may be the 3' overhang of a double-stranded saRNA,
[0096] Thus, the saRNA of the present invention may be single-stranded and consists of
(i) a sequence having at least 80% complementarity to a region of the target antisense RNA transcript; and (ii) a 3' tail of 1 -5 nucleotides, which may comprise uracil residues. The saRNA of the present invention may have complementarity to a region of the target antisense RNA transcript over its whole length, except for the 3' tail, if present. As mentioned above, instead of "complementary to the target antisense RNA transcript" the saRNA of the present invention may also be defined as having "identity" to the coding strand of the target gene. The saRNA of the present invention may be double-stranded and consists of a first strand comprising (i) a first sequence having at least 80% complementarity to a region of the target antisense RNA transcript and (ii) a 3' overhang of 1 -5 nucleotides; and a second strand comprising (i) a second sequence that forms a duplex with the first sequence and (ii) a 3' overhang of 1-5 nucleotides.
[0097] As described herein, the genomic sequence of the target gene may be used to design saRNA of the target gene. The sequence of a target antisense RNA transcript may be determined from the sequence of the target gene for designing saRNA of the target gene.
However, the existence of such a target antisense RNA transcript does not need to be
determined.
[0098] One aspect of the present invention provides an saRNA that modulates the expression of a target gene. Also provided is an saRNA that modulates the level of a target transcript. In some embodiments, the target transcript is a coding transcript, e.g., mRNA.
Another aspect of the present invention provides an saRNA that modulates the level of a protein encoded by the coding target transcript. In one embodiment, the expression of target gene is increased by at least 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80% in the presence of the saRNA of the present invention compared to the expression of target gene in the absence of the saRNA of the present invention. In a further embodiment, the expression of target gene is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, in the presence of the saRNA of the present invention compared to the expression of target gene in the absence of the saRNA of the present invention. The modulation of the expression of target gene may be reflected or determined by the change of mRNA levels encoding the target gene.
[ 0099] Non-limiting examples of target genes which can be modulated by the saRNA of the present invention may be coding genes, including the target genes described in Table 1 such
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
Figure imgf000330_0001
Figure imgf000331_0001
Figure imgf000332_0001
Figure imgf000333_0001
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000348_0001
Figure imgf000349_0001
Figure imgf000350_0001
Figure imgf000351_0001
Figure imgf000352_0001
Figure imgf000353_0001
Figure imgf000354_0001
Figure imgf000355_0001
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Figure imgf000361_0001
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000370_0001
Figure imgf000371_0001
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
Figure imgf000375_0001
Figure imgf000376_0001
Figure imgf000377_0001
Figure imgf000378_0001
Figure imgf000379_0001
Figure imgf000380_0001
Figure imgf000381_0001
Figure imgf000382_0001
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0001
Figure imgf000386_0001
Figure imgf000387_0001
Figure imgf000388_0001
Figure imgf000389_0001
Figure imgf000390_0001
Figure imgf000391_0001
Figure imgf000392_0001
Figure imgf000393_0001
Figure imgf000394_0001
Figure imgf000395_0001
Figure imgf000396_0001
Figure imgf000397_0001
Figure imgf000398_0001
Figure imgf000399_0001
Figure imgf000400_0001
Figure imgf000401_0001
Figure imgf000402_0001
Figure imgf000403_0001
Figure imgf000404_0001
Figure imgf000405_0001
Figure imgf000406_0001
Figure imgf000407_0001
Figure imgf000408_0001
Figure imgf000409_0001
Figure imgf000410_0001
Figure imgf000411_0001
Figure imgf000412_0001
Figure imgf000413_0001
Figure imgf000414_0001
Figure imgf000415_0001
Figure imgf000416_0001
Figure imgf000417_0001
Figure imgf000418_0001
Figure imgf000419_0001
Figure imgf000420_0001
Figure imgf000421_0001
Figure imgf000422_0001
Figure imgf000423_0001
Figure imgf000424_0001
Figure imgf000425_0001
Figure imgf000426_0001
Figure imgf000427_0001
Figure imgf000428_0001
Figure imgf000429_0001
Figure imgf000430_0001
Figure imgf000431_0001
Figure imgf000432_0001
Figure imgf000433_0001
Figure imgf000434_0001
Figure imgf000435_0001
Figure imgf000436_0001
Figure imgf000437_0001
Figure imgf000438_0001
Figure imgf000439_0001
Figure imgf000440_0001
Figure imgf000441_0001
Figure imgf000442_0001
Figure imgf000443_0001
Figure imgf000444_0001
Figure imgf000445_0001
Figure imgf000446_0001
Figure imgf000447_0001
Figure imgf000448_0001
Figure imgf000449_0001
Figure imgf000450_0001
Figure imgf000451_0001
Figure imgf000452_0001
Figure imgf000453_0001
Figure imgf000454_0001
Figure imgf000455_0001
Figure imgf000456_0001
Figure imgf000457_0001
Figure imgf000458_0001
Figure imgf000459_0001
Figure imgf000460_0001
Figure imgf000461_0001
Figure imgf000462_0001
Figure imgf000463_0001
Figure imgf000464_0001
Figure imgf000465_0001
Figure imgf000466_0001
Figure imgf000467_0001
Figure imgf000468_0001
Figure imgf000469_0001
Figure imgf000470_0001
Figure imgf000471_0001
Figure imgf000472_0001
Figure imgf000473_0001
Figure imgf000474_0001
Figure imgf000475_0001
Figure imgf000476_0001
Figure imgf000477_0001
Figure imgf000478_0001
Figure imgf000479_0001
Figure imgf000480_0001
Figure imgf000481_0001
Figure imgf000482_0001
Figure imgf000483_0001
Figure imgf000484_0001
Figure imgf000485_0001
Figure imgf000486_0001
Figure imgf000487_0001
Figure imgf000488_0001
Figure imgf000489_0001
Figure imgf000490_0001
Figure imgf000491_0001
Figure imgf000492_0001
Figure imgf000493_0001
Figure imgf000494_0001
Figure imgf000495_0001
Figure imgf000496_0001
Figure imgf000497_0001
Figure imgf000498_0001
Figure imgf000499_0001
Figure imgf000500_0001
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
Figure imgf000505_0001
Figure imgf000506_0001
Figure imgf000507_0001
Figure imgf000508_0001
Figure imgf000509_0001
Figure imgf000510_0001
Figure imgf000511_0001
Figure imgf000512_0001
Figure imgf000513_0001
Figure imgf000514_0001
Figure imgf000515_0001
Figure imgf000516_0001
Figure imgf000517_0001
Figure imgf000518_0001
Figure imgf000519_0001
Figure imgf000520_0001
Figure imgf000521_0001
Figure imgf000522_0001
Figure imgf000523_0001
Figure imgf000524_0001
Figure imgf000525_0001
Figure imgf000526_0001
Figure imgf000527_0001
Figure imgf000528_0001
Figure imgf000529_0001
Figure imgf000530_0001
Figure imgf000531_0001
Figure imgf000532_0001
Figure imgf000533_0001
Figure imgf000534_0001
Figure imgf000535_0001
Figure imgf000536_0001
Figure imgf000537_0001
Figure imgf000538_0001
Figure imgf000539_0001
Figure imgf000540_0001
Figure imgf000541_0001
Figure imgf000542_0001
Figure imgf000543_0001
Figure imgf000544_0001
Figure imgf000545_0001
Figure imgf000546_0001
Figure imgf000547_0001
Figure imgf000548_0001
Figure imgf000549_0001
Figure imgf000550_0001
Figure imgf000551_0001
Figure imgf000552_0001
Figure imgf000553_0001
Figure imgf000554_0001
Figure imgf000555_0001
Figure imgf000556_0001
Figure imgf000557_0001
Figure imgf000558_0001
Figure imgf000559_0001
Figure imgf000560_0001
Figure imgf000561_0001
Figure imgf000562_0001
Figure imgf000563_0001
Figure imgf000564_0001
Figure imgf000565_0001
Figure imgf000566_0001
Figure imgf000567_0001
Figure imgf000568_0001
Figure imgf000569_0001
Figure imgf000570_0001
Figure imgf000571_0001
Figure imgf000572_0001
Figure imgf000573_0001
Figure imgf000574_0001
Figure imgf000575_0001
Figure imgf000576_0001
Figure imgf000577_0001
Figure imgf000578_0001
Figure imgf000579_0001
Figure imgf000580_0001
Figure imgf000581_0001
Figure imgf000582_0001
Figure imgf000583_0001
Figure imgf000584_0001
Figure imgf000585_0001
Figure imgf000586_0001
Figure imgf000587_0001
Figure imgf000588_0001
Figure imgf000589_0001
Figure imgf000590_0001
Figure imgf000591_0001
Figure imgf000592_0001
Figure imgf000593_0001
Figure imgf000594_0001
Figure imgf000595_0001
Figure imgf000596_0001
Figure imgf000597_0001
Figure imgf000598_0001
Figure imgf000599_0001
Figure imgf000600_0001
Figure imgf000601_0001
Figure imgf000602_0001
Figure imgf000603_0001
Figure imgf000604_0001
Figure imgf000605_0001
Figure imgf000606_0001
Figure imgf000607_0001
Figure imgf000608_0001
Figure imgf000609_0001
Figure imgf000610_0001
Figure imgf000611_0001
Figure imgf000612_0001
Figure imgf000613_0001
Figure imgf000614_0001
Figure imgf000615_0001
Figure imgf000616_0001
Figure imgf000617_0001
Figure imgf000618_0001
Figure imgf000619_0001
Figure imgf000620_0001
Figure imgf000621_0001
Figure imgf000622_0001
Figure imgf000623_0001
Figure imgf000624_0001
Figure imgf000625_0001
Figure imgf000626_0001
Figure imgf000627_0001
Figure imgf000628_0001
Figure imgf000629_0001
Figure imgf000630_0001
Figure imgf000631_0001
Figure imgf000632_0001
Figure imgf000633_0001
Figure imgf000634_0001
Figure imgf000635_0001
Figure imgf000636_0001
Figure imgf000637_0001
Figure imgf000638_0001
Figure imgf000639_0001
Figure imgf000640_0001
Figure imgf000641_0001
Figure imgf000642_0001
Figure imgf000643_0001
Figure imgf000644_0001
Figure imgf000645_0001
Figure imgf000646_0001
Figure imgf000647_0001
Figure imgf000648_0001
Figure imgf000649_0001
Figure imgf000650_0001
Figure imgf000651_0001
Figure imgf000652_0001
Figure imgf000653_0001
Figure imgf000654_0001
Figure imgf000655_0001
Figure imgf000656_0001
Figure imgf000657_0001
Figure imgf000658_0001
Figure imgf000659_0001
Figure imgf000660_0001
Figure imgf000661_0001
Figure imgf000662_0001
Figure imgf000663_0001
Figure imgf000664_0001
Figure imgf000665_0001
Figure imgf000666_0001
Figure imgf000667_0001
Figure imgf000668_0001
Figure imgf000669_0001
Figure imgf000670_0001
Figure imgf000671_0001
Figure imgf000672_0001
Figure imgf000673_0001
Figure imgf000674_0001
Figure imgf000675_0001
Figure imgf000676_0001
Figure imgf000677_0001
Figure imgf000678_0001
Figure imgf000679_0001
Figure imgf000680_0001
Figure imgf000681_0001
Figure imgf000682_0001
Figure imgf000683_0001
Figure imgf000684_0001
Figure imgf000685_0001
Figure imgf000686_0001
Figure imgf000687_0001
Figure imgf000688_0001
Figure imgf000689_0001
Figure imgf000690_0001
Figure imgf000691_0001
Figure imgf000692_0001
Figure imgf000693_0001
Figure imgf000694_0001
Figure imgf000695_0001
Figure imgf000696_0001
Figure imgf000697_0001
Figure imgf000698_0001
Figure imgf000699_0001
Figure imgf000700_0001
Figure imgf000701_0001
Figure imgf000702_0001
Figure imgf000703_0001
Figure imgf000704_0001
Figure imgf000705_0001
Figure imgf000706_0001
Figure imgf000707_0001
Figure imgf000708_0001
Figure imgf000709_0001
Figure imgf000710_0001
Figure imgf000711_0001
Figure imgf000712_0001
Figure imgf000713_0001
Figure imgf000714_0001
Figure imgf000715_0001
Figure imgf000716_0001
Figure imgf000717_0001
Figure imgf000718_0001
Figure imgf000719_0001
Figure imgf000720_0001
Figure imgf000721_0001
Figure imgf000722_0001
Figure imgf000723_0001
Figure imgf000724_0001
Figure imgf000725_0001
Figure imgf000726_0001
Figure imgf000727_0001
Figure imgf000728_0001
Figure imgf000729_0001
Figure imgf000730_0001
Figure imgf000731_0001
Figure imgf000732_0001
Figure imgf000733_0001
Figure imgf000735_0001
Figure imgf000736_0001
Figure imgf000737_0001
Figure imgf000738_0001
Figure imgf000739_0001
Figure imgf000740_0001
Figure imgf000741_0001
Figure imgf000742_0001
Figure imgf000743_0001
Figure imgf000744_0001
Figure imgf000745_0001
Figure imgf000747_0001
Figure imgf000748_0001
Figure imgf000749_0001
Figure imgf000750_0001
Figure imgf000751_0001
Figure imgf000752_0001
Figure imgf000753_0001
Figure imgf000754_0001
Figure imgf000755_0001
Figure imgf000756_0001
Figure imgf000757_0001
Figure imgf000758_0001
Figure imgf000759_0001
Figure imgf000760_0001
Figure imgf000761_0001
Figure imgf000762_0001
Figure imgf000763_0001
Figure imgf000764_0001
Figure imgf000765_0001
Figure imgf000766_0001
Figure imgf000767_0001
Figure imgf000768_0001
Figure imgf000769_0001
Figure imgf000770_0001
Figure imgf000771_0001
Figure imgf000772_0001
Figure imgf000773_0001
Figure imgf000774_0001
Figure imgf000775_0001
Figure imgf000776_0001
Figure imgf000777_0001
Figure imgf000778_0001
Figure imgf000779_0001
Figure imgf000780_0001
Figure imgf000781_0001
Figure imgf000782_0001
Figure imgf000783_0001
Figure imgf000784_0001
Figure imgf000785_0001
Figure imgf000786_0001
Figure imgf000787_0001
Figure imgf000788_0001
Figure imgf000789_0001
Figure imgf000790_0001
Figure imgf000791_0001
Figure imgf000792_0001
Figure imgf000793_0001
Figure imgf000794_0001
Figure imgf000795_0001
Figure imgf000796_0001
Figure imgf000797_0001
Figure imgf000798_0001
Figure imgf000799_0001
Figure imgf000800_0001
Figure imgf000801_0001
Figure imgf000802_0001
Figure imgf000803_0001
Figure imgf000804_0001
Figure imgf000805_0001
Figure imgf000806_0001
Figure imgf000807_0001
Figure imgf000808_0001
Figure imgf000809_0001
Figure imgf000810_0001
Figure imgf000811_0001
Figure imgf000812_0001
Figure imgf000813_0001
Figure imgf000814_0001
Figure imgf000815_0001
Figure imgf000816_0001
Figure imgf000817_0001
Figure imgf000818_0001
Figure imgf000819_0001
Figure imgf000820_0001
Figure imgf000821_0001
Figure imgf000822_0001
Figure imgf000823_0001
Figure imgf000824_0001
Figure imgf000825_0001
Figure imgf000826_0001
Figure imgf000827_0001
Figure imgf000828_0001
Figure imgf000829_0001
Figure imgf000830_0001
Figure imgf000831_0001
Figure imgf000832_0001
Figure imgf000833_0001
Figure imgf000834_0001
Figure imgf000835_0001
Figure imgf000836_0001
Figure imgf000837_0001
Figure imgf000838_0001
Figure imgf000839_0001
Figure imgf000840_0001
Figure imgf000841_0001
Figure imgf000842_0001
Figure imgf000843_0001
Figure imgf000844_0001
Figure imgf000845_0001
Figure imgf000846_0001
Figure imgf000847_0001
Figure imgf000848_0001
Figure imgf000849_0001
Figure imgf000850_0001
Figure imgf000851_0001
Figure imgf000852_0001
Figure imgf000853_0001
Figure imgf000854_0001
Figure imgf000855_0001
Figure imgf000856_0001
Figure imgf000857_0001
Figure imgf000858_0001
Figure imgf000859_0001
Figure imgf000860_0001
Figure imgf000861_0001
Figure imgf000862_0001
Figure imgf000863_0001
Figure imgf000864_0001
Figure imgf000865_0001
Figure imgf000866_0001
Figure imgf000867_0001
Figure imgf000868_0001
Figure imgf000869_0001
Figure imgf000870_0001
Figure imgf000871_0001
Figure imgf000872_0001
Figure imgf000873_0001
Figure imgf000874_0001
Figure imgf000875_0001
Figure imgf000876_0001
Figure imgf000877_0001
Figure imgf000878_0001
Figure imgf000879_0001
Figure imgf000880_0001
Figure imgf000881_0001
Figure imgf000882_0001
Figure imgf000884_0001
Figure imgf000885_0001
Figure imgf000886_0001
Figure imgf000887_0001
Figure imgf000888_0001
Figure imgf000889_0001
Figure imgf000890_0001
Figure imgf000891_0001
Figure imgf000892_0001
Figure imgf000893_0001
Figure imgf000894_0001
Figure imgf000895_0001
Figure imgf000896_0001
Figure imgf000897_0001
Figure imgf000898_0001
Figure imgf000899_0001
Figure imgf000900_0001
Figure imgf000901_0001
Figure imgf000902_0001
Figure imgf000903_0001
Figure imgf000904_0001
Figure imgf000905_0001
Figure imgf000906_0001
Figure imgf000907_0001
Figure imgf000908_0001
Figure imgf000909_0001
Figure imgf000910_0001
Figure imgf000911_0001
Figure imgf000912_0001
Figure imgf000913_0001
Figure imgf000914_0001
Figure imgf000915_0001
Figure imgf000916_0001
Figure imgf000917_0001
Figure imgf000918_0001
Figure imgf000919_0001
Figure imgf000920_0001
Figure imgf000921_0001
Figure imgf000922_0001
Figure imgf000923_0001
Figure imgf000924_0001
Figure imgf000925_0001
Figure imgf000926_0001
Figure imgf000927_0001
Figure imgf000928_0001
Figure imgf000929_0001
Figure imgf000930_0001
Figure imgf000931_0001
Figure imgf000932_0001
Figure imgf000933_0001
Figure imgf000934_0001
Figure imgf000935_0001
Figure imgf000936_0001
Figure imgf000937_0001
Figure imgf000938_0001
Figure imgf000939_0001
Figure imgf000940_0001
Figure imgf000941_0001
Figure imgf000942_0001
Figure imgf000943_0001
Figure imgf000944_0001
Figure imgf000945_0001
Figure imgf000946_0001
Figure imgf000947_0001
Figure imgf000948_0001
Figure imgf000949_0001
Figure imgf000950_0001
Figure imgf000951_0001
Figure imgf000952_0001
Figure imgf000953_0001
Figure imgf000954_0001
Figure imgf000955_0001
Figure imgf000956_0001
Figure imgf000957_0001
Figure imgf000958_0001
Figure imgf000959_0001
Figure imgf000960_0001
Figure imgf000961_0001
Figure imgf000962_0001
Figure imgf000963_0001
Figure imgf000964_0001
Figure imgf000965_0001
Figure imgf000966_0001
Figure imgf000967_0001
Figure imgf000968_0001
Figure imgf000969_0001
Figure imgf000970_0001
Figure imgf000971_0001
Figure imgf000972_0001
Figure imgf000973_0001
Figure imgf000974_0001
Figure imgf000975_0001
Figure imgf000976_0001
Figure imgf000977_0001
Figure imgf000978_0001
Figure imgf000979_0001
Figure imgf000980_0001
Figure imgf000981_0001
Figure imgf000982_0001
Figure imgf000983_0001
Figure imgf000984_0001
Figure imgf000985_0001
Figure imgf000986_0001
Figure imgf000987_0001
Figure imgf000988_0001
Figure imgf000989_0001
Figure imgf000990_0001
Figure imgf000991_0001
Figure imgf000992_0001
Figure imgf000993_0001
Figure imgf000994_0001
Figure imgf000995_0001
Figure imgf000996_0001
Figure imgf000997_0001
Figure imgf000998_0001
Figure imgf000999_0001
Figure imgf001000_0001
Figure imgf001001_0001
Figure imgf001002_0001
Figure imgf001003_0001
Figure imgf001004_0001
Figure imgf001005_0001
Figure imgf001006_0001
Figure imgf001007_0001
Figure imgf001008_0001
Figure imgf001009_0001
Figure imgf001010_0001
Figure imgf001011_0001
Figure imgf001012_0001
Figure imgf001013_0001
Figure imgf001014_0001
Figure imgf001015_0001
Figure imgf001016_0001
Figure imgf001017_0001
Figure imgf001018_0001
Figure imgf001019_0001
Figure imgf001020_0001
Figure imgf001021_0001
Figure imgf001022_0001
Figure imgf001023_0001
Figure imgf001024_0001
Figure imgf001025_0001
Figure imgf001026_0001
Figure imgf001027_0001
Figure imgf001028_0001
Figure imgf001029_0001
Figure imgf001030_0001
Figure imgf001031_0001
Figure imgf001032_0001
Figure imgf001033_0001
Figure imgf001034_0001
Figure imgf001035_0001
Figure imgf001036_0001
Figure imgf001037_0001
Figure imgf001038_0001
Figure imgf001039_0001
Figure imgf001040_0001
Figure imgf001041_0001
Figure imgf001042_0001
Figure imgf001043_0001
Figure imgf001044_0001
Figure imgf001045_0001
Figure imgf001046_0001
Figure imgf001047_0001
Figure imgf001048_0001
Figure imgf001049_0001
Figure imgf001050_0001
Figure imgf001051_0001
Figure imgf001052_0001
Figure imgf001053_0001
Figure imgf001054_0001
Figure imgf001055_0001
Figure imgf001056_0001
Figure imgf001057_0001
Figure imgf001058_0001
Figure imgf001059_0001
Figure imgf001060_0001
Figure imgf001061_0001
Figure imgf001062_0001
Figure imgf001063_0001
Figure imgf001064_0001
Figure imgf001065_0001
Figure imgf001066_0001
Figure imgf001067_0001
Figure imgf001068_0001
Figure imgf001069_0001
Figure imgf001070_0001
Figure imgf001071_0001
Figure imgf001072_0001
Figure imgf001073_0001
Figure imgf001074_0001
Figure imgf001075_0001
Figure imgf001076_0001
Figure imgf001077_0001
Figure imgf001078_0001
Figure imgf001079_0001
Figure imgf001080_0001
Figure imgf001081_0001
Figure imgf001082_0001
Figure imgf001083_0001
Figure imgf001084_0001
Figure imgf001085_0001
Figure imgf001086_0001
Figure imgf001087_0001
Figure imgf001088_0001
Figure imgf001089_0001
Figure imgf001090_0001
Figure imgf001091_0001
Figure imgf001092_0001
Figure imgf001093_0001
Figure imgf001094_0001
Figure imgf001095_0001
Figure imgf001096_0001
Figure imgf001097_0001
Figure imgf001098_0001
Figure imgf001099_0001
Figure imgf001100_0001
Figure imgf001101_0001
Figure imgf001102_0001
Figure imgf001103_0001
Figure imgf001104_0001
Figure imgf001105_0001
Figure imgf001106_0001
Figure imgf001107_0001
Figure imgf001108_0001
Figure imgf001109_0001
Figure imgf001110_0001
Figure imgf001111_0001
Figure imgf001112_0001
Figure imgf001113_0001
Figure imgf001114_0001
Figure imgf001115_0001
Figure imgf001116_0001
Figure imgf001117_0001
Figure imgf001118_0001
Figure imgf001119_0001
Figure imgf001120_0001
Figure imgf001121_0001
Figure imgf001122_0001
Figure imgf001123_0001
Figure imgf001124_0001
Figure imgf001125_0001
Figure imgf001126_0001
Figure imgf001127_0001
Figure imgf001128_0001
Figure imgf001129_0001
Figure imgf001130_0001
Figure imgf001131_0001
Figure imgf001132_0001
Figure imgf001133_0001
Figure imgf001134_0001
Figure imgf001135_0001
Figure imgf001136_0001
Figure imgf001137_0001
Figure imgf001138_0001
Figure imgf001139_0001
Figure imgf001140_0001
Figure imgf001141_0001
Figure imgf001142_0001
Figure imgf001143_0001
Figure imgf001144_0001
Figure imgf001145_0001
Figure imgf001146_0001
Figure imgf001147_0001
Figure imgf001148_0001
Figure imgf001149_0001
Figure imgf001150_0001
Figure imgf001151_0001
Figure imgf001152_0001
Figure imgf001153_0001
Figure imgf001154_0001
Figure imgf001155_0001
Figure imgf001156_0001
Figure imgf001157_0001
Figure imgf001158_0001
Figure imgf001159_0001
Figure imgf001160_0001
Figure imgf001161_0001
Figure imgf001162_0001
Figure imgf001163_0001
Figure imgf001164_0001
Figure imgf001165_0001
Figure imgf001167_0001
Figure imgf001168_0001
Figure imgf001169_0001
Figure imgf001170_0001
Figure imgf001171_0001
Figure imgf001172_0001
Figure imgf001173_0001
Figure imgf001174_0001
Figure imgf001175_0001
Figure imgf001176_0001
Figure imgf001177_0001
Figure imgf001178_0001
Figure imgf001179_0001
Figure imgf001180_0001
Figure imgf001181_0001
Figure imgf001182_0001
Figure imgf001183_0001
Figure imgf001184_0001
Figure imgf001185_0001
Figure imgf001186_0001
Figure imgf001187_0001
Figure imgf001188_0001
Figure imgf001189_0001
Figure imgf001190_0001
Figure imgf001191_0001
Figure imgf001192_0001
Figure imgf001193_0001
Figure imgf001194_0001
Figure imgf001195_0001
Figure imgf001196_0001
Figure imgf001197_0001
Figure imgf001198_0001
Figure imgf001199_0001
Figure imgf001200_0001
Figure imgf001201_0001
Figure imgf001202_0001
Figure imgf001203_0001
Figure imgf001204_0001
Figure imgf001205_0001
Figure imgf001206_0001
Figure imgf001207_0001
Figure imgf001208_0001
Figure imgf001209_0001
Figure imgf001210_0001
Figure imgf001211_0001
Figure imgf001212_0001
Figure imgf001213_0001
Figure imgf001214_0001
Figure imgf001215_0001
Figure imgf001216_0001
Figure imgf001217_0001
Figure imgf001218_0001
Figure imgf001219_0001
Figure imgf001220_0001
Figure imgf001221_0001
Figure imgf001222_0001
Figure imgf001223_0001
Figure imgf001224_0001
Figure imgf001225_0001
Figure imgf001226_0001
Figure imgf001227_0001
Figure imgf001228_0001
Figure imgf001229_0001
Figure imgf001230_0001
Figure imgf001231_0001
Figure imgf001232_0001
Figure imgf001233_0001
Figure imgf001234_0001
Figure imgf001235_0001
Figure imgf001236_0001
Figure imgf001237_0001
Figure imgf001238_0001
Figure imgf001239_0001
Figure imgf001240_0001
Figure imgf001241_0001
Figure imgf001242_0001
Figure imgf001243_0001
Figure imgf001244_0001
Figure imgf001245_0001
Figure imgf001246_0001
Figure imgf001247_0001
Figure imgf001248_0001
Figure imgf001249_0001
Figure imgf001250_0001
Figure imgf001251_0001
Figure imgf001252_0001
Figure imgf001253_0001
Figure imgf001254_0001
Figure imgf001255_0001
Figure imgf001256_0001
Figure imgf001257_0001
Figure imgf001258_0001
Figure imgf001259_0001
Figure imgf001260_0001
Figure imgf001261_0001
Figure imgf001262_0001
Figure imgf001263_0001
Figure imgf001264_0001
Figure imgf001265_0001
Figure imgf001266_0001
Figure imgf001267_0001
Figure imgf001268_0001
Figure imgf001269_0001
Figure imgf001270_0001
Figure imgf001271_0001
Figure imgf001272_0001
Figure imgf001273_0001
Figure imgf001274_0001
Figure imgf001275_0001
Figure imgf001276_0001
Figure imgf001277_0001
Figure imgf001278_0001
Figure imgf001279_0001
Figure imgf001280_0001
Figure imgf001281_0001
Figure imgf001282_0001
Figure imgf001283_0001
Figure imgf001284_0001
Figure imgf001285_0001
Figure imgf001286_0001
Figure imgf001287_0001
Figure imgf001288_0001
Figure imgf001289_0001
Figure imgf001290_0001
Figure imgf001291_0001
Figure imgf001292_0001
Figure imgf001293_0001
Figure imgf001294_0001
Figure imgf001295_0001
Figure imgf001296_0001
Figure imgf001297_0001
Figure imgf001298_0001
Figure imgf001299_0001
Figure imgf001300_0001
Figure imgf001301_0001
Figure imgf001302_0001
Figure imgf001303_0001
Figure imgf001304_0001
Figure imgf001305_0001
Figure imgf001306_0001
Figure imgf001307_0001
Figure imgf001308_0001
Figure imgf001309_0001
Figure imgf001310_0001
Figure imgf001311_0001
Figure imgf001312_0001
Figure imgf001313_0001
Figure imgf001314_0001
Figure imgf001315_0001
Figure imgf001316_0001
Figure imgf001317_0001
Figure imgf001318_0001
Figure imgf001319_0001
Figure imgf001320_0001
Figure imgf001321_0001
Figure imgf001322_0001
Figure imgf001323_0001
Figure imgf001324_0001
Figure imgf001325_0001
Figure imgf001326_0001
Figure imgf001327_0001
Figure imgf001328_0001
Figure imgf001329_0001
Figure imgf001330_0001
Figure imgf001331_0001
Figure imgf001332_0001
Figure imgf001333_0001
Figure imgf001334_0001
Figure imgf001335_0001
Figure imgf001336_0001
Figure imgf001337_0001
Figure imgf001338_0001
Figure imgf001339_0001
Figure imgf001340_0001
Figure imgf001341_0001
Figure imgf001342_0001
Figure imgf001343_0001
Figure imgf001344_0001
Figure imgf001345_0001
Figure imgf001346_0001
Figure imgf001347_0001
Figure imgf001348_0001
Figure imgf001349_0001
Figure imgf001350_0001
Figure imgf001351_0001
Figure imgf001352_0001
Figure imgf001353_0001
Figure imgf001354_0001
Figure imgf001355_0001
Figure imgf001356_0001
Figure imgf001357_0001
Figure imgf001358_0001
Figure imgf001359_0001
Figure imgf001360_0001
Figure imgf001361_0001
Figure imgf001362_0001
Figure imgf001363_0001
Figure imgf001364_0001
Figure imgf001365_0001
Figure imgf001366_0001
Figure imgf001367_0001
Figure imgf001368_0001
Figure imgf001369_0001
Figure imgf001370_0001
Figure imgf001371_0001
Figure imgf001372_0001
Figure imgf001373_0001
Figure imgf001374_0001
Figure imgf001375_0001
Figure imgf001376_0001
Figure imgf001377_0001
Figure imgf001378_0001
Figure imgf001379_0001
Figure imgf001380_0001
Figure imgf001381_0001
Figure imgf001382_0001
Figure imgf001383_0001
Figure imgf001384_0001
Figure imgf001385_0001
Figure imgf001386_0001
Figure imgf001387_0001
Figure imgf001388_0001
Figure imgf001389_0001
Figure imgf001390_0001
Figure imgf001391_0001
Figure imgf001392_0001
Figure imgf001393_0001
Figure imgf001394_0001
Figure imgf001395_0001
Figure imgf001396_0001
Figure imgf001397_0001
Figure imgf001398_0001
Figure imgf001399_0001
Figure imgf001400_0001
Figure imgf001401_0001
Figure imgf001402_0001
Figure imgf001403_0001
Figure imgf001404_0001
Figure imgf001405_0001
Figure imgf001406_0001
Figure imgf001407_0001
Figure imgf001408_0001
Figure imgf001409_0001
Figure imgf001410_0001
Figure imgf001411_0001
Figure imgf001412_0001
Figure imgf001413_0001
Figure imgf001414_0001
Figure imgf001415_0001
Figure imgf001416_0001
Figure imgf001417_0001
Figure imgf001418_0001
Figure imgf001419_0001
Figure imgf001420_0001
Figure imgf001421_0001
Figure imgf001422_0001
Figure imgf001423_0001
Figure imgf001424_0001
Figure imgf001425_0001
Figure imgf001426_0001
Figure imgf001427_0001
Figure imgf001428_0001
Figure imgf001429_0001
Figure imgf001430_0001
Figure imgf001431_0001
Figure imgf001432_0001
Figure imgf001433_0001
Figure imgf001434_0001
Figure imgf001435_0001
Figure imgf001436_0001
Figure imgf001437_0001
Figure imgf001438_0001
Figure imgf001439_0001
Figure imgf001440_0001
Figure imgf001441_0001
Figure imgf001442_0001
Figure imgf001443_0001
Figure imgf001444_0001
Figure imgf001445_0001
Figure imgf001446_0001
Figure imgf001447_0001
Figure imgf001448_0001
Figure imgf001449_0001
Figure imgf001450_0001
Figure imgf001451_0001
Figure imgf001452_0001
Figure imgf001453_0001
Figure imgf001454_0001
Figure imgf001455_0001
Figure imgf001456_0001
Figure imgf001457_0001
Figure imgf001458_0001
Figure imgf001459_0001
Figure imgf001460_0001
Figure imgf001461_0001
Figure imgf001462_0001
Figure imgf001463_0001
Figure imgf001464_0001
Figure imgf001465_0001
Figure imgf001466_0001
Figure imgf001467_0001
Figure imgf001468_0001
Figure imgf001469_0001
Figure imgf001470_0001
Figure imgf001471_0001
Figure imgf001472_0001
Figure imgf001473_0001
Figure imgf001474_0001
Figure imgf001475_0001
Figure imgf001476_0001
Figure imgf001477_0001
Figure imgf001478_0001
Figure imgf001479_0001
Figure imgf001480_0001
Figure imgf001481_0001
Figure imgf001482_0001
Figure imgf001483_0001
Figure imgf001484_0001
Figure imgf001485_0001
Figure imgf001486_0001
Figure imgf001487_0001
Figure imgf001488_0001
Figure imgf001489_0001
Figure imgf001490_0001
Figure imgf001491_0001
Figure imgf001492_0001
Figure imgf001493_0001
Figure imgf001494_0001
Figure imgf001495_0001
Figure imgf001496_0001
Figure imgf001497_0001
Figure imgf001498_0001
Figure imgf001499_0001
Figure imgf001500_0001
Figure imgf001501_0001
Figure imgf001502_0001
Figure imgf001503_0001
Figure imgf001504_0001
Figure imgf001505_0001
Figure imgf001506_0001
Figure imgf001507_0001
Figure imgf001508_0001
Figure imgf001509_0001
Figure imgf001510_0001
Figure imgf001511_0001
Figure imgf001512_0001
Figure imgf001513_0001
Figure imgf001514_0001
Figure imgf001515_0001
Figure imgf001516_0001
Figure imgf001517_0001
Figure imgf001518_0001
Figure imgf001519_0001
Figure imgf001520_0001
Figure imgf001521_0001
[00103] Table 2 below describes non-coding target genes and their TSS locations.
Table 2 (following page). Non-coding Target Genes, Target Transcripts and TSS Locations
Figure imgf001523_0001
Figure imgf001524_0001
Figure imgf001525_0001
Figure imgf001526_0001
Figure imgf001527_0001
Figure imgf001528_0001
Figure imgf001529_0001
Figure imgf001530_0001
Figure imgf001531_0001
Figure imgf001532_0001
Figure imgf001533_0001
Figure imgf001534_0001
Figure imgf001535_0001
Figure imgf001536_0001
Figure imgf001537_0001
Figure imgf001538_0001
Figure imgf001539_0001
Figure imgf001540_0001
Figure imgf001541_0001
Figure imgf001542_0001
Figure imgf001543_0001
Figure imgf001544_0001
Figure imgf001545_0001
Figure imgf001546_0001
Figure imgf001547_0001
Figure imgf001548_0001
Figure imgf001549_0001
Figure imgf001550_0001
Figure imgf001551_0001
Figure imgf001552_0001
Figure imgf001553_0001
Figure imgf001554_0001
Figure imgf001555_0001
Figure imgf001556_0001
Figure imgf001557_0001
Figure imgf001558_0001
Figure imgf001559_0001
Figure imgf001560_0001
Figure imgf001561_0001
Figure imgf001562_0001
Figure imgf001563_0001
Figure imgf001564_0001
Figure imgf001565_0001
Figure imgf001566_0001
Figure imgf001567_0001
Figure imgf001568_0001
Figure imgf001569_0001
Figure imgf001570_0001
Figure imgf001571_0001
Figure imgf001572_0001
Figure imgf001573_0001
Figure imgf001574_0001
Figure imgf001575_0001
Figure imgf001576_0001
Figure imgf001577_0001
Figure imgf001578_0001
Figure imgf001579_0001
Figure imgf001580_0001
Figure imgf001581_0001
Figure imgf001582_0001
Figure imgf001583_0001
Figure imgf001584_0001
Figure imgf001585_0001
Figure imgf001586_0001
Figure imgf001587_0001
Figure imgf001588_0001
Figure imgf001589_0001
Figure imgf001590_0001
Figure imgf001591_0001
Figure imgf001592_0001
Figure imgf001593_0001
Figure imgf001594_0001
Figure imgf001595_0001
Figure imgf001596_0001
Figure imgf001597_0001
Figure imgf001598_0001
Figure imgf001599_0001
Figure imgf001600_0001
Figure imgf001601_0001
Figure imgf001602_0001
Figure imgf001603_0001
Figure imgf001604_0001
Figure imgf001605_0001
Figure imgf001606_0001
Figure imgf001607_0001
Figure imgf001608_0001
Figure imgf001609_0001
Figure imgf001610_0001
Figure imgf001611_0001
Figure imgf001612_0001
Figure imgf001613_0001
Figure imgf001614_0001
Figure imgf001615_0001
Figure imgf001616_0001
Figure imgf001617_0001
Figure imgf001618_0001
Figure imgf001619_0001
Figure imgf001620_0001
Figure imgf001621_0001
Figure imgf001622_0001
Figure imgf001623_0001
Figure imgf001624_0001
Figure imgf001625_0001
Figure imgf001626_0001
Figure imgf001627_0001
Figure imgf001628_0001
Figure imgf001629_0001
Figure imgf001630_0001
Figure imgf001631_0001
Figure imgf001632_0001
Figure imgf001633_0001
Figure imgf001634_0001
Figure imgf001635_0001
Figure imgf001636_0001
Figure imgf001637_0001
Figure imgf001638_0001
Figure imgf001639_0001
Figure imgf001640_0001
Figure imgf001641_0001
Figure imgf001642_0001
Figure imgf001643_0001
Figure imgf001644_0001
Figure imgf001645_0001
Figure imgf001646_0001
Figure imgf001647_0001
Figure imgf001648_0001
Figure imgf001649_0001
Figure imgf001650_0001
Figure imgf001651_0001
Figure imgf001652_0001
Figure imgf001653_0001
Figure imgf001654_0001
Figure imgf001655_0001
Figure imgf001656_0001
Figure imgf001657_0001
Figure imgf001658_0001
Figure imgf001659_0001
Figure imgf001660_0001
Figure imgf001661_0001
Figure imgf001662_0001
Figure imgf001663_0001
Figure imgf001664_0001
Figure imgf001665_0001
Figure imgf001666_0001
Figure imgf001667_0001
Figure imgf001668_0001
Figure imgf001669_0001
Figure imgf001670_0001
Figure imgf001671_0001
Figure imgf001672_0001
Figure imgf001673_0001
Figure imgf001674_0001
Figure imgf001675_0001
Figure imgf001676_0001
Figure imgf001677_0001
Figure imgf001678_0001
Figure imgf001679_0001
Figure imgf001680_0001
Figure imgf001681_0001
Figure imgf001682_0001
Figure imgf001683_0001
Figure imgf001684_0001
Figure imgf001685_0001
Figure imgf001686_0001
Figure imgf001687_0001
Figure imgf001688_0001
Figure imgf001689_0001
Figure imgf001690_0001
Figure imgf001692_0001
Figure imgf001693_0001
Figure imgf001694_0001
Figure imgf001695_0001
Figure imgf001696_0001
Figure imgf001697_0001
Figure imgf001698_0001
Figure imgf001699_0001
Figure imgf001700_0001
Figure imgf001701_0001
Figure imgf001702_0001
Figure imgf001703_0001
Figure imgf001704_0001
Figure imgf001705_0001
Figure imgf001706_0001
Figure imgf001707_0001
Figure imgf001708_0001
Figure imgf001709_0001
Figure imgf001710_0001
Figure imgf001711_0001
Figure imgf001712_0001
Figure imgf001713_0001
Figure imgf001714_0001
Figure imgf001715_0001
Figure imgf001716_0001
Figure imgf001717_0001
Figure imgf001718_0001
Figure imgf001719_0001
Figure imgf001720_0001
Figure imgf001721_0001
Figure imgf001722_0001
Figure imgf001723_0001
Figure imgf001724_0001
Figure imgf001725_0001
Figure imgf001726_0001
Figure imgf001727_0001
Figure imgf001728_0001
Figure imgf001729_0001
Figure imgf001730_0001
Figure imgf001731_0001
Figure imgf001732_0001
Figure imgf001733_0001
Figure imgf001734_0001
Figure imgf001735_0001
Figure imgf001736_0001
Figure imgf001737_0001
Figure imgf001738_0001
Figure imgf001739_0001
Figure imgf001740_0001
Figure imgf001741_0001
Figure imgf001742_0001
Figure imgf001743_0001
Figure imgf001744_0001
Figure imgf001745_0001
Figure imgf001746_0001
Figure imgf001747_0001
Figure imgf001748_0001
Figure imgf001749_0001
Figure imgf001750_0001
Figure imgf001751_0001
Figure imgf001752_0001
Figure imgf001753_0001
Figure imgf001754_0001
Figure imgf001755_0001
Figure imgf001756_0001
Figure imgf001757_0001
Figure imgf001758_0001
Figure imgf001759_0001
Figure imgf001760_0001
Figure imgf001761_0001
Figure imgf001762_0001
Figure imgf001763_0001
Figure imgf001764_0001
Figure imgf001765_0001
Figure imgf001766_0001
Figure imgf001767_0001
Figure imgf001768_0001
Figure imgf001769_0001
Figure imgf001770_0001
Figure imgf001771_0001
Figure imgf001772_0001
Figure imgf001773_0001
Figure imgf001774_0001
Figure imgf001775_0001
Figure imgf001776_0001
Figure imgf001777_0001
Figure imgf001778_0001
Figure imgf001779_0001
Figure imgf001780_0001
Figure imgf001781_0001
Figure imgf001782_0001
Figure imgf001783_0001
Figure imgf001784_0001
Figure imgf001785_0001
Figure imgf001786_0001
Figure imgf001787_0001
Figure imgf001788_0001
Figure imgf001789_0001
Figure imgf001790_0001
Figure imgf001791_0001
Figure imgf001792_0001
Figure imgf001793_0001
Figure imgf001794_0001
Figure imgf001795_0001
Figure imgf001796_0001
Figure imgf001797_0001
Figure imgf001798_0001
Figure imgf001799_0001
Figure imgf001800_0001
Figure imgf001801_0001
Figure imgf001802_0001
Figure imgf001803_0001
Figure imgf001804_0001
Figure imgf001805_0001
Figure imgf001806_0001
Figure imgf001807_0001
Figure imgf001808_0001
Figure imgf001809_0001
Figure imgf001810_0001
Figure imgf001811_0001
Figure imgf001812_0001
Figure imgf001813_0001
Figure imgf001814_0001
Figure imgf001815_0001
Figure imgf001816_0001
Figure imgf001817_0001
Figure imgf001818_0001
Figure imgf001819_0001
Figure imgf001820_0001
Figure imgf001821_0001
Figure imgf001822_0001
Figure imgf001823_0001
Figure imgf001824_0001
Figure imgf001825_0001
Figure imgf001826_0001
Figure imgf001827_0001
Figure imgf001828_0001
Figure imgf001829_0001
Figure imgf001830_0001
Figure imgf001831_0001
Figure imgf001832_0001
Figure imgf001833_0001
Figure imgf001834_0001
Figure imgf001835_0001
Figure imgf001836_0001
Figure imgf001837_0001
Figure imgf001838_0001
Figure imgf001839_0001
Figure imgf001840_0001
Figure imgf001841_0001
Figure imgf001842_0001
Figure imgf001843_0001
Figure imgf001844_0001
Figure imgf001845_0001
Figure imgf001846_0001
Figure imgf001847_0001
Figure imgf001848_0001
Figure imgf001849_0001
Figure imgf001850_0001
Figure imgf001851_0001
Figure imgf001852_0001
Figure imgf001853_0001
Figure imgf001854_0001
Figure imgf001855_0001
Figure imgf001856_0001
Figure imgf001857_0001
Figure imgf001858_0001
Figure imgf001859_0001
Figure imgf001860_0001
Figure imgf001861_0001
Figure imgf001862_0001
Figure imgf001863_0001
Figure imgf001864_0001
Figure imgf001865_0001
Figure imgf001866_0001
Figure imgf001867_0001
Figure imgf001868_0001
Figure imgf001869_0001
Figure imgf001870_0001
Figure imgf001871_0001
Figure imgf001872_0001
Figure imgf001873_0001
Figure imgf001874_0001
Figure imgf001875_0001
Figure imgf001876_0001
Figure imgf001877_0001
Figure imgf001878_0001
Figure imgf001879_0001
Figure imgf001880_0001
Figure imgf001881_0001
Figure imgf001882_0001
Figure imgf001883_0001
Figure imgf001884_0001
Figure imgf001885_0001
Figure imgf001886_0001
Figure imgf001887_0001
Figure imgf001888_0001
Figure imgf001889_0001
Figure imgf001890_0001
Figure imgf001891_0001
Figure imgf001892_0001
Figure imgf001893_0001
Figure imgf001894_0001
Figure imgf001895_0001
Figure imgf001896_0001
Figure imgf001897_0001
Figure imgf001898_0001
Figure imgf001899_0001
Figure imgf001900_0001
Figure imgf001901_0001
Figure imgf001902_0001
Figure imgf001903_0001
Figure imgf001904_0001
Figure imgf001905_0001
Figure imgf001906_0001
Figure imgf001907_0001
Figure imgf001908_0001
Figure imgf001909_0001
Figure imgf001910_0001
Figure imgf001911_0001
Figure imgf001912_0001
Figure imgf001913_0001
Figure imgf001914_0001
Figure imgf001915_0001
Figure imgf001916_0001
Figure imgf001917_0001
Figure imgf001918_0001
Figure imgf001919_0001
Figure imgf001920_0001
Figure imgf001921_0001
Figure imgf001922_0001
Figure imgf001923_0001
Figure imgf001924_0001
Figure imgf001925_0001
Figure imgf001926_0001
Figure imgf001927_0001
Figure imgf001928_0001
Figure imgf001929_0001
Figure imgf001930_0001
Figure imgf001931_0001
Figure imgf001932_0001
Figure imgf001933_0001
Figure imgf001934_0001
Figure imgf001935_0001
Figure imgf001936_0001
Figure imgf001937_0001
Figure imgf001938_0001
Figure imgf001939_0001
Figure imgf001940_0001
Figure imgf001941_0001
Figure imgf001942_0001
Figure imgf001943_0001
Figure imgf001944_0001
Figure imgf001945_0001
Figure imgf001946_0001
Figure imgf001947_0001
Figure imgf001948_0001
Figure imgf001949_0001
Figure imgf001950_0001
Figure imgf001951_0001
Figure imgf001952_0001
Figure imgf001953_0001
Figure imgf001954_0001
Figure imgf001955_0001
Figure imgf001956_0001
[00104] The saRNAs of the present invention may be single-stranded and comprise 14-
30 nucleotides. The sequence of a single-stranded saRNA may have at least 60%, 70%>, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs; 4048-315235, 318727- 584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208- 2585259.
[00105] In one embodiment, the single-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532- 1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259.
[00106] In one embodiment, the saRNA is a single-stranded saRNA which comprises an antisense sequence such as, but not limited to any of the antisense sequences described in the sequence listing referenced at the beginning of this application.
[00107] In one embodiment, the saRNA is a single-stranded saRNA which comprises an antisense sequence such as, but not limited to any of the sense sequences described in the sequence listing referenced at the beginning of this application.
[00108] The single stranded saRNAs of the present invention ma be modified or unmodified.
[00109] In one embodiment, the single-stranded saRNA may have a 3' tail.
[00110] In one embodiment, the saRNAs may be double-stranded. The two strands form a duplex, also known as an saRNA duplex, and each strand comprises 14-30 nucleotides. The first strand of a double-stranded saRNA may have at least 60%, 70%, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062- 913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259. In one embodiment, the first strand of the double-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259. The second strand of a double- stranded saRNA may have at least 60%, 70%o, 80% or 90% identity with a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931, 1564373-1879188, and 1889208-2585259. In one embodiment, the second strand of the double-stranded saRNA comprises a sequence such as, but not limited to, SEQ ID NOs: 4048-315235, 318727-584784, 589062-913309, 917532-1241079, 1245402-1559931 , 1564373- 1879188, and 1889208-2585259. In one embodiment, the double-stranded saRNA may have a 3' overhang on each strand.
[00111] In one embodiment, the saRNA of the present invention is an saRNA duplex.
The saRNA duplex may be a pair of sense and antisense sequences such as, but not limited to, any of the sense sequence and corresponding antisense sequences described in the sequence listing referenced at the beginning of this application. The saRNA of the present invention may be the pair of the sense sequence and antisense sequence described in the sequence listing referenced at the beginning of this application,
[00112] The double-stranded saRNA of the present invention may be modified or unmodified.
Bifunction Oligonucleotides
I ( 01 131 Bifunction or dual-functional oligonucleotides, e.g., saRNA may be designed to up-regulate the expression of a first gene and down-regulate the expression of at least one second gene. One strand of the dual-functional oligonucleotide activates the expression of the first gene and the other strand inhibits the expression of the second gene. Each strand might further comprise a Dicer substrate sequence.
Chemical Modifications of saRNA
[00114] Herein, in saRNA, the terms "modification" or, as appropriate, "modified" refer to structural and/or chemical modifications with respect to A, G, U or C ribonucleotides.
Nucleotides in the saRNAs of the present invention may comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynueleotides. The saRNA of the present invention may include any useful modification, such as to the sugar, the nucleobase, or the intemucleoside linkage (e.g. to a linking phosphate / to a phosphodiester linkage / to the phosphodiester backbone). One or more atoms of a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro). In certain embodiments, modifications (e.g., one or more modifications) are present in each of the sugar and the intemucleoside linkage. Modifications according to the present invention may be modifications of ribonucleic acids (RNAs) to deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof In a non-limiting example, the 2'-OH of U is substituted with 2 ' OMc.
[00115] In one embodiment, the saRNAs of the present invention may comprise at least one modification described herein,
[00116] In another embodiment, the saRNA is an saRNA duplex and the sense strand and antisense sequence may independently comprise at least one modification. As a non-limiting example, the sense sequence may comprises a modification and the antisense strand may be unmodified. As another non-limiting example, the antisense sequence may comprises a modification and the sense strand may be unmodified. As yet another non-limiting example, the sense sequence may comprises more than one modification and the antisense strand may comprise one modification. As a non-limiting example, the antisense sequence may comprises more than one modification and the sense strand may comprise one modification.
[00117] The saRNA of the present invention can include a combination of modifications to the sugar, the nucleobase, and/or the intemueleoside linkage. These combinations can include any one or more modifications described herein or in International Application Publication
WO2013/052523 filed October 3, 2012, in particular Formulas (Ia)-(Ia-5), (Ib)-(If), (Ila)-(IIp), (IIb-1 ), (IIb-2), (IIc-l )-(IIc-2), (IIn-1), (IIn-2), (IVa)-(IVl), and (IXa)-(IXr)), the contents of which are incorporated herein by reference in their entirety.
[00118] The saRNA of the present invention may or may not be uniformly modified along the entire length of the molecule. For example, one or more or all types of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may or may not be uniformly modified in the saRNA of the invention. In some embodiments, all nucleotides X in an saRNA of the invention are modified, wherein X may be any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.
[00119] Different sugar modifications, nucleotide modifications, and/or intemueleoside linkages (e.g., backbone structures) may exist at various positions in an saRNA. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of an saRNA such that the function of saRNA is not substantially decreased. The saRNA of the present invention may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e. any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%).
[00120] In some embodiments, the saRNA of the present invention may be modified to be a spherical nucleic acid (SNA) or a circular nucleic acid. The terminals of the saRNA of the present invention may be linked by chemical reagents or enzymes, producing spherical saRNA that has no free ends. Spherical saRNA is expected to be more stable than its linear counterpart and to be resistant to digestion with RNase R exonuclease. Spherical saRNA may former comprise other structural and/or chemical modifications with respect to A, G, U or C
ribonucleotides.
[00121] In some embodiments, the saRNA of the present invention may comprise inverted deoxy abasic modifications on the passenger strand. The at least one inverted deoxy abasic modification may be on 5' end, or 3' end, or both ends of the passenger strand. The inverted deoxy basic modification may encourage preferential loading of the guide strand,
[00122] The saRNA of the present invention may be modified with any modifications of an oligonucleotide or polynucleotide disclosed in pages 136 to 247 of PCT Publication
W()2()13T 1666 published Oct. 10, 2013, the contents of which are incorporated herein by reference in their entirety.
saRNA Conjugates and Combinations
[00123] Conjugation may result in increased stability and/or half-life and may be particularly useful in targeting the saRNA of the present invention to specific sites in the cell, tissue or organism. The saRNA of the present invention can be designed to be conjugated to other polynucleotides, 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 (e.g. EDTA), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEGJi, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases, proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell, hormones and hormone receptors, non-peptidic species, such as lipids, lectins,
carbohydrates, vitamins, cofactors, or a drag. Suitable conjugates for nucleic acid molecules are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
[00124] According to the present invention, saRNA of the present invention may be administered with, or further include one or more of RNAi agents, small interfering RNAs (siRNAs), small hairpin RNAs (shRNAs), long non-coding RNAs (IncRNAs), enhancer RNAs, enhancer-derived RNAs or enhancer-driven RNAs (eRNAs), mieroRNAs (miRNAs), miRNA binding sites, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, ap tamers or vectors, and the like to achieve different functions. The one or more RNAi agents, small interfering RNAs (siRNAs), small hairpin RNAs (shRNAs), long non- coding RNAs (IncRNA), mieroRNAs (miRNAs), miRNA binding sites, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers or vectors may comprise at least one modification or substitution.
[00125] In some embodiments, the modification is selected from a chemical substitution of the nucleic acid at a sugar position, a chemical substitution at a phosphate position and a chemical substitution at a base position. In other embodiments, the chemical modification is selected from incorporation of a modified nucleotide; 3' capping; conjugation to a high molecular weight, non-immunogenic compound; conjugation to a lipophilic compound; and incorporation of phosphorothioate into the phosphate backbone. In one embodiment, the high molecular weight, non-immunogenic compound is polyalkylene glycol, or polyethylene glycol (PEG).
[00126] In one embodiment, saRNA comprising at least one modification may show efficacy in proliferating cells. [00127] In one embodiment, saRNA of the present invention may be attached to a transgene so it can be co-expressed from an RNA polymerase II promoter. In a non-limiting example, saRNA of the present invention is attached to green fluorescent protein gene (GFP).
[00128] In one embodiment, saRNA of the present invention may be attached to a DNA or RNA aptamer, thereby producing saRNA-aptamer conjugate, Aptamers are oligonucleotides or peptides with high selectivity, affinity and stability. They assume specific and stable three- dimensional shapes, thereby providing highly specific, tight binding to target molecules. An aptamer may be a nucleic acid species that has been engineered through repeated rounds οΐίη vitro selection or equivalently, SELEX (systematic evolution of ligands by exponential enrichment) to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms. Nucleic acid aptamers have specific binding affinity to molecules through interactions other than classic Watson-Crick base pairing. Nucleic acid aptamers, like peptides generated by phage display or monoclonal antibodies (mAhs), are capable of specifically binding to selected targets and, through binding, block their targets' ability to function. In some cases, aptamers may also be peptide aptamers. For any specific molecular target, nucleic acid aptamers can be identified from combinatorial libraries of nucleic acids, e.g. by SELEX. Peptide aptamers may be identified using a yeast two hybrid system. A skilled person is therefore able to design suitable aptamers for delivering the saRNAs or cells of the present invention to target cells such as liver cells. DNA aptamers, RNA aptamers and peptide aptamers are contemplated. Administration of saRNA of the present invention to the liver using liver-specific aptamers is preferred.
[00129] As used herein, a typical nucleic acid aptamer is approximately 10-15 kDa in size (20-45 nucleotides), binds its target with at least nanomolar affinity, and discriminates against closely related targets. Nucleic acid aptamers may be ribonucleic acid, deoxyribonucleic acid, or mixed ribonucleic acid and deoxyribonucleic acid. Aptamers may be single-stranded ribonucleic acid, deoxyribonucleic acid or mixed ribonucleic acid and deoxyribonucleic acid. Aptamers may comprise at least one chemical modification.
[00130] A suitable nucleotide length for an aptamer ranges from about 15 to about 100 nucleotides (nt), and in various other embodiments, 15-30 nt, 20-25 nt, 30-100 nt, 30-60 nt, 25- 70 nt, 25-60 nt, 40-60 nt, 25-40 nt, 30-40 nt, any of 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39 or 40 nt or 40-70 nt in length. However, the sequence can be designed with sufficient flexibility such that it can accommodate interactions of a tamers with two targets at the distances described herein. Aptamers may be further modified to provide protection from nuclease and other enzymatic activities. The aptamcr sequence can be modified by any suitable methods known in the art.
[ 00131] The saRNA-aptamer conjugate ma be formed using any known method for linking two moieties, such as direct chemical bond formation, linkage via a linker such as streptavidin and so on.
[00132] In one embodiment, saRNA of the present invention may be attached to an antibody. Methods of generating antibodies against a target cell surface receptor are well known. The saRNAs of the invention may be attached to such antibodies with known methods, for example using RNA carrier proteins. The resulting complex may then be administered to a subject and taken up by the target cells via receptor-mediated endocytosis.
[00133] In one embodiment, saRNA of the present invention may be conjugated with lipid moieties such as a cholesterol moiety (Letsinger et al, Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), eholie acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4: 1053-1060), a thioether, e.g., beryl-5-tritylthiol (Manoharan et al, Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manoharan et al, Biorg. Med. Chem. Let, 1993, 3:2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Bchmoaras et ah, EMBO J, 1991, 10: 1 1 1 1-1 1 18; Kabanov et al, FEBS Lett., 1990, 259:327-330; Svinarchuk et al, Biochimie, 1993, 75:49-54), a phospholipid, e.g., di- hexadecyl-rac -glycerol or triethyl-ammonium 1 ,2-di-0-hexadecyl-rac-glycero-3-Hphosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36:3651 -3654; Shea et al., Nucl. Acids Res., 1990, 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al, Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manohara et ah, Tetrahedron Lett., 1995, 36:3651 -3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229- 237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et ah, J. Pharmacol. Exp. Ther., 1996, 277:923-937), the content of each of which is herein incorporated by reference in its entirety.
[00134] In one embodiment, the saRNA of the present invention is conjugated with a ligand. In one non-limiting example, the ligand may be any ligand disclosed in US
20130184328 to Manoharan et ah, the contents of which are incorporated herein by reference in their entirety. The conjugate has a formula of Ligand-[linker]0ptionai-[tether]optionai-oligonucleotide agent. The oligonucleotide agent may comprise a subunit having formulae (I) disclosed by US 20130184328 to Manoharan et al., the contents of which are incorporated herein by reference in their entirety. In another non-limiting example, the ligand may be any ligand disclosed in US 20130317081 to Akinc et al., the contents of which are incorporated herein by reference in their entirety, such as a lipid, a protein, a hormone, or a carbohydrate ligand of Formula 1 1 -X XV I . The ligand may be coupled with the saRNA with a bivalent or traivalent branched linker in Formula XXXI-XXXV disclosed in Akinc.
[00135] Representative U.S. patents that teach the preparation of such nucleic acid lipid conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105;
5,525,465; 5,541 ,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,591,584; 5,109,124;
5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044;
4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582;
4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,1 2,963; 5,214,136; 5,245,022;
5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371 ,241 , 5,391,723;
5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142;
5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941 , the content of each of which is herein incorporated by reference in its entirety.
[00136] The saRNA of the present invention may be provided in combination with other active ingredients known to have an effect in the particular method being considered. The other active ingredients may be administered simultaneously, separately, or sequentially with the saRNA of the present invention. In one embodiment, saRNA of the present invention is administered with saRNA modulating a different target gene. Non-limiting examples include saRNA that modulates albumin, insulin or FTNF4A genes. Modulating any gene may be achieved using a single saRNA or a combination of two or more different saRNAs. Non-limiting examples of saRNA that can be administered with saRNA of the present invention include saRNA modulating albumin or BNF4A disclosed in International Publication WO 2012/175958 filed June 20, 2012, saRNA modulating insulin disclosed in International Publications WO 2012/046084 and WO 2012/046085 both filed Oct. 10, 2011, saRNA modulating human progesterone receptor, human major vault protein (IrMVP), E-cadherin gene, p53 gene, or PTEN gene disclosed in US Pat. No. 7,709,456 filed November 13, 2006 and US Pat. Publication US 2010/0273863 filed April 23, 2010, saRNAs targeting p21 gene disclosed in International Publication WO 2006/113246 filed April 1 1, 2006, any nucleic acid disclosed in
WO2012/065143 filed November 12, 2011 that upregulates the expression of genes in Table 8 of WO 2012/065143 or increases the expression of a tumor suppressor, any oligonucleotide that activates target genes in Table 4 of W ( ) 2013 / 173 35 filed May 16, 2013, any oligonucleotide that activates target genes in Table 4 of WO2013/173637 filed May 16, 2013, any
oligonucleotide complementary to a sequence selected from the sequences in SEQ ID NOs: 1- 1212 of WO2013/173652 filed May 16, 2013, any oligonucleotide modulating APOA1 and ABCA1 gene expressions disclosed in WO2013173647 filed May 16, 2013, any oligonucleotide modulating SMN family gene expressions disclosed in WO2013173638 filed May 16, 2013, any oligonucleotide modulating PTEN gene expression disclosed in WO2013173605 filed May 16, 2013, any oligonucleotide modulating MECP2 gene expression disclosed in WO2013173608 filed May 16, 2013, any oligonucleotide modulating ATP2A2 gene expression disclosed in WO2013173598 filed May 16, 2013, any oligonucleotide modulating UTRN gene expression disclosed in WO2013173645 filed May 16, 2013, any nucleic acid molecule that modulates the expression of CD97, TS-a, C/EBP delta, CDC23, P1NK1, HIFla, Gnbp3g, Adrenomedullin AMI receptor, 3-oxoacid CoA transferase, Cathepsin W or BACE1 disclosed in US Pat. No. 8,288,354 filed December 28, 2006, antagoNAT with formula (1) disclosed in US 2013/0245099 filed November 17, 2011, any antagoNAT that upregulates the expression of hemoglobin (HBF/HBG) polynucleotides disclosed in US Pat, No. 8,318,690 filed April 30, 2010, any antisense oligonucleotide that increases the expression of apolipoprotein (ApoAl)
polynucleotide disclosed in US Pat. No. 8,153,696 filed October 2, 2009 (CURNA), the contents of each of which are incorporated herein by reference in their entirety.
[00137] In on embodiment, the saRNA is conjugated with a carbohydrate ligand, such as any carbohydrate ligand disclosed in US Pat No. 8106022 and 8828956 to Manoharan et al. (Alnylam Pharmaceuticals), the contents of which are incorporated herein by reference in their entirety. For example, the carbohydrate ligand may be monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide. These carbohydrate- conjugated RNA agents may target the parenchymal cells of the liver. In one embodiment, the saRNA is conjugated with more than one carbohydrate ligand, preferably two or three. In one embodiment, the saRNA is conjugated with one or more galactose moiety. In another embodiment, the saRNA is conjugated at least one (e.g., two or three or more) lactose molecules (lactose is a glucose coupled to a galactose). In another embodiment, the saRNA is conjugated with at least one (e.g., two or three or more) N-Acetyl-Galaetosamine (GalNAc), N-Ac- Glucosamine (GluNAc), or mannose (e.g., mannose-6-phosphate). In one embodiment, the saRNA is conjugated with at least one mannose ligand, and the conjugated saRNA targets macrophages.
[00138] In one embodiment, saRNA of the present invention is administered with a small interfering RNA or siRNA that inhibits the expression of a gene.
[00139] In one embodiment, saRNA of the present invention is administered with one or more drugs for therapeutic purposes.
II. Composition of the invention
[00140] One aspect of the present invention provides pharmaceutical compositions comprising a small activating RNA (saRNA) that upregulates a target gene, and at least one pharmaceutically acceptable carrier.
Formulation, Delivery, Administration, and Dosing
[00141] Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired. Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference in its entirety). The use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other eomponent(s) of the pharmaceutical composition.
[00142] In some embodiments, compositions are administered to humans, human patients or subjects. For the purposes of the present disclosure, the phrase "active ingredient" generally refers to saRNA to be delivered as described herein. [00143] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the
pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
[00144] in one embodiment, the efficacy of the formulated saRNA described herein may be determined in proliferating cells,
[00145] Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.
[00146] A pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
[00147] Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1 % and 100%, e.g., between .5 and 50%, between 1 -30%, between 5-80%, at least 80% (w/w) active ingredient.
[00148] In some embodiments, the formulations described herein may contain at least one saRNA. As a non-limiting example, the formulations may contain 1 , 2, 3, 4 or 5 saRNAs with different sequences. In one embodiment, the formulation contains at least three saRNAs with different sequences. In one embodiment, the formulation contains at least five saRNAs with different sequences.
[00149] The saRNA of the present invention can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the saRNA); (4) alter the biodistribution (e.g., target the saRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo.
[00150] In addition to traditional excipients such as any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, excipients of the present invention can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core-shell nanoparticles, peptides, proteins, cells transfected with saRNA (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics and combinations thereof. Accordingly, the formulations of the invention can include one or more excipients, each in an amount that together increases the stability of the saRNA and/or increases cell transfection by the saRNA. Further, the saRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles. Pharmaceutically acceptable carriers, excipients, and delivery agents for nucleic acids that may be used in the formulation with the saRNA of the present invention are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
[ 00151] In one embodiment, the saRNA of the present invention comprises two single
RNA strands that are 1 nucleotides in length each that are annealed to form a double-stranded saRNA as the active ingredient. The composition further comprises a salt buffer composed of 50mM Tris-HCl, pH 8.0, lOOmM NaCl and 5mM EDTA.
[00152] In another embodiment, the saRNA of the present invention may be delivered with dendrimers. Dendrimers are highly branched macromolecules. In one embodiment, the saRNA of the present invention is complexed with structurally flexible poly(amidoamine) (PAMAM) dendrimers for targeted in vivo delivery. The complex is called sa R A-d cndri mers . Dendrimers have a high degree of molecular uniformity, narrow molecular weight distribution, specific size and shape characteristics, and a highly-functional izcd terminal surface. The manufacturing process is a series of repetitive steps starting with a central initiator core. Each subsequent growth step represents a new generation of polymers with a larger molecular diameter and molecular weight, and more reactive surface sites than the preceding generation.
[00153] PAMAM dendrimers are efficient nucleotide delivery systems that bear primary amine groups on their surface and also a tertiary amine group inside of the structure. The primary amine group participates in nucleotide binding and promotes their cellular uptake, while the buried tertiary amino groups act as a proton sponge in endosomes and enhance the release of nucleic acid into the cytoplasm. These dendrimers protect the saRNA carried by them from ribonuclease degradation and achieves substantial release of saRNA over an extended period of time via endocytosis for efficient gene targeting. The in vivo efficacy of these nanoparticles have previously been evaluated where biodistribution studies show that the dendrimers preferentially accumulate in peripheral blood mononuclear cells and live with no discernible toxicity {see Zhou et al., Molecular Ther. 201 1 Vol 1 , 2228-2238, the contents of which are incorporated herein by reference in their entirety). PAMAM' dendrimers may comprise a triethanolamine (TEA) core, a diaminobutane (DAB) core, a cystamine core, a diaminohexane (HEX) core, a
diamonododecane (DODE) core, or an ethylenedi amine (EDA) core. In one embodiment, PAMAM dendrimers comprise a TEA core or a DAB core.
Lipidoids
[ 00154] The synthesis of lipidoids has been extensively described and formulations containing these compounds are particularly suited for delivery of oligonucleotides or nucleic acids (see Mahon et al, Bioconjug Chem. 2010 21 :1448-1454; Schroeder et al, J Intern Med. 2010 267:9-21 ; Akinc et al, Nat Biotechnol. 2008 26:561 -569; Love et al, Proc Natl Acad Sci U S A. 2010 107: 1864-1869; Siegwart et al, Proc Natl Acad Sci U S A. 201 1 108:12996-3001 ; all of which are incorporated herein in their entireties).
[ 00155] While these lipidoids have been used to effectively deliver double-stranded small interfering RNA molecules in rodents and non-human primates (see Akinc et al., Nat Biotechnol. 2008 26:5 1 -569; Frank- amenet sky et al., Proc Natl Acad Sci U S A. 2008 105:1 1915-1 1920; Akinc et al, Mol Ther. 2009 17:872-879; Love et al., Proc Natl Acad Sei U S A. 2010 107:1864- 1869; Leusehner et al., Nat Biotechnol. 201 1 29: 1005-1010; all of which is incorporated herein in their entirety), the present disclosure contemplates their formulation and use in delivering saRNA. Complexes, micelles, liposomes or particles can be prepared containing these lipidoids and therefore, can result in an effective delivery of the saRNA following the injection of a lipidoid formulation via localized and/or systemic routes of administration. Lipidoid complexes of saRNA can be administered by various means including, but not limited to, intravenous, intramuscular, or subcutaneous routes,
[00156] In vivo delivery of nucleic acids may be affected by many parameters, including, but not limited to, the formulation composition, nature of particle PEGylation, degree of loading, oligonucleotide to lipid ratio, and biophysical parameters such as, but not limited to, particle size (Akinc et al., Mol Ther. 2009 17:872-879; the contents of which are herein incorporated by reference in its entirety). As an example, small changes in the anchor chain length of
poly(ethylene glycol) (PEG) lipids may result in significant effects on in vivo efficacy.
Formulations with the different lipidoids, including, but not limited to penta[3-(l - laurylaminopropionyl)]-triethylenetetramine hydrochloride (TETA-5LAP; aka 98N12-5, see Murugaiah et al, Analytical Biochemistry, 401 :61 (2010); the contents of which are herein incorporated by reference in its entirety), C 12-200 (including derivatives and variants), and MD1 , can be tested for in vivo activity.
[00157] The lipidoid referred to herein as "98N12-5" is disclosed by Akinc et al., Mol
Ther. 2009 17:872-879 and the contents of which is incorporated by reference in its entirety.
[00158] The lipidoid referred to herein as "CI 2-200" is disclosed by Love et al., Proc
Natl Acad Sci U S A. 2010 107: 1864-1869 and Liu and Huang, Molecular Therapy. 2010 669- 670; the contents of both of which are herein incorporated by reference in their entirety. The lipidoid formulations can include particles comprising either 3 or 4 or more components in addition to the saRNA. As an example, formulations with certain lipidoids, include, but are not limited to, 98N12-5 and may contain 42% lipidoid, 48% cholesterol and 10% PEG (C14 alkyl chain length). As another example, formulations with certain lipidoids, include, but are not limited to, C12-200 and may contain 50% lipidoid, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and 1.5% PEG-DMG. [00159] In one embodiment, an saRNA formulated with a lipidoid for systemic intravenous administration can target the liver. For example, a final optimized intravenous formulation using saRNA and comprising a lipid molar composition of 42% 98N12-5, 48% cholesterol, and 10% PHG-lipid with a final weight ratio of about 7,5 to 1 total lipid to saRNA and a C14 alkyl chain length on the PEG lipid, with a mean particle size of roughly 50-60 nm, can result in the distribution of the formulation to be greater than 90%o to the liver, (see, Akinc et al, Mol Ther. 2009 17:872-879; the contents of which are herein incorporated by reference in its entirety). In another example, an intravenous formulation using a CI 2-200 (see published international application WO2010129709, the contents of which is herein incorporated by reference in their entirety) lipidoid may have a molar ratio of 50/10/38.5/1.5 of C12- 200/disteroylphosphatidyl choline/cholesterol/PEG-DMG, with a weight ratio of 7 to 1 total lipid to nucleic acid and a mean particle size of 80 nm may be effective to deliver saRNA (see, Love et al., Proc Natl Acad Sci U S A. 2010 107: 1864-1869, the contents of which are herein incorporated by reference in its entirety).
[00160] In another embodiment, an MD1 lipidoid-containing formulation may be used to effectively deliver saRNA to hepatocytes in vivo. The characteristics of optimized lipidoid formulations for intramuscular or subcutaneous routes may vary significantly depending on the target cell type and the ability of formulations to diffuse through the extracellular matrix into the blood stream. While a particle size of less than 150 nm may be desired for effective hepatocyte delivery due to the size of the endothelial fenestrae (see, Akinc et al., Mol Ther. 2009 17:872- 879, the contents of which are herein incorporated by reference in its entirety), use of a lipidoid- formulated saRNA to deliver the formulation to other cells types including, but not limited to, endothelial cells, myeloid cells, and muscle cells may not be similarly size-limited.
[00161] Use of lipidoid formulations to deliver siRNA in vivo to other non-hep atocyte cells such as myeloid cells and endothelium has been reported (see Akinc et al, Nat Biotechnol. 2008 26:561-569; Leuschner et al, Nat Biotechnol. 2011 29:1005-1010; Cho et al. Adv. Fund. Mater. 2009 19:31 12-3118; 8th International Judah Folkman Conference, Cambridge, MA October 8-9, 2010; the contents of each of which is herein incorporated by reference in its entirety). Effective delivery to myeloid cells, such as monocytes, lipidoid formulations may have a similar component molar ratio. Different ratios of lipidoids and other components including, but not limited to, disteroylphosphatidyl choline, cholesterol and PEG-DMG, may be used to optimize the formulation of saRNA for delivery to different cell types including, but not limited to, hepatoeytes, myeloid cells, muscle cells, etc. For example, the component molar ratio may include, but is not limited to, 50% C12-200, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and %1.5 PEG-DMG (see Leuschner et al, Nat Biotechnol 2011 29: 1005-1010; the contents of which are herein incorporated by reference in its entirety). The use of lipidoid formulations for the localized delivery of nucleic acids to cells (such as, but not limited to, adipose cells and muscle cells) via either subcutaneous or intramuscular delivery, may not require all of the formulation components desired for systemic delivery, and as such may comprise only the lipidoid and saRNA,
Liposomes, Lipoplexes, and Lipid Nanoparticles
[00162] The saRNA of the invention can be formulated using one or more liposomes, lipoplexes, or lipid nanoparticles. In one embodiment, pharmaceutical compositions of saRNA include liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of nutrients and pharmaceutical formulations. Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter. Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis. Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical formulations.
[00163] The formation of liposomes may depend on the physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped and the liposomal ingredients, the nature of the medium in which the lipid vesicles are dispersed, the effective concentration of the entrapped substance and its potential toxicity, any additional processes involved during the application and/or delivery of the vesicles, the optimization size,
polydispersity and the shelf-life of the vesicles for the intended application, and the batch-to- batch reproducibility and possibility of large-scale production of safe and efficient liposomal products. [00164] In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from 1 ,2-dioleyloxy-JV,iV- dimethylaminopropane (DODMA) liposomes, DiLa2 liposomes from Marina Biotech (Bothell, WA), l ,2-dilinoleyloxy-3-dimethylaminopropane (DLin-DMA), 2 ,2-dilinoleyl-4-(2- dimethylaminoethyl)-[l ,3]-dioxolane (DLin-KC2-DMA), and MC3 (US20100324120; the contents of which are herein incorporated by reference in its entirety) and liposomes which may deliver small molecule drugs such as, but not limited to, DOX1L® from Janssen Biotech, Inc. (Horsham, PA).
[00165] In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271 -281 ; Zhang et al. Gene Therapy. 1999 6: 1438-1447; Jeffs et al. Pharm Res. 2005 22:362-372; Morrissey et al, Nat BiotechnoL 2005 2: 1002-1007; Zimmermann et al., Nature. 2006 441 : 1 1 1 -1 14; Heyes et al. J Contr Rel. 2005 107:276-287; Semple et al. Nature Biotech. 2010 28 :172-176; Judge et al. J Clin Invest. 2009 1 19:661 -673; deFougerolles Hum Gene Ther. 2008 19: 125-132; the contents of each of which are incorporated herein in their entireties). The original manufacture method by Wheeler et al. was a detergent dialysis method, which was later improved by Jeffs et al. and is referred to as the spontaneous vesicle formation method. The liposome formulations may be composed of 3 to 4 lipid components in addition to the saRNA. As an example a liposome can contain, but is not limited to, 55% cholesterol, 20% disteroylphosphatidyl choline (DSPC), 10% PEG-S-DSG, and 15% 1 ,2-dioleyloxy-N.N- dimcthylaminopropane (DODMA), as described by Jeffs et al. In another example, certain liposome formulations may contain, but are not limited to, 48% cholesterol, 20% DSPC, 2% PEG-c-DMA, and 30% cationic lipid, where the cationic lipid can be 1 ,2-distearloxy-JV,N- dimethylaminopropane (DSDMA), DODMA, DLin-DMA, or 1 ,2-dilinolcnv loxy-3 - dimethylaminopropane (DLenDMA), as described by Heyes et al In another example, the nucleic acid-lipid particle may comprise a cationic lipid comprising from about 50 mol % to about 85 mol % of the total lipid present in the particle; a non-cationic lipid comprising from about 13 mol % to about 49.5 mol % of the total lipid present in the particle; and a conjugated lipid that inhibits aggregation of particles comprising from about 0.5 mol % to about 2 mol % of the total lipid present in the particle as described in WO2009 1 27060 to Maelaehlan et al, the contents of which are incorporated herein by reference in their entirety. In another example, the nucleic acid-lipid particle may be any nucleic acid-lipid particle disclosed in US2006008910 to Maelaehlan et al., the contents of which are incorporated herein by reference in their entirety. As a non-limiting example, the nucleic acid-lipid particle may comprise a cationic lipid of Formula I, a non-eationic lipid, and a conjugated lipid that inhibits aggregation of particles.
[00166] In one embodiment, the saRNA of the present invention may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers.
[00167] In one embodiment, the liposome may contain a sugar-modified lipid disclosed in US5595756 to Bally et al., the contents of which are incorporated herein by reference in their entirety. The lipid may be a ganglioside and cerebroside in an amount of about 10 mol percent [00168] In one embodiment, the saRNA of the present invention may be formulated in a liposome comprising a cationic lipid. The liposome may have a molar ratio of nitrogen atoms in the cationic lipid to the phosphates in the saRNA (N:P ratio) of between 1 :1 and 20:1 as described in International Publication No. WO2013006825, the contents of which are herein incorporated by reference in its entirety. In another embodiment, the liposome may have a N;P ratio of greater than 20:1 or less than 1 : 1.
[00169] In one embodiment, the saRNA of the present invention may be formulated in a lipid-polycation complex. The formation of the lipid-polycation complex may be accomplished by methods known in the art and/or as described in U.S. Pub. No. 20120178702, the contents of which are herein incorporated by reference in its entirety. As a non-limiting example, the polycation may include a cationic peptide or a polypeptide such as, but not limited to, polylysine, poly ornithine and/or polyarginine and the cationic peptides described in International Pub. No. WO2012013326; herein incorporated by reference in its entirety. In another embodiment, the saRNA may be formulated in a lipid-polycation complex which may further include a neutral lipid such as, but not limited to, cholesterol or dioleoyl phosphatidylethanolamine (DOPE), [00170] The liposome formulation may be influenced by, but not limited to, the selection of the catiomc lipid component, the degree of cationic lipid saturation, the nature of the
PEGylation, ratio of all components and biophysical parameters such as size. In one example by Semple et al. (Semple et al. Nature Biotech. 2010 28: 172-176; the contents of which are herein incorporated by reference in its entirety), the liposome formulation was composed of 57.1 % catiomc lipid, 7.1% d i almitoylphosphatidylcholine, 34.3 % cholesterol, and 1.4% PEG-c-DMA, [00171] In some embodiments, the ratio of PEG in the lipid nanoparticle (LNP) formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified from C 14 to CI 8 to alter the pharmacokinetics and/or biodistribution of the LNP formulations. As a non-limiting example, LNP formulations may contain 1 -5% of the lipid molar ratio of PEG-c-DOMG as compared to the cationic lipid, DSPC and cholesterol In another embodiment the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG-DSG ( 1 ,2-Distearoyl-sn-g ycerol, methoxypolyethylene glycol) or PEG-DPG ( 1 ,2-Dipalmitoyl-sn -glycerol, methoxypolyethylene glycol). The cationic lipid may be selected from any lipid known in the art such as, but not limited to, DLin-MC3-DMA, D Lin-DM A , C 1 2- 200 and DLin-KC2 -DMA,
[00172] In one embodiment, the saRNA of the present invention may be formulated in a lipid nanoparticle such as the lipid nanopartieles described in International Publication No.
WO2012170930, the contents of which are herein incorporated by reference in its entirety.
[001 3] In one embodiment, the cationic lipid which may be used in formulations of the present invention may be selected from, but not limited to, a cationic lipid described in
International Publication Nos. WO2012040184, WO201 1 153 120, WO201 1 149733,
WO201 1090965, WO201 1043913, WO201 1022460, WO2012061259, WO2012054365, WO2012044638, WO2010080724, WO201021865 and WO2008103276, US Patent Nos.
7,893,302, 7,404,969 and 8,283,333 and US Patent Publication No. US201000361 15 and US20120202871 ; the contents of each of which is herein incorporated by reference in their entirety. In another embodiment, the cationic lipid may be selected from, but not limited to, formula A described in International Publication Nos. WO2012040184, WO201 1 153120, WO201 1 149733, WO201 1090965, WO201 1043913, WO201 1022460, WO2012061259, WO2012054365 and WO2012044638; the contents of each of which is herein incorporated by reference in their entirety. In yet another embodiment, the cationic lipid may be selected from, but not limited to, formula CLI-CLXXIX of International Publication No, WO2008103276, formula CLI-CLXXIX of US Patent No. 7,893,302, formula CLI-CLXXXXII of US Patent No. 7,404,969 and formula I- VI of US Patent Publication No. US201000361 15; the contents of each of which are herein incorporated by reference in their entirety. In yet another embodiment, the cationic lipid may be a multivalent cationic lipid such as the cationic lipid disclosed in US Patent No. 7223887 to Gaucheron et al, the contents of which are incorporated herein by reference in their entirety. The cationic lipid may have a positively-charged head group including two quaternary amine groups and a hydrophobic portion including four hydrocarbon chains as described in US Patent No, 7223887 to Gaucheron et al., the contents of which are incorporated herein by reference in their entirety. In yet another embodiment, the cationic lipid may be biodegradable as the biodegradable lipids disclosed in US20130195920 to Maier et al., the contents of which are incorporated herein by reference in their entirety. The cationic lipid may have one or more biodegradable groups located in a lipidic moiety of the cationic lipid as described in formula I- IV in US 20130195920 to Maier et al, the contents of which are incorporated herein by reference in their entirety.
[00174] As a non-limiting example, the cationic lipid may be selected from (2QZ,23Z)-
N,N-dimethylnonacosa-20,23-dien- 10-amine, ( 17Z,20Z)-N,N-dimemylhexacosa- 17,20-dien-9- amine, ( 1 Z, 19Z)-N5N-dimethy lpentaeosa-1 6, 19-dien-8-amine, ( 13Z, 16Z)-N,N-dimethyldoeosa- 13,16-dien-5-arnine, ( 12Z.15Z)-N,N-dimethylhenieosa-l 2, 15-dien-4-amine, ( 14Z, 17Z)-N,N- dimethyltricosa-14, 17-dien-6-amine, ( 15Z, 18Z)-N,N-dimethyltetraeosa- 15,18-dien-7 -amine, ( 18Z,2 lZ)-N,N-dimethylheptacosa- 18,21 -dien-10-amine, ( 15ZJ 8Z)-N,N-dimethyltetracosa- 15,18-dien-5-amine, ( 14Z, 17Z)-N,N-dimethyltricosa-l 4, 17-dien-4-amine, (19Z,22Z)-N,N- dimeihyloctacosa-19,22-dien-9-amine, ( 18Z.2 1 Z)-N,N-dimethylheptacosa- 18 ,21 -dicn-8 - amine, ( 17Z,20Z)-N,N-dimethylhexaeosa- 17,20-dien-7-amine, (16Z,19Z)-N,N- dimethylpentacosa-16, 1 -dien-6-amine, (22Z,25Z)-N,N-dimethylhentriaconta-22,25-dien-l 0- amine, (21 Z ,24Z)-N,N-dimethyltriaconta-21 ,24-dien-9-amine, (18Z)-N,N-dimetylheptacos-l 8- en- 1 ()-aminc, ( 17Z)-N. -dimcthylhcxacos- 17-cn-9-aminc, ( 19Z,22Z)-N,N-dimethyloctacosa-
19.22- dien-7-amine, N,N-dimethylheptacosan-10-amine, (20Z,23Z)-N-ethyl-N-methylnonacosa-
20.23 - dien-10-amke, 1 -[(1 lZ,14Z)-l-nonylicosa-l 1,14-dien-l-yl] pyrrolidine, (20Z)-N,N- dimethylheptacos-20-en-l 0-amine, (15Z)-N,N-dimethyl eptacos-1 5-en-l. 0-amine, (14Z)-N,N- dimethy lnonacos - 14 -en-10-amine, ( 17Z)-N,N-dimethy lnonacos- 17 -en-10-amine, (24Z)- ,N- dimethyltritriacont-24-en-10-amine, (20Z)-N,N-dimethylnonacos-20-en-l 0-amine, (22Z)-N,N- dimethylhentriacont-22-en-10-amine, { 16Z)-N,N-dimethylpentacos-l 6-en-8-amine, ( 12Z, 15Z)~ N. -dimethyl-2 -nony lhenicosa- 12,15 -dien- 1 -amine, ( 13Z, 16Z)-N ,N -dimethyl - -nonyldocosa- 13, 16-dicn-l-aminc, N,N-dimethyl-l-[(lS,2R)-2-octylcyclopropyl] eptadeean-8-amine. 1 - [( 1 S,2R)-2-hexylcyclopropyl]-N,N-dimethylnonadecan- 10-amine, N,N-dimethyl-l -[( 1 S ,2R)-2- octylcyclopropyljnonadecan- 10-amine, N, -dimcthyl-21 -[(lS.2R)-2 - octylcyclopropyl]henicosan-10-araine,N,N-dimethyl-l -[(l S,2S)-2-{[(lR,2R)-2- pentylcyclopropy 1] methyl } eye lopropyl jnonadecan- 10 - a ι n i n e , , N - d i m e t h y 1 - 1 -[( 1 S,2R)-2- oetyleyelopropyl]hexadeean-8-aniine. N,N-dimethyl-[(lR,2S)-2-undecyIcyclopropyl]tetradecan- 5-amine, N,N-dimethyl-3 - {7-[(l S,2R)-2-octylcyelopropyl]heptyl} dodecan-1 -amine, 1 - [(l R,2S)-2-hepty lcyelopropyl]-N,N-dimethyloctadecan-9-amine, ! -[(! S,2R)-2- decylcyclopropyl]-N,N-dimethylpentadecan-6-amine, N ,N -d imethyl-l -[( IS ,2R)-2 - octylcyelopropyl]pentadeean-8-amine, R-N,N-dimemyl-l -[(9Z, 12Z)-octadeca-9,12-dien-l - yloxy]-3-(oetyloxy)propan-2 -amine, S-N,N-dimethyl-l -[(9Z, 12Z)-octadeca-9,12-dien-l -yloxy]- 3-(octyloxy)propan-2 -amine, 1 - {2-[(9Z, 12Z)-octadeca- , 1 2-dicn- 1 -yloxy] - 1 - [(octyloxy)methyl]ethyl}pyrrolidine, (2S)-N,N-dimethyl-l -[(9Z, 12Z)-octadeca-9,12-dien-l - yloxy]-3-[(5Z)-oct-5-en-l -yloxy]propan-2-amine, 1 - {2-[(9Z, 12Z)-oetadeca-9 , 12-dien- 1 -yloxy]- 1 -[(octyloxy)methyl]ethyl}azetidine, (2S)- 1 -(hexyloxy)-N,N-dimethyl-3-[(9Z, 12Z)-octadeca- 9, 12-dien- 1 -yloxy]propan-2-amine. (2S)- 1 -(he tyloxy)-N ,N -dimethy 1-3 -[(9Z, 12Z)-oetadeca- 9, 12-dien- 1 -yloxy]propan-2-amine, N,N-dimethyl- l -(nonyloxy)-3-[(9Z,12Z)-octadeca-9,12- dien-1 -yloxy]propan-2 -amine, N,N-dimethyl- 1 -[(9Z)-oetadec-9-en- 1 -yloxy]-3 -(oetyloxy)propan- 2-amine; (2S)-N,N-dimethyl- 1 -[(6Z,9Z, 12Z)-octadeea-6,9, 12-trien- 1 -yloxy]-3 -(octyloxy)propan- 2-aminc. (2S)-1 -[(1 lZ, 14Z)-icosa- l l ,14-dien-l -yloxy]-N,N-dimethyl-3-(pentyloxy)propan-2- amine, (2S)-1 -(hexyloxy)-3-[(l l Z,14Z)-ieosa-l 1 , 14-dien-l -yloxy]-N,N-dimethylpropan-2- amine, 1 -[(1 1 Z, 14Z)-ieosa-l 1 , 14-dien-l -yloxy]-N,N-dimethy 1 -3-(oetyloxy)propan-2-amine, 1 - [(13Z, 16Z)-docosa-13,16-dien-l-yloxy]-N,N-dimethyl-3-(octyloxy)propan-2-amine, (2S)-1 - [( 1 3Z.16Z)-doeosa- l 3, 1 6-dicn- l -yloxy ]-3 -( hcxy loxy)- . -d imclhy lpropan -2 -amine. (2S)-1 - [( 13Z)-docos-l 3 -en- 1 -yloxy]-3 -(hexyloxy)-N,N-dimethylpropan-2-amine, I -[( 13Z)-doeos- 13- en- 1 -yloxy] -N,N-dimethyl -3 -(oetyloxy)propan-2-amine, 1 -[(9Z)-hexadec-9-en- 1 -yloxy]-N,N- dimethyl-3-(octyloxy)propan-2-amine5 (2 R)- ,N-dimemyl-I I( 1 -metoylo ctyl)oxy']-3 -[(9Z. 1 2Z )- octadeca-9, 12-dien- 1 -yloxyjpropan-2-amine. (2R)~ 1 - [(3 ,7-dimethyloetyl)oxy] -N,N -dimethy 1-3 - [(9Z, 12Z)-octadeca-9, 12-dien- 1 -yloxy]propan-2-amine, Ν,Ν-dimethy 1- 1 -(oetyloxy)-3 -({ 8- [( 1 S,2S)-2-{[( 1 R,2R)-2-pentylcyclopropy 1] methyl } eye lopropyl] octyl } oxy)propan-2-amine, N.N- dimethyl- l -{[8-(2-oc lyleyelopropyl)octyl]oxy}-3-(octyloxy)propan-2 -amine and (11E,20Z,23Z)~ N,N-dimethylnonacosa-ll,20,2-trien-10-amme or a pharmaceutically acceptable salt or stereoisomer thereof.
[00175] In one embodiment, the lipid may be a eleavable lipid such as those described in
International Publication No. WO20 1 2 1 70889. the contents of which are herein mcorporated by reference in their entirety.
[001 6] In one embodiment, the nanopartieles described herein may comprise at least one cationic polymer described herein and or known in the art.
[00177] In one embodiment, the cationic lipid may be synthesized by methods known in the art and/or as described in International Publication Nos. WO2012040184, WO201 1 153120, WO201 1 149733, WO201 1090965, WO201 1043913, WO201 1022460, WO2012061259, WO2012054365, WO2012044638, WO2010080724 and WO201021865; the contents of each of which is herein incorporated by reference in their entirety.
[00178] In one embodiment, the LNP formulations of the saRNA may contain PEG-c-
DOMG at 3% lipid molar ratio. In another embodiment, the LNP formulations of the saRNA may contain PEG-c-DOMG at 1.5% lipid molar ratio.
[001 9] In one embodiment, the pharmaceutical compositions of the saRNA may include at least one of the PEGylated lipids described in International Publication No. 2012099755, the contents of which is herein incorporated by reference in its entirety.
[00180] In one embodiment, the LNP formulation may contain PEG-DMG 2000 (l ^- dimyristoyl-sn-glycero-3-phophoethanolamine-N-[methoxy(polyethylene glycol)-2000). In one embodiment, the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art and at least one other component. In another embodiment, the LNP formulation may contain PEG-DMG 2000, a cationic lipid known in the art, DSPC and cholesterol. As a non-limiting example, the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol. As another non-limiting example the LNP formulation may contain PEG-DMG 2000, DLin-DMA, DSPC and cholesterol in a molar ratio of 2:40: 10:48 (see e.g., Geall et al, Nonviral delivery of self-amplifying RNA vaccines, PNAS 2012; PMID: 22908294; herein incorporated by reference in its entirety). As another non-limiting example, the saRNA described herein may be formulated in a nanoparticle to be delivered by a parenteral route as described in U.S. Pub. No. 20120207845; the contents of which is herein incorporated by reference in its entirety. The cationic lipid may also be the cationic lipids disclosed in US20130156845 to Manoharan et al. and US 20130129785 to Manoharan et al, WO 2012047656 to Wasan et al., WO 2010144740 to Chen et al, WO 2013086322 to Ansell et al, or WO 2012016184 to Manoharan et al, the contents of each of which are incorporated herein by reference in their entirety,
( 001 11 In one embodiment, the saRNA of the present invention may be formulated with a plurality of cationic lipids, such as a first and a second cationic lipid as described in
US20130017223 to Hope et al, the contents of which are incorporated herein by reference in their entirety. The first cationic lipid can be selected on the basis of a first property and the second cationic lipid can be selected on the basis of a second property, where the properties may be determined as outlined in US20130017223, the contents of which are herein incorporated by reference in its entirety. In one embodiment, the first and second properties are
complementary.
[00182] In another embodiment, the saRNA may be formulated with a lipid particle comprising one or more cationic lipids and one or more second lipids, and one or more nucleic acids, wherein the lipid particle comprises a solid core, as described in US Patent Publication No. US20120276209 to Cullis et al., the contents of which are incorporated herein by reference in their entirety.
[00183] In one embodiment, the saRNA of the present invention may be complexed with a cationic amphiphile in an oil-in-water (o/w) emulsion such as described in EP2298358 to Satishchandran et al., the contents of which are incorporated herein by reference in their entirety. The cationic amphiphile may be a cationic lipid, modified or unmodified spermine, bupivacaine, or benzalkonium chloride and the oil may be a vegetable or an animal oil. As a non-limiting example, at least 10% of the nucleic acid-cationic amphiphile complex is in the oil phase of the oil-in-water emulsion (see e.g., the complex described in European Publication No. EP2298358 to Satishchandran et al, the contents of which are herein incorporated by reference in its entirety).
[00184] In one embodiment, the saRNA of the present invention may be formulated with a composition comprising a mixture of cationic compounds and neutral lipids. As a non-limiting example, the cationic compounds may be formula (I) disclosed in WO 1999010390 to Ansell et al., the contents of which are disclosed herein by reference in their entirety, and the neutral lipid may be selected from the group consisting of diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide and sphingomyelin. In another non-limiting example, the lipid formulation may comprise a eationie lipid of formula A, a neutral lipid, a sterol and a PEG or PEG-modified lipid disclosed in US 20120101 148 to Akinc et al, the contents of which are incorporated herein by reference in their entirety.
[00185] In one embodiment, the LNP formulation may be formulated by the methods described in International Publication Nos. WO2011127255 or WO2008103276, each of which are herein incorporated by reference in their entirety. As a non-limiting example, the saRNA of the present invention may be encapsulated in any of the lipid nanoparticle (LNP) formulations described in WO2011 127255 and/or WO2008103276; the contents of each of which are herein incorporated by reference in their entirety.
[00186] In one embodiment, the LNP formulations described herein may comprise a polycationic composition. As a non-limiting example, the polycationic composition may be selected from formula 1-60 of US Patent Publication No. US20050222064; the contents of which is herein incorporated by reference in its entirety. In another embodiment, the LNP formulations comprising a polycationic composition may be used for the delivery of the saRNA described herein in vivo and/or in vitro,
[00187] In one embodiment, the LNP formulations described herein may additionally comprise a permeability enhancer molecule. Non-limiting permeability enhancer molecules are described in US Patent Publication No. US20050222064; the contents of which is herein incorporated by reference in its entirety.
[00188] In one embodiment, the pharmaceutical compositions may be formulated in liposomes such as, but not limited to, DiLa2 liposomes (Marina Biotech, Bothell. WA), SMARTICLES®/NOV340 (Marina Biotech, Bothell, WA), neutral DOPC (1,2-dioleoyl-sn- glycero-3 -phosphoeholine) based liposomes (e.g., siRNA delivery for ovarian cancer (Landen et al. Cancer Biology & Therapy 2006 5(12)1708-1713); the contents of which is herein incorporated by reference in its entirety) and hyaluronan-coated liposomes (Quiet Therapeutics, Israel).
[00189] In some embodiments, the pharmaceutical compositions may be formulated with any amphoteric liposome disclosed in WO 2008/043575 to Panzner and US 8580297 to Essler et al. (Marina Biotech), the contents of which are incorporated herein by reference in their entirety. The amphoteric liposome may comprise a mixture of lipids including a eationie amphiphile, an anionic amphiphile and optional one or more neutral amphiphiles. The amphoteric liposome may comprise amphoteric compounds based on amphiphilic molecules, the head groups of which being substituted with one or more amphoteric groups. In some embodiments, the
pharmaceutical compositions may be formulated with an amphoteric lipid comprising one or more amphoteric groups having an isoelectric point between 4 and 9, as disclosed in US
20140227345 to Essler et al. (Marina Biotech), the contents of which are incorporated herein by reference in their entirety.
[00190] In some embodiments, the pharmaceutical composition may be formulated with liposomes comprising a sterol derivative as disclosed in US 7312206 to Panzner et al.
(Novosom), the contents of which are incorporated herein by reference in their entirety. In some embodiments, the pharmaceutical composition may be formulated with amphoteric liposomes comprising at least one amphipathic cationic lipid, at least one amphipathic anionic lipid, and at least one neutral lipid, or liposomes comprise at least one amphipathic lipid with both a positive and a negative charge, and at least one neutral lipid, wherein the liposomes are stable at pH 4.2 and pH 7.5, as disclosed in US Pat. No. 7780983 to Panzner et al. (Novosom), the contents of which are incorporated herein by reference in their entirety. In some embodiments, the pharmaceutical composition may be formulated with liposomes comprising a serum-stable mixture of lipids taught in US 201 10076322 to Panzner et al, the contents of which are incorporated herein by reference in their entirety, capable of encapsulating the saRNA of the present invention. The lipid mixture comprises phosphatidylcholine and
phosphatidylethanolamine in a ratio in the range of about 0.5 to about 8. The lipid mixture may also include pH sensitive anionic and cationic amphiphiles, such that the mixture is amphoteric, being negatively charged or neutral at pH 7.4 and positively charged at pH 4. The drug/lipid ratio may be adjusted to target the liposomes to particular organs or other sites in the body. In some embodiments, liposomes loaded with the saRNA of the present invention as cargo, are prepared by the method disclosed in US 20120021042 to Panzner et al, the contents of which are incorporated herein by reference in their entirety. The method comprises steps of admixing an aqueous solution of a polyanionic active agent and an alcoholic solution of one or more amphiphiles and buffering said admixture to an acidic pH, wherein the one or more amphiphiles are susceptible of forming amphoteric liposomes at the acidic pH, thereby to form amphoteric liposomes in suspension encapsulating the active agent. [00191] The nanoparticle formulations may be a carbohydrate nanoparticle comprising a carbohydrate carrier and a nucleic acid molecule (e.g., saR A), As a non-limiting example, the carbohydrate carrier may include, but is not limited to, an anhydride-modified phytoglyeogen or glycogen-type material, phtoglycogen octenyl succinate, phytoglyeogen beta-dextrin, anhydride- modified phytoglyeogen beta-dextrin. (See e.g., International Publication No. WO20 12 1 091 2 1 ; the contents of which is herein incorporated by reference in its entirety).
[00192] Lipid nanoparticle formulations may be improved by replacing the cationic lipid with a biodegradable cationic lipid which is known as a rapidly eliminated lipid nanoparticle (reLNP). lonizable cationic lipids, such as, but not limited to, DLinDMA, DLin-KC2-DMA, and DLin-MC3-DMA, have been shown to accumulate in plasma and tissues over time and may be a potential source of toxicity. The rapid metabolism of the rapidly eliminated lipids can improve the toierabiiity and therapeutic index of the lipid nanoparticles by an order of magnitude from a 1 mg/kg dose to a 10 mg/kg dose in rat. Inclusion of an enzymatically degraded ester linkage can improve the degradation and metabolism profile of the cationic component, while still maintaining the activity of the reLNP formulation. The ester linkage can be internally located within the lipid chain or it may be terminally located at the terminal end of the lipid chain. The internal ester linkage may replace any carbon in the lipid chain.
[00193] In one embodiment, the saRNA may be formulated as a lipoplex, such as, without limitation, the ATUPLEX™ system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom),
STEMFECT™ from STEMGENT® (Cambridge, MA), and polyethylenimine (PEI) or protamine-based targeted and non-targeted delivery of nucleic acids (Aleku et al. Cancer Res. 2008 68:9788-9798; Striimberg et al. Int J Clin Pharmacol Ther 2012 50:76-78; Santel et al, Gene Ther 2006 13:1222-1234; Santel et al, Gene Ther 2006 13 :1360-1370; Gutbier et al, Pulm Pharmacol. Ther. 2010 23:334-344; Kaut'mann et al. Mierovasc Res 2010 80:286-293Weide et al. J Immunother. 2009 32:498-507; Weide et al. J Immunother. 2008 31 : 180488; Pascolo Expert Opin. Biol. Ther. 4: 1285-1294; Fotin-Mleezek et al., 201 1 J. Immunother. 34: 1 -15; Song et al, Nature Biotechnol. 2005, 23:709-717; Peer et al, Proe Natl Acad Sci U S A. 2007 6; 104:4095-4100; deFougerolles Hum Gene Ther. 2008 19:125- 132; the contents of each of which are incorporated herein by reference in its entirety). [00194] In one embodiment such formulations may also be constructed or compositions altered such that they passively or actively are directed to different cell types in vivo, including but not limited to hepatocytes, immune cells, tumor cells, endothelial cells, antigen presenting cells, and leukocytes (Akinc et al, Mol Ther. 2010 18:1357-1364; Song et al., Nat Biotechnol. 2005 23:709-717; Judge et al., J Clin Invest. 2009 1 19:661 -673; Kaufmann et al., Microvasc Res
2010 80:286-293; Santel et al., Gene Ther 2006 13: 1222-1234; Santel et al, Gene Ther 2006 13:1360-1370; Gutbier et al., Pulm Pharmacol. Ther. 2010 23 :334-344; Basha et al., Mol. Ther.
201 1 19:2186-2200; Fenske and Cullis, Expert Opin Drug Deliv. 2008 5:25-44; Peer et al, Science. 2008 319:627-630; Peer and L icberman. Gene Ther. 201 1 18:1 127-1 133 ; the contents of each of which are incorporated herein by reference in its entirety). One example of passive targeting of formulations to liver cells includes the DLin-DMA, DLin-KC2-DMA and DLin- MC3-DMA-based lipid nanoparticle formulations which have been shown to bind to
apolipoprotein E and promote binding and uptake of these formulations into hepatocytes in vivo (Akinc et al. Mol Ther. 2010 18: 1357-1364; the contents of which is herein incorporated by reference in its entirety). Formulations can also be selectively targeted through expression of different ligands on their surface as exemplified by, but not limited by, folate, transferrin, N- aeetylgalactosamine (GalNAc), and antibody targeted approaches (Kolhatkar et al., Curt Drug Discov Technol. 201 1 8: 197-206; Musacchio and Torchilin, Front Biosci. 201 1 16: 1388-1412; Yu et al., Mol Membr Biol. 2010 27:286-298; Patil et al., Crit Rev Ther Drug Carrier Syst. 2008 25:1 -61 ; Benoit et al, Biomacromolecules. 201 1 12:2708-2714; Zhao et al, Expert Opin Drug Deliv. 2008 5:309-319; Akinc et al, Mol Ther. 2010 18: 1357-1364; Srinivasan et al., Methods Mol Biol. 2012 820: 105-1 16; Ben-Arie et al, Methods Mol Biol. 2012 757:497-507; Peer 2010 J Control Release. 20:63-68; Peer et al., Proc Natl Acad Sci U S A. 2007 104:4095-4100; Kim et at, Methods Mol Biol. 201 1 721 :339-353; Subramanya et al, Mol Ther. 2010 18:2028-2037; Song et al., Nat Biotechnol. 2005 23:709-717; Peer et al, Science. 2008 319:627-630; Peer and Lieberman, Gene Ther. 201 1 18: 1 127-1 133; the contents of each of which are incorporated herein by reference in its entirety).
[00195] In one embodiment, the saRNA is formulated as a solid lipid nanoparticle. A solid lipid nanoparticle (SLN) may be spherical with an average diameter between 10 to 1000 urn. SLN possess a solid lipid core matrix that can solubilize lipophilic molecules and may be stabilized with surfactants and/or emulsifiers. In a further embodiment, the lipid nanoparticle may be a self-assembly lipid-polymer nanoparticle (see Zhang et al, ACS Nano, 2008, 2 (8), pp 1696-1702; the contents of which are herein incorporated by reference in its entirety).
[00196] In one embodiment, the saRNA of the present invention can be formulated for controlled release and/or targeted delivery. As used herein, "controlled release" refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome. In one embodiment, the saRNA may be encapsulated into a delivery agent described herein and/or known in the art for controlled release and/or targeted delivery. As used herein, the term "encapsulate" means to enclose, surround or encase. As it relates to the formulation of the compounds of the invention, encapsulation may be substantial, complete or partial The term "substantially encapsulated" means that at least greater than 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.9 or greater than 99.999% of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent. "Partially encapsulated" means that less than 10, 10, 20, 30, 40 50 or less of the pharmaceutical composition or compound of the invention may be enclosed, surrounded or encased within the delivery agent. Advantageously, encapsulation may be determined by measuring the escape or the activity of the pharmaceutical composition or compound of the invention using fluorescence and or electron micrograph. For example, at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the pharmaceutical composition or compound of the invention are encapsulated in the delivery agent.
[00197] In another embodiment, the saRNA may be encapsulated into a lipid
nanoparticle or a rapidly eliminated lipid nanoparticle and the lipid nanoparticles or a rapidly eliminated lipid nanoparticle may then be encapsulated into a polymer, hydrogel and/or surgical sealant described herein and/or known in the art. As a non-limiting example, the polymer, hydrogel or surgical sealant may be PLGA, ethylene vinyl acetate (EVAc), poloxamer,
GELSUE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA),
TISSELL® (Baxter International, Inc., Decrfield. IL), PEG-based sealants, and COSEAL® (Baxter International, Inc., Decrfield, IL).
[00198] In another embodiment, the lipid nanoparticle may be encapsulated into any polymer known in the art which may form a gel when injected into a subject. As another non- limiting example, the lipid nanopartiele may be encapsulated into a polymer matrix which may be biodegradable.
[00199] In one embodiment, the saRNA formulation for controlled release and/or targeted delivery may also include at least one controlled release coating. Controlled release coatings include, but are not limited to, OPADRY®, polyvinylpyrrolidone/ vinyl acetate copolymer, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, EUDRAGIT RL®, EUDRAGIT RS® and cellulose derivatives such as ethylcellulose aqueous dispersions (AQUACOAT® and SURELEASE®).
[00200] In one embodiment, the controlled release and/or targeted delivery formulation may comprise at least one degradable polyester which may contain polycationic side chains. Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-laetide-co-L- lysine), poly(4-hydroxy-L-proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
[00201] In one embodiment, the saRNA of the present invention may be formulated with a targeting lipid with a targeting moiety such as the targeting moieties disclosed in
US20130202652 to Manoharan et al., the contents of which are incorporated herein by reference in their entirety. As a non-limiting example, the targeting moiety of formula I of US
20130202652 to Manoharan et al. may selected in order to favor the lipid being localized with a desired organ, tissue, cell, cell type or subtype, or organelle. Non-limiting targeting moieties that are contemplated in the present invention include transferrin, anisamide, an RGD peptide, prostate specific membrane antigen (PSMA), fueose, an antibody, or an aptamer.
[00202] In one embodiment, the saRNA of the present invention may be encapsulated in a therapeutic nanopartiele. Therapeutic nanoparticles may be formulated by methods described herein and known in the art such as, but not limited to, International Pub Nos. WO2010005740, WO2010030763, WO2010005721 , WO2010005723, WO2012054923, US Pub. Nos.
US201102624 1, US20100104645, US20100087337, US20100068285, US20110274759, US20100068286 and US20120288541 and US Pat No. 8,206,747, 8,293,276, 8,318,208 and 8,318,211 ; the contents of each of which are herein incorporated by reference in their entirety. In another embodiment, therapeutic polymer nanoparticles may be identified by the methods described in US Pub No. US20120140790, the contents of which are herein incorporated by reference in its entirety. [00203] In one embodiment, the therapeutic nanoparticle may be formulated for sustained release. As used herein, "sustained release" refers to a pharmaceutical composition or compound that conforms to a release rate over a specific period of time. The period of time may include, but is not limited to, hours, days, weeks, months and years. As a non-limiting example, the sustained release nanoparticle may comprise a polymer and a therapeutic agent such as, but not limited to, the saRNA of the present invention (see International Pub No. 2010075072 and US Pub No. US20100216804, US201 10217377 and US20120201859, the contents of each of which are herein incorporated by reference in their entirety).
[00204] In one embodiment, the therapeutic nanoparticles may be formulated to be target specific. As a non-limiting example, the therapeutic nanoparticles may include a corticosteroid (see International Pub. No. WO201 1084518; the contents of which are herein incorporated by reference in its entirety), hi one embodiment, the therapeutic nanoparticles may be formulated to be cancer specific. As a non-limiting example, the therapeutic nanoparticles may be formulated in nanoparticles described in International Pub No. WO2008121949, WO2010005726,
WO2010005725, WO201 1084521 and US Pub No. US20100069426, US20120004293 and US20100104655, the contents of each of which are herein incorporated by reference in their entirety.
[00205] In one embodiment, the nanoparticles of the present invention may comprise a polymeric matrix. As a non-limiting example, the nanoparticle may comprise two or more polymers such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides,
polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes,
polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas,
polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co- L-lysine), poly(4-h yd rox y- . -p ro line ester) or combinations thereof.
[00206] I one embodiment, the therapeutic nanoparticle comprises a diblock copolymer.
In one embodiment, the diblock copolymer may include PEG in combination with a polymer such as, but not limited to, polyethylenes, polycarbonates, polyanhydrides, polyhydroxyacids, polypropylfumerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, poly(ethylene imine), poly(serine ester), poly(L-lactide-co-L -lysine), poly(4- hydroxy-L-proline ester) or combinations thereof,
[00207] As a non-limiting example the therapeutic nanopartiele comprises a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No, 8,236,330, each of which is herein incorporated by reference in their entirety). In another non-limiting example, the therapeutic nanopartiele is a stealth nanopartiele comprising a dibloek copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968 and International Publication No.
WO2012 1 66923. the contents of each of which is herein incorporated by reference in its entirety).
[00208] In one embodiment, the therapeutic nanopartiele may comprise a multiblock copolymer such as, but not limited to the multiblock copolymers described in U.S. Pat. No. 8,263,665 and 8,287,910; the contents of each of which are herein incorporated by reference in its entirety.
[00209] In one embodiment, the block copolymers described herein may be included in a polyion complex comprising a non-polymeric micelle and the block copolymer. (See e.g., U.S. Pub. No. 20120076836; the contents of which are herein incorporated by reference in its entirety).
[00210] In one embodiment, the therapeutic nanopartiele may comprise at least one acrylic polymer. Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methaerylate copolymers, ethoxyethyl methacrylates, cyanoethyl methaerylate, amino alkyl methaerylate copolymer, polyiaerylic acid), poly(methacrylic acid), polyeyanoacrylates and combinations thereof.
[00211] In one embodiment, the therapeutic nanoparticles may comprise at least one amine-containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers, poly(beta-amino esters) (See e.g., U.S. Pat. No. 8,287,849; the contents of which are herein incorporated by reference in its entirety) and combinations thereof.
[00212] In one embodiment, the therapeutic nanoparticles may comprise at least one degradable polyester which may contain polycationic side chains. Degradable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), poly(4-hydroxy-L- proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer. [00213] In another embodiment, the therapeutic nanoparticle may include a conjugation of at least one targeting ligand. The targeting ligand may be any ligand known in the art such as, but not limited to, a monoclonal antibody. ( Kirpotin et al, Cancer Res. 2006 66:6732-6740; the contents of which are herein incorporated by reference in its entirety).
[ 00214 ] In one embodiment, the therapeutic nanoparticle may he formulated in an aqueous solution which may be used to target cancer (see International Pub No. WO201 1084513 and US Pub No. US201 10294717, the contents of each of which is herein incorporated by reference in their entirety).
[00215] In one embodiment, the saRNA may be encapsulated in, linked to and/or associated with synthetic nanocarriers. Synthetic nanocarriers include, but are not limited to, those described in International Pub. Nos. WO20 10005740. WO2010030763, WO201213501 , WO2012 149252. WO2 1 2 149255, WO20 12 149259. WO201 2149265, WO2012 149268.
WO2012149282, WO2012149301 , WO2012149393, WO2012149405, WO201214941 1 , WO201 2149454 and WO2013019669, and US Pub. Nos. US201 10262491 , US20100104645, US20100087337 and US20120244222, the contents of each of which are herein incorporated by reference in their entirety. The synthetic nanocarriers may be formulated using methods known in the art and/or described herein. As a non-limiting example, the synthetic nanocarriers may be formulated by the methods described in International Pub Nos. WO2010005740,
W()2() 10030763 and Wt)20 1 2 1 3501 and US Pub. Nos. US201 10262491 . US20100104645, US20100087337 and US2012024422, the contents of each of which are herein incorporated by reference i their entirety. In another embodiment, the synthetic nanocarrier formulations may be lyophilizcd by methods described in International Pub. No. WO201 1072218 and US Pat No. 8,21 1 ,473; the contents of each of which are herein incorporated by reference in their entirety.
[00216] In one embodiment, the synthetic nanocarriers may contain reactive groups to release the saRNA described herein (see International Pub. No. WO20120952552 and US Pub No. US20120171229, the contents of each of which are herein incorporated by reference in their entirety).
[00217] In one embodiment, the synthetic nanocarriers may be formulated for targeted release. In one embodiment, the synthetic nanocarrier may be formulated to release the saRNA at a specified pH and/or after a desired time interval. As a non-limiting example, the synthetic nanoparticle may be formulated to release the saRNA after 24 hours and/or at a pH of 4.5 (see International Pub, Nos. WO2010138193 and WO2010138194 and US Pub Nos. US20110020388 and US20110027217, the contents of each of which is herein incorporated by reference in their entireties).
[00218] In one embodiment, the synthetic nanocarriers may be formulated for controlled and/or sustained release of the saRNA described herein. As a non-limiting example, the synthetic nanocarriers for sustained release may be formulated by methods known in the art, described herein and or as described in International Pub No. WO2010138192 and US Pub No. 20100303850, the contents each of which is herein incorporated by reference in their entirety.
[0021 ] In one embodiment, the nanoparticle may be optimized for oral administration.
The nanoparticle may comprise at least one cationic biopolymer such as, but not limited to, chitosan or a derivative thereof. As a non-limiting example, the nanoparticle may be formulated by the methods described in U.S. Pub. No. 20120282343; the contents of which are herein incorporated by reference in its entirety.
[00220] In one embodiment, the saRNA of the present invention may be formulated in a modular composition such as described in US 8575123 to Manoharan et ah, the contents of which are herein incorporated by reference in their entirety. As a non-limiting example, the modular composition may comprise a nucleic acid, e.g., the saRNA of the present invention, at least one endosomolytic component, and at least one targeting ligand. The modular composition may have a formula such as any formula described in US 8575123 to Manoharan et ah, the contents of which are herein incorporated by reference in their entirety.
[00221] In one embodiment, the saRNA of the present invention may be encapsulated in the lipid formulation to form a stable nucleic acid-lipid particle (SNALP) such as described in US8546554 to de Fougerolles et al., the contents of which are incorporated here by reference in their entirety. The lipid may be cationic or non-cationic. In one non-limiting example, the lipid to nucleic acid ratio (mass/mass ratio) (e.g., lipid to saRNA ratio) will be in the range of from about 1 :1 to about 50:1 , from about 1:1 to about 25:1, from about 3: 1 to about 15:1 , from about 4:1 to about 10:1, from about 5:1 to about 9:1 , or about 6: 1 to about 9:1, or 5: 1, 6:1, 7:1, 8:1, 9:1, 10:1 , or 11 : 1. In another example, the SNALP includes 40% 2,2-Dilinoleyl-4- dimethylaminoethyl-[l ,3]-dioxolane (Lipid A), 10% dioleoylphosphatidylcholine (DSPC), 40% cholesterol, 10% polyethyleneglycol (PEG)-C -DOMG (mole percent) with a particle size of 63.0±2() nm and a 0.027 nucleic acid/lipid ratio. In another embodiment, the saRNA of the present invention may be formulated with a nucleic acid-lipid particle comprising an endosomal membrane destabilizer as disclosed in US 7189705 to Lam et al., the contents of which are incorporated herein by reference in their entirety. As a non-limiting example, the endosomal membrane destabilizer may be a Ca2^ ion,
[00222] In one embodiment, the saRNA of the present invention may be formulated with formulated lipid particles (FLiPs) disclosed in US 8148344 to Akinc et al., the contents of which are herein incorporated by reference in their entirety. Akinc et al. teach that FLiPs may comprise at least one of a single or double-stranded oligonucleotide, where the oligonucleotide has been conjugated to a lipophile and at least one of an emulsion or liposome to which the conjugated oligonucleotide has been aggregated, admixed or associated. These particles have surprisingly been shown to effectively deliver oligonucleotides to heart, lung and muscle disclosed in US 8148344 to Akinc et al., the contents of which are herein incorporated by reference in their entirety.
[00223] In one embodiment, the saRNA of the present invention may be delivered to a cell using a composition comprising an expression vector in a lipid formulation as described in US 6086913 to Tam et al., the contents of which are incorporated herein by reference in their entirety. The composition disclosed by Tam is serum-stable and comprises an expression vector comprising first and second inverted repeated sequences from an adeno associated virus (AAV), a rep gene from AAV, and a nucleic acid fragment. The expression vector in Tam is complexed with lipids.
[00224] In one embodiment, the saRNA of the present invention may be formulated with a lipid formulation disclosed in US 20120270921 to de Fougerolles et al, the contents of which are incorporated herein by reference in their entirety. In one non-limiting example, the lipid formulation may include a cationic lipid having the formula A described in US 20120270921, the contents of which are herein incorporated by reference in its entirety. In another non-limiting example, the compositions of exemplary nucleic acid-lipid particles disclosed in Table A of US 20120270921, the contents of which are incorporated herein by reference in their entirety, may be used with the saRNA of the present invention.
[00225] In one embodiment, the saRNA of the present invention may be fully encapsulated in a lipid particle disclosed in US 20120276207 to Maurer et al, the contents of which are incorporated herein by reference in their entirety. The particles may comprise a lipid composition comprising preformed lipid vesicles, a charged therapeutic agent, and a destabilizing agent to form a mixture of preformed vesicles and therapeutic agent in a
destabilizing solvent, wherein the destabilizing solvent is effective to destabilize the membrane of the preformed lipid vesicles without disrupting the vesicles.
[ 00226] In one embodiment, the saRNA of the present invention may be formulated with a conjugated lipid. In a non-limiting example, the conjugated lipid may have a formula such as described in US 20120264810 to Lin et ai, the contents of which are incorporated herein by reference in their entirety. The conjugate lipid may form a lipid particle which further comprises a eationic lipid, a neutral lipid, and a lipid capable of reducing aggregation.
[00227] In one embodiment, the saRNA of the present invention may be formulated in a neutral liposomal formulation such as disclosed in US 20120244207 to Fitzgerald et al., the contents of which are incorporated herein by reference in their entirety. The phrase "neutral liposomal formulation" refers to a liposomal formulation with a near neutral or neutral surface charge at a physiological pH. Physiological pH can be, e.g., about 7.0 to about 7.5, or, e.g., about 7.5, or, e.g., 7.0, 7.1 , 7.2, 7.3 , 7.4, or 7.5 , or, e.g., 7.3, or, e.g., 7.4. An example of a neutral liposomal formulation is an ionizable lipid nanoparticle (iLNP). A neutral liposomal formulation can include an ionizable eationic lipid, e.g., DLin-KC2 -DMA.
[00228] In one embodiment, the saRNA of the present invention may be formulated with a charged lipid or an amino lipid. As used herein, the term "charged lipid" is meant to include those lipids having one or two fatty acyl or fatty alkyl chains and a quaternary amino head group. The quaternary amine carries a permanent positive charge. The head group can optionally include an ionizable group, such as a primary, secondary, or tertiary amine that may be protonated at physiological pH. The presence of the quaternary amine can alter the pKa of the ionizable group relative to the pKa of the group in a structurally similar compound that lacks the quaternary amine (e.g., the quaternary amine is replaced by a tertiary amine) In some
embodiments, a charged lipid is referred to as an "amino lipid." hi a non-limiting example, the amino lipid may be any amino lipid described in US201 10256175 to Hope et al., the contents of which are incorporated herein by reference in their entirety. For example, the amino lipids may have the structure disclosed in Tables 3-7 of Hope, such as structure (II), DLin-K-C2 -DMA, DLin-K2-DMA, DLin-K6-DMA, etc.. The resulting pharmaceutical preparations may be lyophilized according to Hope. In another non-limiting example, the amino lipids may be any amino lipid described in US 201101 17125 to Hope et al, the contents of which are incorporated herein by reference in their entirety, such as a lipid of structure (I), DLin-K-DMA, DLin-C-DAP, DLin-DAC, D Lin -MA, DLin-S-DMA, etc. In another non-limiting example, the amino lipid may have the structure (I), (II), (III), or (IV), or 4-(R)-DUn-K-DMA (VI), 4-(S)-DUn-K-DMA (V) as described in WO2009132131 to Manoharan et al, the contents of which are incorporated herein by reference in their entirety. In another non-limiting example, the charged lipid used in any of the formulations described herein may be any charged lipid described in EP2509636 to Manoharan et al., the contents of which are incorporated herein by reference in their entirety,
[00229] In one embodiment, the saRNA of the present invention may be formulated with an association complex containing lipids, liposomes, or lipoplexes. In a non-limiting example, the association complex comprises one or more compounds each having a structure defined by formula (I), a PEG-lipid having a structure defined by formula (XV), a steroid and a nucleic acid disclosed in US8034376 to Manoharan et al,, the contents of which are incorporated herein by reference in their entirety. The saRNA may be formulated with any
association complex described in US8034376, the contents of which are herein incorporated by reference in its entirety.
[00230] In one embodiment, the saRNA of the present invention may be formulated with reverse head group lipids. As a non-limiting example, the saRNA may be formulated with a zwitterionic lipid comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the head group, such as a lipid having structure (A) or structure (I) described in WO2011056682 to Leung et al, the contents of which are incorporated herein by reference in their entirety.
[00231] In one embodiment, the saRNA of the present invention may be formulated in a lipid bilayer carrier. As a non-limiting example, the saRNA may be combined with a lipid- detergent mixture comprising a lipid mixture of an aggregation-preventing agent in an amount of about 5 mol% to about 20 mol%, a cationic lipid in an amount of about 0.5 mol% to about 50 mol%, and a fusogenic lipid and a detergent, to provide a nucleic acid-lipid-detergent mixture; and then dialyzing the nucleic acid-lipid-detergent mixture against a buffered salt solution to remove the detergent and to encapsulate the nucleic acid in a lipid bilayer carrier and provide a lipid bilayer-nucleic acid composition, wherein the buffered salt solution has an ionic strength sufficient to encapsulate of from about 40 % to about 80 % of the nucleic acid, described in WO 1999018933 to Cullis et al,, the contents of which are incorporated herein by reference in their entirety.
[00232] In one embodiment, the saRNA of the present invention may be formulated in a nucleic acid-lipid particle capable of selectively targeting the saRNA to a heart, liver, or tumor tissue site. For example, the nucleic acid-lipid particle may comprise (a) a nucleic acid; (b) 1.0 mole % to 45 mole % of a cationic lipid; (c) 0,0 mole % to 90 mole % of another lipid; (d) 1 ,0 mole % to 10 mole % of a bilayer stabilizing component; (e) 0,0 mole % to 60 mole % cholesterol; and (f) 0,0 mole % to 10 mole % of cationic polymer lipid as described in
EP1328254 to Cullis et al, the contents of which are incorporated herein by reference in their entirety. Cullis teaches that varying the amount of each of the cationic lipid, bilayer stabilizing component, another lipid, cholesterol, and cationic polymer lipid can impart tissue selectivity for heart, liver, or tumor tissue site, thereby identifying a nucleic acid-lipid particle capable of selectively targeting a nucleic acid to the heart, liver, or tumor tissue site.
Polymers, Biodegradable Nanoparticles, and Core-Shell Nanopar tides
[00233] The saRNA of the invention can be formulated using natural and/or synthetic polymers. Non-limiting examples of polymers which may be used for delivery include, but are not limited to, DYNAMIC POLYCONJUGATE® (Arrowhead Research Corp., Pasadena, CA) formulations from M1RUS® Bio (Madison, WI) and Roche Madison (Madison, WI),
PHASERX™ polymer formulations such as, without limitation, SMARTT POLYMER
TECHNOLOGY™ (PHASERX®, Seattle, WA), DMRI/DOPE, poloxamer, VAXFECTIN® adjuvant from Vical (San Diego, CA), chitosan, cyelodextrin from Calando Pharmaceuticals (Pasadena, CA), dendrimers and poly(lactic-co-glycolie acid) (PLGA) polymers. RON DEL i M (RNAi Oligonucleotide Nanoparticle Delivery) polymers (Arrowhead Research Corporation, Pasadena, CA) and pH responsive co-block polymers such as, but not limited to, PHASERX® (Seattle, WA).
[00234] A non-limiting example of chitosan formulation includes a core of positively charged chitosan and an outer portion of negatively charged substrate (U.S. Pub. No.
20120258176; herein incorporated by reference in its entirety). Chitosan includes, but is not limited to N-trimethyl chitosan, mono-N-carboxymethyl chitosan (MCC), N-palmitoyl chitosan (NPCS), EDTA-chitosan, low molecular weight chitosan, chitosan derivatives, or combinations thereof. [00235] In one embodiment, the polymers used in the present invention have undergone processing to reduce and/or inhibit the attachment of unwanted substances such as, but not limited to, bacteria, to the surface of the polymer. The polymer may be processed by methods known and/or described in the art and/or described in International Pub. No. WO2012150467, herein incorporated by reference in its entirety .
[00236] A non-limiting example of PLGA formulations include, hut are not limited to,
PLGA injectable depots (e.g., ELIGARD® which is formed by dissolving PLGA in 66% N- methyl-2-pyrrolidone (N MP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).
[00237] Many of these polymer approaches have demonstrated efficacy in delivering oligonucleotides in vivo into the cell cytoplasm (reviewed in de Fougerolles Hum Gene Ther. 2008 19:125-132; herein incorporated by reference in its entirety). Two polymer approaches that have yielded robust in vivo delivery of nucleic acids, in this case with small interfering RNA (siRNA), are dynamic polyconjugates and cyclodextrin-based nanoparticles. The first of these delivery approaches uses dynamic polyconjugates and has been shown in vivo in mice to effectively deliver siRNA and silence endogenous target mRNA in hepatoeytes (Rozema et ah, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; herein incorporated by reference in its entirety). This particular approach is a multicomponent polymer system whose key features include a membrane-active polymer to which nucleic acid, in this case siRNA, is covalently coupled via a disulfide bond and where both PEG (for charge masking) and N- acetylgalactosamine (for hepatocyte targeting) groups are linked via pH-sensitive bonds
(Rozema et al., Proc Natl Acad Sci U S A. 2007 104: 12982- 12887; herein incorporated by reference in its entirety). On binding to the hepatocyte and entry into the endosome, the polymer complex disassembles in the low-pH environment, with the polymer exposing its positive charge, leading to endosomal escape and cytoplasmic release of the siRNA from the polymer. Through replacement of the N-acetylgalactosamine group with a mannose group, it was shown one could alter targeting from asialoglycoprotein receptor-expressing hepatoeytes to sinusoidal
endothelium and Kupffer cells. Another polymer approach involves using transferrin-targeted cyclodextrin-containing polycation nanoparticles. These nanoparticles have demonstrated targeted silencing of the EWS-FLI1 gene product in transferrin receptor-expressing Ewing's sarcoma tumor cells (Hu-Lieskovan et al., Cancer Res.2005 65: 8984-8982; herein incorporated by reference in its entirety) and siRNA formulated in these nanoparticles was well tolerated in non-human primates {Heidel et al., Proe Natl Acad Sci USA 2007 104:5715-21; herein incorporated by reference in its entirety). Both of these delivery strategies incorporate rational approaches using both targeted delivery and endosomal escape mechanisms.
[00238] The polymer formulation can permit the sustained or delayed release of saRNA
(e.g., following intramuscular or subcutaneous injection). The altered release profile for the saRNA can result in, for example, translation of an encoded protein over an extended period of time. Biodegradable polymers have been previously used to protect nucleic acids from
degradation and been shown to result in sustained release of payloads in vivo (Rozcma et al, Proc Natl Acad Sci U S A. 2007 104: 12982-12887; Sullivan et al., Expert Opin Drug Deliv. 2010 7: 1433-1446; Convertine et al., Biomacromolecules. 2010 Oct 1 ; Chu et al, Ace Chem Res. 2012 Jan 13; Manganiello et al., Biomaterials. 2012 33:2301-2309; Benoit et al,
Biomacromolecules. 2011 12:2708-2714; Singha et al., Nucleic Acid Tfaer. 2011 2:133-147; de Fougerolles Hum Gene Ther. 2008 19:125-132; Seliaffert and Wagner, Gene Thcr. 2008
16:1 131 -1138; Chaturvedi et al, Expert Opin Drug Deliv. 201 1 8:1455-1468; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464:1067-1070; each of which is herein incorporated by reference in its entirety).
[00239] In one embodiment, the pharmaceutical compositions may be sustained release formulations. In a further embodiment, the sustained release formulations may be for subcutaneous delivery. Sustained release formulations may include, but are not limited to, PLGA microspheres, ethylene vinyl acetate (EVAc), poloxamer, GELSFTE® (Nanotherapeutics, Inc. Alachua, FL), HYLENEX® (Halozyme Therapeutics, San Diego CA), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, GA), TISSELL® (Baxter Intemational, Inc Deer fie Id, IL), PEG-based sealants, and COSEAL® (Baxter International, Inc Deerfield, IL).
[00240] As a non-limiting example saRNA may be formulated in PLGA microspheres by preparing the PLGA microspheres with tunable release rates (e.g., days and weeks) and encapsulating the saRNA in the PLGA microspheres while maintaining the integrity of the saRNA during the encapsulation process. EVAc are non-biodegradeable, biocompatible polymers which are used extensively in pre-clinical sustained release implant applications (e.g., extended release products Ocusert a pilocarpine ophthalmic insert for glaucoma or progestasert a sustained release progesterone intrauterine device; transdermal delivery systems Testoderm, Duragesic and Selegiline; catheters). Poloxamer F-407 NF is a hydrophilic, non-ionic surfactant tribloek copolymer of polyoxyethylene-polyoxypropylene-polyoxyethylene having a low viscosity at temperatures less than 5°C and forms a solid gel at temperatures greater than 15°C. PEG-based surgical sealants comprise two synthetic PEG components mixed in a delivery device which can be prepared in one minute, seals in 3 minutes and is reabsorbed within 30 days.
GELSITE® and natural polymers are capable of in-situ gelation at the site of administration. They have been shown to interact with protein and peptide therapeutic candidates through ionic ineraction to provide a stabilizing effect.
[00241] Polymer formulations can also be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N- acetylgalactosamine (GalNAc) (Benoit et al., Biomacromolecules. 201 1 12:2708-2714; Rozema et al, Proc Natl Acad Sci U S A. 2007 104; 12982-12887; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464: 1067-1070; each of which is herein incorporated by reference in its entirety).
[00242] The saRNA of the invention may be formulated with or in a polymeric compound. The polymer may include at least one polymer such as, but not limited to, polyethen.es, polyethylene glycol (PEG), poly(l-lysine)(PLL), PEG grafted to PEL, cationic lipopolymer, biodegradable cationic lipopolymer, polyethyleneimine (PE1), cross-linked branched polyialkylene imines), a polyaminc derivative, a modified poloxamer, a biodegradable polymer, elastic biodegradable polymer, biodegradable block copolymer, biodegradable random copolymer, biodegradable polyester copolymer, biodegradable polyester block copolymer, biodegradable polyester block random copolymer, multiblock copolymers, linear biodegradable copolymer, poly[a-(4-aminobutyl)-L-glycolic acid) (PAGA), biodegradable cross-linked cationic multi-block copolymers, polycarbonates, polyanhydrides, polyhydroxyacids,
polypropylramerates, polycaprolactones, polyamides, polyacetals, polyethers, polyesters, poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polycyanoacrylates, polyureas, polystyrenes, polyamines, polylysine, polyiethylene imine), poly(serine ester), poly(L-lactide-co-L-lysine), poly(4- hydroxy-L-proline ester), acrylic polymers, amine-containing polymers, dextran polymers, dextran polymer derivatives or combinations thereof . [00243] As a non-limiting example, the saR NA of the invention may be formulated with the polymeric compound of PEG grafted with PLL as described in U.S. Pat. No, 6,177,274; herein incorporated by reference in its entirety. The formulation may be used for transfecting cells in vitro or for in vivo delivery of the saRNA, In another example, the saRNA may be suspended in a solution or medium with a cationic polymer, in a dry pharmaceutical composition or in a solution that is capable of being dried as described in U.S. Pub. Nos, 20090042829 and 20090042825; each of which are herein incorporated by reference in their entireties.
[00244] As another non-limiting example the saRNA of the invention may be formulated with a PLGA-PEG block copolymer (see US Pub. No. US20120004293 and US Pat No.
8,236,330, herein incorporated by reference in their entireties) or PLGA-PEG-PLGA block copolymers (See U.S. Pat. No. 6,004,573, herein incorporated by reference in its entirety). As a non-limiting example, the saRNA of the invention may be formulated with a diblock copolymer of PEG and PLA or PEG and PLGA (see US Pat No 8,246,968, herein incorporated by reference in its entirety),
[00245] A polyamine derivative may be used to deliver nucleic acids or to treat and/or prevent a disease or to be included in an implantable or injectable device (U.S. Pub. No.
20100260817 herein incorporated by reference in its entirety). As a non-limiting example, a pharmaceutical composition may include the saRNA and the polyamine derivative described in U.S. Pub. No. 20100260817 (the contents of which are incorporated herein by reference in its entirety. As a non-limiting example the saRNA of the present invention may be delivered using a polyaminde polymer such as, but not limited to, a polymer comprising a 1 ,3 -dipolar addition polymer prepared by combining a carbohydrate diazide monomer with a dilkyne unite comprising oligoamines (U.S. Pat. No. 8,236,280; herein incorporated by reference in its entirety).
[00246] In one embodiment, the saRNA of the present invention may be formulated with at least one polymer and/or derivatives thereof described in International Publication Nos.
WO201 1 1 15862, WO20 12082574 and WO2012068187 and U.S. Pub. No. 20120283427, the contents of each of which are herein incorporated by reference in their entireties. In another embodiment, the saRNA of the present invention may be formulated with a polymer of formula Z as described in WO201 1 1 15862, herein incorporated by reference in its entirety. In yet another embodiment, the saRNA may be formulated with a polymer of formula Z, Z' or Z" as described in International Pub. Nos. WO201 2082574 or WO20 1 2068 1 87 and U.S. Pub. No. 2012028342, the contents of each of which are herein incorporated by reference in their entireties. The polymers formulated with the saRNA of the present invention may be
synthesized by the methods described in International Pub. Nos. WO2012082574 or
WO2012068187, the contents of each of which are herein incorporated by reference in their entireties.
[00247] The saRNA of the invention may be formulated with at least one acrylic polymer. Acrylic polymers include but are not limited to, acrylic acid, methacrylic acid, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl
methacrylates, cyanoethyl methacrylate, amino alkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), polycyanoacrylates and combinations thereof.
[00248] Formulations of saRNA of the invention may include at least one amine- containing polymer such as, but not limited to polylysine, polyethylene imine, poly(amidoamine) dendrimers or combinations thereof.
[00249] For example, the saRNA of the invention may he formulated in a pharmaceutical compound including a poly(alkylene imine), a biodegradable cationic lipopolymer, a
biodegradable block copolymer, a biodegradable polymer, or a biodegradable random
copolymer, a biodegradable polyester block copolymer, a biodegradable polyester polymer, a biodegradable polyester random copolymer, a linear biodegradable copolymer, PAGA, a biodegradable cross-linked cationic multi-block copolymer or combinations thereof The biodegradable cationic lipopolymer may be made by methods known in the art and/or described in U.S. Pat. No. 6,696,038, U.S. App. Nos. 20030073619 and 20040142474 each of which is herein incorporated by reference in their entireties. The poly(alkylene imine) may be made using methods known in the art and/or as described in U.S. Pub. No. 20100004315, herein
incorporated by reference in its entirety. The biodegradable polymer, biodegradable block copolymer, the biodegradable random copolymer, biodegradable polyester block copolymer, biodegradable polyester polymer, or biodegradable polyester random copolymer may be made using methods known in the art and/or as described in U.S. Pat. Nos. 6,517,869 and 6,267,987, the contents of which are each incorporated herein by reference in their entirety. The linear biodegradable copolymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,652,886. The PAGA polymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,217,912 herein incorporated by reference in its entirety. The PAGA polymer may be copolymerized to form a copolymer or block copolymer with polymers such as but not limited to, poly-L-lysine, polyargine, polyornithine, histones, avidin, protamines, polylactides and poly(lactide-co-glycolides). The biodegradable cross-linked cationic multi-block copolymers may be made my methods known in the art and/or as described in U.S. Pat. No. 8,057,821 or U.S. Pub. No. 2012009145 each of which are herein incorporated by reference in their entireties. For example, the multi-block copolymers may be synthesized using linear polyethyleneimine (LPEI) blocks which have distinct patterns as compared to branched polyethyleneimines. Further, the composition or pharmaceutical composition may be made by the methods known in the art, described herein, or as described in U.S. Pub. No.
20100004315 or U.S. Pat. Nos. 6,267,987 and 6,217,912 each of which are herein incorporated by reference in their entireties.
[00250] The saRNA of the invention may be formulated with at least one degradable polyester which may contain polycationic side chains. Degradeable polyesters include, but are not limited to, poly(serine ester), poly(L-lactide-co-L-lysine), p o 1 y (4 -h y dro x y- L -proline ester), and combinations thereof. In another embodiment, the degradable polyesters may include a PEG conjugation to form a PEGylated polymer.
[00251 ] The saRNA of the invention may be formulated with at least one crosslinkable polyester. Crosslinkable polyesters include those known in the art and described in US Pub. No. 20120269761, herein incorporated by reference in its entirety.
[00252] In one embodiment, the polymers described herein may be conjugated to a lipid- terminating PEG. As a non-limiting example, PLGA may be conjugated to a lipid-terminating PEG forming PLGA-DSPE-PEG. As another non-limiting example, PEG conjugates for use with the present invention are described in International Publication No. WO2008103276, herein incorporated by reference in its entirety. The polymers may be conjugated using a ligand conjugate such as, but not limited to, the conjugates described in U.S. Pat. No. 8,273,363, herein incorporated by reference in its entirety.
[00253] In one embodiment, the saRNA described herein may be conjugated with another compound. Non-limiting examples of conjugates are described in US Patent Nos.
7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties. In another embodiment, saRNA of the present invention may be conjugated with conjugates of formula 1 -122 as described in US Patent Nos, 7,964,578 and 7,833,992, each of which are herein incorporated by reference in their entireties. The saRNA described herein may be conjugated with a metal such as, but not limited to, gold. (See e.g., Giljohann et al Joum. Amer. Chem. Soc. 2009 131(6): 2072-2073; herein incorporated by reference in its entirety). In another
embodiment, the saRNA described herein may be conjugated and/or encapsulated in gold- nanopartieles. (International Pub. No. WO201216269 and U.S. Pub. No. 20120302940; each of which is herein incorporated by reference in its entirety).
[00254] As described in U.S. Pub. No. 20100004313, herein incorporated by reference in its entirety, a gene delivery composition may include a nucleotide sequence and a poloxamer. For example, the saRNA of the present invention may be used in a gene delivery composition with the poloxamer described in U.S. Pub. No. 20100004313.
[00255] In one embodiment, the polymer formulation of the present invention may be stabilized by contacting the polymer formulation, which may include a cationic carrier, with a cationie lipopolymer which may be covalently linked to cholesterol and polyethylene glycol groups. The polymer formulation may be contacted with a cationic lipopolymer using the methods described in U.S. Pub. No. 20090042829 herein incorporated by reference in its entirety.
[00256] The cationic carrier may include, but is not limited to, polyethylenimine, poly(trimethylenimine), poly(tetramethylenimine), polypropylenimine, aminoglyeoside- polyamine, dideoxy-diamino -b-cyclodextrin, spermine, spermidine, poly(2-dimethylamino)ethyl methacrylate, poly(lysine), poly(histidine), poly(arginine), cationized gelatin, dendrimers, ehitosan, 1 ,2-Dioleoyl-3-Trimethylammomum-Pf opane(DOTAP), N- [ 1 -(2,3 - dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), 1 -[2-(oleoyloxy)ethyl]-2- oleyl-3-(2-hydroxyethyl)imidazolinium chloride (DOT1M), 2,3-dioleyloxy-N- [2(sperminecarboxamido)ethyl]-N,N-dimethyl- 1 -propanaminium trifluoroacetate (DOSPA), 3 B- [N— (N',N'-Dimethylaminoethane)-carbamoyl]Cholesterol Hydrochloride (DC-Cholesterol HC1) diheptadecylamidoglycyl spermidine (DOGS), N.N-distearyl-N.N-dimethylammonium bromide (DDAB), N-( 1 ,2-dimyristyloxyprop-3 -yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide (DMRIE). N . N -d io 1 ey 1 -X .N -d i me thy 1 am m oni u m chloride DODAC) and combinations thereof.
[00257] The saRNA of the invention may be formulated in a polyplex of one or more polymers (U.S. Pub. No. 20120237565 and 20120270927; each of which is herein incorporated by reference in its entirety). In one embodiment, the polyplex comprises two or more cationic polymers. The cationic polymer may comprise a poly(ethylene imine) (PEI) such as linear PEL [00258] The saRNA of the invention can also be formulated as a nanoparticle using a combination of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphate. Components may be combined in a core-shell, hybrid, and/or layer-by-layer architecture, to allow for fine-tuning of the nanoparticle so to delivery of the saRNA may be enhanced (Wang et al, Nat Mater. 2006 5 :791 -796; Fuller et al., Biomaterials. 2008 29:1526- 1532; DeKokcr et al, Adv Drug Deliv Rev. 201 1 63:748-761 ; Endres et al, Biomaterials. 201 1 32:7721 -7731 ; Su et al, Mol Pharm. 201 1 Jun 6;8(3):774-87; herein incorporated by reference in its entirety). As a non-limiting example, the nanoparticle may comprise a plurality of polymers such as, but not limited to hydrophilic -hydrophobic polymers (e.g., PEG-PLGA), hydrophobic polymers (e.g., PEG) and/or hydrophilic polymers (International Pub. No, WO201 20225 129; herein incorporated by reference in its entirety).
[00259] Biodegradable calcium phosphate nanoparticles in combination with lipids and/or polymers may be used to deliver saRNA in vivo. In one embodiment, a lipid coated calcium phosphate nanoparticle, which may also contain a targeting ligand such as anisamide, may be used to deliver the saRNA of the present invention. For example, to effectively deliver siRNA in a mouse metastatic lung model a lipid coated calcium phosphate nanoparticle was used (Li et al, J Contr Rel. 2010 142: 416-421 ; Li et al, J Contr Rel. 2012 158:108-1 14; Yang et al, Mol Ther, 2012 20:609-615; herein incorporated by reference in its entirety). This delivery system combines both a targeted nanoparticle and a component to enhance the endosomal escape, calcium phosphate, in order to improve delivery of the siRNA.
[00260] In one embodiment, calcium phosphate with a PEG-polyanion block copolymer may be used to delivery saRNA (Kazikawa et al, J Contr Rel. 2004 97:345-356; Kazikawa et al., J Contr Rel. 2006 1 1 1 :368-370; herein incorporated by reference in its entirety).
[00261] In one embodiment, a PEG-charge-conversional polymer (Pitella et al.,
Biomaterials. 201 1 32:3106-31 14) may be used to form a nanoparticle to deliver the saRNA of the present invention. The PEG-charge-conversional polymer may improve upon the PEG- polyanion block copolymers by being cleaved into a polycation at acidic pH. thus enhancing endosomal escape. [00262] The use of core-shell nanoparticles has additionally focused on a high- throughput approach to synthesize cationic cross-linked nanogel cores and various shells (Siegwart et al, Proc Natl Acad Sci U S A. 201 1 108:12996-13001). The complexation, delivery, and internalization of the polymeric nanoparticles can be precisely controlled by altering the chemical composition in both the core and shell components of the nanoparticle. For example, the core-shell nanoparticles may efficiently deliver saRNA to mouse hepatocytes after they covalently attach cholesterol to the nanoparticle,
[00263] In one embodiment, a hollow lipid core comprising a middle PLGA layer and an outer neutral lipid layer containing PEG may be used to delivery of the saRNA of the present invention. As a non-limiting example, in mice bearing a luciferase-expressing tumor, it was determined that the l ipid-polymer-l ipid hybrid nanoparticle significantly suppressed luciferase expression, as compared to a conventional lipoplex (Shi et al, Angew Chem Int Ed. 201 1 50:7027-7031 ; herein incorporated by reference in its entirety).
[00264] In one embodiment, the lipid nanoparticles may comprise a core of the saRNA disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the modified nucleic acids in the core.
[00265] Core-shell nanoparticles for use with the saRNA of the present invention may be formed by the methods described in U.S. Pat. No. 8,313,777 herein incorporated by reference in its entirety.
[00266] In one embodiment, the core-shell nanoparticles may comprise a core of the saRNA disclosed herein and a polymer shell. The polymer shell may be any of the polymers described herein and are known in the art. In an additional embodiment, the polymer shell may be used to protect the saRNA in the core. As a non-limiting example, the core-shell nanoparticle may be used to treat an eye disease or disorder (See e.g. US Publication No. 20120321719, herein incorporated by reference in its entirety).
[00267] In one embodiment, the polymer used with the formulations described herein may be a modified polymer (such as, but not limited to, a modified polyacetal) as described in International Publication No. WO201 1 120053, herein incorporated by reference in its entirety. Delivery
[ 00268] The present disclosure encompasses the delivery of saRNA for any of therapeutic, prophylactic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.
[00269] The saRNA of the present invention may be delivered to a cell naked. As used herein in, "naked" refers to delivering saRNA free from agents which promote transfection. For example, the saRNA delivered to the cell may contain no modifications. The naked saRNA may be delivered to the cell using routes of administration known in the art and described herein.
[00270] The saRNA of the present invention may be formulated, using the methods described herein. The formulations may contain saRNA which may be modified and/or unmodified. The formulations may further include, but are not limited to, cell penetration agents, a pharmaceutically acceptable carrier, a delivery agent, a bioerodible or biocompatible polymer, a solvent, and a sustained-release delivery depot. The formulated saRNA may be delivered to the cell using routes of administration known in the art and described herein.
[00271] The compositions may also be formulated for direct delivery to an organ or tissue in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with the compositions, and the like. The saRNA of the present invention may also be cloned into a retroviral replicating vector (RRV) and transduced to cells.
Administration
[00272] The saRNA of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavemous injection, ( into the base of the penis), intravaginal administration, intrauterine, extra-amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), or in ear drops. In specific embodiments, compositions may be administered in a way which allows them cross the blood-brain barrier, vascular barrier, or other epithelial barrier. Routes of
administration disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety, may be used to administer the saRNA of the present invention.
Dosage Forms
[00273] A pharmaceutical composition described herein can be formulated into a dosage form described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous). Liquid dosage forms, injectable preparations, pulmonary forms, and solid dosage forms described in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety may be used as dosage forms for the saRNA of the present invention.
III. Methods of Use
[00274] One aspect of the present invention provides methods of using saRNA of the present invention and pharmaceutical compositions comprising the saRNA and at least one pharmaceutically acceptable carrier. The saRNA of the present invention modulates the expression of its target gene. In one embodiment is provided a method of regulating the expression of a target gene in vitro and/or in vivo comprising administering the saRNA of the present invention. In one embodiment, the expression of the target gene is increased by at least 5, 10, 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80% in the presence of the saRNA of the present invention compared to the expression of the target gene in the absence of the saRNA of the present invention. In a further embodiment, the expression of the target gene is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, in the presence of the saRNA of the present invention compared to the expression of the target gene in the absence of the saRNA of the present invention. [00275] In one embodiment, the increase in gene expression of the saRNA descried herein is shown in proliferating cells.
[00276] The target gene may be any gene of the human genome. As a non-limiting example, the target gene may be any of the coding target genes listed in Table 1 or any of the non-coding genes listed in Table 2,
[00277]
[00278] In one embodiment, the saRNA described herein may be used as a spacer in a
CRISPR (clustered regularly interspaced palindromic repeats) system, such as a CRI PR Cas9 system. The CRISPR system comprising saRNA described herein may be used to cleave and edit a target gene.
[00279] In one embodiment, the saRNA described herein may be used to treat blood disorders such as disorders affecting red blood cells (e.g., anemia, Thalassemia, sickle cell anemia and polycythemia vera), disorder affecting white blood cells (e.g., lymphoma, leukemia, multiple myeloma and myelodysplasitc syndrome), disorders affecting platelets (e.g., thrombocytopenia, idiopathic thrombocytopenic purpura, hcparin-induced thrombocytopenia, thrombotic thrombocytopenic purpura and essential thrombocytosis), disorders affecting blood plasma (e.g., sepsis, hemophilia, von Willebrand disease, hypercoagulable state, deep venous thrombosis and disseminated intravascular coagulation (DIC)).
[00280] In one embodiment, the saRNA described herein may be used to treat albumin disorders such as analbuminaemia, bisalbuminemia, familial dysalbuminemic hyperthyroxinemia and hypoalbuminemia.
[00281] In one embodiment, the saRNA described herein may be used to treat coagulopathies such as haemophilia, activated protein C resistance, amyloid purpura, antiphospholipid syndrome, antithrombin III deficiency, Bernard-Soulier syndrome, bleeding diathesis, cavernous sinus thrombosis, coagulopathy, congenital afibrinogenemia, congenital amegakaryocytic thrombocytopenia, deep vein thrombosis, disseminated intravascular coagulation, dyiibrinogcncmia, essential thrombocythaemia, Evans syndrome, factor I deficiency, factor V leiden, factor XIII deficiency, Fechtners syndrome, giant platelet disorder, Glanzmann' s thrombasthenia, gray platelet syndrome, Harris platelet syndrome, heparain- induced thrombocytopenia, I lermansky-Pudlak Syndrome, hyperprothrombinemia,
hypoprothrombinemia, idiopathic thrombocytopenic purpura, Kasabach-Merritt syndrome, May- Hegglin anomaly, neonatal alloimmune thrombocytopenia, Onyalai, phlebothrombosis, platelet storage pool deficiency, platelet transfusion refractoriness, Prett Test, primary fibrinogenolysis, protein C deficiency, protein S deficiency, prothrombin G2021 OA, pscudo gray platelet syndrome, purpura fulminans, Quebec platelet disorder, Scott syndrome, sticky platelet syndrome, superficial vein thrombosis, thrombocytopenia, thrombocytopenic purpura, thrombocytosis, thrombophilia, thrombotic microangiopathy, thrombotic thrombocytopenic purpura, Upshaw-Sehulman syndrome and Von Willebrand disease.
[00282] In one embodiment, the saRNA described herein may be used to treat blood disease and disorders such as anemia, leukemia, lymphoma, multiple myeloma, sepsis and sickle-cell disease, atypical hemolytic uremic syndrome, chomeia, cytopenia, elliptocyte, fibrinogenolysis, hematolic disease, Hemolytic -uremic syndrome, hepatic veno-occlusive disease, hypervisocity syndrome, leukemoid reaction, leukocytosis, leukopenia, light chain deposition diease, macroglobulinemia, omithinaemia, pancytopenia, panmyelosis,
pseudothrombocytopenia, Shwachman-Dίamond syndrome and tumor lysis syndrome.
[00283] In one embodiment, the saRNA described herein may be used to treat hemorrhagic and hematological disorders of the fetus and newborn such as ABO, anti-Kell, anti- Rhc, anti-RhE, kernicterus, neonatal jaundice, placental insufficiency, polycythemia and Rh disease.
[00284] In one embodiment, the saRNA described herein may be used to treat hematologic neoplasms such as hematologic malignant neoplasms (e.g., leukemia, lymphoma, multiple myeloma) and myeloid neoplasia (e.g., myeloid leukemia).
[00285] In one embodiment, the saRNA described herein may be used to treat histoeytosis related conditions such as Erdheim-Chester disease, Hemophagocytic
lymphohistiocytosis, Langerhans cell histiocytosis, malignant histiocytosis, Non-Langerhans cell histiocytosis, Non-X histiocytosis, and X-type histiocytosis.
[00286] In one embodiment, the saRNA described herein may be used to treat lymphocytic disorders such as lymphocytic immune system disorders (e.g., autoimmune lymphoproliferative syndrome, hypergammaglobulinemia, hypogammaglobulinemia, lymphorproiiferative disorders, paraproteinemia, persistent polyclonal B-ceil lymphocytosis and RASopathy) lymphocytic leukemia (e.g., acute lymphocytic leukemia and chronic lymphocytic leukemia) and lymphoma. [00287] In one embodiment, the saRNA described herein may be used to treat monocyte and granulocyte disorders such as Agranulocytosis, Autoimmune neutropenia, Bandemia, Basopenia, Basophilia, Eosinopenia, Eosinophilia, Febrile neutropenia, Granulocytosis, Hypereosinophiiia, Hypereosinophilic syndrome, Hypersegmented neutrophil, Monocytopenia, Monocytosis, Morning pseudoneutropenia, Neutropenia and Neutrophilia.
[00288] In one embodiment, the saRNA described herein may be used to treat red blood cell disorders such as anemia, African iron overload, Aldolase A deficiency, Atransferrinemia, Bahima disease, Congenital dyserythropoietic anemia, Erythroid dysplasia, Haemochromatosis type 3, Hemoglobin Lepore syndrome, Hemoglobin variants, Hemoglobinemia,
Hemosiderinuria, Hereditary pyropoikilocytosis, HFE hereditary haemochromatosis, Iron deficiency, Latent iron deficiency, McLeod syndrome, Methemoglobinemia, Microeytosis, Myomatous erythrocytosis syndrome, Poikilocytosis, Polychromasia, Polycythemia, Porphyria, Reticulocytopenia, Rh deficiency syndrome, Sick cell syndrome, Spherocytosis,
Sulfhemoglobinemia, and Transient erythroblastopenia of childhood,
[00289] In one embodiment, the saRNA described herein may be used to treat breast disorders such as breast neoplasia (e.g., breast cancer), breast cyst, breast disease, breast engorgement, breast hematoma, breast lump, Duct ectasia of breast, Hypertrophy of breast, Inverted nipple, Fissure of the nipple, Mammary myofibroblastoma, Mastitis, Mastodynia, Mondor's disease, Nonpuerperal mastitis, Pseudoangiomatous stromal hyperplasia, Ptosis (breasts), Subareolar abscess, Tuberous breasts, and Zuska's disease
[00290] In one embodiment, the saRNA described herein may be used to treat cardiovascular diseases such as cardiogenetic disorders, congenital disorders of the circulatory system (e.g., congenital heart disease, congenital vascular defects), pericardium disease (e.g., mesothelial hyperplasia), heart disease and vascular disease.
[00291] In one embodiment, the saRNA described herein may be used to treat cardiogenetic disorders such as Alagille syndrome, Brugada syndrome, dilated cardiomyopathy, Familial atrial fibrillation. Hypertrophic cardiomyopathy, Long QT syndrome, McLeod syndrome, Sakati-Nyhan-Tisdale syndrome, Short QT syndrome, Smith Martin Dodd syndrome and Timothy syndrome.
[00292] In one embodiment, the saRNA described herein may be used to treat heart disease such as cardiac dysrhythmia, cardiomegaly, cardiomyopathy, cardiopulmonary resuscitation, chronic rheumatic heart disease, congenital heart disease, heart neoplasia, ischemic heart disease, pericardial disorder, valvular heart disease, Acute decompensated heart failure, Aneurysm of heart, Arteriosclerotic heart disease, Athletic heart syndrome, Atrioventricular fistula, Autoimmune heart disease, Brown atrophy of the heart, Cardiac amyloidosis, Cardiac asthma, Cardiac contractility modulation, Cardiac syndrome X, Cardiogenic shock,
Cardiophobia, Cardiorenal syndrome, Cardiotoxicity, Carditis, Coronary artery aneurysm, Coronary artery anomaly, Coronary artery disease, Coronary artery ectasia, Coronary occlusion, Coronary thrombosis, Coronary vasospasm, Coeur en sabot, Coxsackievirus-induced
cardiomyopathy, Diastolic heart failure, Dressler's syndrome, Duroziez's disease, Eisenmenger's syndrome, Embryoeardia, Endocardial fibroelastosis, Heart failure, High-output cardiac failure, Hyperdynamic precordium. Idiopathic giant-cell myocarditis, Interventricular dyssynchrony, Intraventricular dyssynchrony, Isolated atrial amyloidosis, Keshan disease, Kounis syndrome, Mydicar, Myocardial bridge, Myocardial disarray, Myocardial rupture, Myocardial scarring, Myocardial stunning, Myocarditis, Nonbacterial thrombotic endocarditis, Ostial disease, Phosphorus and non-atherosclerotic heart disease, Postpericardiotomy syndrome, Right axis deviation, Roemheld syndrome, Shone's syndrome, Traumatic cardiac arrest, Ventricular aneurysm, Viral cardiomyopathy, and Waon therapy.
[00293] In one embodiment, the saRNA described herein may be used to treat vascular disease such as cerebrovascular disease, congenital vascular defect, disease of the arteries, arterioles and capillaries, disease of the aorta, disease of veins, lymphatic vessels and lymph nodes, hypertension, ischemia, vascular neoplasia, Aggressive angiomyxoma, Anemic infarct, Aneurysm, Angiodysplasia, Angiopathy, Annuloaortic ectasia, Aortitis, Aortoiliac occlusive disease, Arterial stiffness, Arteriolosclerosis, Arteriosclerosis, Atheroma, Atherosclerosis, Brain ischemia, Thromboangiitis obliterans, Capillaritis, Carotid artery stenosis, Carotid bruit, Cerebral amyloid angiopathy, Cholesterol embolism, Chronic cerebrospinal venous insufficiency, Chronic venous insufficiency, Circulating endothelial cell, CLOVES syndrome, Collapsed vein, Corona phlebectatica, Degos disease, Diabetic angiopathy, Diabetic nephropathy, Dural arteriovenous fistula, Endothelial dysfunction, Endotheliitis, Familial aortic dissection, Fatty streak, Fibro- Adipose Vascular Anomaly, Fibromuscular dysplasia, Flammer syndrome, Hemorrhagic infarct, Hereditary cystatin C amyloid angiopathy, Hollenhorst plaque, Hot aches, Hyaline
arteriolosclerosis, Hyperplastic arteriolosclerosis, Hypotension, Inferior vena cava syndrome, Intermittent claudication, Intradural pseudoaneurysm, Jugular vein ectasia, Kawasaki disease, Loeys Dictz syndrome, Lupus vasculitis, Macrovascular disease, Metanephric dysplastic hematoma of the sacral region, Monckeberg's arteriosclerosis, Obliterating endarteritis,
Orthostatic hypertension, Orthostatic hypotension, Orthostatic intolerance, Paget Schroettcr disease, Paradoxical embolism, Peripheral vasculopathy, Phlebitis, Phlegmasia alba dolens, Phlegmasia cerulea dolens, Portacaval anastomosis, Portal vein thrombosis, Postural orthostatic tachycardia syndrome, Pscudothrombophlcbitis. Pulmonic regurgitation, Pylephlebitis,
Rasmussen's aneurysm, Sack Barabas syndrome, Stewart-Treves syndrome, Superior mesenteric artery syndrome, Susac's syndrome, Takayasu's arteritis, Thoracic aortic aneurysm, Thrombophlebitis, Varicocele, Vascular anomaly, Vascular disease, Vascular malformation, Vaso-ocelusive crisis, Vegetative-vascular dystonia, Venous ulcer, Vertebral artery dissection, and Vertebrobasilar insufficiency,
[00294] In one embodiment, the saRNA described herein may be used to treat cutaneous conditions such as acneiform eruptions, Autoinflammatory syndromes, , Chronic blistering cutaneous conditions, Conditions of the mucous membranes, Conditions of the skin appendages , Conditions of the subcutaneous fat , Connective tissue diseases (e.g., cutaneous lupus erythematosus and systemic connective tissue disorders), Cutaneous congenital anomalies (e.g., genodermatoses), Dermal and subcutaneous growths , Dermatitis (e.g., atopic dermatitis, contact dermatitis, eczema, pustular dermatitis, seborrheic dermatitis), Disturbances of human pigmentation , Drug eruptions, Endocrine-related cutaneous conditions , Eosinophilic cutaneous conditions , Epidermal nevi, Epidermal neoplasms, Epidermal cysts, Erythemas, Integumentary neoplasia , Lymphoid-related cutaneous conditions, Melanocytic nevi and neoplasms (e.g., melanoma), Monocyte- and macrophage-related cutaneous conditions, Mucinoses ,
Neurocutaneous conditions , Noninfectious immunodeficiency-related cutaneous conditions, Papulosquamous disorders (e.g., Lichenoid eruptions and psoriasis), Papulosquamous hyperkeratotic cutaneous conditions (e.g., palmoplantar keratodermas), Pregnancy-related cutaneous conditions, Pruritic skin conditions, Reactive neutrophilic cutaneous conditions, Recalcitrant palmoplantar eruptions , Skin conditions resulting from errors in metabolism , Skin conditions resulting from physical factors (e.g., bums, immersion foot syndrome, ionizing radiation-induced cutaneous conditions), Urticaria and angioedema, Vascular-related cutaneous conditions, atrophia maculosa varioliformis cutis, and dermatopathic lymphadenopathy. [00295] In one embodiment, the saRNA described herein may be used to treat digestive diseases such as accessory digestive gland disorders (e.g., diseases of the liver, gallbladder, biliary tract and pancreas, Alagille syndrome, biliary fever, Dub in -John son syndrome and Gilbert's syndrome), gastrointestinal cancer, tongue disorders, mesothelial hyperplasia, colitis, diseases of the appendix, diseases of the intestine, disease of the oesophagus, stomac and duodenum, steatorrhea-related diseases, peritoneum disorders, , Alvarez' syndrome, Heyde's syndrome, Reynolds syndrome, Rocmhcld syndrome, Abdominal epilepsy, Aerophagia, Ameboma, Anorectal abscess, Anismus, Anorectal disorder, Bezoar, Bile acid malabsorption, Blind loop syndrome, Bowel infarction, Bowel obstruction, Callous ulcer, Cameron lesions, Campylobacteriosis, Cholera, Coeliae disease, Cronkhite-Canada syndrome, Cryptitis,
Dientamoebiasis, Distal intestinal obstruction syndrome, Duodenal atresia, Dysphagia, Enteric neuropathy, Enterolith, Enteropathy, Eosinophilic gastroenteritis, Epiploic appendagitis, Fibrosing colonopathy, Functional constipation, Functional gastrointestinal disorder, Fundie gland polyposis, Gallstone ileus, Gastrointestinal bleeding, Gastrojejunocolic fistula, Globus pharyngis. Hematochezia, High-altitude flatus expulsion, Hirschsprung's disease,
Hyperehlorhydria, Ileosigmoid knot, Inflammatory bowel disease, Intestinal metaplasia, Intestinal neuronal dysplasia, Intestinal spirochetosis, Intestinal varices, Kcriorrhea. Lower gastrointestinal bleeding, Malabsorption, Megacolon, Megarectum, Melanosis coli,
Microsporidiosis, Microvillous inclusion disease, Mucositis, Necrotizing enterocolitis,
Pancolitis, Perianal hematoma, Peutz-Jeghers syndrome, Pharyngitis, Pneumatosis intestinalis, Pouchitis, Proctocolitis, Protein losing enteropathy, Pseudodiarrhea, Radiation enteropathy, Radiation proctitis, Rectal bleeding, Rectal hemorrhage, Sandifer syndrome, Splenic flexure syndrome, Steatorrhea, Stercoral perforation, Stercoral ulcer, Stress ulcer, Superior mesenteric artery syndrome, Tropical sprue and Whipple's disease.
[00296] In one embodiment, the saRNA described herein may be used to treat diseases of the ear and mastoid process such as Autoimmune inner ear disease, Balance disorder, Bilateral vestibulopathy, Cauliflower ear, Cryptotia, Ear disease, Earache, Enlarged vestibular aqueduct, Hcmotympanum. Hyperacusis, Macrotia, Mastoidectomy, Mastoiditis, Microtia, Misophonia, Mondini dysplasia, Otalgia, Otomycosis, Sopite syndrome, Superior canal dehiscence, Surfer's ear, Tinnitus, Usher syndrome, and Vestibular hyperacusis. [00297] In one embodiment, the saRNA described herein may be used to treat endocrine disease such as adrenal gland disorders, congenital disorders of the endocrine system, disorders of the endocrine pancreas (e.g., diabetes), endocrine gonad disorders, endocrine neoplasia (e.g., thyroid cancer), endocrine-related cutaneous conditions, hypothalamus disorders, parathyroid disorders, pituitary disorders, thyroid disease (e.g., thyroid cancer and thyroid tumor), Acute infectious thyroiditis, Autoimmune thyroiditis, Colloid nodule, Congenital hypothyroidism, Cretinism, Dc Quervain's thyroiditis, Euthyroid, Euthyroid sick syndrome, Familial
dysalhuminemic hyperthyroxmemia, Goitre, Goitrogen, Graves' disease, Graves'
ophthalmopathy, Hashimoto's thyroiditis, Hashitoxicosis, Hyperthyroidism, Hyperthyroxmemia, Hypothyroidism, Maternal hypothyroidism, Myxedema coma, Myxedematous psychosis, Ord's thyroiditis, Palpation thyroiditis, Polar T3 syndrome, Postpartum thyroiditis, Radiation-induced thyroiditis, Radioactive iodine uptake test, Riedel's thyroiditis, Subacute lymphocytic thyroiditis, Subacute thyroiditis, Thyroid disease, Thyroid dyshormonogenesis, Thyroid hormone resistance, Thyroid neoplasm, Thyroid nodule, Thyroid storm, Thyroidectomy, Thyroiditis, Thyroidologist, Thyrotoxic periodic paralysis, Thyrotoxicosis factitia, Toxic multinodular goitre, Toxic nodular goiter, Van Wyk and G rum bach syndrome, Winter-over syndrome, Congenital hyperinsulinism, Hyperglycemia, Hyperinsulinemia, Hyperinsulinemic hypoglycemia, Hyperinsulinism,
Hyperproinsulinemia, Hypoglycemia, Idiopathic hypoglycemia, Impaired fasting glucose, Impaired glucose tolerance, Insulitis, Ketotic hypoglycemia, Nesidioblastosis,
Oxyhyperglyeemia, Reactive hypoglycemia, Spontaneous hypoglycemia, Stress hyperglycemia, Zollinger-Ellison syndrome, Achard-Thiers syndrome, Adenosis, Adipsia, Autoimmune polyendocrine syndrome, Autoimmune polyendocrine syndrome type 1 , Autoimmune polyendocrine syndrome type 2, Autoimmune polyendocrine syndrome type 3, Autosomal dominant hypophosphatemia rickets, Cerebral salt-wasting syndrome, Chondrodysplasia Blomstrand, Diabetes insipidus, Endemic goitre, Endocrine bone disease, Galactorrhea hyperprolactinemia, Goitre, Gynecomastia, Idiopathic short stature, hicidentaloma, Metabolic syndrome, Morgagni Stewart Morel syndrome, Osteoporosis, Pickardt syndrome, Pituitary Pars Intermedia Dysfimction, POEMS syndrome, Postural orthostatic tachycardia syndrome, and Pseudo-Cushing's syndrome.
[00298] In one embodiment, the saRNA described herein may be used to treat metabolic diseases such as, acid-base disturbances, albinism, amyloidosis, electrolyte disturbances, gout, inborn errors of metabolism (e.g., amino acid metabolism disorders, cholesterol and steroid metabolism disorders, eieosanoid metabolism disorder, fatty-aeid metabolism disorders, glycoprotein metabolism disorders, glycoprotein metabolism disorders, heme metabolism disorders, lipid metabolism disorders, lysosomal storage diseases, phospholipid metabolism disorders, proteoglycan metabolism disorders, TCA and ETC metabolism disorders),
leukodysrophies, lipid disorders (e.g., lipid metabolism disorders and lipid storage disorders), obesity, senescence, Abnormal basal metabolic rate, Danon disease, Exercise-associated hyponatremia, High anion gap metabolic acidosis, MDP syndrome, Metabolic myopathy, Pansteatitis, Refeeding syndrome, Winchester syndrome, X-linked hypophosphatemia,
Combined hyperlipidemia, Dyslipidemia, Hypcrl ipidemia. Hypocholesterolemia,
Hypolipoproteinemia, Acid lipase disease, Batten disease, Canavan disease, Cerebrotendineous xanthomatosis, Cholesteryl ester storage disease, Fabry disease, Farber disease, Gangliosidosis, Gaucher's disease, GM 1 gangliosidoses, GM2 gangliosidoses, Infantile neuronal ceroid lipofuscinosis, Jansky-B ielschowsky disease, Krabbc disease, Lysosomal Acid Lipase
Deficiency, Metachromatic leukodystrophy, Multiple sulfatase deficiency, Neuronal ceroid lipofuscinosis, Niemann-Pick disease (e.g., Niemann-Pick disease, SMPD 1 -associated or Niemann Pick disease, type C), Pelizaeus-Merzbacher disease, Sandhoff disease, Schindlcr disease, Sphingolipidoses, Sulfatidosis, Tay-Sachs disease, and Wolman disease.
[00299] In one embodiment, the saRNA described herein may be used to treat lysosomal storage diseases such as Activator Deficiency/GM2 Gangliosidosis, Alpha-mannosidosis, Aspartylglucosaminuria, Cholesteryl ester storage disease, Chronic Hexosaminidase A
Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis,
Galactosialidosis, Gaucher Disease (e.g., Gaucher Disease Type I, Gaucher Disease Type II, Gaucher Disease Type III), GM1 gangliosidosis (e.,g Infantile, Late infantile/Juvenile,
Adult/Chronic), I -Cell disease/Mucolipidosis II, Infantile Free Sialic Acid Storage Disease/ISSD, Juvenile Hexosaminidase A Deficiency, Krabbc disease (e.g., Infantile Onset, Late Onset), Lysosomal acid lipase deficiency (e.g., Early onset, Late onset), Metachromatic Leukodystrophy, Mucopolysaccharidoses disorders (e.g., Pseudo-Hurler polydystrophy/Mucolipidosis IIIA, MPSI Hurler Syndrome, MPSI Scheie Syndrome, MPS I Hurler-Scheie Syndrome, MPS II Hunter syndrome, Sanfilippo syndrome Type A/MPS III A, Sanfilippo syndrome Type B/MPS III B , Sanfilippo syndrome Type C/MPS III C, Sanfilippo syndrome Type D/MPS III D, Morquio Type A MPS EVA, Morquio Type B/MPS IVB, MPS IX Myaluronidase Deficiency, MPS VI
Maroteaux-Lamy. MPS VII Sly Syndrome, Mucolipidosis I/Sialidosis, Mucolipidosis IIIC, Mucolipidosis type IV), Multiple sulfatase deficiency, Niemann-Piek Disease (e.g., Type A, Type B, Type C), Neuronal Ceroid Lipofuscinoses (e.g., CLN6 disease - Atypical Late Infantile, Late Onset variant, Early Juvenile, Batten-Spielmeyer-Vogt/Juvenile NCL/CLN3 disease, Finnish Variant Late Infantile CLN5, Jansky-B ielschowsky disease/Late infantile CLN2/TPP1 Disease, Kufs/Adult-onset NCL/CLN4 disease, Northern Epilepsy/variant late infantile CLN8, Santavuori-I lalti a I n tan tile CLN1/PPT disease, Beta-mannosidosis), Pompe disease/Glycogen storage disease type II, Pyenodysostosis, Sandhoff disease/ Adult Onset/GM2 Gangliosidosis, Sandhoff disease/GM2 gangliosidosis - Infantile, Sandhoff disease/GM2 gangliosidosis - Juvenile, Schindler disease, Salla disease/Sialic Acid Storage Disease, Tay-Sachs/GM2 gangliosidosis and Wolman disease.
[00300] In one embodiment, the saRNA described herein may be used to treat nutritional diseases such as nutritional deficiencies, hyperalimentation and malnutrition,
[00301] In one embodiment, the saRNA described herein may be used to treat diseases of the eye and adnexa such as disorders of the choroid and retina, congenital disorders of the eyes, disorders of the conjunctiva, disorders of the eyelid, lacrimal system and orbit, glaucoma, disorders of the iris and ciliary body, disorders of the lens, disorders of the ocular muscles, binocular movement, accommodation and refraction, ocular neoplasia, disorders of the optic nerve and visual pathways, periorbital conditions, disorders of the sclera and cornea, disorders of the vitreous body and globe, Actinic conjunctivitis, Acute hemorrhagic conjunctivitis, Acute posterior multifocal placoid pigment epitheliopathy, Acute zonal occult outer retinopathy, Amaurosis, Amaurosis fugax, Angioid streaks, Anisocoria, Anterior segment mesenchymal dysgenesis, Argyll Robertson pupil, Arlt's line, Arteritic anterior ischemic optic neuropathy, Asteroid hyalosis, Autoimmune optic neuropathy, Balint's syndrome, Bardet Bicdl syndrome, Bitot's spots, Blast-related ocular trauma, Boehm syndrome, Boston's sign, Brushfield spots, Bullous keratopathy, Buphthalmos, Cerebral achromatopsia, Chemosis, Chiasmal syndrome, Closed-eye hallucination, Cogan syndrome, Coloboma, Coloboma of optic nerve, Computer vision syndrome, Cone dystrophy, Conjugate gaze palsy, Corectopia, Cornea plana 1 , Cornea plana 2, Cornea verticillata, Corneal abrasion, Corneal ectatic disorders, Corneal hydrops, Corneal perforation, Dacryocystocele, Dermatochalasis, Dermoid cyst, Diffuse lamellar keratitis, Dissociated vertical deviation, Distorted vision, Drusen, Eales disease, Endophthalmitis, Panophthalmia, Hpiblepharon, Periorbital dark circles, Eye disease, Eye injuries during general anaesthesia, Eye injury, Fixation disparity, Flammer syndrome, Fleischer ring, Flexner- Wintersteiner rosette, Foster-Kennedy syndrome, Fuchs heterochromic iridocyclitis, Fuchs spot, Giant-cell arteritis, Graves' ophthalmopathy, Heterochromia iridum, Homer's syndrome, Hypopyon, Hypotelorism, Idiopathic intracranial hypertension, Idiopathic orbital inflammatory disease, Infantile esotropia, Infiltrative ophthalmopathy, Interstitial keratitis, Intraocular hemorrhage, Intraocular schwannoma, Intraretinal microvascular abnormalities, Iridocorneal endothelial syndrome, Iridoplegia, Irvine-Gass syndrome, Kcratoconus. Keratomalacia, Khodadoust line, Krukenberg's spindle, Late congenital syphilitic oculopathy, Lattice degeneration, Lecithin cholesterol aeyltransferase deficiency, Leukoeoria, Limbal nodule, Loa loa fdariasis, Macropsia, Macular hole, Macular hypoplasia, Macular telangiectasia,
Maeulopathy, Marcus Gunn pupil, Marshall syndrome, Mechanical anisoeoria, Microcoria, Micropsia, Miosis, Mirhosseini-Holmes- Walton syndrome, Mitochondrial optic neuropathies, Monotixation syndrome, Moore's lightning streaks, Munson's sign, Mydriasis, Neonatal conjunctivitis, Nuclear sclerosis, Nystagmus, Ocular albinism type 1 , Ocular larva migrans, Ocular melanosis, Ocular myasthenia, Ocular neurosis, Ocular rosacea, Oculomotor apraxia, Optic disc pit, Optic nerve hypoplasia, Optic neuropathy, Optic papillitis, Panophthalmitis, Parinaud's syndrome, Pelopsia, Persistent hyperplastic primary vitreous, Persistent tunica vasculosa lentis, Phantom eye syndrome, Phlyctenule, Photopsia, Physiological anisoeoria, Pigment dispersion syndrome, Pneumosinus dilatans, Polycoria, Post-LASIK ectasia, Posterior ischemic optic neuropathy, Posterior vitreous detachment, Presumed ocular histoplasmosis syndrome, Progressive outer retinal necrosis, Proliferative vitreoretinopathy, Pseudoexfoliation syndrome, Pseudomyopia, Pseudophakic photic phenomena, Pseudostrabismus, Punctate epithelial erosions, Recurrent corneal erosion, Retinal degeneration (rhodopsin mutation), Retinal dysplasia, Roth's spot, Sclerocomea, Septo-optic dysplasia, Siegrist streaks, Skew deviation, Solar retinopathy, Staphyloma, Stargardt disease, Stickler syndrome, Sudden acquired retinal degeneration, Superior limbic keratoconjunctivitis, Superior oblique myokymia,
Sympathetic ophthalmia, Synchysis seintillans, Teleopsia, Tenonitis, Terrien's marginal degeneration, Terson syndrome, Toxic anterior segment syndrome, Trachoma, Trochleitis, Uveoparotitis, Vernal keratoconjunctivitis, Vision loss, Visual impairment, Visual looming syndrome, Viircorctinopathy. Vitreous touch syndrome, Vossius ring, Wagner's disease, Weill- Marchesani syndrome, White dot syndromes, cataract, persistent fetal vasculature, and X-l inked congenital stationary night blindness.
[00302] In one embodiment, the saRNA described herein may be used to treat foot diseases such as immersion foot syndrome, trench foot, Achilles bursitis, Ainhum, Athlete's foot, Blue toe syndrome, Brachymetatarsia, Bunion, Burning feet syndrome, Calcaneal spur, Callus, Chilblains, Equinovalgus, Haglund's deformity, Haglund's syndrome, Hallux varus, Janeway lesion, Metatarsophalangeal joint sprain, Morton's neuroma, Morton's toe, Palmoplantar keratoderma, Plantar calcaneal bursitis, Plantar fasciitis, Plantar wart, Ship foot, Syndesmosis procedure, Tailor's bunion, Tarsal coalition, Tarsal tunnel syndrome and Toe walking,
[00303] In one embodiment, the saRNA described herein may be used to treat genitourinary system disease such as gynecologic disorders, male genital disorders, urological conditions, bulbar urethral necrosis, Frasier syndrome and reproductive system disease.
[00304] In one embodiment, the saRNA described herein may be used to treat gynecologic disorders such as congential disorders of the female genital organs, gynecologic neoplasia (e.g., gynecological cancer), infertility, inflammatory diseases of the female pelvic organ, noninflammatory disorders of the female genital tract (e.g., menstrual disorders) and sexually transmitted diseases and infections, Asherman's syndrome, Breakthrough bleeding, Cervical motion tenderness, Cryptomenorrhea, Dermoid cyst, Endosalpingiosis, Esthiomene, Female genital disease, Fitz-Hugh-Curtis syndrome, Gynecologic hemorrhage, Hematocolpos, Hyperestrogenism, Hypergonadism, Hypogonadism, Labial fusion, Lipschutz ulcer, Meigs' syndrome, Metrophlebitis, Mixed gonadal dysgenesis, Ovarian apoplexy, Ovarian vein syndrome, Papillary hidradenoma, Persistent genital arousal disorder, Polycystic ovary syndrome, Pelvic floor dysfunction, Premature ovarian failure, Premenstrual dysphoric disorder, Rokitansky nodule, Round ligament pain, XY gonadal dysgenesis, Tight hymenal ring,
Vaginismus, Vulvar vestibulitis, Vulvodynia and XX gonadal dysgenesis.
[00305] In one embodiment, the saRNA described herein may be used to treat gynecological cancer such as Adenomyoma, Chorioblastoma, Chorioepithelioma,
Dysgerminoma, Embryonal carcinoma, Endometrial intraepithelial neoplasia, Gestational choriocarcinoma, Gonadoblastoma, Hereditary leiomyomatosis and renal cell cancer syndrome, Mixed Mullcrian tumor, Mucinous tumor, Ovarian fibroma, Ovarian serous cystadenoma, Peutz- Jeghers syndrome, Polyembryoma, Thecoma, Trophoblastic neoplasm, Uterine fibroid, Vaginal intraepithelial neoplasia, and Vulvar intraepithelial neoplasia,
[00306] In one embodiment, the saRNA described herein may be used to treat noninflammatory disorders of the female genital tract such as menstrual disorders, Adenomyosis, Asherman's syndrome, Atrophic vaginitis, Atypical polypoid adenomyoma, Cervical
incompetence, Cervical intraepithelial neoplasia, Cervical polyp, Corpus luteum cyst, Cystocele, Dysfunctional uterine bleeding, Dyspareunia, Dysplasia, Endometrial hyperplasia, Endometrial polyp, Endometriosis, Endometriosis of ovary, Enterocele, Fallopian tube obstruction, Female genital prolapse, Female infertility, Follicular cyst of ovary, Gonadal torsion, Recurrent miscarriage, Hematometra, Hematosalpinx, Kraurosis vulvae, Leukorrhea, Mittelschmerz, Obstetric fistula, Ovarian apoplexy, Ovarian cyst, Ovarian disease, Ovarian hyperstimulation syndrome. Ovarian torsion, Paraovarian cyst, Pelvic congestion syndrome, Persistent genital arousal disorder, Rectocele, Rectovaginal fistula, Retroverted uterus, Salpingitis isthmica nodosa, Sigmoidocele, Stenosis of uterine cervix, Theca lutein cyst, Urethrocele, Uterine prolapse, Vaginal bleeding, Vaginismus, Van Wyk and Grumbaeh syndrome and Vesicovaginal fistula.
[00307] In one embodiment, the saRNA described herein may be used to treat male genital disorders such as congenital disorders of the male genital organs, epididymis disorders, male genital neoplasia (e.g., prostate cancer), penis disorders, prostate disorders, sexually transmitted diseases and infections and testicle disorders.
[00308] In one embodiment, the saRNA described herein may be used to treat male genital neoplasia such as Bowen's disease, Penile cancer, Choriocarcinoma, Embryoma,
Embryonal carcinoma, Endodermal sinus tumor, Extramammary Paget's disease, Germ cell tumor, Germinoma, Gonadoblastoma, Granulosa cell tumor, Gynandroblastoma, High-grade prostatic intraepithelial neoplasia, Intratubular germ cell neoplasia, Leydig cell tumour, Mule spinners' cancer, Prostate cancer, Reinke crystals, Seminoma, Sertoli cell tumour, Sertoli-Leydig cell tumour, Sex cord-gonadal stromal tumour, Spermatocyte seminoma and Teratoma.
[00309] In one embodiment, the saRNA described herein may be used to treat urological conditions such as Bladder stone, Emphysematous cystitis, Enuresis, Eosinophilic cystitis, Foumier gangrene, Hydrocele, Kidney stone, Loin pain hematuria syndrome, Lower urinary tract symptoms, Malakoplakia, Micturition syncope, Nephrogenic adenoma, Obstructive uropathy, Ovarian vein syndrome, Paruresis, Pelvic myoneuropathy, Peutz-Jeghers syndrome, Pneumaturia, Pseudodyssynergia, Purohit-Blaivas Staging System, Renal colic, Renal stone formation in space, Retroperitoneal fibrosis, Sedoanalgesia, Transurethral resection of the prostate syndrome, Urinary tract infection, Urinary tract obstruction, Urinoma, Urolithiasis, Uropathy, Vesieouretic reflux, and Zellweger syndrome.
[00310] In one embodiment, the saRNA described herein may be used to treat hair disease such as Alopecia areata, Alopecia totalis, Alopecia universalis, Androgenic alopecia, Black piedra, Bubble hair deformity, Hair disease, Hair follicle nevus, Hair loss, Hypertrichosis, Hypotrichosis, Kinking hair, Loose anagen syndrome, Monilethrix, Pili annulati, Pili torti, Plica neuropathica, Telogen effluvium, Tinea capitis and Trichotillomania.
[00311] In one embodiment, the saRNA described herein may be used to treat immune system disorder such as asthma, autoimmune diseases, chronic fatigue syndrome, human MHC mediate diseases, hypersensitivity, immunodeficiency lymphocytic immune system disorders, Aagenaes syndrome, Animal allergy, Arthus reaction, Asplenia, Autoimmune heart disease, Autosplenectomy, Caspase-8 deficiency state, Castleman's disease, Chronic fatigue syndrome, Cryofibrinogenemia, Cutaneous small-vessel vasculitis, Cytokine release syndrome, Cytokine storm, Dermic Morgan fold, Ectopic thymus, Extracutaneous mastocytoma, Familial
Mediterranean fever, Gleich's syndrome, Graft-versus-host disease, Gulf War syndrome, Heavy chain disease, History of chronic fatigue syndrome, HLA-B27, Hyper-IgM syndrome type 1 , Hyper-lgM syndrome type 5, Ichthyosis acquisita, Idiopathic CD4+ lymphocytopenia, Immune disorder, Immune dysregulation, Immune reconstitution inflammatory syndrome, Immune- mediated inflammatory diseases, Immunoproliferative disorder, Kimura's disease,
Lymphocytopenia, Mastocytosis, Mortimer's disease, Oral allergy syndrome, Paraneoplastic syndrome, RAS -associated autoimmune leukoproliferative disorder, Seabather's eruption, Systemic inflammatory response syndrome, T cell deficiency, Thymus hyperplasia, Transplant rejection and XMEN disease.
[00312] In one embodiment, the saRNA described herein may be used to treat asthma such as Acute severe asthma, Aspirin-induced asthma, Asthmagen, Baker's asthma, Brittle asthma, Bronchial thermoplasty, Bronchospasm, Cough-variant asthma, Dynamic hyperinflation, Exercise-induced bronchoconstriction, Occupational asthma, and Reactive airway disease. [003 13] In one embodiment, the saRNA described herein may be used to treat mouth diseases such as chronic sclerosing sialadenitis and sialadenitis.
[00314] In one embodiment, the saRNA described herein may be used to treat musculoskeletal disorders such as chrondropathies, congenital disorders of the muscoskeletal system (e.g., arthrogryposis and phocemelia), crystal deposition diseases, joint disorders (e.g., gout), muscular disorders, myoneural junction and neuromuscular diseases (e.g., motor neuron disease and muscular dystrophy), osteopathies (e.g., osteitis and osteonecrosis), skeletal disorders (e.g., osseous and chrondromatous neoplasia), soi tissue disorders, systemic connective tissue disorders, Achard syndrome, Acropachy, Ankylosing hyperostosis, Arterial tortuosity syndrome, Attenuated patella alta, Baker's cyst, BlackBerry thumb, Bone cyst, Bone disease, Cervical spinal stenosis, Cervical spine disorder, Chondroealeinosis, Condylar resorption, Copenhagen disease, Costochondritis, Dead arm syndrome, Dentomandibular Sensorimotor Dysfunction, Diffuse idiopathic skeletal hyperostosis, Disarticulation,
Dolichostenomelia, Du Bois sign, Enthesopathy, Enthesophyte, FACES syndrome, Facet syndrome, Foot drop, Genu reeurvatum, Giant-cell tumor of the tendon sheath, Grisel's syndrome, Hanhart syndrome, Heavy legs, Hill-Sachs lesion, Injection fibrosis, Intersection syndrome, Intervertebral disc disorder, Joint effusion, Khan Kinetic Treatment, Knee effusion, Knee pain, Lumbar disc disease, Meromelia, Microtrauma, Myelonecrosis, Neuromeehanics of idiopathic scoliosis, Osteolysis, PeUegrini-Stieda syndrome, Pigmented villonodular synovitis, Piriformis syndrome, Potassium-aggravated myotonia, Primary Spine Practitioner,
Pseudarthrosis, Radial tunnel syndrome, Radicular pain, List of repetitive strain injury software, Repetitive strain injury, Saber shin, Sacroiliac joint dysfunction, Seal finger, Sever's disease, Snapping hip syndrome, Spinal decompression, Spinal enthesopathy, Spondyloarthropathy, Sternal cleft, Subungual exostosis, Synovial sarcoma, Tailor's bunion, Temporomandibular joint dysfunction, Tension myositis syndrome, Tietze syndrome, Traumatic spondylopathy, Ulnar nerve entrapment, Ulnar tunnel syndrome, Unicameral bone cyst, Winged scapula, Yoga foot drop and Zenker's paralysis.
[00315] In one embodiment, the saRNA described herein may be used to treat neurological disorders such as epilepsy, headaches, sleep disorders, stroke, central nervous system disorders, peripheral nervous system disorders, Tay-Sachs disease, dyslexia, Tourettes syndrome, Acute flaccid myelitis, Adrenoleukodystrophy, Aicardi syndrome, Alexander disease, Amblyaudia. Asperger syndrome, User:ParanoidLemmings sandbox. Autism, Batten disease, Canavan disease, Cerebral palsy, Childhood disintegrative disorder, Epilepsy, Idiopathic childhood occipital epilepsy of Gastaut, Krabbc disease, Leigh's disease, Lysosomal storage disease, Metachromatic leukodystrophy, Myoclonic astatic epilepsy, Panayiotopoulos syndrome, Paroxysmal tonic upgaze, Pervasive developmental disorder, Pleomorphic xanthoastrocytoma, Progressive rubella panencephalitis, Rett syndrome, Sensory Dysfunction Disorder, Severe myoclonic epilepsy of infancy, Stereotypic movement disorder, Tay-Sachs disease and Transient hyperammonemia of the newborn,
[00316] In one embodiment, the saRNA described herein may be used to treat central nervous system disorders such as brain disorders, demyelmating diseases of CNS and spinal cord disorders, Anterior horn disease, Cavernous sinus thrombosis, Central nervous system
depression, Central nervous system disease, Central nervous system fatigue, Choroid plexus cyst, Encephalomyelitis, Hippocampal sclerosis, Meningoencephalitis, Misophonia, G.H. Monrad- Krohn, Musical hallucinations, Periventricular leukomalacia, Pineal gland cyst, and
Thalamocortical dysrhythmia.
[00317] In one embodiment, the saRNA described herein may be used to treat brain disorders such as agnosia, aphasias, brain tumor, hypothalamus disorders, leukodystrophies, neurological brain disorder, pituitary disorder, Acquired brain injury, Acute cerebellar ataxia of childhood, Aqueductal stenosis, Basal ganglia disease, Brain abscess, Cerebellopontine angle syndrome, Cerebral amyloid angiopathy, Cerebral hypoxia, Cerebral softening, Cerebral vasospasm, Cerebritis, Cerebrospinal fluid leak, Childhood acquired brain injury, Cortical blindness, Cortical visual impairment, Encephalopathy, Ethylmalonie encephalopathy,
Frontotemporal dementia and parkinsonism linked to chromosome 17, Glial scar, Gliosis, Hashimoto's encephalopathy, Hepatic encephalopathy, Holmes rebound phenomenon,
Hypertensive leukoencephalopathy, La Crosse encephalitis, Leukoencephalopathy,
Leukoencephalopathy with neuroaxonal spheroids, Medial medullary syndrome,
Meningioangiomatosis, Micropolygyria, Normal pressure hydrocephalus, Olivopontocerebellar atrophy, Pediatric acquired brain injury, Pleomorphic xanthoastrocytoma, Primary and secondary brain injury, Progressive multifocal leukoencephalopathy, Right hemisphere brain damage, Samat staging, Sports-related traumatic brain injury, Temporal lobe necrosis, and Ventriculitis. [00318] In one embodiment, the saRNA described herein may be used to treat demyelinating diseases of CNS such as multiple sclerosis, CNS demyelinating autoimmune diseases, Adrenoleukodystrophy, Alexander disease, Alpers' disease, Balo concentric sclerosis, CAMFAK syndrome, Canavan disease, Central pontine myelinolysis, Experimental autoimmune encephalomyelitis, Hereditary CNS demyelinating disease, Krabbe disease,
Leukoencephalopathy with vanishing white matter, Marchiafava-Bignami disease,
Megalencephalic leukoencephalopathy with subcortical cysts, Metachromatic leukodystrophy, Neuromyelitis optica, Pelizaeus-Merzbacher disease, and Diffuse myelinoclastic sclerosis.
[00319] In one embodiment, the saRNA described herein may be used to treat peripheral nervous system disorders such as cranial nerve disorders, nerve disorder, nerve root disorder, plexus disorders, PNS neoplasia, Accessory nerve disorder, Anesthesia dolorosa, Anti-MAG peripheral neuropathy, Autonomic dysreflexia, Axillary nerve dysfunction, Axillary nerve palsy, Charcot-Marie-Tooth disease, Chemotherapy-induced peripheral neuropathy, Chronic solvent- induced encephalopathy, CMV polyradieulomyelopathy, Congenital insensitivity to pain with anhidrosis, Denervation, Diabetic neuropathy, Dysautonomia, Facial nerve paralysis, Familial dysautonomia, Hereditary sensory and autonomic neuropathy, Hereditary sensory and autonomic neuropathy type I, Horner's syndrome. Multiple system atrophy, Nerve compression syndrome, Nerve injury, Orthostatic hypotension, Orthostatic intolerance, Paroxysmal sympathetic hyperactivity, Peripheral neuropathy, Piriformis syndrome, Plexopathy, Polyneuropathy, Postural orthostatic tachycardia syndrome, Primary autonomic failure, Pronator teres syndrome, Proximal diabetic neuropathy, Pudendal nerve entrapment, Pure autonomic failure, Radial neuropathy, Radiculopathy, Sciatica, Thoracic outlet syndrome, Ulnar neuropathy, Vegetative-vascular dystonia, Villaret's syndrome, and Wallenberg's Syndrome.
[00320] In one embodiment, the saRNA described herein may be used to treat orthopedic problems such as Articular cartilage damage, Bankart lesion, Blount's disease, Calcific tendinitis, Dactylitis, Diastematomyelia, Failed back syndrome, Flat feet, Hill-Sachs lesion, Knee pain, Larsen syndrome, Myelopathy, Perthes Lesion, Pigeon toe, Shoulder problem and Spondylitis.
[00321] In one embodiment, the saRNA described herein may be used to treat respiratory diseases such as congenital disorders of the respiratory system, diseases of pleura (e.g., mesothelioma, pleura neoplasia), lower respiratory tract diseases (e.g., asthma, acute lower respiratory diseases, bronchus disorders, chronic lower respiratory disease, lung disorders), pulmonary heart disease and disease of pulmonary circulation, pulmonary lesion, pulmonary tumor, respiratory and cardiovascular disorders specific to the perinatal period, respiratory system neoplasia and upper respiratory track disease.
[00322] In one embodiment, the saRNA described herein may be used to treat lung disorders such as tuberculosis, Alcoholic lung disease, Alpha 1 -antitrypsin deficiency, Alveolar capillary dysplasia, Alveolar lung disease, Antisynthetase syndrome, Asbestosis, Aspergilloma, Atypical pulmonary carcinoid tumor, Baritosis, Brown induration, Cavitation (biology),
Chalicosis, Chiirg Strauss syndrome, Combined pulmonary fibrosis and emphysema, Cystic fibrosis, Flash pulmonary edema, Flock worker's lung, Ghon's complex, Granulomatosis with polyangiitis, Hepatization of lungs, High-altitude pulmonary edema, Honeycombing, Idiopathic pulmonary haemosiderosis, Indium lung, Interstitial lung disease, Lung abscess, Lung disease, Lyeoperdonosis, Lymphangioleiomyomatosis, Obstructive lung disease, Occupational lung disease, Pneumonia alba, Progressive massive fibrosis, Pulmonary alveolar proteinosis,
Pulmonary aspiration, Pulmonary capillary hemangiomatosis, Pulmonary contusion, Pulmonary cyst, Pulmonary fibrosis, Pulmonary hematoma, Pulmonary hemorrhage, Pulmonary hyperplasia, Pulmonary laceration, Pulmonary toxicity, Pulmonary-renal syndrome, Respiratory bronchiolitis interstitial lung disease, Restrictive lung disease, Rheumatoid lung disease, Sarcoidosis,
Surfactant metabolism dysfunction, Swimming-induced pulmonary edema, Swyer- James syndrome, Transfusion-related acute lung injury, Tumor-like disorders of the lung pleura, Ventilation perfusion mismatch, and Ventilator-associated lung injury.
[00323] In one embodiment, the saRNA described herein may be used to treat sexual disorders such as erectile dysfunction, persistent genital arousal disorder, sexual masochism disorder, sexual sadism disorder and spermatorrhea.
[00324] In one embodiment, the saRNA described herein may be used to treat voice disorders such as Bogart-Bacall syndrome, chroditis, laryngitis, muteness, Reinke's edema, spasmodic dysphonia, vocal cord paresis, vocal fold cyst and vocal fold nodule.
[00325] In one embodiment, the saRNA described herein may be used to treat immune system disorders such as asthma, autoimmune diseases, chronic fatigue syndrome, human MHC mediate disease, hypersensitivity, immunodeficiency and lymphocytic immune system disorders.
[00326] In one embodiment, the saRNA described herein may be used to treat autoimmune diseases such as Acute disseminated encephalomyelitis, Alopecia areata, Alopecia universalis, Ankylosing spondylitis, Antiphospholipid syndrome, Antisynthetase syndrome, Aplastic anemia, Apolipoprotein H, Arthritis mutilans, Atrophic gastritis, Autoimmune enteropathy, Autoimmune gastrointestinal dysmotility, Autoimmune hemolytic anemia, Autoimmune hypophysitis, Autoimmune inner ear disease, Autoimmune Oophoritis,
Autoimmune pancreatitis, Autoimmune polyendocrine syndrome, Autoimmune polyendocrine syndrome type 1, Autoimmune polyendocrine syndrome type 2, Autoimmune polyendocrine syndrome type 3, Autoimmune/inflammatory syndrome induced by adjuvants, Balo concentric sclerosis, Behcet's disease, BENTA disease, Bickerstaff s encephalitis, Bullous pemphigoid, Chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, Cicatricial pemphigoid, CNS demyelinating autoimmune diseases, Coeliae disease, Cogan syndrome, Cold agglutinin disease, CREST syndrome, Crohn's disease, Cutaneous manifestations of sarcoidosis, Dermatitis, Dermatomyositis, Diabetes Health, Diabetes mellitus type 1 , Drug-induced lupus erythematosus, Eosinophilic fasciitis, Erythema nodosum, Gestational pemphigoid, Goodpasture syndrome, Granulomatosis with polyangiitis, Graves' disease, Graves' ophthalmopathy, Guillain- Barre syndrome, Hashimoto's encephalopathy, Hashimoto's thyroiditis, Heerfordt's syndrome, Henoch-Schonlein purpura, Hughes-Stovin syndrome, Idiopathic granulomatous hepatitis, Idiopathic inflammatory demyelinating diseases of the central nervous system, IgA nephropathy, Inflammatory bowel disease, IPEX syndrome, Lambert-Eaton myasthenic syndrome, Latent autoimmune diabetes of adults, Pathophysiology of multiple sclerosis, Lichen planus, Linear IgA bullous dermatosis, Lofgren syndrome, Lupus erythematosus, Lupus pernio, Marburg's variant multiple sclerosis, Marie Antoinette syndrome, McDonald criteria, Mixed connective tissue disease, Morphea, Multiple sclerosis , Myasthenia gravis, Neuro-Behcet's disease, Neurological fatigue, Neuromyelitis optica, Neurosarcoidosis, Ocular myasthenia, Opsoclonus myoclonus syndrome, Optic neuritis Ord's thyroiditis, Overlap syndrome, Palindromic rheumatism, Paraneoplastic pemphigus, Pemphigoid, Pemphigus, Poser criteria, Primary biliary cirrhosis, Primary sclerosing cholangitis, Proctitis, Progressive inflammatory neuropathy, Psoriatic arthritis, RAS-associated autoimmune leukoproliferative disorder, Raynaud's phenomenon, Reactive arthritis, Relapsing polychondritis, Reynolds syndrome, Rheumatoid arthritis, Riedel's thyroiditis, Samoyed hereditary glomerulopathy, Sarcoidosis, Satoyoshi syndrome, Diffuse myelinoclastic sclerosis, Scleroderma, Scleromyositis, Sebaceous adenitis, Sjogren's syndrome, Susac's syndrome, Sympathetic ophthalmia, Graves' disease, Systemic lupus erythematosus, Systemic scleroderma. Thrombotic thrombocytopenic purpura, Transverse myelitis, Uhthoff s phenomenon, Ulcerative colitis, Undifferentiated connective tissue disease, Vitiligo and Warm autoimmune hemolytic anemia.
[00327] In one embodiment, the saRNA described herein may be used to treat lymphoma such as Adult T-cell leukemia/ lymphoma, Aggressive lymphoma, AIDS-rclated lymphoma, ALK+ large B-cell lymphoma, Anaplastic large-cell lymphoma, Angioimmunoblastic T-cell lymphoma, B-eell CLL/lymphoma, B-cell lymphoma, Blastic NK cell lymphoma, Burkitt's lymphoma, CD30+ cutaneous T-cell lymphoma, Cutaneous B-cell lymphoma, Cutaneous lymphoma, Cutaneous T cell lymphoma, Diffuse large B-cell lymphoma, Enteropathy-associated T-cell lymphoma, Extranodal NK/T-cell lymphoma, Follicular large-cell lymphoma, Follicular lymphoma, Gastric lymphoma, Germinal center B-cell like diffuse large B-cell lymphoma, Gray zone lymphoma, Hepatosplenic T-cell lymphoma, Intraocular lymphoma, Intravascular large B- cell lymphoma, Large B-cell lymphoma arising in HHV8-associated multicentric Castleman's disease, Large-cell lymphoma, Lennert lymphoma, Lymphoblastic lymphoma, Lymphoma cutis, MALT lymphoma, Mantle cell lymphoma, Marginal zone B-cell lymphoma, Mixed-cell lymphoma, Nodal marginal zone B cell lymphoma, Non-Hodgkin lymphoma, Non-mycosis fungoides CD30- cutaneous large T-cell lymphoma, Orbital lymphoma, Peripheral T-cell lymphoma, Plasmablastic lymphoma, Post-transplant lymphoproliferative disorder, Precursor T- lymphoblastic lymphoma, Primary central nervous system lymphoma, Primary cutaneous follicle center lymphoma, Primary cutaneous immunocytoma, Primary cutaneous marginal zone lymphoma, Primary effusion lymphoma, Primary mediastinal (thymic) large B cell lymphoma, Primary mediastinal B-cell lymphoma, Endoexoenteric, Secondary cutaneous CD30+ large-cell lymphoma, Sezary disease, Splenic lymphoma with villous lymphocytes, Splenic marginal zone lymphoma, Splenic tumor, T-cell lymphoma, T-lymphoblastic leukemia/lymphoma, Thyroid lymphoma and Waldenstrom's macroglobulinemia.
[00328] In one embodiment, the saRNA described herein may be used to treat cancers such as Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML),
Adrenocortical Carcinoma, AIDS-Related Cancers (e.g., Kaposi Sarcoma and AIDS-Related Lymphoma), Anal Cancer, Appendix Cancer, Astrocytomas, Childhood, Atypical
Teratoid/Rhabdoid Tumor, Childhood, Central Nervous System, Basal Cell Carcinoma , Extrahepatic Bile Duct Cancer , Bladder Cancer, Bone Cancer (e.g., Ewing Sarcoma Family of Tumors, Osteosarcoma and Malignant Fibrous Histiocytoma), Brain Stem Glioma, Childhood, Brain Tumor (e.g., Astrocytoma, Brain Stem Glioma, Central Nervous System Atypical
Teratoid/Rhabdoid Tumor, Central Nervous System Embryonal Tumors, Central Nervous System Germ Cell Tumors, Craniopharyngioma, Ependymoma), Breast Cancer , Bronchial Tumors, Burkitt Lymphoma , Carcinoid Tumor (e.g., childhood, Gastrointestinal), Carcinoma of Unknown Primary, Cardiac (Heart) Tumors, Childhood, Central Nervous System (e.g., Atypical Teratoid/Rhabdoid Tumor, Embryonal Tumors, Germ Cell Tumor and Lymphoma), Cervical Cancer, Chordoma, Childhood, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Myeloproliferative Neoplasms, Colon Cancer, Colorectal Cancer, Craniopharyngioma, Cutaneous T-Cell Lymphoma , Duct, Bile, Extrahepatic, Ductal Carcinoma In Situ (DC1S), Embryonal Tumors, Central Nervous System, Endometrial Cancer,
Ependymoma, Esophageal Cancer, Esthesi ©neuroblastoma, Childhood, Ewing Sarcoma, Extracranial Germ Cell Tumor, Childhood, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer (e.g., Intraocular Melanoma and Retinoblastoma), Fallopian Tube Cancer, Fibrous Histiocytoma of Bone, Malignant, and Osteosarcoma, Gallbladder Cancer, Gastric (Stomach) Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Stromal Tumors (GIST), Germ Cell Tumor (e.g., Central Nervous System, Childhood, Extracranial, Childhood, Extragonadal, Ovarian, Testicular), Gestational Trophoblastic Disease, Glioma , Hairy Cell Leukemia, Head and Neck Cancer, Heart Cancer, Hepatocellular (Liver) Cancer, Histiocytosis, Langerhans Cell, Hodgkin Lymphoma, Hypopharyngeal Cancer, , Intraocular Melanoma, Pancreatic Neuroendocrine Tumors, Islet Cell Tumors , Kaposi Sarcoma, Kidney (e.g., Renal Cell and Wilms Tumor and Other Childhood Kidney Tumors), Langerhans Cell Histiocytosis, Laryngeal Cancer, Leukemia (Acute Lymphoblastic (ALL), Acute Myeloid (AML), Chronic Lymphocytic (CLL), Chronic Myelogenous (CML) and Hairy Cell), Lip and Oral Cavity Cancer, Liver Cancer , Lung Cancer (e.g., Childhood, Non-Small Cell, Small Cell), Lymphoma (e.g., AIDS-Related, Burkitt, Cutaneous T-Cell, Hodgkm, Non-Hodgkin and Primary Central Nervous System (CNS)), Macroglobulinemia, Waldenstrom, Male Breast Cancer, Malignant Fibrous Histiocytoma of Bone and Osteosarcoma, Melanoma, Melanoma Intraocular (Eye), Merkel Cell Carcinoma, Malignant Mesothelioma, Metastatic Squamous Neck Cancer with Occult Primary, Midline Tract Carcinoma Involving NUT Gene, Mouth Cancer, Multiple Endocrine Neoplasia Syndromes, Childhood, Multiple Myeloma/Plasma Cell Neoplasm, Mycosis Fungoides, Myelodysplasia Syndromes, Myelodysplastic/Myeloproliferative Neoplasms, Myelogenous Leukemia, Chronic (CML), Myeloid Leukemia, Acute (AML), Multiple Myeloma, Chronic Myeloproliferative Neoplasms, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin Lymphoma, Non-Small Cell Lung Cancer, Oral Cancer, Lip and Oral Cavity Cancer, Oropharyngeal Cancer, Osteosarcoma and Malignant Fibrous Histiocytoma of Bone, Ovarian Cancer (e.g., Childhood, Epithelial, Germ Cell Tumor, Low Malignant Potential Tumor), Pancreatic Cancer, Pancreatic Neuroendocrine Tumors (Islet Cell Tumors), Papillomatosis, Childhood, Paraganglioma, Paranasal Sinus and Nasal Cavity Cancer, Parathyroid Cancer, Penile Cancer, Pharyngeal Cancer, Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple Myeloma, Childhood Plcuropulmonary Blastema, Primary Central Nervous System (CNS) Lymphoma, Primary Peritoneal Cancer, Prostate Cancer, Rectal Cancer, Renal Cell (Kidney) Cancer, Retinoblastoma, Childhood Rhabdomyosarcoma, Salivary Gland Cancer, Childhood, Sarcoma (e.g., Ewing, Kaposi, Osteosarcoma (Bone Cancer), Rhabdomyosarcoma, Soft Tissue and Uterine), Sezary Syndrome, Skin Cancer (e.g., childhood, Melanoma, Merkel Cell Carcinoma and Nonmelanoma), Small Cell Lung Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Cell Carcinoma , Metastatic Squamous Neck Cancer with Occult Primary, Stomach (Gastric) Cancer, Cutaneous T-Cell Lymphoma, Testicular Cancer, Throat Cancer, Thymoma and Thymic Carcinoma, Thyroid Cancer,
Transitional Cell Cancer of the Renal Pelvis and Ureter, Transitional Cell Cancer of the Ureter and Renal Pelvis, Urethral Cancer, Endometrial Uterine Cancer , Uterine Sarcoma, Vaginal Cancer, Vulvar Cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.
[00329] Additional disease and disorders which may be treated using the saRNA of the present invention including, but are not limited to, Pel-Ebstein fever, plasma cell dyscrasia, plasmacytoma, smouldering myeloma, Waldenstrom's macroglobulinemia, Chromosome 5q deletion syndrome, Myelofibrosis, Refractory anemia, Refractory cytopenia, Sideroblastic anemia, Bing-Neel syndrome, Actinic granuloma, Annular elastolytic giant-cell granuloma, Benign cephalic histiocytosis, Birbeck granules, Congenital self-healing reticulohistiocytosis, Darier-Roussy disease, Darier-Roussy saroid, Eruptive histiocytoma, Generalized eruptive histiocytoma, Generalized franuloma annulare, Giant cell elastophagocytosis, Giant-cell retichlohistiocytoma, Granuloma nulare, Granuloma multiforme, Hand-Sehuller-Christian disease, Hashimoto -Pritzker disease, Heerfordt's syndrome, Hereditary progressive mucinous histiocytosis, Indeterminate cell histiocytosis, Letterer-Siew disease, Localized granuloma annulare, Lofgren syndrome, Macular granuloma annulare, Meischer's granuloma, Montgomery syndrome, Multicentric reticulohistiocytosis, Necrobiotic xanthogranuloma, O'Brien granuloma, Papular xanthoma, Patch-type granuloma annulare, Perforating granuloma annulare, Progressive nodular histiocytosis, Pseudorheumatoid nodule, Reticulohistiocytoma, Reticulohistiocytosis, Sea-blue histiocytosis, Solitary reticulohistiocytoma, Solitary reticulohistiocytosis, Subcutaneous granuloma annulare, Systemic sarcoidosis, Cellular fibroadenoma, Cyst of Montgomery, Fibroadenoma, Fibrocystic breast changes, Fibrosclerosis of breast, Galactocele, and
Galactorrhea, Granulomatous mastitis.
IV, Kits and Devices
Kits
[00330] The invention provides a variety of kits for conveniently and/or effectively carrying out methods of the present invention. Typically kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.
[00331] In one embodiment, the present invention provides kits for regulate the expression of genes in vitro or in vivo, comprising saRNA of the present invention or a combination of saRNA of the present invention, saRNA modulating other genes, siRNAs, miRNAs or other oligonucleotide molecules.
[00332] The kit may fiirther comprise packaging and instructions and/or a delivery agent to form a formulation composition. The delivery agent may comprise a saline, a buffered solution, a lipidoid, a dendrimer or any delivery agent disclosed herein.
[00333] Non-limiting examples of genes are described herein in Table 1 .
[00334] In one embodiment, the kits comprising saRNA described herein may be used with proliferating cells to show efficacy.
[00335] In one non-limiting example, the buffer solution may include sodium chloride, calcium chloride, phosphate and/or EDTA. In another non-limiting example, the buffer solution may include, but is not limited to, saline, saline with 2mM calcium, 5% sucrose, 5% sucrose with 2mM calcium, 5% Mannitol, 5% Mannitol with 2mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2mM calcium and mannose (See U.S. Pub, No. 20120258046; herein incorporated by reference in its entirety). In yet another non-limiting example, the buffer solutions may be precipitated or it may be lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of saRNA in the buffer solution over a period of time and/or under a variety of conditions.
Devices
[00336] The present invention provides for devices which may incorporate saRNA of the present invention. These devices contain in a stable formulation available to be immediately delivered to a subject in need thereof, such as a human patient,
[00337] Non-limiting examples of the devices include a pump, a catheter, a needle, a transdermal patch, a pressurized olfactory delivery device, iontophoresis devices, multi-layered microfluidic devices. The devices may be employed to deliver saRNA of the present invention according to single, multi- or split-dosing regiments. The devices may be employed to deliver saRNA of the present invention across biological tissue, intradermal, subcutaneously, or intramuscularly. More examples of devices suitable for delivering oligonucleotides are disclosed in International Publication WO 2013/090648 filed December 14, 2012, the contents of which are incorporated herein by reference in their entirety.
Definitions
[00338] For convenience, the meaning of certain terms and phrases used in the specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, the definition in this section shall prevail.
[00339] About: As used herein, the term "about" means +/- 10% of the recited value.
[00340] Administered in combination: As used herein, the term "administered in combination" or "combined administration" means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. In some embodiments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently close together such that a combinatorial (e.g., a synergistic) effect is achieved.
[00341] Amino acid: As used herein, the terms "amino acid" and "amino acids" refer to all naturally occurring L-alpha-amino acids. The amino acids are identified by either the onc- letter or three-letter designations as follows: aspartic acid (Asp:D), isoleucine (Ile:I), threonine (ThnT), leucine (Leu:L), serine (Ser:S), tyrosine (Tyr:Y), glutamic acid (Glu:E), phenylalanine (Phe:F), proline (Pro:P), histidine (His:H), glycine (Gly:G), lysine (Lys:K), alanine (Ala: A), arginine (Arg:R), cysteine (Cys:C), tryptophan (Trp:W), valine (Val:V), glutamine (Gln:Q) methionine (Met:M), asparagines (Asn:N), where the amino acid is listed first followed parenthetically by the three and one letter codes, respectively,
[00342] Animal: As used herein, the term "animal" refers to any member of the animal kingdom. In some embodiments, "animal" refers to humans at any stage of development. In some embodiments, "animal" refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
[00343] Approximately: As used herein, the term "approximately" or "about," as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11 %, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%>, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
[00344] Associated with: As used herein, the terms "associated with," "conjugated,"
"linked," "attached," and "tethered," when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions. An "association" need not be strictly through direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity sufficiently stable such that the "associated" entities remain physically associated.
[00345] Bifunction or Bifunctional: As used herein, the terms "bifunction" and
"bifunctional" refers to any substance, molecule or moiety which is capable of or maintains at least two functions. The functions may affect the same outcome or a different outcome. The structure that produces the function may be the same or different. For example, bifunctional saRNA of the present invention may comprise a cytotoxic peptide (a first function) while those nucleosides which comprise the saRNA are, in and of themselves, cytotoxic (second function),
[00346] Biocompatible: As used herein, the term "biocompatible" means compatible with living cells, tissues, organs or systems posing little to no risk of injury, toxicity or rejection by the immune system.
[00347] Biodegradable: As used herein, the term "biodegradable" means capable of being broken down into innocuous products by the action of living things.
[00348] Biologically active: As used herein, the phrase "biologically active" refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active. In particular embodiments, the saRNA of the present invention may be considered biologically active if even a portion of the saRNA is biologically active or mimics an activity considered biologically relevant.
[00349] Cancer: As used herein, the term "cancer" in an individual refers to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Often, cancer cells will be in the form of a tumor, but such cells may exist alone within an individual, or may circulate in the blood stream as independent cells, such as leukemic cells.
[00350] Cell growth: As used herein, the term "cell growth" is principally associated with growth in cell numbers, which occurs by means of cell reproduction (i.e. proliferation) when the rate of the latter is greater than the rate of cell death (e.g. by apoptosis or necrosis), to produce an increase in the size of a population of cells, although a small component of that growth may in certain circumstances be due also to an increase in cell size or cytoplasmic volume of individual cells. An agent that inhibits cell growth can thus do so by either inhibiting proliferation or stimulating cell death, or both, such that the equilibrium between these two opposing processes is altered. [00351] Celt type: As used herein, the term "cell type" refers to a cell from a given source (e.g., a tissue, organ) or a cell in a given state of differentiation, or a cell associated with a given pathology or genetic makeup.
[00352] Chromosome; As used herein, the term "chromosome" refers to an organized structure of DNA and protein found in cells.
[00353] Complementary: As used herein, the term "complementary" as it relates to nucleic acids refers to hybridization or base pairing between nucleotides or nucleic acids, such as, for example, between the two strands of a double-stranded DNA molecule or between an oligonucleotide probe and a target are complementary.
[00354] Condition : As used herein, the term "condition" refers to the status of any cell, organ, organ system or organism. Conditions may refect a disease state or simply the physiologic presentation or situation of an entity. Conditions may be characterized as phenotypic conditions such as the macroscopic presentation of a disease or genotypic conditions such as the underlying gene or protein expression profiles associated with the condition. Conditions may be benign or malignant.
[00355] Controlled Release: As used herein, the term "controlled release" refers to a pharmaceutical composition or compound release profile that conforms to a particular pattern of release to effect a therapeutic outcome,
[00356] Cytostatic: As used herein, "cytostatic" refers to inhibiting, reducing, suppressing the growth, division, or multiplication of a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
[00357] Cytotoxic: As used herein, "cytotoxic" refers to killing or causing injurious, toxic, or deadly effect on a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.
[00358] Delivery: As used herein, "delivery" refers to the act or manner of delivering a compound, substance, entity, moiety, cargo or payload.
[00359] Delivery Agent: As used herein, "delivery agent" refers to any substance which facilitates, at least in part, the in vivo delivery of an saRNA of the present invention to targeted cells. [00360] Destabilized: As used herein, the term "destable," "destabilize," or "destabilizing region" means a region or molecule that is less stable than a starting, wild-type or native form of the same region or molecule.
[00361] Detectable label: As used herein, "detectable label" refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity that is readily detected by methods known in the art including radiography, fluorescence, chemiluminescence, enzymatic activity, absorb ancc and the like. Detectable labels include radioisotopes, fluorophores, chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin, streptavidin and haptens, quantum dots, and the like. Detectable labels may be located at any position in the oligonucleotides disclosed herein. They may be within the nucleotides or located at the 5' or 3' terminus.
[00362] Encapsulate: As used herein, the term "encapsulate" means to enclose, surround or encase.
[00363] Engineered: As used herein, embodiments of the invention are "engineered" when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule,
[00364] Equivalent subject: As used herein, "equivalent subject" may be e.g. a subject of similar age, sex and health such as liver health or cancer stage, or the same subject prior to treatment according to the invention. The equivalent subject is "untreated" in that he does not receive treatment with an saRNA according to the invention. However, he may receive a conventional anti-cancer treatment, provided that the subject who is treated with the saRNA of the invention receives the same or equivalent conventional anti-cancer treatment.
[00365] Exosome: As used herein, "exosome" is a vesicle secreted by mammalian cells.
[00366] Expression: As used herein, "expression" of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3 ' end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post-translational modification of a polypeptide or protein.
[00367] Feature: As used herein, a "feature" refers to a characteristic, a property, or a distinctive element. [00368] Formulation: As used herein, a "formulation" includes at least one saRNA of the present invention and a delivery agent.
[00369] Fragment: A "fragment," as used herein, refers to a portion. For example, fragments of proteins may comprise polypeptides obtained by digesting full-length protein isolated from cultured cells. Fragments of oligonucleotides may comprise nucleotides, or regions of nucleotides.
[00370] Functional: As used herein, a "functional" biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.
[00371] Gene: As used herein, the term "gene" refers to a nucleic acid sequence that comprises control and most often coding sequences necessary for producing a polypeptide or precursor. Genes, however, may not be translated and instead code for regulatory or structural RNA molecules.
[00372] A gene may be derived in whole or in part from any source known to the art, including a plant, a fungus, an animal, a bacterial genome or episome, eukaryotic, nuclear or plasmid DNA, cDNA, viral DNA, or chemically synthesized DNA. A gene may contain one or more modifications in either the coding or the untranslated regions that could affect the biological activity or the chemical structure of the expression product, the rate of expression, or the manner of expression control. Such modifications include, but are not limited to, mutations, insertions, deletions, and substitutions of one or more nucleotides. The gene may constitute an uninterrupted coding sequence or it may include one or more introns, bound by the appropriate splice junctions.
[00373] Gene expression: As used herein, the term "gene expression" refers to the process by which a nucleic acid sequence undergoes successful transcription and in most instances translation to produce a protein or peptide. For clarity, when reference is made to measurement of "gene expression", this should be understood to mean that measurements may be of the nucleic acid product of transcription, e.g., RNA or mRNA or of the amino acid product of translation, e.g., polypeptides or peptides. Methods of measuring the amount or levels of RNA, mRNA, polypeptides and peptides are well known in the art.
[00374] Genome: The term "genome" is intended to include the entire DNA complement of an organism, including the nuclear DNA component, chromosomal or extrachromosomal DNA, as well as the cytoplasmic domain (e.g., mitochondrial DNA). [00375] Homology: As used herein, the term "homology" refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some
embodiments, polymeric molecules are considered to be "homologous" to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar. The term "homologous" necessarily refers to a comparison between at least two sequences (polynucleotide or polypeptide sequences). In accordance with the invention, two polynucleotide sequences are considered to be homologous if the polypeptides they encode are at least about 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids. In some embodiments, homologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. For polynucleotide sequences less than 60 nucleotides in length, homology is determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. In accordance with the invention, two protein sequences are considered to be homologous if the proteins are at least about 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least about 20 amino acids.
[00376] The term "hyperproliferative cell" may refer to any cell that is proliferating at a rate that is abnormally high in comparison to the proliferating rate of an equivalent healthy cell (which may be referred to as a "control"). An "equivalent healthy" cell is the normal, healthy counterpart of a cell. Thus, it is a cell of the same type, e.g. from the same organ, which performs the same functions(s) as the comparator cell. For example, proliferation of a hyperproliferative hepatocyte should be assessed by reference to a healthy hepatocyte, whereas proliferation of a hyperproliferative prostate cell should be assessed by reference to a healthy prostate cell.
[00377] By an "abnormally high" rate of proliferation, it is meant that the rate of proliferation of the hyperproliferative cells is increased by at least 20, 30, 40%, or at least 45, 50, 55, 60, 65, 70, 75%, or at least 80%, as compared to the proliferative rate of equivalent, healthy (non-hyperproliferative) cells. The "abnormally high" rate of proliferation may also refer to a rate that is increased by a factor of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, or by a factor of at least 15, 20, 25, 30, 35, 40, 45, 50, or by a factor of at least 60, 70, 80, 90, 100, compared to the proliferative rate of equivalent, healthy cells. [00378] Hyperproliferative disorder: As used herein, a "hyperproliferative disorder" may be any disorder which involves hyperproliferative cells as defined above. Examples of hyperproliferative disorders include neoplastic disorders such as cancer, psoriatic arthritis, rheumatoid arthritis, gastric hyperproliferative disorders such as inflammatory bowel disease, skin disorders including psoriasis, Reiter's syndrome, pityriasis rubra pilaris, and
hyperproliferative variants of the disorders of keratinization.
[00379] The skilled person is fully aware of how to identify a hyperproliferative cell. The presence of hyperproliferative cells within an animal may be identifiable using scans such as X- rays, MRI or CT scans. The hyperproliferative cell may also be identified, or the proliferation of cells may be assayed, through the culturing of a sample in vitro using cell proliferation assays, such as MTT, XTT. MTS or WST-1 assays. Cell proliferation in vitro can also be determined using flow cytometry,
[00380] Identity: As used herein, the term "identity" refers to the overall relatedness between polymeric molecules, e.g., between oligonucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g. , gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes), hi certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%», at least 50%», at least 60%, at least 70%», at least 80%», at least 90%, at least 95%>, or 100%» of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M , ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W.. cd.. Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds,, Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is incorporated herein by reference. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CAB IDS, 1989, 4: 1 1 - 17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H , and Lipman, D., SIAM J Applied Math., 48:1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer programs. Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et at, Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altsehul, S. F. et al., J. Molec. Biol, 215, 403 (1990)).
[00381] Inhibit expression of a gene: As used herein, the phrase "inhibit expression of a gene" means to cause a reduction in the amount of an expression product of the gene. The expression product can be an RNA transcribed from the gene (e.g., an mRNA) or a polypeptide translated from an mRNA transcribed from the gene. Typically a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom. The level of expression may be determined using standard techniques for measuring mRNA or protein.
[00382] In vitro: As used herein, the term ";*« vitro'7 refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (eg., animal, plant, or microbe).
[00383] In vivo: As used herein, the term "in vivon refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof).
[00384] Isolated: As used herein, the term "isolated" refers to a substance or entity that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, isolated agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is "pure" if it is substantially free of other components. Substantially isolated: By "substantially isolated" is meant that the compound is substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
[00385] Label: The term "label" refers to a substance or a compound which is incorporated into an object so that the substance, compound or object may be detectable.
[00386] Linker: As used herein, a linker refers to a group of atoms, e.g., 10-1 ,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached to a modified nucleoside or nucleotide on the nucleobase or sugar moiety at a first end, and to a payload, e.g., a detectable or therapeutic agent, at a second end. The linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence. The linker can be used for any useful purpose, such as to form saRNA conjugates, as well as to administer a payload, as described herein.
[00387] Examples of chemical groups that can be incorporated into the linker include, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, thioether, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein. Examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or tetraethylene glycol), and dextran polymers and derivatives thereof. Other examples include, but are not limited to, cleavable moieties within the linker, such as, for example, a disulfide bond (-S-S-) or an azo bond (-N=N-), which can be cleaved using a reducing agent or photolysis. Non-limiting examples of a selectively cleavable bond include an aniido bond can be cleaved for example by the use of tris(2 -carboxy ethy l)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond can be cleaved for example by acidic or basic hydrolysis,
[00388] Metastasis: As used herein, the term "metastasis" means the process by which cancer spreads from the place at which it first arose as a primary tumor to distant locations in the body. Metastasis also refers to cancers resulting from the spread of the primary tumor. For example, someone with breast cancer may show metastases in their lymph system, liver, bones or lungs.
[00389] Modified: As used herein "modified" refers to a changed state or structure of a molecule of the invention. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the saRNAs of the present invention are modified by the introduction of non-natural nucleosides and/or nucleotides.
[00390] Naturally occurring: As used herein, "naturally occurring" means existing in nature without artificial aid.
[00391] Nucleic acid: The term "nucleic acid" as used herein, refers to a molecule comprised of one or more nucleotides, i.e., ribonucleotides, deoxyribonucleotides, or both. The term includes monomers and polymers of ribonucleotides and deoxyribonucleotides, with the ribonucleotides and/or deoxyribonucleotides being bound together, in the case of the polymers, via 5' to 3' linkages. The ribonucleotide and deoxyribonucleotide polymers may be single or double-stranded. However, linkages may include any of the linkages known in the art including, for example, nucleic acids comprising 5' to 3' linkages. The nucleotides may be naturally occurring or may be synthetically produced analogs that are capable of forming base-pair relationships with naturally occurring base pairs. Examples of non-naturally occurring bases that are capable of forming base-pairing relationships include, but are not limited to, aza and deaza pyrimidine analogs, aza and deaza purine analogs, and other heterocyclic base analogs, wherein one or more of the carbon and nitrogen atoms of the pyrimidine rings have been substituted by heteroatoms, e.g., oxygen, sulfur, selenium, phosphorus, and the like. [00392] Patient: As used herein, "patient" refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained professional for a particular disease or condition.
[00393] Peptide: As used herein, "peptide" is less than or equal to 50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
[00394] Pharmaceutically acceptable: The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[00395] Pharmaceutically acceptable excipients: The phrase "pharmaceutically acceptable excipient," as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
Excipients may include, for example; antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmeliose, crosslmked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose,
hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (com), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.
[00396] Pharmaceutically acceptable salts: The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. As used herein,
"pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsuifate, ethanesuifonate, fiimarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride,
hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3 -pheny lpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium,
tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington 's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth (eds.), Wiley- VCH, 2008, and Berge et al, Journal of Pharmaceutical Science, 66, 1 - 19 (1977), each of which is incorporated herein by reference in its entirety.
[00397] Pharmaceutically acceptable solvate: The term "pharmaceutically acceptable solvate," as used herein, means a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-. and tri-hydrates), N-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), Ν,Ν'- dimethylformamide (DMF), Ν,Ν'-dimethylacetamide (DMAC), l ,3-dimethyl-2-imidazolidmone (DMEU), l ,3-dimethyl-3,4,5,6-tetrahydro-2-(lH)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrol idone. benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a "hydrate."
[00398] Pharmacologic effect: As used herein, a "pharmacologic effect" is a measurable biologic phenomenon in an organism or system which occurs after the organism or system has been contacted with or exposed to an exogenous agent. Pharmacologic effects may result in therapeutically effective outcomes such as the treatment, improvement of one or more symptoms, diagnosis, prevention, and delay of onset of disease, disorder, condition or infection. Measurement of such biologic phenomena may be quantitative, qualitative or relative to another biologic phenomenon. Quantitative measurements may be statistically significant. Qualitative measurements may be by degree or kind and may be at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more different. They may be observable as present or absent, better or worse, greater or less. Exogenous agents, when referring to pharmacologic effects are those agents which are, in whole or in part, foreign to the organism or system. For example, modifications to a wild type biomolecule, whether structural or chemical, would produce an exogenous agent. Likewise, incorporation or combination of a wild type molecule into or with a compound, molecule or substance not found naturally in the organism or system would also produce an exogenous agent.
[00399] The saRNA of the present invention, comprises exogenous agents. Examples of pharmacologic effects include, but are not limited to, alteration in cell count such as an increase or decrease in neutrophils, reticulocytes, granulocytes, erythrocytes (red blood cells), megakaryocytes, platelets, monocytes, connective tissue macrophages, epidermal langerhans cells, osteoclasts, dendritic cells, microglial cells, neutrophils, eosinophils, basophils, mast cells, helper T cells, suppressor T cells, cytotoxic T cells, natural killer T cells, B cells, natural killer cells, or reticulocytes. Pharmacologic effects also include alterations in blood chemistry, pH, hemoglobin, hematocrit, changes in levels of enzymes such as, but not limited to, liver enzymes AST and ALT, changes in lipid profiles, electrolytes, metabolic markers, hormones or other marker or profile known to those of skill in the art,
[00400] Physicochemical: As used herein, "physicoehemieal" means of or relating to a physical and/or chemical property.
[00401] Preventing: As used herein, the term "preventing" refers to partially or completely delaying onset of an infection, disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.
[00402] Prodrug: The present disclosure also includes prodrugs of the compounds described herein. As used herein, "prodrugs" refer to any substance, molecule or entity which is in a form predicate for that substance, molecule or entity to act as a therapeutic upon chemical or physical alteration. Prodrugs may by covalently bonded or sequestered in some way and which release or are converted into the active drug moiety prior to, upon or after administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol, 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
[00403] Prognosing: As used herein, the term "prognosing" means a statement or claim that a particular biologic event will, or is very likely to, occur in the future.
[00404] Progression: As used herein, the term "progression" or "cancer progression" means the advancement or worsening of or toward a disease or condition. [00405] Proliferate: As used herein, the term "proliferate" means to grow, expand or increase or cause to grow, expand or increase rapidly, "Proliferative" means having the ability to proliferate. " Anti-pro liferative" means having properties counter to or inapposite to proliferative properties.
[00406] Protein: A "protein" means a polymer of amino acid residues linked together by peptide bonds. The term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function. Typically, however, a protein will be at least 50 amino acids long, in some instances the protem encoded is smaller than about 50 amino acids. In this case, the polypeptide is termed a peptide. If the protein is a short peptide, it will be at least about 10 amino acid residues long. A protein may be naturally occurring, recombinant, or synthetic, or any combination of these. A protein may also comprise a fragment of a naturally occurring protein or peptide, A protein may be a single molecule or may be a multi-molecular complex. The term protein may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analogue of a corresponding naturally occurring amino acid.
[00407] Protein expression: The term "protein expression" refers to the process by which a nucleic acid sequence undergoes translation such that detectable levels of the amino acid sequence or protein are expressed.
[00408] Purified: As used herein, "purify," "purified," "purification" means to make substantially pure or clear from unwanted components, material defilement, admixture or imperfection.
[00409] Regression: As used herein, the term "regression" or "degree of regression" refers to the reversal, either phenotypically or genotypicaily, of a cancer progression. Slowing or stopping cancer progression may be considered regression.
[00410] Sample: As used herein, the term "sample" or "biological sample" refers to a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen). A sample further may include a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs, A sample further refers to a medium, such as a nutrient broth or gel, which may contain cellular components, such as proteins or nucleic acid molecule.
[00411] Signal Sequences: As used herein, the phrase "signal sequences" refers to a sequence which can direct the transport or localization of a protein.
[00412] Single unit dose: As used herein, a "single unit dose" is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single
administration event.
[00413] Similarity: As used herein, the term "similarity" refers to the overall relatedness between polymeric molecules, e.g. between polynucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art.
[00414] Split dose: As used herein, a "split dose" is the division of single unit dose or total daily dose into two or more doses.
[00415] Stable: As used herein "stable" refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and in one embodiment, capable of formulation into an efficacious therapeutic agent.
[00416] Stabilized: As used herein, the term "stabilize", "stabilized," "stabilized region" means to make or become stable.
[00417] Subject: As used herein, the term "subject" or "patient" refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
[00418] Substantially: As used herein, the term "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially" is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena. [00419] Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2%.
[00420] Substantially simultaneously: As used herein and as it relates to plurality of doses, the term means within 2 seconds.
[00421] Suffering from: An individual who is "suffering from" a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
[00422] Susceptible to: An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition (for example, cancer) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic
polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
[00423] Sustained release: As used herein, the term "sustained release" refers to a pharmaceutical composition or compound release profile that conforms to a release rate over a specific period of time.
[00424] Synthetic: The term "synthetic" means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present invention may be chemical or enzymatic.
[00425] Targeted Cells: As used herein, "targeted cells" refers to any one or more cells of interest. The cells may be found in vitro, in vivo, in situ or in the tissue or organ of an organism. The organism may be an animal, in one embodiment, a mammal, or a human and most In one embodiment, a patient.
[00426] Therapeutic Agent: The term "therapeutic agent" refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
[00427] Therapeutically effective amount: As used herein, the term "therapeutically effective amount" means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when admimstered to a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.
[00428] Therapeutically effective outcome: As used herein, the term "therapeutically effective outcome" means an outcome that is sufficient in a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.
[00429] Total daily dose: As used herein, a "total daily dose" is an amount given or prescribed in 24 hour period. It may be administered as a single unit dose.
[00430] Transcription factor: As used herein, the term "transcription factor" refers to a
DNA-binding protein that regulates transcription of DNA into RNA, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.
[00431] Treating: As used herein, the term "treating" refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular infection, disease, disorder, and/or condition. For example, "treating" cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition,
[00432] The phrase "a method of treating" or its equivalent, when applied to, for example, cancer refers to a procedure or course of action that is designed to reduce, eliminate or prevent the number of cancer cells in an individual, or to alleviate the symptoms of a cancer. "A method of treating" cancer or another proliferative disorder does not necessarily mean that the cancer cells or other disorder will, in fact, be completely eliminated, that the number of cells or disorder will, in fact, be reduced, or that the symptoms of a cancer or other disorder will, in fact, be alleviated. Often, a method of treating cancer will be performed even with a low likelihood of success, but which, given the medical history and estimated survival expectancy of an individual, is nevertheless deemed an overall beneficial course of action.
[00433] Tumor growth: As used herein, the term "tumor growth" or "tumor metastases growth", unless otherwise indicated, is used as commonly used in oncology, where the term is principally associated with an increased mass or volume of the tumor or tumor metastases, primarily as a result of tumor cell growth.
[00434] Tumor Burden: As used herein, the term "tumor burden" refers to the total
Tumor Volume of all tumor nodules with a diameter in excess of 3mm carried by a subject.
[00435] Tumor Volume: As used herein, the term "tumor volume" refers to the size of a tumor. The tumor volume in mm3 is calculated by the formula: volume = (width)2 x length/2.
[00436] Unmodified: As used herein, "unmodified" refers to any substance, compound or molecule prior to being changed in any way. Unmodified may, but does not always, refer to the wild type or native form of a biomolecule. Molecules may undergo a series of modifications whereby each modified molecule may serve as the "unmodified" starting molecule for a subsequent modification.
Equivalents and Scope
[00437] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the invention described herein. The scope of the present invention is not intended to be limited to the above Description, but rather is as set forth in the appended claims. [00438] In the claims, articles such as "a," "an," and "the" may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.
[00439] it is also noted that the term "comprising" is intended to be open and permits the inclusion of additional elements or steps.
[00440] Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.
[00441] In addition, it is to be understood that any particular embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the compositions of the invention (e.g., any nucleic acid or protein encoded thereby; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.
[00442] All cited sources, for example, references, publications, databases, database entries, and art cited herein, are incorporated into this application by reference, even if not expressly stated in the citation. In case of conflicting statements of a cited source and the instant application, the statement in the instant application shall control.
[00443] The invention is further illustrated by the following non-limiting examples. EXAMPLES
Example 1. Upreeulation of a target gene expression through transfection with its saRNA in vitro
[00444] 2()nM. 50nM or ΙΟΟηΜ of saRNA of an saRNA for a target gene is transfected onto a monolayer of cells using lipofectamine 2000 (Life Technologies, US) following the manufacturer's instructions. The target genes may be any of those described herein such as in the target genes listed in Table 1. The saRNA may be single stranded or double-stranded. The cells may be HepG2 cells. This process is repeated three times before cells are harvested for isolation of total RNA for mRNA analysis.
[00445] After transfection, the isolation of total RNA is performed using the
RNAqueous-Miero kit (Ambion, UK) following the manufacturer's instructions. Briefly, the cells are gently eentrifuged followed by 3 pulses of sonication at Output 3 in lysis buffer (AMBION, UK). The cell lysates are then processed through an RNA binding column, followed by multiple washes and elution. The total RNA isolated is quantified by a NANODROP 2000 spectrophotometer. 500ng of total extracted RNA is processed for elimination of genomic DNA followed by reverse transcription using the QUANT1TECT® Reverse Transcription kit from QIAGEN.
[00446] The isolated RNA extracts are analyzed using quantitative reverse transcriptase
(qRT-PCR). Briefly, the extracts are reverse transcribed using First Strand cDNA synthesis kit (Qiagen). The cDNA is then amplified for quantitative analysis using QuantiFast® SYBR® Green PCT Kit from Qiagen. Amplification is performed using Applied Biosystems 7900HT FAST-Real-Time System. Amplified products are then analyzed using Applied Biosystems RQ Manager 1.2.1. 5 independent experiments are amplified in triplicates for quantitative analysis. Student T-Test scoring is performed at 99% confidence intervals.
[00447] mRNA production of the target gene is determined within the cells through the use of ELISA. Briefly, the cells are grown in phenol-red free RPMI media in the presence of charcoal stripped FCS. Following three sets of saRNA transfections at 8hrs, 16hrs and 24hrs, the culture media is collected for total ELISA.
[00448] An increase in mRNA levels only occurred in cells transfected with saRNAs, not in control cells transfected with scramble saRNA. Example 2. Upregulation of target gene expression through the transfection with saRNA in mouse in vivo
[00449] Ten male C57BI6/J, 8 week old mice are used for the experiment (control group
N=5). The target genes may be any of those described herein such as in the target genes listed in Table 1 .
[00450] saRNA of the target gene reconstituted with 100 μL ofRNase/Dnase free H20;
50 μL of complex A and 50 μL of complex B (InvivoFectamine, Invitrogen, CA, USA) are mixed, incubated at 50 °C for 30 minutes and are used for tail vein injections. Control animals are injected with equal volume of PBS while a positive control animal received siRNA against the target gene; a total of 5 control and 5 experimental animals are injected.
[00451] After administration, the total RNA is isolated. Frozen tissue sections from the mice are placed into scintillation vials containing Trizol and homogenised for 30 seconds. The tissue may he from the liver of the mice. The homogcnatc is then transferred in Falcon tubes for a further 2 minutes of homogenisation. Chloroform is then added to this and mixed by vortexing followed by a centrifugation step at 12,000 rpm for 15 minutes at 4°C. The aqueous upper phase is then transferred into a fresh microfuge tube where RNA is precipitated using 5 mg/ml of linear acrylamide (Ambion) and isopropanol overnight at -20 °C. The RNA is pelleted by
centrifugation at 12,000 rpm for 15 minutes at 4 °C and washed with ice cold 70% ethanol. The RNA is pelleted again at 7,500 rpm for 5 minutes at 4 °C. The supernatant is removed immediately and the RNA pellet allowed to air dry. The RNA is dissolved in nuclease free water for immediate analysis for RNA integrity using a Bionanalyser,
[00452] The isolated RNA is analyzed using qRT-PCR as described in Example 1.
mRNA production of the target gene is determined using an ELISA as described in Example 1. The administration of saRNA using a dendrimer delivery vehicle to a mouse leads to a significant increase in the mRNA encoded by the target gene within the blood circulation.
[00453] The effects of administration of saRNA on overall mice organ function may be studied. As a non-limiting example, the effects of administration of saRNA on overall liver function are determined according to the liver function markers gamma glutamyl transpeptidase, alanine aminotransferase, and aspartate aminotransferase or bilirubin.
[00454] saRNA also affects other the expression of other genes according to the mRNA levels of these genes. Tissue samples from the treated mice are used to measure the expression levels of various genes. The mRNA transcript levels of these genes are measured by RT-PCT mRNA expression level.
OTHER EMBODIMENTS
[00455] It is to be understood that while the present disclosure has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the present disclosure, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

Claims
1. A synthetic isolated saRNA which up-regulates expression of a target protein selected from the group consisting of any protein expressed by a target gene in Table 1.
2. The saRNA of claim 1, wherein the saRNA is single-stranded and is 14-30 nucleotides in length.
3. The saRNA of claim 2, wherein the saRNA is at least 80% identical to the reverse complement of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 1- 4047, 315236-318726, 584785-589061, 913310-917531, 1241080-1245401, and 1559932- 1564372.
4. The saRNA of claim 1 or claim 2, wherein the saRNA comprises a 3' overhang.
5. The saRNA of claim 2, wherein the saRNA comprises an antisense sequence selected from the group consisting of the antisense sequences described in the sequence listing referenced at the beginning of this application.
6. The saRNA of any of claims 2-5, wherein the saRNA comprises a 3' tail.
7. The saRNA of any of claims 2-6, wherein the saRNA comprises at least one
modification.
8. The saRNA of claim 1, wherein the saRNA is an saRNA duplex comprising an antisense strand and a sense strand wherein each strand is independently 14-30 nucleotides in length.
9. The saRNA of claim 8, wherein the antisense strand of the saRNA duplex is at least 80% identical to the reverse complement of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 1-4047 , 315236-318726, 584785-589061, 913310-917531, 1241080-1245401, and 1559932-1564372.
10. The saRNA of claim 8 or claim 9, wherein the antisense strand or the sense strand of the saRNA independently comprises a 3' overhang.
11. The saRNA of claim 8, wherein the saRNA comprises a sense sequence and an antisense sequence selected from the group of sequences consisting of the sense sequences and corresponding antisense sequences described in the sequence listing referenced at the beginning of this application.
12. The saRNA of any of claims 8-11, wherein the saRNA comprises a 3' tail.
13. The saRNA of any of claims 8-12, wherein the antisense strand and the sense strand independently comprise at least one modification.
14. A pharmaceutical composition comprising the saRNA of any of claims 1-13 and at least one pharmaceutically acceptable excipient.
15. A method of modulating the expression of a target protein, comprising administering the saRNA of any of claims 1-13.
16. The method of claim 15, wherein the expression of the target protein is increased.
17. The method of claim 16, wherein the expression of the target protein is increased by at least 30%.
18. The method of claim 17, wherein the expression of the target protein is increased by at least 50%.
19. A synthetic isolated saRNA which up-regulates expression of a target gene selected from the group consisting of any non-coding target gene in Table 2.
20. The saRNA of claim 19, wherein the saRNA is single-stranded and is 14-30 nucleotides in length.
21. The saRNA of claim 20, wherein the saRNA is at least 80% identical to the reverse complement of a nucleotide sequence selected from the group consisting of SEQ ID NOs:
1879189-1889207.
22. The saRNA of claim 19 or claim 20, wherein the saRNA comprises a 3' overhang.
23. The saRNA of claim 20, wherein the saRNA comprises an antisense sequence selected from the group consisting of the antisense sequences described in the sequence listing referenced at the beginning of this application.
24. The saRNA of any of claims 20-23, wherein the saRNA comprises a 3' tail.
25. The saRNA of any of claims 20-24, wherein the saRNA comprises at least one modification.
26. The saRNA of claim 19, wherein the saRNA is an saRNA duplex comprising an antisense strand and a sense strand wherein each strand is independently 14-30 nucleotides in length.
27. The saRNA of claim 26, wherein the antisense strand of the saRNA duplex is at least 80% identical to the reverse complement of a nucleotide sequence selected from the group consisting of SEQ ID NOs: 1879189-1889207.
28. The saRNA of claim 26 or claim 27, wherein the antisense strand or the sense strand of the saRNA independently comprises a 3' overhang.
29. The saRNA of claim 26, wherein the saRNA comprises a sense sequence and an antisense sequence selected from the group of sequences consisting of the sense sequences and corresponding antisense sequences described in the sequence listing referenced at the beginning of this application.
30. The saRNA of any of claims 26-29, wherein the saRNA comprises a 3' tail.
31. The saRNA of any of claims 26-30, wherein the antisense strand and the sense strand independently comprise at least one modification.
32. A pharmaceutical composition comprising the saRNA of any of claims 19-31 and at least one pharmaceutically acceptable excipient.
33. A method of modulating the expression of a target protein, comprising administering the saRNA of any of claims 19-31.
34. The method of claim 33, wherein the expression of the target protein is increased.
35. The method of claim 34, wherein the expression of the target protein is increased by at least 30%.
36. The method of claim 35, wherein the expression of the target protein is increased by at least 50%.
PCT/GB2016/051116 2015-04-22 2016-04-21 Sarna compositions and methods of use WO2016170348A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2017555233A JP2018512876A (en) 2015-04-22 2016-04-21 saRNA compositions and methods of use
US15/568,046 US20180305689A1 (en) 2015-04-22 2016-04-21 Sarna compositions and methods of use
EP16735927.2A EP3286318A2 (en) 2015-04-22 2016-04-21 Sarna compositions and methods of use
US17/319,744 US20210363525A1 (en) 2015-04-22 2021-05-13 Sarna compositions and methods of use

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
US201562150895P 2015-04-22 2015-04-22
US201562150904P 2015-04-22 2015-04-22
US201562150897P 2015-04-22 2015-04-22
US201562150892P 2015-04-22 2015-04-22
US201562150893P 2015-04-22 2015-04-22
US201562150908P 2015-04-22 2015-04-22
US201562150900P 2015-04-22 2015-04-22
US62/150,893 2015-04-22
US62/150,900 2015-04-22
US62/150,904 2015-04-22
US62/150,895 2015-04-22
US62/150,908 2015-04-22
US62/150,897 2015-04-22
US62/150,892 2015-04-22

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/568,046 A-371-Of-International US20180305689A1 (en) 2015-04-22 2016-04-21 Sarna compositions and methods of use
US17/319,744 Continuation US20210363525A1 (en) 2015-04-22 2021-05-13 Sarna compositions and methods of use

Publications (3)

Publication Number Publication Date
WO2016170348A2 true WO2016170348A2 (en) 2016-10-27
WO2016170348A3 WO2016170348A3 (en) 2017-04-13
WO2016170348A8 WO2016170348A8 (en) 2017-11-30

Family

ID=56363867

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2016/051116 WO2016170348A2 (en) 2015-04-22 2016-04-21 Sarna compositions and methods of use

Country Status (4)

Country Link
US (2) US20180305689A1 (en)
EP (1) EP3286318A2 (en)
JP (3) JP2018512876A (en)
WO (1) WO2016170348A2 (en)

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106834288A (en) * 2016-12-16 2017-06-13 南京医科大学 A kind of long non-coding RNA and its application in diagnosis/treatment stomach cancer
CN106929508A (en) * 2017-02-17 2017-07-07 张灏 The saRNA and its transport vehicle of a kind of activation PTPRO gene expressions
CN107937550A (en) * 2018-01-08 2018-04-20 北京泱深生物信息技术有限公司 A kind of and the relevant biomarker of breast cancer occurrence and development and its application
CN107988223A (en) * 2017-12-06 2018-05-04 张灏 A kind of saRNA of activation PTPRO gene expressions and its application in tumor stem cell treatment
CN108085389A (en) * 2017-12-29 2018-05-29 北京泱深生物信息技术有限公司 A kind of and the relevant lncRNA of breast cancer and its application
CN108130368A (en) * 2017-12-21 2018-06-08 中国医学科学院北京协和医院 Applications of the lncRNA in diagnosis or indication adolescent idiopathic scoliosis product is prepared
CN108424969A (en) * 2018-06-06 2018-08-21 深圳市颐康生物科技有限公司 A kind of biomarker, the method for diagnosing or estimating mortality risk
CN109097466A (en) * 2018-09-21 2018-12-28 王赞鑫 SNP site relevant to dissection of aorta disease and its application
CN109097465A (en) * 2018-09-19 2018-12-28 蒲佐 The application of the SNP site of CLIP3 gene
CN109097464A (en) * 2018-09-19 2018-12-28 蒲佐 The application of the SNP site of CFAP43 gene
CN109182503A (en) * 2018-09-28 2019-01-11 新乡医学院 Atherosclerosis molecular marker and its application
WO2019051312A1 (en) * 2017-09-07 2019-03-14 University Of Massachusetts Selenium cancer therapy
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019068099A1 (en) * 2017-09-29 2019-04-04 The Broad Institute, Inc. Methods and compositions for modulating suppression of lymphocyte activity
WO2019113472A1 (en) * 2017-12-08 2019-06-13 University Of Connecticut Compositions and methods for treating disorders of genomic imprinting
CN109996895A (en) * 2016-11-28 2019-07-09 卡斯滕·科思 The method and biomarker of in-vitro diagnosis phrenoblabia
CN110106180A (en) * 2019-05-29 2019-08-09 中国医科大学附属盛京医院 A kind of lncRNA molecule and its application in Treatment for Glioma/prognosis evaluation
CN110577988A (en) * 2019-07-19 2019-12-17 南方医科大学 Prediction model for fetal growth restriction
WO2020061591A1 (en) * 2018-09-21 2020-03-26 President And Fellows Of Harvard College Methods and compositions for treating diabetes, and methods for enriching mrna coding for secreted proteins
CN111020029A (en) * 2018-10-10 2020-04-17 上海康派尼恩医疗科技有限公司 Method, primer and kit for detecting PD-L1 gene expression level by real-time fluorescent quantitative PCR
WO2020086967A1 (en) * 2018-10-25 2020-04-30 Quadrant Biosciences Inc. Methods and machine learning for disease diagnosis
CN111118154A (en) * 2020-01-16 2020-05-08 华南协同创新研究院 Application of LINC01272 in preparation of tumor detection reagent and/or treatment drug
WO2020128163A1 (en) 2018-12-20 2020-06-25 Itä-Suomen Yliopisto Synthetic microrna mimics
CN111394351A (en) * 2020-03-18 2020-07-10 昆明医科大学 siRNA for inhibiting DICER1-AS1 expression and application thereof
CN111655869A (en) * 2017-11-30 2020-09-11 梅约医学教育与研究基金会 Detection of breast cancer
CN111718956A (en) * 2020-06-30 2020-09-29 山东农业大学 Preparation method and application of chicken-derived TRIM25 gene recombinant fluorescent expression plasmid
WO2021032777A1 (en) 2019-08-19 2021-02-25 Mina Therapeutics Limited Oligonucleotide conjugate compositions and methods of use
CN112575076A (en) * 2020-11-02 2021-03-30 深圳恒生医院 LncRNA marker for detecting and evaluating early spontaneous abortion and product using LncRNA marker
CN112661826A (en) * 2020-12-25 2021-04-16 中山大学 Small peptide ERpeptide and application thereof in acute myelogenous leukemia
US11058767B2 (en) 2018-02-21 2021-07-13 Bristol-Myers Squibb Company CAMK2D antisense oligonucleotides and uses thereof
CN113151471A (en) * 2021-04-28 2021-07-23 复旦大学附属中山医院 Gene composition, kit and method for detecting KRAS mutation of lung adenocarcinoma
US20210246506A1 (en) * 2018-08-04 2021-08-12 Imperial College Of Science, Technology And Medicine Method of identifying a subject having kawasaki disease
EP3778892A4 (en) * 2018-04-10 2022-01-19 Ractigen Therapeutics Novel small activating rna
CN114540500A (en) * 2022-03-21 2022-05-27 深圳市陆为生物技术有限公司 Product for evaluating overall survival of breast cancer patients
CN114574493A (en) * 2022-04-02 2022-06-03 中国科学院遗传与发育生物学研究所 sgRNA combination for editing sheep SOCS2 gene, primers for amplification and application
WO2022140264A1 (en) * 2020-12-23 2022-06-30 Eli Lilly And Company Rna therapeutics and methods of use thereof
US11427624B2 (en) * 2017-06-20 2022-08-30 Immunocore Limited T cell receptors
WO2022200810A1 (en) * 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022217283A1 (en) 2021-04-08 2022-10-13 Joslin Diabetes Center, Inc. Methods of diagnosing and predicting renal decline
US11549146B2 (en) 2016-05-20 2023-01-10 Cedars-Sinai Medical Center Diagnosis of inflammatory bowel disease based on genes
WO2022245734A3 (en) * 2021-05-17 2023-02-09 Corlieve Therapeutics Methods and compositions for treating epilepsy
US11597930B2 (en) 2020-04-02 2023-03-07 Mirecule, Inc. Targeted inhibition using engineered oligonucleotides
EP3894559A4 (en) * 2018-12-03 2023-04-05 Triplet Therapeutics, Inc. Methods for the treatment of trinucleotide repeat expansion disorders associated with mlh3 activity
WO2023111337A1 (en) * 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Antisense oligonucleotide
EP3919618A4 (en) * 2019-01-30 2023-11-08 Ractigen Therapeutics Oligonucleotide molecule and application thereof in tumor therapy
US11845937B2 (en) 2020-09-11 2023-12-19 Arrowhead Pharmaceuticals, Inc. RNAi agents for inhibiting expression of DUX4, compositions thereof, and methods of use
WO2024001172A1 (en) * 2022-06-27 2024-01-04 Ractigen Therapeutics Oligonucleotide modulators activating complement factor h expression

Families Citing this family (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010062960A2 (en) 2008-11-26 2010-06-03 Cedars-Sinai Medical Center METHODS OF DETERMINING RESPONSIVENESS TO ANTI-TNFα THERAPY IN INFLAMMATORY BOWEL DISEASE
CA2908553A1 (en) 2013-03-27 2014-10-02 Cedars-Sinai Medical Center Mitigation and reversal of fibrosis and inflammation by inhibition of tl1a function and related signaling pathways
WO2015010108A1 (en) 2013-07-19 2015-01-22 Cedars-Sinai Medical Center Signature of tl1a (tnfsf15) signaling pathway
WO2017112921A1 (en) * 2015-12-24 2017-06-29 The Arizona Board Of Regents On Behalf Of The University Of Arizona Systems and methods for characterizing sepsis
EP3411066A1 (en) * 2016-02-01 2018-12-12 Eli Lilly and Company Parathyroid hormone anti-rankl antibody fusion compounds
KR20180127416A (en) 2016-03-17 2018-11-28 세다르스-신나이 메디칼 센터 Diagnosis of Inflammatory Bowel Disease through RNASET2
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
GB201611738D0 (en) 2016-07-05 2016-08-17 Cambridge Entpr Ltd Biomarkers for inflammatory bowel disease
UY37376A (en) * 2016-08-26 2018-03-23 Amgen Inc ARNI CONSTRUCTIONS TO INHIBIT EXPRESSION OF ASGR1 AND METHODS FOR USE
CN110381980A (en) 2017-01-06 2019-10-25 艾维迪提生物科学有限责任公司 The method that nucleic acid-peptide composition and inducing exon are skipped
US11306311B2 (en) * 2017-06-08 2022-04-19 Chandrasekhar Kanduri Long non-coding RNA in cancer
US11168322B2 (en) 2017-06-30 2021-11-09 Arbor Biotechnologies, Inc. CRISPR RNA targeting enzymes and systems and uses thereof
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
MX2020005860A (en) 2017-12-06 2020-09-09 Avidity Biosciences Inc Compositions and methods of treating muscle atrophy and myotonic dystrophy.
US20210361693A1 (en) * 2018-02-09 2021-11-25 Empirico Inc. Process to inhibit or eliminate eosinophilic diseases of the airway and related conditions
SG11202007644PA (en) 2018-02-14 2020-09-29 Deep Genomics Incorporated Oligonucleotide therapy for wilson disease
US20210130824A1 (en) * 2018-02-16 2021-05-06 Crispr Therapeutics Ag Compositions and methods for gene editing by targeting fibrinogen-alpha
CN108192973B (en) * 2018-03-02 2021-06-04 中南大学湘雅医院 Application of long-chain non-coding RNA LINC00559 as biomarker in preparation of lung adenocarcinoma prognosis detection preparation
CN108192974B (en) * 2018-03-02 2021-11-05 中南大学湘雅医院 Application of long-chain non-coding RNA LINC00842 as biomarker in preparation of lung adenocarcinoma prognosis detection preparation
CN108192975B (en) * 2018-03-02 2021-06-04 中南大学湘雅医院 Application of long-chain non-coding RNA LINC00559 as biomarker in preparation of lung squamous carcinoma prognosis detection preparation
CN108192976B (en) * 2018-03-02 2021-06-04 中南大学湘雅医院 Application of long-chain non-coding RNA LINC00336 as biomarker in preparation of lung squamous carcinoma prognosis detection preparation
US11566246B2 (en) * 2018-04-12 2023-01-31 Mina Therapeutics Limited SIRT1-saRNA compositions and methods of use
CN110468200B (en) * 2018-05-10 2023-02-28 复旦大学附属肿瘤医院 Multi-gene detection kit for cancer prognosis
US20210198669A1 (en) * 2018-06-01 2021-07-01 Children's National Medical Center Compositions and methods for treating and preventing muscular disorders and dystrophies, steroid side effects, and inflammation
CN108753967B (en) * 2018-06-08 2022-06-17 复旦大学附属中山医院 Gene set for liver cancer detection and panel detection design method thereof
CN109207585A (en) * 2018-11-01 2019-01-15 中国人民解放军第Oo医院 Immune thrombocytopenia lncRNA marker and kit and application
AU2019377855A1 (en) 2018-11-08 2021-06-03 Greenlight Biosciences, Inc. Control of insect infestation
CN109207598A (en) * 2018-11-09 2019-01-15 汕头大学医学院附属肿瘤医院 A kind of long-chain non-coding RNA is preparing the application in esophagus cancer diagnosis reagent or therapeutic agent
EP3881233A4 (en) * 2018-11-15 2022-11-23 Ampel Biosolutions, LLC Machine learning disease prediction and treatment prioritization
CN109355391A (en) * 2018-12-05 2019-02-19 山东省千佛山医院 Application of the LINC01876 as the molecular marker of diagnosing liver cancer
CN110093348B (en) * 2018-12-05 2022-11-29 西北工业大学 shRNA for enhancing mouse NPFFR2 gene expression
IL297818B1 (en) 2018-12-21 2024-05-01 Avidity Biosciences Inc Anti-transferrin receptor antibodies and uses thereof
JP2022515881A (en) * 2018-12-29 2022-02-22 ラクティゲン セラピューティクス Oligonucleic acid molecules and their applications
CN109813912B (en) * 2019-01-04 2021-12-28 深圳大学 Application of group of serum differential protein combinations in preparation of reagent for detecting autism
CN109837336B (en) * 2019-01-09 2022-07-19 中国人民解放军军事科学院军事医学研究院 Radiation sensitive gene marker and application thereof in identification of high LET radiation
CN109652532A (en) * 2019-01-11 2019-04-19 中国人民解放军总医院 A kind of marker detecting drug for cardiovascular disease
JP7241185B2 (en) * 2019-01-17 2023-03-16 エフ. ホフマン-ラ ロシュ アーゲー Protein target of E3 ubiquitin ligase (UBE3A)
WO2020150606A1 (en) * 2019-01-18 2020-07-23 The General Hospital Corporation Methods and compositions for modulating immune dysregulation
KR20210131322A (en) * 2019-01-24 2021-11-02 락티젠 세러퓨틱스 Oligomeric Nucleic Acids for Skin Protection
US20220290122A1 (en) * 2019-02-19 2022-09-15 Whitehead Institute For Biomedical Research Methods and agents for modulating mitochondrial nad levels
US20220146531A1 (en) * 2019-03-07 2022-05-12 Reti Mark Co., Ltd. A combination of biomarkers for diagnosing of diabetic retinopathy and use thereof
CA3162618A1 (en) 2019-03-20 2020-09-24 Peter Jungsoo PARK Antisense oligonucleotide-based progranulin augmentation therapy in neurodegenerative diseases
AU2020247700A1 (en) * 2019-03-25 2021-10-14 The Board Of Trustees Of The Leland Stanford Junior University Signature for diagnosis of bacterial VS viral infections
KR20200113556A (en) * 2019-03-25 2020-10-07 황인후 Composition for preventing or treating diabetes and method for screening antidiabetic agents using ST8SIA1
CN109880828B (en) * 2019-03-26 2021-02-05 河南师范大学 siRNA for interfering Mroh7 gene expression and application, interference method and medicament thereof
WO2020214820A2 (en) * 2019-04-18 2020-10-22 University Of Massachusetts Aim2 inhibitors and uses thereof
CN111849968A (en) * 2019-04-30 2020-10-30 中美瑞康核酸技术(南通)研究院有限公司 Oligonucleotide molecule and application thereof in acute intermittent porphyria treatment
CN114127300A (en) * 2019-05-09 2022-03-01 费城儿童医院 Compositions and methods for treating leukodystrophy and whole animal and cell models for identifying effective drugs for treating leukodystrophy
CN110079601B (en) * 2019-05-23 2022-04-19 青岛市中心医院 Diagnosis and treatment marker for radioactivity related diseases and application thereof
CN110117659B (en) * 2019-06-18 2022-10-11 上海奕谱生物科技有限公司 Novel tumor marker STAMP-EP10 and application thereof
CN110184358A (en) * 2019-06-25 2019-08-30 台州市立医院 The OIT3 gene of thyroid cancer early diagnosis and its application
WO2020264177A1 (en) * 2019-06-26 2020-12-30 Fred Hutchinson Cancer Research Center Methods and compositions comprising brd9 activating therapies for treating cancers and related disorders
CN110456060B (en) * 2019-07-31 2022-09-09 四川大学华西医院 Application of EFCAB13 autoantibody detection reagent in preparation of lung cancer screening kit
CN110541027A (en) * 2019-08-21 2019-12-06 昆明医科大学第一附属医院 Application of lncRNA HIF1A-AS1 in resisting deep vein thrombosis
CN112444627B (en) * 2019-09-05 2022-03-29 四川大学华西医院 High-risk chest pain screening kit
EP4055175A4 (en) * 2019-11-04 2023-11-29 Decibel Therapeutics, Inc. Cochlear inner hair cell promoters and uses thereof
CN110876752B (en) * 2019-11-18 2021-04-30 暨南大学 Application of long-chain non-coding RNA NRON functional motif in preparation of medicines for inhibiting bone resorption and preventing and treating osteoporosis
US20220412993A1 (en) * 2019-12-05 2022-12-29 The General Hospital Corporation Multi-Biomarker Prediction Models for Multiple Infection Episodes Following Blunt Trauma
CN111197073A (en) * 2019-12-19 2020-05-26 武汉艾米森生命科技有限公司 Method for extracting DNA sample from excrement and methylation detection method of colorectal cancer related gene
CN112795579B (en) * 2019-12-25 2022-03-18 四川省人民医院 Keshan disease gene screening kit
CN113092774B (en) * 2020-01-09 2022-08-19 中国科学院生物物理研究所 Use of mtEF4 protein as biomarker for hair growth and hair loss
CN113134010B (en) * 2020-01-20 2023-09-01 上海市生物医药技术研究院 Micro RNA of targeted estrogen receptor alpha and anti-tumor application thereof
US20230047844A1 (en) * 2020-01-24 2023-02-16 Arizona Board Of Regents On Behalf Of The University Of Arizona Compounds and methods for treating or reducing pruritus
CN111214482B (en) * 2020-02-21 2022-11-11 东莞市第八人民医院(东莞市儿童医院) Application of linc00467 gene-targeted siRNA in drug resistance of leukemia
CN111254193A (en) * 2020-02-27 2020-06-09 上海交通大学 Application of DNA methylation marker PCYT1A in preparation of kit for diagnosing PCOS
CN111440867B (en) * 2020-03-06 2020-12-22 河北医科大学第二医院 Application of biomarker in diagnosis and treatment of liver cancer
AU2021237465A1 (en) 2020-03-19 2022-10-13 Avidity Biosciences, Inc. Compositions and methods of treating Facioscapulohumeral muscular dystrophy
US11446387B2 (en) 2020-03-27 2022-09-20 Avidity Biosciences, Inc. Compositions and methods of treating muscle dystrophy
CN111424091B (en) * 2020-04-20 2022-02-08 中国医学科学院北京协和医院 Marker for differential diagnosis of benign and malignant thyroid follicular tumor and application thereof
CN111458510B (en) * 2020-04-30 2020-10-30 郑州大学第一附属医院 Early esophageal cancer and high risk group screening marker and related joint inspection card
US20230416822A1 (en) * 2020-05-19 2023-12-28 Falcon Bioscience, Llc Detection and treatment of conditions characterized by perfusion shortage
CN111562394B (en) * 2020-06-02 2021-07-02 西安交通大学医学院第一附属医院 Application of heat shock factor 2binding protein in liver ischemia reperfusion injury and drug-induced liver injury
CN111593127A (en) * 2020-06-30 2020-08-28 北京泱深生物信息技术有限公司 Non-coding RNA closely related to laryngeal squamous cell carcinoma
CN111910000B (en) * 2020-07-02 2022-07-01 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Tumor microenvironment component marker combination and system for predicting nasopharyngeal carcinoma prognosis
CN111778562A (en) * 2020-07-08 2020-10-16 复旦大学附属华山医院 DNA library for obtaining information related to mutation of pathogenic gene of cranial carotid interlayer and application
CN112063707B (en) * 2020-07-09 2022-04-22 浙江大学 Use of long non-coding RNA LOC102724238 in diagnosis and treatment of primary pterygium
CN113930423B (en) * 2020-07-14 2024-05-10 北京大学 SaRNA for protecting myocardial cells from stress injury and application thereof
WO2022015998A1 (en) * 2020-07-15 2022-01-20 University Of Connecticut Gene panels and methods of use thereof for screening and diagnosis of congenital heart defects and diseases
CN111733249B (en) * 2020-07-21 2020-12-11 北京泱深生物信息技术有限公司 Molecular marker related to occurrence and development of laryngeal carcinoma
WO2022035984A1 (en) * 2020-08-12 2022-02-17 Skyhawk Therapeutics, Inc. Antisense oligonucleotides for treatment of conditions and diseases related to trem2
CN112094896B (en) * 2020-08-27 2024-02-09 上海市公共卫生临床中心 Active tuberculosis diagnosis marker, kit and application thereof
CN112143813B (en) * 2020-10-28 2021-06-29 镇江市第一人民医院 PRR34-AS1 AS novel molecular marker and quantitative detection method and application thereof
CN112251513B (en) * 2020-11-26 2023-02-17 中国人民解放军总医院第一医学中心 Esophageal cancer and/or gastric cancer molecular marker Lnc5q31.1, primer, kit and application
CN112553199A (en) * 2020-11-30 2021-03-26 深圳市人民医院 Construction method and application of snhg17-KO gene knockout mouse model
CN112553333B (en) * 2020-12-08 2022-03-08 南方医科大学深圳医院 Application of miR-1207 and target gene thereof in detection of laryngeal squamous cell carcinoma
CN112641797B (en) * 2020-12-30 2021-12-17 温州医科大学 Target and diagnostic marker for inhibiting colorectal cancer growth and metastasis and application thereof
EP4277999A1 (en) * 2021-01-14 2023-11-22 Loma Linda University Methods for evaluation of early stage oral squamous cell carcinoma
CN114958844A (en) * 2021-01-27 2022-08-30 青岛市妇女儿童医院 Application of gene regulator in inhibiting oral cancer
CN112941076B (en) * 2021-02-05 2023-07-07 中国药科大学 FXR-targeted saRNA and application thereof
CN112961914B (en) * 2021-02-07 2022-08-30 安徽省立医院(中国科学技术大学附属第一医院) Application of miR-3074-5p as rheumatoid arthritis marker and kit thereof
CN112725471B (en) * 2021-03-12 2022-09-16 中国大熊猫保护研究中心 Molecular marker evaluation method for wild panda training immune adaptation
WO2022212208A1 (en) * 2021-03-29 2022-10-06 University Of Massachusetts Oligonucleotides for syngr-3 modulation
WO2022215066A1 (en) * 2021-04-05 2022-10-13 Itayandbiond Ltd Small activating rna increasing shank expression and method of treating intellectual disabilities and associated comorbidities associated with shank haploinsufficiency
WO2022214635A1 (en) * 2021-04-08 2022-10-13 Stichting Vu Nucleic acid molecules for compensation of stxbp1 haploinsufficiency and their use in the treatment of stxbp1-related disorders
WO2022226381A1 (en) * 2021-04-22 2022-10-27 H. Lee Moffitt Cancer Center And Research Institute, Inc. Diagnostic to support clinical trial matching and exploratory biomarker analyses in cancer patients
CN112921098B (en) * 2021-04-27 2023-05-05 深圳市人民医院 Marker for detecting triple negative breast cancer, detection reagent and application thereof
CN113234834A (en) * 2021-07-09 2021-08-10 北京泱深生物信息技术有限公司 Group of genes for pancreatic cancer prognosis and application thereof
CN113430273B (en) * 2021-08-17 2022-05-27 广州齐凯生物科技有限公司 Application of long-chain non-coding RNA LINC01565 in acute myelogenous leukemia prognosis
CA3230433A1 (en) * 2021-09-16 2023-03-23 Barbara MURPHY Methods for treating and diagnosing risk of renal allograft fibrosis and rejection
AU2022345098A1 (en) 2021-09-16 2024-04-04 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy
WO2023064604A1 (en) * 2021-10-14 2023-04-20 New York Stem Cell Foundation, Inc. Modified cells and methods of use thereof
KR20230059641A (en) * 2021-10-26 2023-05-03 사회복지법인 삼성생명공익재단 Composition for treatment and metastasis inhibition of colorectal cancer and use thereof
WO2023077016A1 (en) * 2021-10-28 2023-05-04 Baylor College Of Medicine Targeting neuronal sirpα for treatment and prevention of neurological disorders
CN114134147B (en) * 2021-11-16 2023-11-14 山东大学 Non-coding RNA for regulating FZD9 and application thereof
CN114214322A (en) * 2021-11-23 2022-03-22 中国辐射防护研究院 LincR-0003 and application thereof as molecular marker for early diagnosis of radiation damage
WO2023117965A1 (en) * 2021-12-20 2023-06-29 Freie Universität Berlin Methods and agents for increasing rbm3 expression
CN114196675A (en) * 2021-12-24 2022-03-18 复旦大学附属肿瘤医院 Antisense oligonucleotide targeting LINC00624 and application thereof in breast cancer treatment
CN114288315B (en) * 2022-01-13 2022-08-16 暨南大学附属第一医院(广州华侨医院) Pharmaceutical composition containing CCP110 inhibitor and application thereof
CN116486918A (en) * 2022-01-14 2023-07-25 天士力干细胞产业平台有限公司 Stem cell quality evaluation method
CN115247176B (en) * 2022-01-17 2023-06-20 郑州大学第一附属医院 Long-chain non-coding RNA and application thereof
WO2023178386A1 (en) * 2022-03-23 2023-09-28 PYC Therapeutics Limited Methods of treating glaucoma
CN114774445B (en) * 2022-03-29 2024-01-02 中国人民解放军西部战区总医院 Application of DYNLT5 gene in atherosclerosis and tissue repair
US20230313191A1 (en) * 2022-03-29 2023-10-05 Direct Biologics Llc Rna comprising secretomes and methods of their use
WO2023191957A1 (en) * 2022-03-30 2023-10-05 Mirimus, Inc. Compositions and methods of generating novel amirna
WO2023194586A2 (en) * 2022-04-08 2023-10-12 Secarna Pharmaceuticals Gmbh & Co. Kg Oligonucleotide for reducing the expression of leucine-rich repeat kinase 2 (lrrk2) and its use for preventing and/or treating human diseases
WO2023201276A2 (en) * 2022-04-12 2023-10-19 Orthobio Therapeutics, Inc. Lipid nanoparticles for gene editing systems
WO2023233437A1 (en) * 2022-06-01 2023-12-07 Universita' Degli Studi Di Trento Non-coding rna sequences capable of increasing the expression of chd8 and chd2 proteins
CN114959041B (en) * 2022-06-19 2023-08-22 瓯江实验室 Novel target and diagnostic marker for inhibiting colorectal cancer proliferation metastasis and application thereof
WO2024005158A1 (en) * 2022-06-30 2024-01-04 武田薬品工業株式会社 Oligonucleotide
WO2024006994A2 (en) * 2022-07-01 2024-01-04 The University Of Chicago Compositions and methods for the treatment of cancer by targeting the brca1 pseudogene 1 (brca1p1)
TW202403054A (en) * 2022-07-04 2024-01-16 大陸商江蘇鵾遠生物科技股份有限公司 Cancer-specific methylation markers and their use
CN115725587B (en) * 2022-09-14 2023-07-18 中国人民解放军军事科学院军事医学研究院 Oligonucleotide aptamer group for specifically recognizing radiation-sensitive protein TP53I3, kit and detection method
WO2024074668A1 (en) * 2022-10-06 2024-04-11 Stichting Radboud Universitair Medisch Centrum Antisense oligonucleotides for treatment of usher 2a. exons 30-31
WO2024074670A1 (en) * 2022-10-06 2024-04-11 Stichting Radboud Universitair Medisch Centrum Antisense oligonucleotides for treatment of usher 2a. exon 68
CN115948561B (en) * 2023-02-10 2024-01-02 武汉艾米森生命科技有限公司 Reagent and detection kit for esophageal squamous carcinoma diagnosis or auxiliary diagnosis and application thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7709566B2 (en) 2005-10-20 2010-05-04 Asahi Glass Company, Limited Polytetrafluoroethylene aqueous dispersion and its product
US20100210707A1 (en) 2005-04-15 2010-08-19 Longcheng Li Small Activating RNA Molecules and Methods of Use
WO2012065143A1 (en) 2010-11-12 2012-05-18 The General Hospital Corporation Polycomb-associated non-coding rnas
US8288354B2 (en) 2005-12-28 2012-10-16 The Scripps Research Institute Natural antisense and non-coding RNA transcripts as drug targets
US8324181B2 (en) 2005-11-17 2012-12-04 Board Of Regents, The University Of Texas System Modulation of gene expression by oligomers targeted to chromosomal DNA
US20130164846A1 (en) 2010-06-23 2013-06-27 Mina Therapeutics Limited Rna molecules and uses thereof

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1255550A (en) * 1998-11-30 2000-06-07 张良平 Clone of antisense gene resisting heteroplastic transplantation and in-situ synthetase
US7250496B2 (en) * 2002-11-14 2007-07-31 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory genes and uses thereof
PT2284266E (en) * 2002-11-14 2013-12-17 Thermo Fisher Scient Biosciences Inc Sirna targeting tp53
WO2004070062A2 (en) * 2003-02-04 2004-08-19 Wyeth Compositions and methods for diagnosing and treating cancers
US8178503B2 (en) * 2006-03-03 2012-05-15 International Business Machines Corporation Ribonucleic acid interference molecules and binding sites derived by analyzing intergenic and intronic regions of genomes
US7901882B2 (en) * 2006-03-31 2011-03-08 Affymetrix, Inc. Analysis of methylation using nucleic acid arrays
RU2619185C2 (en) * 2009-12-23 2017-05-12 Курна, Инк. Treatment of diseases associated with uncoupling proteins 2 (ucp2), by inhibiting of natural antisense transcript to ucp2
US8916534B2 (en) * 2010-10-08 2014-12-23 Mina Therapeutics Limited Methods of inducing insulin production
US9164590B2 (en) * 2010-12-24 2015-10-20 Kevadiya, Inc. System and method for automated capture and compaction of instructional performances
WO2014179765A2 (en) * 2013-05-02 2014-11-06 Thomas Jefferson University Novel human mirnas for use in diagnosis, prognosis and therapy of human conditions and diseases
WO2023099884A1 (en) * 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100210707A1 (en) 2005-04-15 2010-08-19 Longcheng Li Small Activating RNA Molecules and Methods of Use
US7709566B2 (en) 2005-10-20 2010-05-04 Asahi Glass Company, Limited Polytetrafluoroethylene aqueous dispersion and its product
US8324181B2 (en) 2005-11-17 2012-12-04 Board Of Regents, The University Of Texas System Modulation of gene expression by oligomers targeted to chromosomal DNA
US8288354B2 (en) 2005-12-28 2012-10-16 The Scripps Research Institute Natural antisense and non-coding RNA transcripts as drug targets
US20130164846A1 (en) 2010-06-23 2013-06-27 Mina Therapeutics Limited Rna molecules and uses thereof
WO2012065143A1 (en) 2010-11-12 2012-05-18 The General Hospital Corporation Polycomb-associated non-coding rnas

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JANOWSKI ET AL., NATURE CHEMICAL BIOLOGY, vol. 3, 2007, pages 166 - 173
VOUTILA ET AL., MOL THER NUCLEIC ACIDS, vol. 1, 2012, pages E35
WU ET AL., PLOS ONE, vol. 6, no. 12, 2011, pages E28580

Cited By (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11549146B2 (en) 2016-05-20 2023-01-10 Cedars-Sinai Medical Center Diagnosis of inflammatory bowel disease based on genes
CN109996895A (en) * 2016-11-28 2019-07-09 卡斯滕·科思 The method and biomarker of in-vitro diagnosis phrenoblabia
CN106834288A (en) * 2016-12-16 2017-06-13 南京医科大学 A kind of long non-coding RNA and its application in diagnosis/treatment stomach cancer
CN106834288B (en) * 2016-12-16 2020-08-21 南京医科大学 Long non-coding RNA and application thereof in diagnosis/treatment of gastric cancer
CN106929508A (en) * 2017-02-17 2017-07-07 张灏 The saRNA and its transport vehicle of a kind of activation PTPRO gene expressions
US11427624B2 (en) * 2017-06-20 2022-08-30 Immunocore Limited T cell receptors
US11718657B2 (en) 2017-06-20 2023-08-08 Immunocore Limited T cell receptors
WO2019051312A1 (en) * 2017-09-07 2019-03-14 University Of Massachusetts Selenium cancer therapy
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
EP4219715A2 (en) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019068099A1 (en) * 2017-09-29 2019-04-04 The Broad Institute, Inc. Methods and compositions for modulating suppression of lymphocyte activity
CN111655869A (en) * 2017-11-30 2020-09-11 梅约医学教育与研究基金会 Detection of breast cancer
CN107988223B (en) * 2017-12-06 2021-08-13 张灏 SaRNA for activating PTPRO gene expression and application thereof in tumor stem cell treatment
CN107988223A (en) * 2017-12-06 2018-05-04 张灏 A kind of saRNA of activation PTPRO gene expressions and its application in tumor stem cell treatment
WO2019113472A1 (en) * 2017-12-08 2019-06-13 University Of Connecticut Compositions and methods for treating disorders of genomic imprinting
JP2021505200A (en) * 2017-12-08 2021-02-18 ユニヴァーシティ オブ コネチカット Compositions and Methods for Treating Genomic Imprinting Disorders
CN108130368A (en) * 2017-12-21 2018-06-08 中国医学科学院北京协和医院 Applications of the lncRNA in diagnosis or indication adolescent idiopathic scoliosis product is prepared
CN108130368B (en) * 2017-12-21 2021-12-14 中国医学科学院北京协和医院 Application of lncRNA in preparation of product for diagnosing or predicting adolescent idiopathic scoliosis
CN108085389B (en) * 2017-12-29 2020-06-09 青岛泱深生物医药有限公司 LncRNA related to breast cancer and application thereof
CN108085389A (en) * 2017-12-29 2018-05-29 北京泱深生物信息技术有限公司 A kind of and the relevant lncRNA of breast cancer and its application
CN107937550A (en) * 2018-01-08 2018-04-20 北京泱深生物信息技术有限公司 A kind of and the relevant biomarker of breast cancer occurrence and development and its application
CN107937550B (en) * 2018-01-08 2020-06-02 青岛泱深生物医药有限公司 Biomarker related to breast cancer occurrence and development and application thereof
US11058767B2 (en) 2018-02-21 2021-07-13 Bristol-Myers Squibb Company CAMK2D antisense oligonucleotides and uses thereof
EP3778892A4 (en) * 2018-04-10 2022-01-19 Ractigen Therapeutics Novel small activating rna
CN108424969A (en) * 2018-06-06 2018-08-21 深圳市颐康生物科技有限公司 A kind of biomarker, the method for diagnosing or estimating mortality risk
US20210246506A1 (en) * 2018-08-04 2021-08-12 Imperial College Of Science, Technology And Medicine Method of identifying a subject having kawasaki disease
CN109097464A (en) * 2018-09-19 2018-12-28 蒲佐 The application of the SNP site of CFAP43 gene
CN109097465A (en) * 2018-09-19 2018-12-28 蒲佐 The application of the SNP site of CLIP3 gene
CN109097464B (en) * 2018-09-19 2021-12-14 中国医学科学院阜外医院深圳医院(深圳市孙逸仙心血管医院) Application of SNP (single nucleotide polymorphism) site of CFAP43 gene
CN113056289A (en) * 2018-09-21 2021-06-29 哈佛学院校长同事会 Methods and compositions for treating diabetes and methods for enriching mRNA encoding secreted proteins
CN109097466A (en) * 2018-09-21 2018-12-28 王赞鑫 SNP site relevant to dissection of aorta disease and its application
WO2020061591A1 (en) * 2018-09-21 2020-03-26 President And Fellows Of Harvard College Methods and compositions for treating diabetes, and methods for enriching mrna coding for secreted proteins
CN109182503A (en) * 2018-09-28 2019-01-11 新乡医学院 Atherosclerosis molecular marker and its application
CN111020029A (en) * 2018-10-10 2020-04-17 上海康派尼恩医疗科技有限公司 Method, primer and kit for detecting PD-L1 gene expression level by real-time fluorescent quantitative PCR
CN111020029B (en) * 2018-10-10 2023-09-29 江苏普瑞悉恩生物科技有限公司 Method, primer and kit for detecting PD-L1 gene expression quantity by real-time fluorescence quantitative PCR
WO2020086967A1 (en) * 2018-10-25 2020-04-30 Quadrant Biosciences Inc. Methods and machine learning for disease diagnosis
EP3894559A4 (en) * 2018-12-03 2023-04-05 Triplet Therapeutics, Inc. Methods for the treatment of trinucleotide repeat expansion disorders associated with mlh3 activity
WO2020128163A1 (en) 2018-12-20 2020-06-25 Itä-Suomen Yliopisto Synthetic microrna mimics
EP3919618A4 (en) * 2019-01-30 2023-11-08 Ractigen Therapeutics Oligonucleotide molecule and application thereof in tumor therapy
CN110106180B (en) * 2019-05-29 2020-10-16 中国医科大学附属盛京医院 lncRNA molecule and application thereof in glioma treatment/prognosis evaluation
CN110106180A (en) * 2019-05-29 2019-08-09 中国医科大学附属盛京医院 A kind of lncRNA molecule and its application in Treatment for Glioma/prognosis evaluation
CN110577988B (en) * 2019-07-19 2022-12-20 南方医科大学 Fetal growth restriction prediction model
CN110577988A (en) * 2019-07-19 2019-12-17 南方医科大学 Prediction model for fetal growth restriction
WO2021032777A1 (en) 2019-08-19 2021-02-25 Mina Therapeutics Limited Oligonucleotide conjugate compositions and methods of use
CN111118154B (en) * 2020-01-16 2022-10-18 华南协同创新研究院 Application of LINC01272 in preparation of tumor detection reagent and/or treatment drug
CN111118154A (en) * 2020-01-16 2020-05-08 华南协同创新研究院 Application of LINC01272 in preparation of tumor detection reagent and/or treatment drug
CN111394351B (en) * 2020-03-18 2023-11-07 济南爱新卓尔医学检验有限公司 siRNA for inhibiting DICER1-AS1 expression and application thereof
CN111394351A (en) * 2020-03-18 2020-07-10 昆明医科大学 siRNA for inhibiting DICER1-AS1 expression and application thereof
US11795459B2 (en) 2020-04-02 2023-10-24 Mirecule, Inc. Targeted inhibition using engineered oligonucleotides
US11597930B2 (en) 2020-04-02 2023-03-07 Mirecule, Inc. Targeted inhibition using engineered oligonucleotides
CN111718956A (en) * 2020-06-30 2020-09-29 山东农业大学 Preparation method and application of chicken-derived TRIM25 gene recombinant fluorescent expression plasmid
CN111718956B (en) * 2020-06-30 2022-11-04 山东农业大学 Preparation method and application of chicken-derived TRIM25 gene recombinant fluorescent expression plasmid
US11845937B2 (en) 2020-09-11 2023-12-19 Arrowhead Pharmaceuticals, Inc. RNAi agents for inhibiting expression of DUX4, compositions thereof, and methods of use
CN112575076A (en) * 2020-11-02 2021-03-30 深圳恒生医院 LncRNA marker for detecting and evaluating early spontaneous abortion and product using LncRNA marker
WO2022140264A1 (en) * 2020-12-23 2022-06-30 Eli Lilly And Company Rna therapeutics and methods of use thereof
CN112661826B (en) * 2020-12-25 2022-03-29 中山大学 Small peptide ERpeptide and application thereof in acute myelogenous leukemia
CN112661826A (en) * 2020-12-25 2021-04-16 中山大学 Small peptide ERpeptide and application thereof in acute myelogenous leukemia
WO2022200810A1 (en) * 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022217283A1 (en) 2021-04-08 2022-10-13 Joslin Diabetes Center, Inc. Methods of diagnosing and predicting renal decline
CN113151471B (en) * 2021-04-28 2022-08-12 复旦大学附属中山医院 Gene composition, kit and method for detecting KRAS mutation of lung adenocarcinoma
CN113151471A (en) * 2021-04-28 2021-07-23 复旦大学附属中山医院 Gene composition, kit and method for detecting KRAS mutation of lung adenocarcinoma
WO2022245734A3 (en) * 2021-05-17 2023-02-09 Corlieve Therapeutics Methods and compositions for treating epilepsy
WO2023111337A1 (en) * 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Antisense oligonucleotide
CN114540500A (en) * 2022-03-21 2022-05-27 深圳市陆为生物技术有限公司 Product for evaluating overall survival of breast cancer patients
CN114574493A (en) * 2022-04-02 2022-06-03 中国科学院遗传与发育生物学研究所 sgRNA combination for editing sheep SOCS2 gene, primers for amplification and application
WO2024001172A1 (en) * 2022-06-27 2024-01-04 Ractigen Therapeutics Oligonucleotide modulators activating complement factor h expression

Also Published As

Publication number Publication date
EP3286318A2 (en) 2018-02-28
WO2016170348A8 (en) 2017-11-30
JP2021035360A (en) 2021-03-04
WO2016170348A3 (en) 2017-04-13
JP2023040138A (en) 2023-03-22
US20180305689A1 (en) 2018-10-25
US20210363525A1 (en) 2021-11-25
JP2018512876A (en) 2018-05-24

Similar Documents

Publication Publication Date Title
WO2016170348A2 (en) Sarna compositions and methods of use
US20180258429A1 (en) Sarna compositions and methods of use
ES2932516T3 (en) Combinations of mRNA encoding immunomodulatory polypeptides and uses thereof
AU2019210578B2 (en) C/EBP alpha saRNA compositions and methods of use
US11447773B2 (en) Stabilized HNF4A saRNA compositions and methods of use
US20240018519A1 (en) Stabilized saRNA Compositions and Methods of Use
US20240002850A1 (en) SIRT1-saRNA Compositions and Methods of Use
WO2023099884A1 (en) Pax6 sarna compositions and methods of use
US20220211740A1 (en) Sirt1-sarna compositions and methods of use
US20240175033A1 (en) TMEM173 saRNA Compositions and Methods of Use
WO2023170435A1 (en) Il10 sarna compositions and methods of use
JP2024511092A (en) TMEM173saRNA composition and method of use
CN117337330A (en) TMEM173 saRNA compositions and methods of use
WO2022047105A2 (en) Nucleic acids for inhibiting tert expression
WO2023004150A2 (en) Compositions and methods for treatment of neurodegenerative diseases

Legal Events

Date Code Title Description
REEP Request for entry into the european phase

Ref document number: 2016735927

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017555233

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15568046

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 16735927

Country of ref document: EP

Kind code of ref document: A2