NZ714577B2 - Tmprss6 irna compositions and methods of use thereof - Google Patents

Tmprss6 irna compositions and methods of use thereof Download PDF

Info

Publication number
NZ714577B2
NZ714577B2 NZ714577A NZ71457714A NZ714577B2 NZ 714577 B2 NZ714577 B2 NZ 714577B2 NZ 714577 A NZ714577 A NZ 714577A NZ 71457714 A NZ71457714 A NZ 71457714A NZ 714577 B2 NZ714577 B2 NZ 714577B2
Authority
NZ
New Zealand
Prior art keywords
kim
annotation
nucleotides
strand
tmprss6
Prior art date
Application number
NZ714577A
Other versions
NZ714577A (en
Inventor
Brian Bettencourt
James Butler
Klaus Charisse
Martin Maier
Kallanthottathil G Rajeev
Original Assignee
Alnylam Pharmaceuticals Inc
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 Alnylam Pharmaceuticals Inc filed Critical Alnylam Pharmaceuticals Inc
Priority claimed from PCT/US2014/039149 external-priority patent/WO2014190157A1/en
Publication of NZ714577A publication Critical patent/NZ714577A/en
Publication of NZ714577B2 publication Critical patent/NZ714577B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/316Phosphonothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/343Spatial arrangement of the modifications having patterns, e.g. ==--==--==--
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3521Methyl
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3533Halogen
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)

Abstract

The invention relates to double-stranded RNAi agents targeting the TMPRSS6 gene, wherein the antisense strand thereof comprises at least 15 contiguous nucleotides differing no more than 3 nucleotides from the sequence AGAAUGAACCAGAAGAAGCAGGU (SEQ ID NO: 187) and wherein all the nucleotides of the sense and antisense strands are modified nucleotides and each strand is between 15 to 30 nucleotides in length. The invention further relates to methods of using such RNAi agents to inhibit expression of TMPRSS6 and methods of treating subjects having a TMPRSS6 associated disorder, e.g., an iron overload associated disorder, such as ?-thalassemia or hemochromatosis. The invention additionally relates to a pharmaceutical composition and an isolated cell comprising such RNAi agents. nse and antisense strands are modified nucleotides and each strand is between 15 to 30 nucleotides in length. The invention further relates to methods of using such RNAi agents to inhibit expression of TMPRSS6 and methods of treating subjects having a TMPRSS6 associated disorder, e.g., an iron overload associated disorder, such as ?-thalassemia or hemochromatosis. The invention additionally relates to a pharmaceutical composition and an isolated cell comprising such RNAi agents.

Description

TMPRSS6 iRNA COMPOSITIONS AND METHODS OF USE THEREOF Related Applications This application claims the benefit of priority to US. Provisional Patent Application No. 61/826,178, filed on May 22, 2013 and US. Provisional Patent Application No. 61/912,988, filed on December 6, 2013. This application is related to US. ional Application No. 61/561,710, filed on November 18, 2011, and , filed on November 16, 2012. The entire contents of each of the foregoing applications are hereby incorporated herein by reference.
Sequence Listing The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on May 21, 2014, is named 121301—00720_SL.txt and is 449,620 bytes in size.
Background of the Invention TMPRSS6 (Transmembrane Protease, Serine 6) gene encodes 6, also known as matriptase—2, a type II serine protease. It is ily expressed in the liver, although high levels of TMPRSS6 mRNA are also found in the kidney, with lower levels in the uterus and much smaller amounts detected in many other tissues y et al., Haematologica (2009), 94(6), 840—849). TMPRSS6 plays a role in iron homeostatis by binding and proteolytically degrading the hepcidin activator and BMP co—receptor H]V (hemojuvelin), which causes egulation of hepcidin levels. 6 consists of a short inal intracytoplasmic tail, a type II transmembrane , a stem region composed of two extracellular CUB (complement factor Cls/Clr, urchin embryonic growth factor and BMP (bone morphogenetic protein)) domains, three LDLR (low—density—lipoprotein receptor class A) domains, and a C-terminal trypsin— like serine protease domain. There are also consensus sites for N—glycosylation in the extracellular domain, and a potential phosphorylation site in the intracytoplasmic tail region.
Numberous disorders can be associated with iron overload, a ion characterized by increased levels of iron. Iron overload can result in excess iron deposition in various tissues and can lead to tissue and organ damage. Accordingly, methods for effective treatment of disorders associated with iron overload are currently needed. y of the ion The present invention provides compositions sing RNAi agents, e.g., double— stranded iRNA agents, targeting TMPRSS6. The t ion also provides methods [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM using the compositions of the invention for inhibiting TMPRSS6 expression and for treating TMPRSS6 associated disorders, e.g., iron ad associated disorders, such as thalassemia, e.g., B—thalassemia, or hemochromatosis.
Accordingly, in one aspect, the present invention provides RNAi agents, e.g., double— stranded RNAi agents, e of inhibiting the expression of TMPRSS6 ptase—2) in a cell, wherein the double stranded RNAi agent comprises a sense strand and an antisense strand forming a double—stranded region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 3 tides from any one of the nucleotide sequences of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5, and the antisense strand comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the nucleotide sequences of SEQ ID NO:6, SEQ ID NO:7, or SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO:10, wherein substantially all of the tides of the sense strand and substantially all of the nucleotides of the antisense strand are modified nucleotides, and wherein the sense strand is conjugated to a ligand attached at the minus.
In one embodiment, all of the nucleotides of said sense strand and all of the nucleotides of said antisense strand are ed nucleotides.
In one embodiment, the sense strand and the antisense strand comprise a region of mentarity which comprises at least 15 contiguous tides differing by no more than 3 nucleotides from any one of the antisense sequences listed in any one of Tables 1, 2, 4, , 8, 10, and 12.
In one embodiment, at least one of the modified nucleotides is selected from the group consisting of a 3’-terminal deoxy-thymine (dT) nucleotide, a ethyl modified nucleotide, a 2'—fluoro modified tide, a 2'—deoxy—modified nucleotide, a locked nucleotide, an abasic nucleotide, a 2’-amino-modified nucleotide, a 2’-alkyl-modified nucleotide, a morpholino nucleotide, a phosphoramidate, a non-natural base comprising nucleotide, a tide comprising a 5'—phosphorothioate group, a nucleotide comprising a ’ phosphate or 5’ phosphate mimic (see, 6.5)., PCT Publication No. ), and a terminal nucleotide linked to a cholesteryl derivative or a noic acid bisdecylamide group.
In one embodiment, at least one strand comprises a 3’ overhang of at least 1 nucleotide. In another embodiment, at least one strand comprises a 3’ overhang of at least 2 nucleotides. In another aspect, the present invention provides RNAi agents, e.g., double— stranded RNAi agents, capable of inhibiting the expression of TMPRSS6 (matriptase—2) in a cell, wherein the double stranded RNAi agent ses a sense strand complementary to an antisense strand, wherein the antisense strand comprises a region complementary to part of an mRNA encoding TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in lengthénerein the double stranded RNAi agent is represented by formula (III): [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM sense: 5' np —Na —(X X X) i—Nb —Y Y Y —Nb —(Z Z Z)j —Na — nq 3' antisense: 3' np'—Na’—(X'X'X')k—Nb'-Y’Y’Y'—Nb'—(Z'Z'Z')1—Na’— nq' 5' (III) wherein: i, j, k, and l are each independently 0 or 1; p, p’, q, and q’ are each independently 0—6; each Na and Na' independently represents an oligonucleotide sequence comprising 0— nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two differently modified nucleotides; each Nb and Nb’ independently represents an oligonucleotide sequence comprising 0— 10 nucleotides which are either modified or unmodified or combinations f; each np, np', nq, and nq', each of which may or may not be present, independently represents an overhang nucleotide; XXX, YYY, ZZZ, X’X'X’, Y'Y'Y', and Z’Z'Z' each independently represent one motif of three identical modifications on three consecutive nucleotides; cations on Nb differ from the modification on Y and modifications on Nb' differ from the modification on Y'; and wherein the sense strand is conjugated to at least one ligand.
In one embodiment, i is O;j is 0; i is l;j is 1; both i andj are 0; or both i andj are 1.
In another embodiment, k is 0; l is 0; k is 1; l is 1; both k and l are 0; or both k and l are 1.
In one embodiment, XXX is complementary to X'X'X’, YYY is complementary to Y’Y'Y’, and ZZZ is complementary to Z’Z'Z’.
In one embodiment, YYY motif occurs at or near the cleavage site of the sense strand.
In one embodiment, Y’Y’Y' motif occurs at the 11, 12 and 13 positions of the antisense strand from the 5'—end.
In one ment, Y’ is 2’—O—methyl.
In one embodiment, formula (III) is represented by formula : sense: 5' np —Na —Y Y Y —Na - nq 3' antisense: 3' an—Nal— Y'Y'Y'— Nal- nq/ 5' (IIIa).
In another embodiment, formula (III) is ented by formula (IIIb): sense: 5' np —Na —Y Y Y —Nb -Z Z Z —Na — nq 3' antisense: 3' npr—Na/— Y’Y’Y’—NbI—Z’Z’Z’— Na/— an 5' (IIIb) wherein each Nb and Nb’ independently represents an oligonucleotide sequence comprising 1-5 modified tides.
In yet another embodiment, formula (III) is ented by a (IIIc): sense: 5' np -Na —X X X -Nb -Y Y Y -Na - nq 3' nse: 3' npr—Na/— X’X’X’—Nbl— Y’Y’Y’— Na/— nql 5' (IIIc) [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM wherein each Nb and Nb’ independently ents an oligonucleotide sequence comprising 1—5 modified nucleotides.
In one embodiment, formula (III) is represented by formula (IIId): sense: 5' np —Na —X X X— Nb —Y Y Y —Nb —Z Z Z —Na — nq 3' antisense: 3' an—Na/— X'X’X’— NbI—Y’Y’Y'—Nb/—Z'Z’Z’— Nal— nq/ 5' (HM) wherein each Nb and Nb’ independently represents an oligonucleotide sequence comprising 1—5 modified nucleotides and each Na and Na' independently represents an oligonucleotide sequence comprising 2—10 modified tides.
In one embodiment, the double—stranded region is 15—30 nucleotide pairs in length. In another embodiment, the double—stranded region is 17—23 nucleotide pairs in length. In yet another embodiment, the double—stranded region is 17—25 nucleotide pairs in length. In one embodiment, the double—stranded region is 23—27 tide pairs in length. In another embodiment, the double—stranded region is 19—21 nucleotide pairs in length. In another embodiment, the double—stranded region is 21—23 nucleotide pairs in . In one embodiment, each strand has 15—30 tides. In another embodiment, each strand has 19— nucleotides.
In one embodiment, the modifications on the nucleotides are selected from the group consisting of LNA, HNA, CeNA, 2’—methoxyethyl, 2’—O—alkyl, 2’—O—allyl, 2’—C— allyl, 2’— fluoro, 2’—deoxy, 2’—hydroxyl, and combinations thereof. In another embodiment, the modifications on the nucleotides are 2’—O—methyl or 2’—fluoro cations.
In one embodiment, the ligand is one or more GalNAc derivatives ed through a bivalent or trivalent branched linker. In another embodiment, the ligand is O H H HO O ACHN O\/\/\n/N\/\/N OH K HO o H H AcHN \/\/\n/ \/\/ \H/\/0%" O 0 O HO O\/\/\n/N/V\N o AcHN H H In one embodiment, the ligand is attached to the 3’ end of the sense .
In one embodiment, the RNAi agent is conjugated to the ligand as shown in the ing schematic [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM HO E ACHN OMNWNWO O O O 0 HOfio N’\/‘N o AcHN wherein X is O or S. In a specific embodiment, X is O.
In one embodiment, the agent further comprises at least one phosphorothioate or methylphosphonate internucleotide linkage.
In one embodiment, the phosphorothioate or phosphonate internucleotide linkage is at the 3’—terminus of one strand. In one embodiment, the strand is the antisense . In another embodiment, the strand is the sense .
In one embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is at the 5’—terminus of one strand. In one embodiment, the strand is the antisense . In r embodiment, the strand is the sense strand.
In one embodiment, the phosphorothioate or methylphosphonate internucleotide linkage is at the both the 5’— and 3’—terminus of one strand. In one embodiment, the strand is the antisense strand.
In one embodiment, the RNAi agent comprises 6—8 phosphorothioate internucleotide linkages.
In one embodiment, the antisense strand ses two phosphorothioate internucleotide es at the minus and two phosphorothioate internucleotide es at the 3’—terminus, and the sense strand comprises at least two phosphorothioate ucleotide linkages at either the 5’—terminus or the 3’—terminus.
In one embodiment, the base pair at the 1 position of the 5’-end of the antisense strand of the duplex is an AU base pair.
In one embodiment, the Y nucleotides contain a 2’—fluoro modification.
In one embodiment, the Y’ nucleotides contain a 2’—O—methyl modification.
In one embodiment, p’>0. In another embodiment, p’=2.
In one embodiment, q’=0, p20, q=0, and p’ overhang nucleotides are complementary to the target mRNA. In another embodiment, q’=0, p=0, q=0, and p’ overhang nucleotides are non—complementary to the target mRNA.
In one embodiment, the sense strand has a total of 21 nucleotides and the antisense strand has a total of 23 nucleotides.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM In one embodiment, at least one np' is linked to a neighboring nucleotide via a orothioate linkage.
In one embodiment, all np’ are linked to neighboring nucleotides via phosphorothioate linkages.
In one embodiment, the RNAi agent is selected from the group of RNAi agents listed in any one of Tables 1, 2, 4, 5, 8, 10, and 12.
In one embodiment, the RNAi agent is AD—59743. In r embodiment, the RNAi agent is AD—60940.
In one aspect, the t invention provides double stranded RNAi agents for inhibiting expression of TMPRSS6 in a cell, wherein the double stranded RNAi agent comprises a sense strand and an antisense strand forming a double ed region, wherein the sense strand comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the nucleotide sequences of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5, and the antisense strand comprises at least 15 contiguous tides differing by no more than 3 nucleotides from any one of the nucleotide sequences of SEQ ID NO:6, SEQ ID NO:7, or SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NOle, wherein substantially all of the nucleotides of the sense strand comprise a 2O modification selected from the group consisting of a 2’—O—methyl modification and a 2’— fluoro modification, wherein the sense strand comprises two phosphorothioate intemucleotide linkages at the 5’—terminus, wherein substantially all of the nucleotides of the antisense strand comprise a modification ed from the group consisting of a 2’—O—methyl modification and a 2’— fluoro cation, wherein the antisense strand ses two phosphorothioate intemucleotide linkages at the 5’—terminus and two phosphorothioate intemucleotide linkages at the minus, and wherein the sense strand is conjugated to one or more GalNAc derivatives attached through a branched bivalent or trivalent linker at the 3’—terminus.
In one embodiment, all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand se a modification.
In another aspect, the present invention provides RNAi agents, e.g., double stranded RNAi agents, capable of inhibiting the expression of TMPRSS6 ptase-2) in a cell, wherein the double stranded RNAi agent comprises a sense strand mentary to an antisense strand, wherein the antisense strand ses a region complementary to part of an mRNA encoding TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in lengthfierein the double stranded RNAi agent is represented by formula (III): [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM sense: 5' np —Na —(X X X) i—Nb —Y Y Y —Nb —(Z Z Z)j —Na — nq 3' antisense: 3' np'—Na’—(X'X'X')k—Nb'-Y’Y’Y'—Nb'—(Z'Z'Z')1—Na’— nq' 5' (111) wherein: i, j, k, and l are each independently 0 or 1; p, p’, q, and q’ are each independently 0—6; each Na and Na' independently ents an oligonucleotide sequence comprising 0— nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two ently modified nucleotides; each Nb and Nb’ independently represents an oligonucleotide sequence comprising 0— 10 nucleotides which are either ed or unmodified or ations thereof; each np, np', nq, and nq', each of which may or may not be present independently represents an overhang nucleotide; XXX, YYY, ZZZ, X’X'X’, Y'Y'Y', and Z’Z'Z' each independently represent one motif of three identical modifications on three consecutive nucleotides, and wherein the modifications are 2’—O—methyl or ro modifications; cations on Nb differ from the modification on Y and modifications on Nb' differ from the modification on Y'; and wherein the sense strand is ated to at least one ligand.
In yet another aspect, the t invention provides RNAi agents, e.g., double stranded RNAi agents, capable of inhibiting the expression of TMPRSS6 (matriptase—Z) in a cell, wherein the double ed RNAi agent comprises a sense strand mentary to an antisense strand, wherein the antisense strand comprises a region complementary to part of an mRNA ng TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in length, wherein the double ed RNAi agent is represented by formula (III): sense: 5' np —Na —(X X X) i—Nb —Y Y Y —Nb —(Z Z Z)J~ —Na — nq 3' antisense: 3' np'—Na'-(X'X'X')k—Nb'-Y'Y'Y'—Nb'—(Z'Z'Z')1—Na'- nq' 5' (111) i, j, k, and l are each independently 0 or 1; each np, nq, and nq', each of which may or may not be present, independently represents an overhang nucleotide; p, q, and q’ are each independently 0—6; np’ >0 and at least one np’ is linked to a neighboring nucleotide via a phosphorothioate linkage; each Na and Na’ independently represents an oligonucleotide sequence comprising 0- 25 nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two differently modified nucleotides; each Nb and Nb’ independently represents an oligonucleotide sequence comprising 0— nufitides which are either modified or unmodified or combinations thereof; [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM XXX, YYY, ZZZ, X'X'X', Y'Y'Y’, and Z’Z'Z' each independently represent one motif of three identical cations on three consecutive nucleotides, and n the modifications are 2’—O—methyl or ro modifications; modifications on Nb differ from the modification on Y and modifications on Nb' differ from the modification on Y'; and wherein the sense strand is conjugated to at least one .
In a further aspect, the present invention provides RNAi agents, e.g., double stranded RNAi agents, capable of inhibiting the expression of TMPRSS6 (matriptase—2) in a cell, wherein the double stranded RNAi agent comprises a sense strand complementary to an antisense strand, wherein the antisense strand comprises a region mentary to part of an mRNA encoding TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in length, wherein the double stranded RNAi agent is represented by formula (III): sense: 5' np —Na —(X X X) i—Nb —Y Y Y —Nb —(Z Z Z)J~ —Na — nq 3' antisense: 3' ’—(X'X'X’)k—Nb'-Y'Y’Y'-Nb'—(Z'Z’Z')1—Na’— nq' 5' (111) n: i, j, k, and l are each ndently 0 or 1; each np, nq, and nq', each of which may or may not be present, independently represents an overhang nucleotide; p, q, and q’ are each independently 0—6; np' >0 and at least one np' is linked to a neighboring nucleotide via a phosphorothioate linkage; each Na and Na' independently represents an oligonucleotide sequence comprising 0— tides which are either modified or unmodified or ations thereof, each sequence comprising at least two differently modified nucleotides; each Nb and Nb’ independently represents an oligonucleotide sequence comprising 0— nucleotides which are either ed or unmodified or combinations thereof; XXX, YYY, ZZZ, X’X'X’, Y'Y'Y', and Z’Z'Z' each independently represent one motif of three identical modifications on three utive nucleotides, and wherein the modifications are 2’—O—methyl or 2’—fluoro modifications; modifications on Nb differ from the modification on Y and modifications on Nb’ differ from the modification on Y'; and wherein the sense strand is conjugated to at least one ligand, wherein the ligand is one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.
In another aspect, the present invention provides RNAi agents, e. g., double stranded RNAi agents capable of inhibiting the expression of TMPRSS6 (matriptase—2) in a cell, n the double stranded RNAi agent comprises a sense strand mentary to an antisense , wherein the antisense strand comprises a region complementary to part of an mRNficoding TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM length, wherein the double stranded RNAi agent is represented by formula (III): sense: 5' np —Na —(X X X) i—Nb -Y Y Y -Nb —(Z Z Z)J~ —Na — nq 3' antisense: 3' np'—Na'—(X'X'X')k—Nb'—Y’Y’Y’—Nb'—(Z'Z'Z')1—Na’— nq' 5' (III) wherein: i, j, k, and l are each independently 0 or 1; each np, nq, and nq', each of which may or may not be present, independently represents an overhang nucleotide; p, q, and q' are each independently 0—6; np' >0 and at least one np' is linked to a neighboring nucleotide via a phosphorothioate linkage; each Na and Na' independently represents an oligonucleotide sequence comprising 0— nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two differently modified tides; each Nb and Nb’ independently represents an oligonucleotide sequence comprising 0— 10 nucleotides which are either modified or unmodified or combinations thereof; XXX, YYY, ZZZ, X’X'X', Y'Y'Y’, and Z'Z’Z' each ndently ent one motif of three identical modifications on three utive nucleotides, and wherein the cations are 2’-O—methyl or 2’-fluoro modifications; modifications on Nb differ from the modification on Y and modifications on Nb' differ from the modification on Y'; wherein the sense strand ses at least one phosphorothioate linkage; and wherein the sense strand is conjugated to at least one ligand, n the ligand is one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.
In yet another aspect, the present invention provides RNAi agents, e.g., double stranded RNAi , e of inhibiting the expression of TMPRSS6 (matriptase—2) in a cell, wherein the double stranded RNAi agent comprises a sense strand complementary to an antisense strand, wherein the antisense strand comprises a region mentary to part of an mRNA encoding TMPRSS6, wherein each strand is about 14 to about 30 nucleotides in length, wherein the double stranded RNAi agent is represented by formula (III): sense: 5' np —Na -Y Y Y - Na— nq 3' antisense: 3' np'—Na'— Y’Y'Y'— Na'— nq' 5' (111a) wherein: each np, nq, and nq', each of which may or may not be t, independently represents an overhang nucleotide; p, q, and q’ are each independently 0—6; np’ >0 and at least one np' is linked to a oring nucleotide via a phosphorothioate linkage; Each Na and Na’ independently represents an oligonucleotide sequence comprising 0- [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two differently modified nucleotides; YYY and Y'Y'Y' each independently ent one motif of three cal modifications on three consecutive nucleotides, and wherein the modifications are 2’—O— methyl or 2’—fluoro modifications; wherein the sense strand comprises at least one phosphorothioate linkage; and wherein the sense strand is conjugated to at least one , wherein the ligand is one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.
In one embodiment, the present invention provides RNAi agent selected from the group of RNAi agents listed in any one of Tables 1, 2, 4, 5, 8, l9, and 12.
In one aspect, the present ion provides compositions comprising a modified antisense polynucleotide agent, wherein the agent is e of inhibiting the expression of TMPRSS6 in a cell, and comprises a sequence complementary to a sense sequence selected from the group of the sequences listed in any one of Tables 1, 2, 4, 5, 8, 10, and 12, wherein the polynucleotide is about 14 to about 30 nucleotides in length.
The present invention also es cells, vectors, host cells, and pharmaceutical itions comprising, e.g., the double ed RNAi agents of the invention.
In some embodiments, the RNAi agent is administered using a pharmaceutical composition. 2O In red embodiments, the RNAi agent is stered in a solution. In some such embodiments, the siRNA is administered in an unbuffered on. In one embodiment, the siRNA is administered in water. In other embodiments, the siRNA is administered with a buffer solution, such as an acetate buffer, a citrate buffer, a prolamine buffer, a carbonate buffer, or a phosphate buffer or any combination f. In some embodiments, the buffer solution is phosphate buffered saline (PBS).
In one embodiment, the pharmaceutical compositions further se a lipid formulation. In one embodiment, the lipid formulation comprises a LNP, or XTC. In another embodiment, the lipid formulation comprises a MC3.
In one aspect, the present invention provides methods of inhibiting TMPRSS6 expression in a cell. The methods include ting the cell with an RNAi agent, e.g., a double stranded RNAi agent, or a modified antisense cleotide agent of the invention, or vector of the invention, or a pharmaceutical composition of the invention; and maintaining the cell ed in step (a) for a time sufficient to obtain degradation of the mRNA transcript of a TMPRSS6 gene, thereby inhibiting expression of the TMPRSS6 gene in the cell.
In one embodiment, the cell is within a subject.
In one embodiment, the subject is a human.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In one embodiment, the 6 expression is inhibited by at least about 30%, %, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 100%.
In another embodiment, hepcidin gene expression is increased by at least about 1.5— fold, about 2—fold, about 3—fold, about 4—fold, or about 5—fold.
In yet r embodiment, serum hepcidin concentration is increased by at least about 10%, about 25%, about 50%, about 100%, about 150%, about 200%, about 250%, or about 300%.
In one embodiment, serum iron concentration is decreased by at least about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%.
In another embodiment,a percent errin saturation is decreased by at least about %, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%.
In r aspect, the t invention provides methods of treating a subject having a disorder mediated by, or associated with, TMPRSS6 expression. The methods include administering to the t a therapeutically ive amount of an RNAi agent, e.g., a double ed RNAi agent, of the invention, or a modified antisense cleotide agent of the invention, or a vector of the invention, or a pharmaceutical composition of the invention, thereby treating the subject.
In one aspect, the present ion provides methods of treating a subject having a TMPRSS6—associated disorder. The methods include subcutaneously administering to the subject a therapeutically effective amount of a double stranded RNAi agent, wherein the double stranded RNAi agent comprises a sense strand and an antisense strand forming a double stranded region, wherein the sense strand comprises at least 15 uous nucleotides differing by no more than 3 nucleotides from any one of the nucleotide sequences of SEQ ID NO: 1, SEQ ID NO:2, or SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5, and the antisense strand comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from any one of the nucleotide sequences of SEQ ID NO:6, SEQ ID NO:7, or SEQ ID N028, SEQ ID N029, or SEQ ID NO:10, wherein substantially all of the nucleotides of the antisense strand comprise a cation selected from the group consisting of a ethyl modification and a 2’— fluoro modification, wherein the antisense strand comprises two phosphorothioate internucleotide linkages at the 5’—terminus and two phosphorothioate internucleotide linkages at the 3’—terminus, wherein substantially all of the tides of the sense strand comprise a modification selected from the group consisting of a 2’—O-methyl modification and a 2’— fluorogdification, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM wherein the sense strand comprises two phosphorothioate intemucleotide es at the 5’—terminus and, wherein the sense strand is conjugated to one or more GalNAc derivatives attached through a branched bivalent or trivalent linker at the 3’—terminus, thereby treating the t.
In one embodiment, all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand comprise a modification.
In one embodiment, the subject is a human.
In one ment, the subject has a disorder associated with iron overload, e.g., hereditary hemochromatosis, B—thalassemia (e.g., B—thalassemia major and B—thalassemia iedia) erythropoietic porphyria, Parkinson’s Disease, Alzheimer’s Disease or Friedreich’s Ataxia.
In one embodiment, the RNAi agent, e.g., double stranded RNAi agent, is administered at a dose of about 0.01 mg/kg to about 10 mg/kg, about 1 mg/kg to about 10 mg/kg, about 2 mg/kg to about 10 mg/kg, about 3 mg/kg to about 10 mg/kg, about 4 mg/kg to about 10 mg/kg, about 5 mg/kg to about 15 mg/kg, about 6 mg/kg to about 15 mg/kg, about 7 mg/kg to about 15 mg/kg, about 8 mg/kg to about 15 mg/kg, about 9 mg/kg to about 15 mg/kg, about 10 mg/kg to about 20 mg/kg, about 12 mg/kg to about 20 mg/kg, about 13 mg/kg to about 20 mg/kg, about 14 mg/kg to about 20 mg/kg, about 15 mg/kg to about 20 mg/kg, about 16 mg/kg to about 20 mg/kg or about 18 mg/kg to about 20 mg/kg. In particular embodiments, the double stranded RNAi agent is stered at a dose of about 0.1 mg/kg, about 1.0 mg/kg, or about 3.0 mg/kg.
In one ment, the RNAi agent, e.g., double stranded RNAi agent, is administered subcutaneously or intravenously.
In one embodiment, the RNAi agent is administered in two or more doses. In a specific embodiment, the RNAi agent is administered at intervals selected from the group consisting of once every about 12 hours, once every about 24 hours, once every about 48 hours, once every about 72 hours, once every about 96 hours, once about every 7 days, or once about every 14 days. In particular embodiments, the RNAi agent is administered once a week for up to 2 weeks, up to 3 weeks, up to 4 weeks, up to 5 weeks, or longer.
In yet another aspect, the present invention provides methods of treating an iron overload associated disorder in a subject. The methods include stering to the subject a therapeutically effective amount of an RNAi agent, e.g., a double ed RNAi agent, or the vector of the invention, thereby treating the subject.
In one ment, the iron overload associated disorder is hemochromatosis. In another ment, the iron overload associated er is a thalassemia, e.g., B— thalassemia (e.g., B-thalassemia major and B-thalassemia intermiedia), or erythropoietic porphyria. In yet another embodiment, the iron overload associated disorder is a neurological diseasgg., Parkinson’s se, Alzheimer’s Disease or Friedreich’s Ataxia.
[Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM In one embodiment, the subject is a e or rodent. In r embodiment, the subject is a human.
In one embodiment, the RNAi agent, e.g., double stranded RNAi agent, is administered at a dose of about 0.01 mg/kg to about 10 mg/kg, about 0.5 mg/kg to about 50 mg/kg, about 10 mg/kg to about 30 mg/kg, about 10 mg/kg to about 20 mg/kg, about 15 mg/kg to about 20 mg/kg, about 15 mg/kg to about 25 mg/kg, about 15 mg/kg to about 30 mg/kg, or about 20 mg/kg to about 30 mg/kg.
In one embodiment, the RNAi agent, e.g., double stranded RNAi agent, is administered subcutaneously or intravenously.
In one embodiment, the RNAi agent is administered in two or more doses. In a specific embodiment, the RNAi agent is administered at intervals selected from the group consisting of once every about 12 hours, once every about 24 hours, once every about 48 hours, once every about 72 hours, once every about 96 hours, once about every 7 days, or once about every 14 days.
In one embodiment, administering results in a decrease in iron , ferritin level and/or transferrin saturation level in the subject.
In one embodiment, the s further comprise ining the iron level in the subject.
In one embodiment, the methods of the invention which include stering an iRNA agent of the invention (or pharmaceutical composition of the invention) to a subject are practiced in combination with administration of additional pharmaceuticals and/or other eutic methods. In one embodiment, the methods of the invention further comprise administering an iron chelator, e.g., deferiprone, deferoxamine, and deferasirox, to a subject.
The present invention is further illustrated by the following detailed description and drawings.
Brief Description of the Drawings Figure 1 is a graph showing relative levels of TMPRSS6 mRNA in the liver of wild- type mice following administration of a single dose of 1 mg/kg, 3 mg/kg or 10 mg/kg of the iRNA agent AD—59743.
Figure 2 is a graph showing relative levels of hepcidin mRNA in the liver of wild— type mice following administration of a single dose of 1 mg/kg, 3 mg/kg or 10 mg/kg of the iRNA agent AD—59743.
Figures 3A-3E show the levels of hepatic TMPRSS6 mRNA (Figure 3A), hepatic in mRNA (Figure 3B), serum hepcidin (Figure 3C), total serum iron (Figure 3D), and percerfiansferrin saturation (Figure 3E) in 6 mice at various time points following a [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM single subcutaneous injection of AD—60940 at a dose of 0.3 mg/kg, 1.0 mg/kg or 3.0 mg/kg, or PBS alone (control). Each data point represents the mean value from three mice. The standard deviation of the mean is represented by error bars. Figure 3F demonstrates the relative hepatic TMPRSS6 mRNA concentration as a function of AD—60940 dose at 11 days following administration. Each data point represents the maximum suppression of TMPRSS6 mRNA concentration ed at each dose level. The data were fit to the Hill on.
Figure 4A is a schematic depicting the adminstration regimen of one dose per week for three weeks followed by sacrifice of the mice at day 21. Figure 4B is a graph showing the levels of c TMPRSS6 mRNA, hepatic hepcidin mRNA, and percent transferrin saturation in C57BL/6 mice administered a subcutaneous injection of AD—60940 at a dose of 0.3 mg/kg, 1.0 mg/kg, or PBS (control) according to the regimen shown in Figure 4A. Each bar represents the mean value from three mice. The standard deviation of the mean is represented by error bars. Figure 4C demonstrates the relative c TMPRSS6 mRNA concentration as a function of AD—60940 dose. The data were fit to the Hill on.
Figures 5A-5D are graphs g the relationships n serum hepcidin concentration and relative TMPRSS6 mRNA levels (Figure 5A), between percent transferrin saturation and relative TMPRSS6 mRNA levels (Figure 5B), between serum hepcidin concentration and relative hepcidin mRNA levels (Figure 5C) and n percent transferrin saturation and serum hepcidin concentration (Figure 5D).
Figure 6 is a graph showing ve levels of TMPRSS6 mRNA in the liver of C57BL/6 mice following administration of a single subcutaneous dose of 3 mg/kg of the indicated iRNA agent or PBS (control). The bars represent the mean from three mice and the error bars ent the standard deviation of the mean.
Figure 7 is a graph g relative levels of TMPRSS6 mRNA in the liver of C57BL/6 mice following a subcutaneous dose of 0.3 mg/kg or 1.0 mg/kg of the indicated iRNA agent, or PBS (control), once a week for three weeks. The bars represent the mean from three mice and the error bars represent the standard deviation of the mean.
Figure 8 shows the nucleotide sequence of Homo sapiens TMPRSS6 (SEQ ID NO:1).
Figure 9 shows the nucleotide sequence of Mus musculus TMPRSS6 (SEQ ID NO:2).
Figure 10 shows the nucleotide sequence of Rattus norvegicus TMPRSS6 (SEQ ID NO:3).
Figure 11 shows the nucleotide sequence of Macaca mulatta TMPRSS6 (SEQ ID NO:4).
Figure 12 shows the nucleotide sequence of Macaca mulatta TMPRSS6 (SEQ ID NO:5).
Figure 13 shows the reverse complement of SEQ ID NO:1 (SEQ ID NO:6).
Figure 14 shows the reverse complement of SEQ ID NO:2 (SEQ ID NO:7). figure 15 shows the e complement of SEQ ID NO:3 (SEQ ID NO:8).
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Figure 16 shows the reverse ment of SEQ ID NO:4 (SEQ ID NO:9).
Figure 17 shows the reverse complement of SEQ ID NO:5 (SEQ ID NO: 10).
Detailed Description of the Invention The present invention provides itions comprising RNAi agents, e. g., double— stranded iRNA agents, targeting TMPRSS6. The present invention also provides methods using the compositions of the invention for ting TMPRSS6 expression and for treating TMPRSS6 associated disorders, e.g., B—thalassemia or hemochromatosis.
TMPRSS6 plays an important role in iron homeostasis as an inhibitor of HAMP gene sion. The HAMP gene encodes the liver hormone hepcidin, which is a central regulator of iron tasis. Hepcidin binds to the iron exporter protein ferroportin (FPNl), which is zed mainly on absorptive enterocytes, hepatocytes and macrophages. Hepcidin binding to the extracellular domain of ferroportin leads to the internalization and degradation of ferroportin, thus decreasing the absorption of dietary iron from the intestine, and the e of iron from macrophages and hepatocytes. HAMP gene expression can be stimulated in response to iron through Bone Morphogenetic Protein (BMP)/Sons of Mothers Against Decapentaplegic (SMAD)—dependent signal transduction cascade mediated by the BMP—co— receptor hemojuvelin (HJV). The key role of TMPRSS6 in HAMP regulation is in the inhibition of diated HAMP upregulation. 6 inhibits BMP—mediated HAMP upregulation by cleaving the BMP co—receptor H]V, which is essential for BMP—mediated HAMP upregulation; thus preventing BMP signaling, SMAD translocation to the nucleus, and HAMP transcriptional activation. l human and mouse studies have confirmed the role of TMPRSS6 in HAMP regulation and iron homeostasis (Du et al. Science 2008, Vol. 320, pp1088-1092; Folgueras et al. Blood 2008, Vol. 112, pp2539—45). Studies have shown that loss of on mutations in TMPRSS6 can lead to the upregulation of in expression, causing an inherited iron deficiency anemia called iron refractory iron deficiency anemia (IRIDA) (Finberg. Seminars in Hematology 2009, Vol. 46, pp378—86), which is characterized by elevated hepcidin levels, romic microcytic anemia, low mean cular volume (MCV), low errin tion, poor absorption of oral iron, and incomplete response to parenteral iron. However, loss of function mutations in positive regulators of HAMP (e.g., BMPl, BMP4, and HFE) have been shown to downregulate hepcidin expression and cause iron overload disorders (Milet et al. Am JHum Gen 2007, Vol. 81, 807; Finberg et al. Blood 2011, Vol. 117, pp4590—9). In the primary iron overload disorders, collectively called hereditary hemochromatosis (HH), in anemias characterized by massive ctive hematopoiesis, and in iron overload (secondary romatosis), such as B-thalassemia intermedia (TI), hepcidin levels are low despite elevated serum iron concentrations and iron stores. A mouse modefiB-thalassemia intermedia has demonstrated that the loss of TMPRSS6 expression [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM leads to elevated levels of hepcidin (Finberg 2010 Oral Presentation: "TMPRSS6, an inhibitor of Hepatic BMP/Smad Signaling, is required for in Suppression and Iron Loading in a Mouse Model of B-Thalassemia." American Society of Hematology Annual Meeting 2010, Abstract No.: 164).
The present invention describes iRNA agents, compositions and methods for modulating the expression of a TMPRSS6 gene. In certain embodiments, expression of TMPRSS6 is reduced or inhibited using a 6—specific iRNA agent, thereby leading to increase HAMP expression, and decreased serum iron levels. Thus, inhibition of TMPRSS6 gene expression or ty using the iRNA compositions featured in the invention can be a useful approach to therapies aimed at reducing the iron levels in a t. Such inhibition can be useful for ng iron overload associated disorders, such as hemochromatosis or thalassemia, e.g., B—thalassemia (e.g., assemia major and B—thalassemia intermiedia). 1. Definitions In order that the present ion may be more readily tood, certain terms are first defined. In addition, it should be noted that whenever a value or range of values of a parameter are recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this invention.
The articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at 2O least one) of the grammatical object of the article. By way of example, "an t" means one element or more than one element, 6.57., a ity of elements.
The term "including" is used herein to mean, and is used interchangeably with, the phrase "including but not limited to".
The term "or" is used herein to mean, and is used interchangeably with, the term "and/or," unless context clearly indicates otherwise.
As used herein, "TMPRSS6" refers to the type II plasma membrane serine protease (TTSP) gene or protein. TMPRSS6 is also known as matriptase—2, IRIDA (iron refractory iron—deficiency anemia), transmembrane protease serine 6, type II transmembrane serine protease 6, and membrane—bound mosaic serine proteinase matriptase—2. TMPRSS6 is a serine protease Type II transmembrane protein of approximately 899 amino acids in length.
TMPRSS6 contains multiple domains, 6.57., a short endo domain, a transmembrane , a sea urchin sperm protein/enteropeptidase domain/agrin (SEA) domain, two complement factor/urchin nic growth factor/BMP domains (CUB), three LDL—R class a domains (LDLa), and a trypsin-like serine protease domain with conserved His—Asp-Ser triad (HDS).
The term "TMPRSS6" includes human TMPRSS6, the amino acid and nucleotide sequence of which may be found in, for example, GenBank ion No. GI:56682967; mouse TMPRSS6, the amino acid and nucleotide sequence of which may be found in, for e, GenBfiAccession No. GI: 125656151; rat TMPRSS6, the amino acid and nucleotide ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM ed set by KIM sequence of which may be found in, for example, GenBank Accession No. GI: 194474097; rhesus TMPRSS6, the amino acid and nucleotide sequence of which may be found in, for example, GenBank Accession No. XM_001085203.2 (Glz297260989) and XM_001085319.l (GI: 109094061). Additional examples of AGT mRNA sequences are y ble using publicly available databases, e.g., GenBank, UniProt, OMIM, and the Macaca genome project web site.
The term"TMPRSS6," as used herein, also refers to naturally occurring DNA sequence variations of the 6 gene, such as a single nucleotide polymorphism (SNP) in the TMPRSS6 gene. Exemplary SNPs may be found in the dbSNP database ble at wwwncbi .nlm.nih . oov/ r'o 'ects/SN P.
As used herein, "target sequence" refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of a TMPRSS6 gene, including mRNA that is a product of RNA processing of a primary transcription t.
As used herein, the term "strand comprising a sequence" refers to an ucleotide comprising a chain of tides that is described by the sequence referred to using the standard tide nomenclature.
"G," "C," "A" and "U" each generally stand for a nucleotide that ns guanine, cytosine, adenine, and uracil as a base, respectively. "T" and "dT" are used interchangeably herein and refer to a deoxyribonucleotide wherein the nucleobase is thymine, e.g., deoxyribothymine, xythymidine or thymidine. However, it will be tood that the term "ribonucleotide" or "nucleotide" or "deoxyribonucleotide" can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. The skilled person is well aware that guanine, cytosine, adenine, and uracil may be replaced by other moieties t substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or . Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of the invention by a nucleotide ning, for example, inosine.
Sequences comprising such replacement moieties are embodiments of the invention.
The terms "iRNA", "RNAi agent," "iRNA agent,", "RNA interference agent" as used interchangeably herein, refer to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript via an RNA-induced silencing complex (RISC) pathway. iRNA directs the sequence—specific degradation of mRNA through a process known as RNA interference (RNAi). The iRNA modulates, e.g., inhibits, the expression of TMPRSS6 in a cell, e.g., a cell within a subject, such as a mammalian subject.
In one embodiment, an RNAi agent of the invention includes a single stranded RNA that irficts with a target RNA sequence, e.g., a TMPRSS6 target mRNA sequence, to direct [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM the cleavage of the target RNA. Without wishing to be bound by , it is believed that long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclease known as Dicer (Sharp et al. (2001) Genes Dev. 15:485). Dicer, a ribonuclease— III—like enzyme, processes the dsRNA into 19—23 base pair short interfering RNAs with characteristic two base 3' overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporated into an RNA—induced silencing complex (RISC) where one or more ses unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the target to induce silencing (Elbashir, et al., (2001) Genes Dev. 15: 188). Thus, in one aspect the invention relates to a single stranded RNA (siRNA) generated within a cell and which promotes the formation of a RISC complex to effect silencing of the target gene, i.e., a TMPRSS6 gene.
Accordingly, the term "siRNA" is also used herein to refer to an RNAi as described above.
In another embodiment, the RNAi agent may be a single—stranded siRNA that is introduced into a cell or organism to inhibit a target mRNA. Single— stranded RNAi agents bind to the RISC endonuclease Argonaute 2, which then cleaves the target mRNA. The single—stranded siRNAs are lly 15—30 nucleotides and are chemically modified. The design and testing of single—stranded siRNAs are described in U.S. Patent No. 8,101,348 and in Lima et al., (2012) Cell 150: 883—894, the entire contents of each of which are hereby incorporated herein by reference. Any of the antisense nucleotide sequences bed herein may be used as a single— stranded siRNA as described herein or as chemically modified by the methods described in Lima er al., (2012) Cell 150;:883—894.
In yet another embodiment, the t invention provides single—stranded antisense oligonucleotide molecules targeting TMPRSS6. A "single-stranded antisense ucleotide molecule" is complementary to a sequence within the target mRNA (i.e., TMPRSS6). - ed nse oligonucleotide molecules can inhibit translation in a stoichiometric manner by base pairing to the mRNA and ally obstructing the translation machinery, see Dias, N. et al., (2002) Mol Cancer Ther 1:347—355. atively, the single— stranded antisense oligonucleotide molecules inhibit a target mRNA by hydridizing to the target and cleaving the target through an RNaseH cleavage event. The single-stranded antisense oligonucleotide molecule may be about 10 to about 30 nucleotides in length and have a ce that is complementary to a target sequence. For example, the single— stranded antisense oligonucleotide molecule may comprise a sequence that is at least about , 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more uous nucleotides from any one of the antisense nucleotide sequences described herein, e.g., the sequences provided in any one of Tables, 1, 2, 4, 5, 8, 10, and 12, or bind any of the target sites described herein. The - stranded nse oligonucleotide molecules may se modified RNA, DNA, or a combifi'on thereof.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In another embodiment, an "iRNA" for use in the compositions, uses, and methods of the invention is a double— ed RNA and is referred to herein as a "double ed RNAi agent," "double—stranded RNA (dsRNA) molecule," "dsRNA agent," or "dsRNA". The term "dsRNA", refers to a complex of ribonucleic acid molecules, having a duplex ure comprising two anti—parallel and substantially complementary nucleic acid strands, referred to as having "sense" and "antisense" orientations with respect to a target RNA, i.e., a TMPRSS6 gene. In some embodiments of the invention, a double—stranded RNA (dsRNA) triggers the degradation of a target RNA, e.g., an mRNA, through a post—transcriptional gene— silencing mechanism referred to herein as RNA interference or RNAi.
In general, the majority of tides of each strand of a dsRNA molecule are ribonucleotides, but as described in detail herein, each or both strands can also include one or more non—ribonucleotides, 6.57., a deoxyribonucleotide and/or a modified nucleotide. In on, as used in this specification, an "RNAi agent" may include ribonucleotides with chemical modifications; an RNAi agent may include substantial modifications at multiple nucleotides. Such cations may include all types of cations disclosed herein or known in the art. Any such modifications, as used in a siRNA type molecule, are encompassed by "RNAi agent" for the purposes of this specification and claims.
The two strands forming the duplex structure may be different portions of one larger RNA molecule, or they may be separate RNA molecules. Where the two s are part of one larger molecule, and therefore are ted by an uninterrupted chain of nucleotides between the 3’—end of one strand and the 5’—end of the respective other strand forming the duplex structure, the connecting RNA chain is referred to as a "hairpin loop." Where the two strands are connected covalently by means other than an uninterrupted chain of nucleotides between the 3’—end of one strand and the 5’-end of the respective other strand forming the duplex structure, the connecting structure is ed to as a "linker." The RNA s may have the same or a different number of nucleotides. The m number of base pairs is the number of tides in the shortest strand of the dsRNA minus any overhangs that are present in the . In addition to the duplex structure, an RNAi agent may comprise one or more nucleotide ngs.
In one embodiment, an RNAi agent of the invention is a dsRNA of 24—30 nucleotides that interacts with a target RNA sequence, e.g., a TMPRSS6 target mRNA sequence, to direct the cleavage of the target RNA. Without wishing to be bound by theory, long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclease known as Dicer (Sharp et a1. (2001) Genes Dev. 15:485). Dicer, a ribonuclease-III—like enzyme, processes the dsRNA into 19—23 base pair short ering RNAs with characteristic two base 3' overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporated into an duced silencing complex (RISC) where one or more helicases unwinfie siRNA duplex, enabling the complementary antisense strand to guide target [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM recognition (Nykanen, et a1., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleave the target to induce ing (Elbashir, et a1., (2001) Genes Dev. 15: 188). As used herein, a "nucleotide overhang" refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure of an RNAi agent when a 3'—end of one strand of the RNAi agent s beyond the 5'—end of the other , or vice versa. "Blunt" or "blunt end" means that there are no unpaired nucleotides at that end of the double stranded RNAi agent, i.e., no nucleotide overhang. A "blunt ended" RNAi agent is a dsRNA that is double—stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule. The RNAi agents of the invention include RNAi agents with nucleotide overhangs at one end (i.e., agents with one overhang and one blunt end) or with nucleotide ngs at both ends.
The term "antisense strand" refers to the strand of a double stranded RNAi agent which includes a region that is substantially complementary to a target sequence (e.g., a human TMPRSS6 mRNA). As used , the term n complementary to part of an mRNA encoding transthyretin" refers to a region on the antisense strand that is substantially complementary to part of a TMPRSS6 mRNA sequence. Where the region of mentarity is not fully complementary to the target sequence, the mismatches are most tolerated in the terminal regions and, if present, are lly in a terminal region or regions, e.g., within 6, 5, 4, 3, or 2 nucleotides of the 5’ and/or 3’ us.
The term "sense strand," as used herein, refers to the strand of a dsRNA that includes a region that is substantially complementary to a region of the antisense .
As used herein, the term "cleavage region" refers to a region that is located immediately adjacent to the cleavage site. The cleavage site is the site on the target at which cleavage occurs. In some embodiments, the cleavage region comprises three bases on either end of, and immediately adjacent to, the cleavage site. In some embodiments, the cleavage region comprises two bases on either end of, and immediately nt to, the cleavage site.
In some embodiments, the cleavage site specifically occurs at the site bound by nucleotides and 11 of the antisense strand, and the cleavage region comprises nucleotides 11, 12 and As used herein, and unless otherwise indicated, the term "complementary," when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or cleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can, for example, be stringent conditions, where stringent conditions may e: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50°C or 70°C for 12—16 hours followed by washing. Other ions, such as physiologically relevant condi'fis as may be encountered inside an organism, can apply. For example, a [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., RNAi. The skilled person will be able to determine the set of conditions most appropriate for a test of mentarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.
Sequences can be "fully complementary" with respect to each when there is base— g of the nucleotides of the first tide sequence with the nucleotides of the second nucleotide ce over the entire length of the first and second nucleotide sequences.
However, where a first sequence is referred to as "substantially complementary" with respect to a second sequence herein, the two sequences can be fully mentary, or they may form one or more, but generally not more than 4, 3 or 2 mismatched base pairs upon hybridization, while retaining the ability to hybridize under the conditions most relevant to their ultimate application. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such ngs shall not be regarded as mismatches with regard to the ination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as "fully complementary" for the purposes described herein.
"Complementary" sequences, as used herein, may also include, or be formed entirely 2O from, non—Watson—Crick base pairs and/or base pairs formed from non—natural and modified nucleotides, in as far as the above requirements with respect to their y to hybridize are fulfilled. Such non—Watson-Crick base pairs includes, but not limited to, G:U Wobble or Hoogstein base pairing.
The terms "complementary," "fully complementary" and "substantially complementary" herein may be used with t to the base matching between the sense strand and the antisense strand of a dsRNA, or n the antisense strand of a dsRNA and a target sequence, as will be understood from the context of their use.
As used herein, a polynucleotide that is "substantially complementary to at least part of" a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRNA encoding TMPRSS6) including a 5’ UTR, an open reading frame (ORF), or a 3’ UTR. For example, a polynucleotide is mentary to at least a part of a TMPRSS6 mRNA if the sequence is substantially complementary to a non—interrupted portion of an mRNA encoding TMPRSS6.
The term "inhibiting," as used herein, is used interchangeably with "reducing," "silencing," egulating,77 4‘suppressing" and other r terms, and includes any level of inhibition.
The phrase "inhibiting expression of a TMPRSS6," as used herein, includes inhibition of expfiion of any TMPRSS6 gene (such as, e. g., a mouse TMPRSS6 gene, a rat [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM TMPRSS6 gene, a monkey TMPRSS6 gene, or a human TMPRSS6 gene) as well as variants, (e.g., naturally occurring ts), or mutants of a TMPRSS6 gene. Thus, the TMPRSS6 gene may be a wild—type TMPRSS6 gene, a mutant 6 gene, or a enic TMPRSS6 gene in the context of a genetically manipulated cell, group of cells, or organism.
"Inhibiting expression of a 6 gene" includes any level of inhibition of a TMPRSS6 gene, e.g., at least partial ssion of the expression of a TMPRSS6 gene, such as an inhibition of at least about 5%, at least about 10%, at least about 15%, at least about %, at least about 25%, at least about 30%, at least about 35%,at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%. at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%.
The expression of a TMPRSS6 gene may be assessed based on the level of any variable associated with TMPRSS6 gene expression, e.g., TMPRSS6 mRNA level, TMPRSS6 protein level, hepcidin mRNA level, hepcidin protein level, or iron levels in tissues or serum. Inhibition may be ed by a se in an absolute or relative level of one or more of these variables compared with a control level. The control level may be any type of control level that is utilized in the art, e.g., a pre—dose baseline level, or a level determined from a similar subject, cell, or sample that is untreated or treated with a control (such as, e.g., buffer only control or inactive agent control).
The phrase "contacting a cell with a double stranded RNAi agent," as used herein, includes contacting a cell by any possible means. Contacting a cell with a double stranded RNAi agent es contacting a cell in vitra with the RNAi agent or ting a cell in viva with the RNAi agent. The contacting may be done directly or indirectly. Thus, for example, the RNAi agent may be put into physical contact with the cell by the individual performing the method, or alternatively, the RNAi agent may be put into a situation that will permit or cause it to subsequently come into contact with the cell.
Contacting a cell in vitra may be done, for example, by incubating the cell with the RNAi agent. Contacting a cell in viva may be done, for example, by injecting the RNAi agent into or near the tissue where the cell is located, or by injecting the RNAi agent into another area, the bloodstream or the subcutaneous space, such that the agent will subsequently reach the tissue where the cell to be contacted is located. For example, the RNAi agent may contain and/or be coupled to a ligand, e.g., a GalNAc3 ligand, that directs the RNAi agent to a site of interest, e.g., the liver. Combinations of in vitra and in viva s of ting are also possible. In connection with the methods of the ion, a cell might also be contacted in vitra with an RNAi agent and subsequently transplanted into a subject.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM A "patient" or "subject," as used herein, is intended to include either a human or non— human animal, preferably a , e.g., human or a monkey. Most preferably, the t or patient is a human.
A S6 associated disorder", as used herein, is intended to include any disorder that can be treated or prevented, or the symptoms of Which can be alleviated, by inhibiting the expression of TMPRSS6. In some embodiments, the TMPRSS6 associated disorder is also associated with iron ad, a condition characterized by elevated iron levels, or iron dysregulation. Iron overload may be caused, for example, by hereditary conditions, by elevated iron uptake from diet, or by excess iron administered parenterally that includes intravenous injection of excess iron, and transfusional iron overload.
TMPRSS6 ated disorders include, but are not limited to, hereditary hemochromatosis, idiopathic hemochromatosis, primary hemochromatosis, secondary hemochromatosis, severe juvenile romatosis, neonatal hemochromatosis, sideroblastic , hemolytic anemia, dyserythropoietic anemia, sickle—cell anemia, obinopathy, thalassemia (e.g., B—thalassemia and (it—thalassemia), chronic liver diseases, porphyria cutanea tarda, erythropoietic ria, atransferrinemia, hereditary tyrosinemia, cerebrohepatorenal syndrome, idiopathic pulmonary hemosiderosis, renal derosis.
TMPRSS6 associated disorders e disorders associated With oral administration 2O of excess iron, transfusional iron overload and intravenous injection of excess iron.
TMPRSS6 associated disorders also include disorders With symptoms that are associated with or may be caused by iron overload. Such symptoms include increased risk for liver disease (cirrhosis, cancer), heart attack or heart failure, diabetes mellitus, osteoarthritis, osteoporosis, metabolic syndrome, hypothyroidism, hypogonadism, and in some cases premature death. In one ment, 6 associated disorders include neurodegenerative ers associated with iron overload and/or iron dysregulation, such as Alzheimer's Disease, Parkinson's Disease, Huntington's e, Friedreich’s Ataxia, epilepsy and multiple sclerosis. Administration of an iRNA that targets TMPRSS6, e.g., an iRNA described in any one of Tables 1, 2, 4, 5, 8, 10, and 12 can treat one or more of these symptoms, or prevent the development or progression of a disease or er that is aggravated by increased iron levels.
In one embodiment, a TMPRSS6 associated disorder is a B-thalassemia. A [3— thalassemia is any one of a group of hereditary disorders characterized by a genetic deficiency in the synthesis of beta—globin chains. In the homozygous state, beta thalassemia assemia major") causes severe, transfusion—dependent . In the heterozygous state, the beta thalassemia trait ("thalassemia ) causes mild to moderate microcytic anemia. ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM "Thalassemia intermedia" is a B—thalassemia that results in subjects in whom the clinical severity of the disease is somewhere between the mild symptoms of B—thalassemia minor and the assemia major. The diagnosis is a clinical one that is based on the patient maintaining a actory hemoglobin (Hb) level of at least 6—7 g/dL at the time of diagnosis without the need for regular blood transfusions.
In one ment, a B—thalassemia is thalassemia major. In another embodiment, a B—thalassemia is thalassemia intermedia.
"Therapeutically effective amount," as used herein, is intended to include the amount of an RNAi agent that, when stered to a patient for treating a TMPRSS6 associated disease, is sufficient to effect treatment of the disease (6.5)., by diminishing, ameliorating or maintaining the existing e or one or more symptoms of disease). The "therapeutically effective amount" may vary depending on the RNAi agent, how the agent is administered, the e and its severity and the history, age, weight, family history, genetic makeup, stage of pathological processes mediated by TMPRSS6 expression, the types of ing or concomitant treatments, if any, and other individual characteristics of the patient to be treated.
"Prophylactically effective amount," as used herein, is intended to include the amount of an RNAi agent that, when administered to a subject who does not yet experience or display symptoms of a 6—associated disease, but who may be predisposed to the disease, is sufficient to prevent or ameliorate the e or one or more symptoms of the disease. Ameliorating the disease includes slowing the course of the disease or reducing the severity of later—developing e. The "prophylactically effective amount" may vary depending on the RNAi agent, how the agent is administered, the degree of risk of disease, and the history, age, weight, family history, genetic makeup, the types of preceding or concomitant treatments, if any, and other individual characteristics of the patient to be treated.
A peutically—effective amount" or "prophylacticaly effective amount" also includes an amount of an RNAi agent that produces some desired local or ic effect at a reasonable benefit/risk ratio applicable to any treatment. RNAi gents employed in the methods of the present invention may be administered in a sufficient amount to produce a reasonable benefit/risk ratio applicable to such treatment.
The term "sample," as used herein, es a tion of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject. es of biological fluids e blood, serum and serosal fluids, plasma, cerebrospinal fluid, ocular fluids, lymph, urine, saliva, and the like. Tissue samples may include samples from tissues, organs or localized regions. For example, samples may be derived from particular organs, parts of organs, or fluids or cells within those organs. In certain emborfients, samples may be derived from the liver (e.g., whole liver or certain segments of [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM liver or certain types of cells in the liver, such as, e.g., hepatocytes). In preferred embodiments, a "sample derived from a subject" refers to blood or plasma drawn from the subject. In further embodiments, a "sample derived from a subject" refers to liver tissue (or subcomponents thereof) derived from the subject. 11. iRNAs of the ion Described herein are improved double—stranded RNAi agents which inhibit the expression of a TMPRSS6 gene in a cell, such as a cell within a subject, e.g., a mammal, such as a human having a TMPRSS6 associated disorder, e.g., B—thalassemia (e.g., B—thalassemia major and assemia iedia) or hemochromatosis, and uses of such double—stranded RNAi agents.
Accordingly, the invention provides double—stranded RNAi agents with chemical modifications capable of inhibiting the expression of a target gene (i.e., a 6 gene) in vivo. In certain aspects of the ion, substantially all of the nucleotides of an iRNA of the invention are modified. In other embodiments of the invention, all of the nucleotides of an iRNA of the invention are modified. iRNAs of the invention in which "substantially all of the nucleotides are modified" are y but not wholly modified and can include not more than , 4, 3, 2, or 1 unmodified nucleotides.
The RNAi agent ses a sense strand and an antisense strand. Each strand of the 2O RNAi agent may range from 12—30 nucleotides in length. For example, each strand may be between 14—30 nucleotides in length, 17—30 nucleotides in length, 19—30 tides in length, —30 nucleotides in length, 27—30 nucleotides in length, 17-23 nucleotides in length, 17—21 nucleotides in length, l7—l9 nucleotides in length, 19—25 nucleotides in length, 19—23 nucleotides in , 19—21 tides in length, 21—25 tides in length, or 21—23 nucleotides in length.
The sense strand and antisense strand typically form a duplex double stranded RNA ("dsRNA"), also referred to herein as an "RNAi agent." The duplex region of an RNAi agent may be 12—30 nucleotide pairs in length. For example, the duplex region can be between 14— nucleotide pairs in length, 17—30 nucleotide pairs in length, 27—30 nucleotide pairs in length, 17 — 23 nucleotide pairs in length, 17-21 nucleotide pairs in length, 17—19 nucleotide pairs in length, 19—25 nucleotide pairs in length, 19—23 nucleotide pairs in length, 19— 21 nucleotide pairs in length, 21-25 tide pairs in length, or 21—23 nucleotide pairs in length. In another example, the duplex region is selected from l5, l6, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, and 27 nucleotides in length.
In one embodiment, the RNAi agent may contain one or more ng regions and/or capping groups at the 3’-end, 5’-end, or both ends of one or both s. The overhang can be l—6 nucleotides in length, for instance 2—6 nucleotides in length, l—5 nuclefis in length, 2-5 nucleotides in length, 1-4 nucleotides in , 2-4 nucleotides in [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM length, 1—3 nucleotides in length, 2—3 nucleotides in length, or 1—2 nucleotides in . The overhangs can be the result of one strand being longer than the other, or the result of two strands of the same length being staggered. The overhang can form a mismatch with the target mRNA or it can be complementary to the gene sequences being targeted or can be another sequence. The first and second strands can also be joined, e.g., by additional bases to form a hairpin, or by other non—base linkers.
In one embodiment, the nucleotides in the overhang region of the RNAi agent can each independently be a ed or unmodified nucleotide including, but no limited to 2’- sugar modified, such as, 2-F, 2’—O—methyl, thymidine (T), 2‘—O—methoxyethyl—5— methyluridine (Teo), 2‘—O—methoxyethyladenosine (Aeo), 2‘—O—methoxyethyl—5— methylcytidine (m5Ceo), and any combinations thereof. For example, TT can be an overhang sequence for either end on either . The overhang can form a mismatch with the target mRNA or it can be complementary to the gene sequences being targeted or can be another sequence.
The 5’— or 3’— overhangs at the sense strand, nse strand or both strands of the RNAi agent may be phosphorylated. In some ments, the overhang region(s) contains two nucleotides having a phosphorothioate between the two nucleotides, where the two nucleotides can be the same or different. In one embodiment, the overhang is present at the 3’—end of the sense strand, antisense strand, or both strands. In one embodiment, this 3’— overhang is present in the antisense strand. In one embodiment, this 3’—overhang is present in the sense strand.
The RNAi agent may contain only a single overhang, which can strengthen the interference activity of the RNAi, without affecting its l stability. For example, the -stranded overhang may be located at the 3'—terminal end of the sense strand or, alternatively, at the 3'—terminal end of the antisense strand. The RNAi may also have a blunt end, located at the 5’-end of the antisense strand (or the 3’—end of the sense strand) or vice versa. Generally, the antisense strand of the RNAi has a nucleotide ng at the 3’—end, and the 5’—end is blunt. While not wishing to be bound by theory, the asymmetric blunt end at the 5’—end of the antisense strand and 3’-end overhang of the antisense strand favor the guide strand loading into RISC process.
Any of the nucleic acids featured in the invention can be synthesized and/or ed by methods well established in the art, such as those described in nt protocols in nucleic acid chemistry," Beaucage, S.L. et al. ), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Modifications include, for example, end modifications, e.g., 5’—end modifications horylation, conjugation, inverted linkages) or 3’-end modifications (conjugation, DNA nucleotides, inverted linkages, etc); base modifications, e.g., replacement with stabilizing bases, ilizing bases, or basesa base pair with an ed repertoire of partners, removal of bases (abasic [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM tides), or conjugated bases; sugar modifications (e.g., at the 2’—position or 4’—position) or replacement of the sugar; and/or backbone modifications, including modification or replacement of the phosphodiester linkages. Specific examples of iRNA compounds useful in the embodiments described herein include, but are not limited to RNAs ning modified backbones or no natural intemucleoside linkages. RNAs haVing modified backbones e, among , those that do not have a orus atom in the backbone.
For the purposes of this specification, and as sometimes referenced in the art, modified RNAs that do not have a phosphorus atom in their intemucleoside backbone can also be considered to be oligonucleosides. In some embodiments, a modified iRNA will have a phosphorus atom in its intemucleoside backbone.
Modified RNA backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, otriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'—alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates ing no phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, alkylphosphotriesters, and boranophosphates having normal 3'—5' linkages, 2'—5'—linked analogs of these, and those haVing inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'—5' to 5'—3' or 2'—5' to 5'—2'. Various salts, mixed salts and free acid forms are also ed.
Representative US. patents that teach the preparation of the above phosphorus— containing es include, but are not d to, US. Patent Nos. 808; 4,469,863; 301; 5,023,243; 5,177,195; 897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; ,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 677; 5,476,925; 5,519,126; ,536,821; 5,541,316; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,625,050; 6,028,188; 6,124,445; 6,160,109; 6,169,170; 6,172,209; 6, 239,265; 603; 6,326,199; 6,346,614; 6,444,423; 6,531,590; 6,534,639; 6,608,035; 167; 6,858,715; 6,867,294; 6,878,805; 7,015,315; 7,041,816; 7,273,933; 7,321,029; and US Pat RE39464, the entire contents of each of which are hereby incorporated herein by reference.
Modified RNA backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl intemucleoside linkages, mixed atoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic intemucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others haVing mixed N, O, S and Cfiomponent parts.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM ation] KIM Unmarked set by KIM Representative US. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Patent Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; ,216,141; 5,235,033; 5,64,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; ,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 046; 5,610,289; 5,618,704; ,623,070; 5,663,312; 5,633,360; 437; and, 5,677,439, the entire contents of each of which are hereby incorporated herein by reference.
In other embodiments, suitable RNA mimetics are contemplated for use in iRNAs, in which both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are ed with novel groups. The base units are maintained for hybridization with an riate nucleic acid target compound. One such oligomeric nd, an RNA mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar backbone of an RNA is ed with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative US. patents that teach the preparation of PNA compounds include, but are not limited to, US Patent Nos. 5,539,082; 5,714,331; and 5,719,262, the entire contents of each of which are hereby incorporated herein by reference. Additional PNA compounds suitable for use in the iRNAs of the ion are described in, for example, in Nielsen et al., Science, 1991, 254, 1497—1500.
Some embodiments featured in the invention include RNAs with phosphorothioate nes and ucleosides with heteroatom backbones, and in ular ——CH2——NH—— CH2—, ——CH2--N(CH3)——O——CH2——[known as a methylene (methylimino) or MMI backbone], —— ——N(CH3)——CH2——, —-CH2——N(CH3)—-N(CH3)——CH2-- and --N(CH3)-—CH2--CH2-- [wherein the native phosphodiester backbone is represented as ——O--P——O——CH2——] of the above—referenced US. Patent No. 5,489,677, and the amide backbones of the above— referenced US. Patent No. 5,602,240. In some ments, the RNAs featured herein have morpholino backbone structures of the above—referenced US. Patent No. 5,034,506.
Modified RNAs can also contain one or more substituted sugar moieties. The iRNAs, e.g., , featured herein can include one of the following at the ition: OH; F; 0-, S—, or N—alkyl; O—, S—, or N—alkenyl; 0-, S— or N—alkynyl; or O—alkyl-O—alkyl, wherein the alkyl, alkenyl and alkynyl can be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Exemplary suitable modifications include O[(CH2)nO] InCH3, O(CH2).HOCH3, O(CH2)HNH2, O(CH2) I1CH3, O(CH2)HONH2, and O(CH2)HON[(CH2)HCH3)]2, where n and m are from 1 to about 10. In other embodiments, dsRNAs include one of the following at the 2' position: C1 to C10 lower alkyl, substituted lower alkyl, l, aralkyl, O— alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, $02CH3, ONOZ, N02, N3, NHZ, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substifid silyl, an RNA cleaving group, a reporter group, an intercalator, a group for ation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ing the pharmacokinetic properties of an iRNA, or a group for improving the pharmacodynamic properties of an iRNA, and other substituents having similar properties. In some embodiments, the modification includes a 2'-methoxyethoxy (2'—O——CH2CHZOCH3, also known as 2'—O—(2—methoxyethyl) or 2'—MOE) (Martin et al., Helv. Chim. Acta, 1995, 78:486- 504) i.e., an alkoxy—alkoxy group. Another exemplary modification is 2'— dimethylaminooxyethoxy, i.e., a O(CH2)20N(CH3)2 group, also known as 2'—DMAOE, as described in examples herein below, and 2'—dimethylaminoethoxyethoxy (also known in the art as 2'—O—dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'—O—-CH2——O——CH2——N(CH2)2.
Other modifications include 2'—methoxy (2'—OCH3), 2'—aminopropoxy (2'— OCHzCH2CH2NH2) and 2'—fluoro (2'—F). Similar modifications can also be made at other positions on the RNA of an iRNA, ularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'—5' linked dsRNAs and the 5' position of 5' terminal nucleotide. iRNAs can also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
Representative US. patents that teach the preparation of such modified sugar structures include, but are not limited to, US. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; ,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; ,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and ,700,920, certain of which are commonly owned with the t application,. The entire contents of each of the foregoing are hereby incorporated herein by reference.
An iRNA can also include nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used , "unmodified" or "natural" nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and l nucleobases such as deoxy—thymine (dT), 5—methylcytosine C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2—aminoadenine, 6—methy1 and other alkyl derivatives of adenine and guanine, 2—propyl and other alkyl tives of adenine and guanine, 2—thiouracil, 2— thiothymine and 2—thiocytosine, uracil and cytosine, 5—propynyl uracil and cytosine, 6— azo , cytosine and thymine, 5—uracil (pseudouracil), 4-thiouracil, 8—halo, o, 8— thiol, 8—thioalkyl, 8-hydroxyl anal other 8—substituted adenines and guanines, 5—halo, particularly 5-bromo, 5—trifluoromethyl and other 5-substituted uracils and cytosines, 7— methylguanine and 7—methyladenine, 8—azaguanine and 8—azaadenine, 7—deazaguanine and 7— daazaadenine and 3—deazaguanine and 3—deazaadenine. Further nucleobases include those disclosed in US. Pat. No. 3,687,808, those disclosed in Modified Nucleosides in Biochemistry, hnology and Medicine, Herdewijn, P. ed. Wiley—VCH, 2008; those disclosed in The Concise Encyclopedia Of Polymer Science And ering, pages 858- 859, witz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch er al., Angewandte Chemie, ational Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chgr 15, dsRNA Research and ations, pages 289-302, , S. T. and Lebleu, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM B., Ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds featured in the invention. These include 5— substituted pyrimidines, 6—azapyrimidines and N—2, N—6 and 0—6 substituted purines, ing 2—aminopropyladenine, 5—propynyluracil and 5—propynylcytosine. 5—methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 2°C (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276—278) and are exemplary base substitutions, even more particularly when combined with 2'-O—methoxyethyl sugar modifications.
Representative US. patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted US. Patent Nos. 3,687,808, 4,845,205; 5,130,30; 5,134,066; 5,175,273; ,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; ,587,469; 121, 5,596,091; 5,614,617; 5,681,941; 5,750,692; 6,015,886; 6,147,200; 6,166,197; 6,222,025; 6,235,887; 368; 6,528,640; 6,639,062; 6,617,438; 7,045,610; 7,427,672; and 7,495,088, the entire contents of each of which are hereby incorporated herein by nce.
The RNA of an iRNA can also be modified to include one or more locked nucleic acids (LNA). A locked nucleic acid is a nucleotide having a modified ribose moiety in which the ribose moiety comprises an extra bridge ting the 2' and 4' carbons. This structure effectively "locks" the ribose in the 3'—endo structural conformation. The addition of locked nucleic acids to siRNAs has been shown to se siRNA stability in serum, and to reduce off—target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(1):439—447; Mook, OR. et al., (2007) Mol Canc Ther 6(3):833—843; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3l85—3193). entative US. s that teach the preparation of locked nucleic acid nucleotides include, but are not limited to, the following: US. Patent Nos. 490; 6,670,461; 6,794,499; 6,998,484; 7,053,207; 7,084,125; and 7,399,845, the entire contents of each of which are hereby incorporated herein by reference.
Potentially izing cations to the ends of RNA molecules can include N— laminocaproyl)-4—hydroxyprolinol (Hyp—C6—NHAc), N—(caproyl—4—hydroxyprolinol (Hyp—C6), N—(acetyl—4—hydroxyprolinol (Hyp—NHAc), thymidine—2'—0—deoxythymidine (ether), N—(aminocaproyl)—4—hydroxyprolinol 6—amino), 2—docosanoyl—uridine—3"— ate, inverted base dT(idT) and others. Disclosure of this modification can be found in PCT ation No. .
A. ed iRNAs Comprising Motifs of the Invention In certain aspects of the invention, the double-stranded RNAi agents of the invention include agents with chemical modifications as disclosed, for example, in US. Provisional Applifi'on No. 61/561,710, filed on November 18, 2011, or in PCT/U82012/065691, filed [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM ed set by KIM on November 16, 2012, the entire contents of each of which are incorporated herein by reference.
As shown herein and in Provisional Application No. 61/561,710, a superior result may be obtained by introducing one or more motifs of three identical modifications on three consecutive nucleotides into a sense strand and/or antisense strand of a RNAi agent, ularly at or near the cleavage site. In some embodiments, the sense strand and antisense strand of the RNAi agent may otherwise be completely modified. The introduction of these motifs interrupts the modification pattern, if present, of the sense and/or antisense strand.
The RNAi agent may be optionally ated with a GalNAc derivative ligand, for instance on the sense strand. The resulting RNAi agents present superior gene silencing activity.
More specifically, it has been singly ered that when the sense strand and antisense strand of the double—stranded RNAi agent are modified to have one or more motifs of three identical modifications on three consecutive nucleotides at or near the cleavage site of at least one strand of an RNAi agent, the gene silencing acitivity of the RNAi agent was superiorly enhanced.
In one embodiment, the RNAi agent is a double ended bluntmer of 19 nucleotides in length, wherein the sense strand ns at least one motif of three 2’—F modifications on three consecutive nucleotides at positions 7, 8, 9 from the 5’end. The antisense strand contains at least one motif of three ethyl modifications on three consecutive 2O nucleotides at positions 11, 12, 13 from the 5’end.
In another embodiment, the RNAi agent is a double ended bluntmer of 20 nucleotides in length, wherein the sense strand contains at least one motif of three 2’—F cations on three consecutive nucleotides at positions 8, 9, 10 from the 5’end. The antisense strand ns at least one motif of three 2’—O—methyl modifications on three utive nucleotides at positions 11, 12, 13 from the 5’end.
In yet another embodiment, the RNAi agent is a double ended bluntmer of 21 nucleotides in length, wherein the sense strand contains at least one motif of three 2’—F modifications on three consecutive nucleotides at ons 9, 10, 11 from the 5’end. The antisense strand contains at least one motif of three 2’—O—methyl modifications on three consecutive nucleotides at positions 11, 12, 13 from the 5’end.
In one embodiment, the RNAi agent comprises a 21 nucleotide sense strand and a 23 nucleotide antisense strand, wherein the sense strand contains at least one motif of three 2’-F modifications on three consecutive nucleotides at positions 9, 10, 11 from the 5’end; the antisense strand contains at least one motif of three 2’—O—methy1 modifications on three consecutive nucleotides at positions 11, 12, 13 from the 5’end, wherein one end of the RNAi agent is blunt, while the other end comprises a 2 nucleotide overhang. Preferably, the 2 nucleotide overhang is at the 3’—end of the antisense . When the 2 nucleotide overhang is at tlfi’-end of the nse strand, there may be two orothioate internucleotide [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM linkages between the terminal three nucleotides, wherein two of the three tides are the overhang nucleotides, and the third nucleotide is a paired nucleotide next to the overhang nucleotide. In one embodiment, the RNAi agent additionally has two phosphorothioate intemucleotide linkages between the terminal three nucleotides at both the 5’—end of the sense strand and at the 5’—end of the antisense strand. In one embodiment, every nucleotide in the sense strand and the antisense strand of the RNAi agent, including the nucleotides that are part of the motifs are modified tides. In one embodiment each residue is independently modified with a 2’—O-methyl or 3’—fluoro, e.g., in an alternating motif.
Optionally, the RNAi agent further comprises a ligand (preferably GalNAC3).
In one embodiment, the RNAi agent comprises sense and antisense strands, wherein the RNAi agent comprises a first strand having a length which is at least 25 and at most 29 nucleotides and a second strand having a length which is at most 30 nucleotides with at least one motif of three 2’—O—methyl modifications on three consecutive tides at position ll, l2, 13 from the 5’ end; n the 3’ end of the first strand and the 5’ end of the second strand form a blunt end and the second strand is 1—4 tides longer at its 3’ end than the first strand, wherein the duplex region region which is at least 25 tides in length, and the second strand is sufficiently complemenatary to a target mRNA along at least 19 tide of the second strand length to reduce target gene sion when the RNAi agent is introduced into a mammalian cell, and wherein dicer cleavage of the RNAi agent 2O preferentially results in an siRNA comprising the 3’ end of the second strand, thereby reducing expression of the target gene in the mammal. Optionally, the RNAi agent further comprises a ligand.
In one embodiment, the sense strand of the RNAi agent contains at least one motif of three identical modifications on three consecutive nucleotides, where one of the motifs occurs at the cleavage site in the sense strand.
In one embodiment, the antisense strand of the RNAi agent can also contain at least one motif of three identical modifications on three consecutive tides, where one of the motifs occurs at or near the cleavage site in the antisense strand For an RNAi agent having a duplex region of 17—23 nucleotide in length, the cleavage site of the antisense strand is typically around the 10, 11 and 12 positions from the 5’—end.
Thus the motifs of three identical modifications may occur at the 9, 10, ll positions; 10, ll, 12 positions; ll, l2, 13 positions; l2, l3, 14 ons; or l3, 14, 15 positions of the antisense strand, the count starting from the lSt nucleotide from the 5’—end of the nse strand, or, the count starting from the 1St paired nucleotide within the duplex region from the 5’— end of the antisense strand. The cleavage site in the antisense strand may also change according to the length of the duplex region of the RNAi from the .
The sense strand of the RNAi agent may contain at least one motif of three identical modiffiions on three consecutive nucleotides at the cleavage site of the strand; and the [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM antisense strand may have at least one motif of three identical cations on three consecutive nucleotides at or near the cleavage site of the strand. When the sense strand and the antisense strand form a dsRNA duplex, the sense strand and the antisense strand can be so aligned that one motif of the three nucleotides on the sense strand and one motif of the three nucleotides on the antisense strand have at least one tide overlap, i.e., at least one of the three nucleotides of the motif in the sense strand forms a base pair with at least one of the three nucleotides of the motif in the antisense strand. Alternatively, at least two nucleotides may overlap, or all three nucleotides may p.
In one embodiment, the sense strand of the RNAi agent may contain more than one motif of three identical modifications on three consecutive tides. The first motif may occur at or near the cleavage site of the strand and the other motifs may be a wing cation. The term "wing modification" herein refers to a motif occurring at another portion of the strand that is separated from the motif at or near the cleavage site of the same strand. The wing modification is either adajacent to the first motif or is separated by at least one or more nucleotides. When the motifs are immediately adjacent to each other then the try of the motifs are distinct from each other and when the motifs are separated by one or more nucleotide than the chemistries can be the same or different. Two or more wing modifications may be present. For ce, when two wing modifications are present, each wing modification may occur at one end relative to the first motif which is at or near cleavage site or on either side of the lead motif.
Like the sense strand, the antisense strand of the RNAi agent may n more than one motifs of three cal modifications on three consecutive nucleotides, with at least one of the motifs occurring at or near the cleavage site of the strand. This antisense strand may also contain one or more wing modifications in an alignment similar to the wing modifications that may be present on the sense strand.
In one embodiment, the wing modification on the sense strand or antisense strand of the RNAi agent typically does not include the first one or two terminal nucleotides at the 3’— end, 5’—end or both ends of the strand.
In another embodiment, the wing modification on the sense strand or antisense strand of the RNAi agent typically does not include the first one or two paired nucleotides within the duplex region at the 3’—end, 5’—end or both ends of the strand.
When the sense strand and the antisense strand of the RNAi agent each contain at least one wing modification, the wing modifications may fall on the same end of the duplex region, and have an overlap of one, two or three nucleotides.
When the sense strand and the antisense strand of the RNAi agent each contain at least two wing modifications, the sense strand and the antisense strand can be so aligned that two modifications each from one strand fall on one end of the duplex region, having an overlaEf one, two or three nucleotides; two modifications each from one strand fall on the [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM other end of the duplex region, having an overlap of one, two or three nucleotides; two modifications one strand fall on each side of the lead motif, having an overlap of one, two or three nucleotides in the duplex region.
In one embodiment, every nucleotide in the sense strand and antisense strand of the RNAi agent, including the nucleotides that are part of the motifs, may be modified. Each nucleotide may be ed with the same or different modification which can include one or more alteration of one or both of the non—linking phosphate oxygens and/or of one or more of the linking phosphate oxygens; alteration of a tuent of the ribose sugar, e.g., of the 2’ hydroxyl on the ribose sugar; wholesale replacement of the phosphate moiety with "dephospho" linkers; modification or replacement of a lly occurring base; and replacement or modification of the ribose—phosphate backbone.
As nucleic acids are polymers of subunits, many of the cations occur at a position which is repeated within a nucleic acid, e.g., a modification of a base, or a phosphate moiety, or a non—linking O of a ate moiety. In some cases the modification will occur at all of the subject positions in the nucleic acid but in many cases it will not. By way of example, a modification may only occur at a 3’ or 5’ al position, may only occur in a terminal region, 6.57., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand. A modification may occur in a double strand , a single strand , or in both. A modification may occur only in the double strand region of a RNA or 2O may only occur in a single strand region of a RNA. For example, a orothioate modification at a non—linking 0 position may only occur at one or both termini, may only occur in a terminal region, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand, or may occur in double strand and single strand regions, particularly at termini. The 5’ end or ends can be phosphorylated.
It may be possible, e.g., to enhance stability, to include particular bases in overhangs, or to include modified nucleotides or nucleotide surrogates, in single strand overhangs, e.g., in a 5’ or 3’ overhang, or in both. For example, it can be desirable to include purine tides in overhangs. In some embodiments all or some of the bases in a 3’ or 5’ overhang may be modified, e.g., with a modification described herein. Modifications can include, e.g., the use of modifications at the 2’ position of the ribose sugar with modifications that are known in the art, e.g., the use of deoxyribonucleotides, or , xy—2’—fluoro (2’—F) 2’—O-methyl modified instead of the ribosugar of the nucleobase and modifications in the phosphate group, e.g., phosphorothioate cations. Overhangs need not be homologous with the target sequence.
In one embodiment, each residue of the sense strand and antisense strand is independently modified with LNA, HNA, CeNA, 2’-methoxyethyl, 2’- O-methyl, 2’-O-allyl, 2’—C— allyl, 2’—deoxy, 2’—hydroxyl, or 2’—fluoro. The strands can contain more than one [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM modification. In one embodiment, each e of the sense strand and antisense strand is independently modified with 2’— O—methyl or 2’—fluoro.
At least two different modifications are typically present on the sense strand and antisense . Those two modifications may be the 2’— O—methyl or 2’—fluoro modifications, or others.
In one embodiment, the Na and/or Nb comprise modifications of an alternating pattern.
The term "alternating motif’ as used herein refers to a motif having one or more modifications, each cation occurring on alternating nucleotides of one strand. The alternating tide may refer to one per every other nucleotide or one per every three nucleotides, or a similar pattern. For example, if A, B and C each represent one type of modification to the nucleotide, the alternating motif can be BABABAB. . .," "AABBAABBAABB. . .," "AABAABAABAAB. . .," "AAABAAABAAAB. . .," "AAABBBAAABBB. . .," or "ABCABCABCABC. . .," etc.
The type of modifications contained in the alternating motif may be the same or different. For example, if A, B, C, D each represent one type of modification on the nucleotide, the ating pattern, i.6., modifications on every other nucleotide, may be the same, but each of the sense strand or antisense strand can be selected from several ilities of modifications within the alternating motif such as "ABABAB. . .", "ACACAC..." "BDBDBD..." or D. . .," etc.
In one embodiment, the RNAi agent of the invention comprises the cation pattern for the alternating motif on the sense strand relative to the cation pattern for the alternating motif on the antisense strand is shifted. The shift may be such that the modified group of nucleotides of the sense strand corresponds to a differently ed group of nucleotides of the antisense strand and vice versa. For example, the sense strand when paired with the nse strand in the dsRNA duplex, the alternating motif in the sense strand may start with "ABABAB" from 5’—3’ of the strand and the alternating motif in the antisense strand may start with "BABABA" from 5’—3’of the strand within the duplex region. As another example, the alternating motif in the sense strand may start with "AABBAABB" from 5’—3’ of the strand and the alternating motif in the antisenese strand may start with "BBAABBAA" from 5’—3’ of the strand within the duplex , so that there is a complete or partial shift of the modification patterns between the sense strand and the antisense strand.
In one embodiment, the RNAi agent comprises the pattern of the alternating motif of 2'—O—methyl modification and 2’—F modification on the sense strand initially has a shift relative to the pattern of the alternating motif of 2'—O-methyl modification and 2’—F modification on the antisense strand initially, i.e., the 2'—O—methyl modified nucleotide on the sense strand base pairs with a 2'-F modified nucleotide on the antisense strand and vice versa.
The 1 position of the sense strand may start with the 2'—F modification, and the 1 position of the an'finse strand may start with the 2'- O-methyl modification.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM The introduction of one or more motifs of three identical cations on three consecutive nucleotides to the sense strand and/or antisense strand interrupts the initial modification pattern present in the sense strand and/or nse strand. This interruption of the modification pattern of the sense and/or antisense strand by introducing one or more motifs of three identical modifications on three consecutive nucleotides to the sense and/or nse strand surprisingly es the gene silencing acitivty to the target gene.
In one embodiment, when the motif of three cal cations on three consecutive nucleotides is introduced to any of the strands, the modification of the nucleotide next to the motif is a different modification than the modification of the motif. For example, the portion of the ce containing the motif is . .NaYYYNb. . .," where "Y" represents the cation of the motif of three identical modifications on three utive nucleotide, and "Na" and "Nb" represent a modification to the nucleotide next to the motif "YYY" that is different than the modification of Y, and where Na and Nb can be the same or different modifications. Altnernatively, Na and/or Nb may be present or absent when there is a wing modification present.
The RNAi agent may further comprise at least one phosphorothioate or methylphosphonate internucleotide linkage. The phosphorothioate or methylphosphonate ucleotide linkage modification may occur on any nucleotide of the sense strand or antisense strand or both strands in any position of the strand. For instance, the internucleotide linkage modification may occur on every nucleotide on the sense strand and/or antisense strand; each internucleotide linkage modification may occur in an alternating pattern on the sense strand and/or antisense strand; or the sense strand or antisense strand may n both internucleotide linkage modifications in an alternating pattern. The ating pattern of the internucleotide linkage modification on the sense strand may be the same or different from the antisense strand, and the alternating pattern of the internucleotide linkage cation on the sense strand may have a shift relative to the alternating n of the internucleotide linkage modification on the antisense strand.
In one embodiment, the RNAi comprises a phosphorothioate or methylphosphonate internucleotide linkage modification in the overhang region. For example, the overhang region may contain two nucleotides having a phosphorothioate or methylphosphonate internucleotide linkage between the two nucleotides. Intemucleotide e modifications also may be made to link the overhang nucleotides with the terminal paired nucleotides within the duplex region. For example, at least 2, 3, 4, or all the overhang nucleotides may be linked through phosphorothioate or methylphosphonate internucleotide linkage, and ally, there may be onal phosphorothioate or methylphosphonate internucleotide linkages linking the overhang nucleotide with a paired nucleotide that is next to the overhang nucleotide. For instance, there may be at least two phosphorothioate internucleotide linkages betwefihe terminal three nucleotides, in which two of the three nucleotides are overhang [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM nucleotides, and the third is a paired nucleotide next to the overhang nucleotide. These terminal three nucleotides may be at the 3’—end of the antisense strand, the 3’—end of the sense strand, the 5’—end of the antisense strand, and/or the 5’end of the nse strand.
In one embodiment, the 2 nucleotide overhang is at the 3’—end of the antisense strand, and there are two phosphorothioate internucleotide linkages n the terminal three nucleotides, wherein two of the three nucleotides are the overhang nucleotides, and the third nucleotide is a paired nucleotide next to the overhang tide. Optionally, the RNAi agent may additionally have two phosphorothioate internucleotide linkages between the terminal three nucleotides at both the 5’—end of the sense strand and at the 5’—end of the nse .
In one embodiment, the RNAi agent comprises mismatch(es) with the target, within the duplex, or combinations thereof. The mistmatch may occur in the overhang region or the duplex region. The base pair may be ranked on the basis of their propensity to promote dissociation or melting (6.57., on the free energy of association or dissociation of a particular pairing, the st approach is to examine the pairs on an individual pair basis, though next or or similar analysis can also be used). In terms of promoting dissociation: A:U is preferred over G:C; G:U is preferred over G:C; and LC is preferred over G:C sine). ches, e.g., non—canonical or other than canonical pairings (as described ere herein) are preferred over canonical (A:T, A:U, G:C) pairings; and pairings which include a universal base are preferred over canonical pairings.
In one embodiment, the RNAi agent comprises at least one of the first 1, 2, 3, 4, or 5 base pairs within the duplex regions from the 5’— end of the antisense strand independently selected from the group of: A:U, G:U, I:C, and mismatched pairs, e.g., nonical or other than canonical pairings or pairings which include a universal base, to promote the dissociation of the antisense strand at the 5’—end of the duplex.
In one embodiment, the nucleotide at the 1 position within the duplex region from the ’—end in the antisense strand is selected from the group consisting of A, dA, dU, U, and dT.
Alternatively, at least one of the first 1, 2 or 3 base pair within the duplex region from the 5’— end of the antisense strand is an AU base pair. For example, the first base pair within the duplex region from the 5’— end of the antisense strand is an AU base pair.
In one embodiment, the sense strand sequence may be represented by a (I): ' np—Na-(X X X )i—Nb-Y Y Y -Nb—(Z Z Z )j—Na-nq 3' (I) wherein: i and j are each ndently 0 or 1; p and q are each independently 0—6; each Na independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence sing at least two differently modified nuclefis; ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM each Nb independently ents an oligonucleotide sequence comprising 0—10 modified nucleotides; each np and nq independently represent an overhang nucleotide; wherein Nb and Y do not have the same modification; and XXX, YYY and ZZZ each independently represent one motif of three identical modifications on three consecutive nucleotides. Preferably YYY is all 2’—F modified nucleotides.
In one embodiment, the Na and/or Nb comprise cations of alternating pattern.
In one embodiment, the YYY motif occurs at or near the cleavage site of the sense . For example, when the RNAi agent has a duplex region of 17—23 nucleotides in length, the YYY motif can occur at or the vicinity of the cleavage site (e.g.: can occur at positions 6, 7, 8, 7, 8, 9, 8, 9, 10, 9, 10, 11, 10, 11,12 or 11, 12, 13) of — the sense strand, the count starting from the 1St nucleotide, from the 5’—end; or ally, the count ng at the 1St paired nucleotide within the duplex region, from the 5’— end.
In one embodiment, i is 1 and j is 0, or i is 0 and j is 1, or both i and j are l. The sense strand can therefore be ented by the following formulas: ' np—Na-YYY-Nb-ZZZ-Na-nq 3' (Ib); ' np—Na—XXX—Nb—YYY—Na—nq 3' (lo); or ' np—Na-XXX-Nb-YYY-Nb—ZZZ-Na-nq 3' (Id). 2O When the sense strand is represented by formula (Ib), Nb ents an oligonucleotide sequence comprising 0—10, 0—7, 0—5, 0—4, 0—2 or 0 ed nucleotides. Each Na independently can represent an oligonucleotide sequence comprising 2—20, 2—15, or 2— 10 modified nucleotides.
When the sense strand is represented as formula (Ic), Nb represents an oligonucleotide sequence comprising 0—10, 0—7, 0—10, 0—7, 0—5, 0—4, 0—2 or 0 modified nucleotides. Each Na can independently represent an oligonucleotide sequence comprising 2—20, 2—15, or 2—10 modified nucleotides.
When the sense strand is represented as formula (Id), each Nb independently represents an oligonucleotide sequence comprising 0—10, 0—7, 0—5, 0—4, 0—2 or 0 modified nucleotides. ably, Nb is 0, 1, 2, 3, 4, 5 or 6 Each Na can independently represent an oligonucleotide sequence comprising 2—20, 2—15, or 2—10 modified nucleotides.
Each of X, Y and Z may be the same or different from each other.
In other embodiments, i is 0 and j is 0, and the sense strand may be represented by the formula: 5' YYY— Na—nq 3' (Ia).
When the sense strand is represented by formula (Ia), each Na independently can represent an oligonucleotide sequence comprising 2—20, 2—15, or 2—10 modified nucleotides.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In one ment, the antisense strand sequence of the RNAi may be represented by formula (II): ' nq~—Na’-(Z’Z’Z’)k-Nb’-Y’Y'Y’-Nb'-(X’X’X')1-N’a-np’ 3' (II) k and l are each independently 0 or 1; p’ and q’ are each independently 0—6; each Na’ independently represents an oligonucleotide sequence comprising 0—25 modified nucleotides, each sequence sing at least two differently modified tides; each Nb’ independently represents an oligonucleotide sequence comprising 0— 10 modified nucleotides; each np’ and nq’ independently represent an overhang nucleotide; wherein Nb’ and Y’ do not have the same modification; X’X'X’, Y’Y'Y’ and Z’Z’Z’ each independently represent one motif of three identical cations on three consecutive nucleotides.
In one ment, the Na’ and/or Nb’ se modifications of alternating pattern.
The Y’Y'Y' motif occurs at or near the cleavage site of the antisense strand. For example, when the RNAi agent has a duplex region of ucleotidein , the Y’Y'Y’ 2O motif can occur at positions 9, 10, 11;10, 11, 12; 11, 12, 13; 12, 13, 14 ; or 13, 14, 15 of the antisense strand, with the count starting from the 1St nucleotide, from the 5’—end; or optionally, the count starting at the 1St paired nucleotide within the duplex region, from the ’— end. Preferably, the Y’Y’Y’ motif occurs at positions 11, 12, 13.
In one embodiment, Y’Y'Y' motif is all 2’—OMe modified nucleotides.
In one embodiment, k is 1 and l is 0, or k is 0 and l is 1, or both k and l are 1.
The antisense strand can therefore be represented by the following formulas: ' nq,-Na’-Z’Z’Z’-Nb’—Y’Y’Y'—Na’-np~ 3' (IIb); ' nq’-Na'—Y'Y'Y'—Nb'-X'X'X'-nps 3' (KC); or ' nq’-Na’- Z'Z’Z’-Nb'-Y’Y’Y’-Nb’- X’X’X’-Na’-np~ 3' (11d).
When the antisense strand is represented by formula (IIb), Nb, represents an oligonucleotide sequence comprising 0—10, 0—7, 0—10, 0—7, 0—5, 0—4, 0—2 or 0 modified nucleotides. Each Na’ independently represents an oligonucleotide sequence comprising 2- , 2—15, or 2—10 modified nucleotides.
When the antisense strand is represented as formula (IIc), Nb’ ents an oligonucleotide sequence comprising 0—10, 0—7, 0—10, 0—7, 0—5, 0—4, 0—2 or 0 modified nucleotides. Each Na’ independently represents an oligonucleotide sequence comprising 2- , 2—15, or 2—10 modified nucleotides.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM When the antisense strand is represented as formula (IId), each Nb’ independently represents an ucleotide sequence comprising 0—10, 0-7, 0—10, 0—7, 0—5, 0—4, 0—2 or 0 modified nucleotides. Each Na’ independently represents an oligonucleotide sequence comprising 2—20, 2—15, or 2-10 modified nucleotides. Preferably, Nb is 0, l, 2, 3, 4, 5 or 6.
In other embodiments, k is 0 and l is 0 and the nse strand may be represented by the formula: ' np’—Na~-Y’Y’Y’- Naa-nq~ 3' (la).
When the antisense strand is represented as formula (Ila), each Na’ independently represents an oligonucleotide sequence comprising 2—20, 2—15, or 2—10 modified nucleotides.
Each of X', Y’ and Z' may be the same or different from each other.
Each nucleotide of the sense strand and antisense strand may be independently ed with LNA, HNA, CeNA, 2’-methoxyethyl, 2’-O—methyl, 2’-O-allyl, 2’—C- allyl, 2’- yl, or 2’—fluoro. For example, each nucleotide of the sense strand and antisense strand is independently ed with 2’—O—methyl or 2’—fluoro. Each X, Y, Z, X', Y’ and Z’, in particular, may represent a 2’—O—methyl modification or a 2’—fluoro modification.
In one embodiment, the sense strand of the RNAi agent may contain YYY motif occurring at 9, 10 and ll ons of the strand when the duplex region is 21 nt, the count starting from the ISI nucleotide from the 5’—end, or optionally, the count starting at the lSt paired nucleotide within the duplex region, from the 5’— end; and Y ents 2’—F 2O cation. The sense strand may additionally contain XXX motif or ZZZ motifs as wing modifications at the opposite end of the duplex region; and XXX and ZZZ each independently represents a 2’—OMe modification or 2’—F modification.
In one embodiment the antisense strand may contain Y’Y’Y’ motif occurring at positions ll, l2, 13 of the strand, the count starting from the lSt nucleotide from the 5’—end, or optionally, the count starting at the lSt paired nucleotide within the duplex region, from the ’— end; and Y’ represents 2’-O—methyl cation. The antisense strand may additionally contain X’X'X’ motif or Z’Z’Z’ motifs as wing modifications at the opposite end of the duplex region; and X'X'X' and Z'Z'Z’ each independently represents a 2’—OMe modification or 2’-F modification.
The sense strand represented by any one of the above formulas (Ia), (Ib), (Ic), and (Id) forms a duplex with a antisense strand being ented by any one of formulas (IIa), (IIb), (11c), and (11d), respectively.
Accordingly, the RNAi agents for use in the methods of the ion may comprise a sense strand and an antisense strand, each strand having 14 to 30 nucleotides, the RNAi duplex represented by formula (III): sense: 5' np -Na-(X X X)i -Nb- Y Y Y -Nb -(Z Z Z)J~-Na-nq 3' antisense: 3' npi—Na’—(X’X’X’)k—Nb’—Y’Y’Y’—Nb’-(Z’Z’Z’)1—Na’—nq, 5' (III) ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM wherein: i, j, k, and l are each independently 0 or 1; p, p', q, and q’ are each independently 0—6; each Na and Na, independently represents an oligonucleotide ce comprising 0— modified nucleotides, each sequence comprising at least two differently modified nucleotides; each Nb and Nb, independently represents an oligonucleotide sequence comprising 0— modified nucleotides; wherein each np’, np, nq’, and nq, each of which may or may not be present, independently represents an overhang tide; and XXX, YYY, ZZZ, X’X’X', , and Z'Z'Z’ each independently represent one motif of three identical modifications on three consecutive nucleotides.
In one embodiment, i is 0 andj is 0; or i is l andj is 0; or i is 0 andj is l; or both i and j are 0; or both i andj are 1. In another embodiment, k is 0 and l is 0; or k is l and l is 0; k is 0 and l is l; or both k and l are 0; or both k andl are l.
Exemplary combinations of the sense strand and antisense strand g a RNAi duplex include the formulas below: ' np — Na —Y Y Y —Na—nq 3' 3' npi-Nai—Y'Y’Y’ —Na‘nq’ 5' (Illa) ' np-Na-Y Y Y -Nb-Z Z Z -Na-nq 3' 3' np,—Na'—Y'Y’Y’—Nb5—Z’Z’Z'—Nagnqg 5' (IIIb) 5' np—Na- X X X -Nb -Y Y Y - Na-nq 3' 3' npi-Nai—X’X’X'—Nb’-Y'Y'Y'-Na'—nqi 5' (IIIc) ' np -Na -X X X -Nb-Y Y Y -Nb- Z Z Z -Na—nq 3' 3' np,—Na'—X'X’X’—Nb,—Y’Y’Y’—Nb'—Z'Z’Z’—Na—nq' 5' (IIId) When the RNAi agent is represented by formula (IIIa), each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 ed nucleotides.
When the RNAi agent is represented by formula (IIIb), each Nb independently represents an oligonucleotide sequence comprising 1-10, 1-7, 1—5 or 1—4 modified nucleotides. Each Na independently represents an oligonucleotide sequence comprising 2—20, 2-15, or 2- 10 modified tides.
When the RNAi agent is represented as formula , each Nb, Nb’ independently fi an oligonucleotide sequence comprising 0-10, 0-7, 0-10, 0-7, 0-5, 0-4, 0-2 or [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 0modified tides. Each Na independently represents an oligonucleotide sequence comprising 2—20, 2—15, or 2-10 ed nucleotides.
When the RNAi agent is represented as formula (Illd), each Nb, Nb’ independently represents an oligonucleotide sequence comprising O—lO, 0-7, 0—10, 0—7, 0—5, 0—4, 0—2 or 0modified nucleotides. Each Na, Na, independently represents an oligonucleotide sequence comprising 2—20, 2—15, or 2-10 modified nucleotides. Each of Na, Na’, Nb and Nb, independently comprises modifications of alternating pattern.
Each of X, Y and Z in formulas (III), (Illa), (IIIb), (IIIc), and (IIId) may be the same or different from each other.
When the RNAi agent is represented by formula (III), (Illa), (IIIb), (IIIc), and (Illd), at least one of the Y nucleotides may form a base pair with one of the Y’ nucleotides.
Alternatively, at least two of the Y nucleotides form base pairs with the corresponding Y’ nucleotides; or all three of the Y nucleotides all form base pairs with the corresponding Y' tides.
When the RNAi agent is represented by formula (IIIb) or (IIId), at least one of the Z nucleotides may form a base pair with one of the Z' nucleotides. Alternatively, at least two of the Z nucleotides form base pairs with the corresponding Z’ nucleotides; or all three of the Z nucleotides all form base pairs with the ponding Z' nucleotides.
When the RNAi agent is ented as formula (IIIc) or (IIId), at least one of the X nucleotides may form a base pair with one of the X' nucleotides. Alternatively, at least two of the X nucleotides form base pairs with the corresponding X’ nucleotides; or all three of the X nucleotides all form base pairs with the corresponding X' nucleotides.
In one embodiment, the modification on the Y tide is different than the modification on the Y’ nucleotide, the modification on the Z nucleotide is different than the modification on the Z’ nucleotide, and/or the modification on the X nucleotide is different than the cation on the X’ nucleotide.
In one embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2’-O—methyl or ro modifications. In another embodiment, when the RNAi agent is represented by formula (Illd), the Na modifications are 2’—O—methyl or 2’— fluoro modifications and np' >0 and at least one np' is linked to a neighboring nucleotide a via phosphorothioate linkage. In yet another embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2’-O—methyl or ro ations >0 and , np' at least one np’ is linked to a neighboring tide via phosphorothioate e, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker. In another embodiment, when the RNAi agent is ented by a (IIId), the Na modifications are ethyl or 2’-fluoro modifications >0 and at , np' least one np' is linked to a neighboring nucleotide via phosphorothioate linkage, the sense ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM strand comprises at least one phosphorothioate linkage, and the sense strand is ated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.
In one embodiment, when the RNAi agent is ented by formula (Illa), the Na modifications are 2’-O—methyl or 2’-fluoro cations >0 and at least one np’ is linked , np’ to a oring nucleotide via phosphorothioate linkage, the sense strand ses at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives ed through a bivalent or trivalent branched .
In one embodiment, the RNAi agent is a multimer containing at least two duplexes represented by formula (III), (Illa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non—cleavable. ally, the multimer further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.
In one embodiment, the RNAi agent is a multimer containing three, four, five, six or more duplexes represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non—cleavable.
Optionally, the er further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.
In one embodiment, two RNAi agents ented by formula (III), (IIIa), , (1110), and (IIId) are linked to each other at the 5’ end, and one or both of the 3’ ends and are optionally conjugated to to a ligand. Each of the agents can target the same gene or two different genes; or each of the agents can target same gene at two ent target sites.
Various publications describe multimeric RNAi agents that can be used in the methods of the invention. Such publications include W02007/091269, US Patent N0. 7858769, W02010/141511, W02007/ll7686,W02009/014887 and WOZOl 1/031520 the entire contents of each of which are hereby incorporated herein by reference.
The RNAi agent that contains ations of one or more ydrate moieties to a RNAi agent can optimize one or more properties of the RNAi agent. In many cases, the carbohydrate moiety will be attached to a modified subunit of the RNAi agent. For example, the ribose sugar of one or more ribonucleotide subunits of a dsRNA agent can be replaced with r moiety, 6.57., a non—carbohydrate (preferably cyclic) carrier to which is attached a ydrate ligand. A ribonucleotide subunit in which the ribose sugar of the subunit has been so replaced is referred to herein as a ribose replacement modification subunit (RRMS).
A cyclic carrier may be a carbocyclic ring system, 226., all ring atoms are carbon atoms, or a heterocyclic ring system, i.e., one or more ring atoms may be a heteroatom, e.g., nitrogen, oxygen, sulfur. The cyclic carrier may be a monocyclic ring system, or may contain two or more rings, 6.g. fused rings. The cyclic carrier may be a fully saturated ring system, or it may contaifie or more double bonds.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM The ligand may be ed to the polynucleotide via a carrier. The carriers include (i) at least one one ment point," preferably two "backbone ment points" and (ii) at least one "tethering attachment point." A "backbone attachment point" as used herein refers to a functional group, 6. g. a hydroxyl group, or generally, a bond available for, and that is suitable for incorporation of the carrier into the backbone, e.g., the phosphate, or modified phosphate, e. g., sulfur containing, backbone, of a ribonucleic acid. A "tethering attachment point" (TAP) in some embodiments refers to a constituent ring atom of the cyclic carrier, e.g., a carbon atom or a heteroatom (distinct from an atom which provides a backbone attachment point), that connects a selected moiety. The moiety can be, e.g., a carbohydrate, e. g. monosaccharide, disaccharide, trisaccharide, tetrasaccharide, accharide and polysaccharide. Optionally, the selected moiety is connected by an intervening tether to the cyclic carrier. Thus, the cyclic carrier will often include a functional group, e.g., an amino group, or generally, provide a bond, that is suitable for incorporation or tethering of another chemical entity, 6.57., a ligand to the constituent ring.
The RNAi agents may be conjugated to a ligand via a carrier, wherein the carrier can be cyclic group or acyclic group; preferably, the cyclic group is selected from pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, [1,3]dioxolane, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, zinonyl, tetrahydrofuryl and and n; preferably, the acyclic group is selected from serinol backbone or diethanolamine ne.
In certain specific embodiments, the RNAi agent for use in the methods of the invention is an agent selected from the group of agents listed in any one of Tables 1, 2, 4, 5, 8, 10, and 12. In one embodiment, when the agent is an agent listed in Table 12, the agent may lack a terminal dT.
The present invention further es —stranded RNAi agents comprising any one of the sequences listed in any one of Tables 1, 2, 4, 5, 8, 10, and 12 which comprise a 5’ phosphate or phosphate mimetic on the antisense strand (see, e.g., PCT Publication No. WO 2011005860). Further, the present invention includes double—stranded RNAi agents comprising any one of the sequences listed in any one of Tables 1, 2, 4, 5, 8, 10, and 12 which include a 2’fluoro group in place of a 2’—OMe group at the S’end of the sense .
These agents may further comprise a ligand.
In one embodiment, the agent is 40 (sense strand: CfsusGfgUfanuUfoCqungngfaCfaAfL96; antisense strand: usUfngfaCchqufggaAfanaCchfgsasg).
A. Ligands The double-stranded RNA (dsRNA) agents of the invention may optionally be conjugated to one or more ligands. The ligand can be attached to the sense strand, antisense strandfiboth s, at the 3’-end, 5’-end or both ends. For instance, the ligand may be [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM ated to the sense strand. In preferred embodiments, the ligand is conjgated to the 3’— end of the sense strand. In one preferred embodiment, the ligand is a GalNAc ligand. In particularly preferred embodiments, the ligand is GalNAC3: O H H HO O ACHN O\/\/\n/N\/\/N OH : HO o H H AcHN \/\/\n/ \/\/ \H/\/0%" O O O Ho O\/\/\n/N/\/\N o AcHN H H In some embodiments, the ligand, e.g., GalNAc ligand, is attached to the 3’ end of the RNAi agent. In one embodiment, the RNAi agent is conjugated to the ligand, e.g., GalNAc ligand, as shown in the ing schematic HO%&o N N O AcHN WEI—IMH wherein X is O or S. In one embodiment, X is O.
A wide variety of entities can be coupled to the RNAi agents of the present invention.
Preferred moieties are ligands, which are coupled, preferably covalently, either directly or indirectly via an intervening .
In preferred ments, a ligand alters the bution, targeting or lifetime of the molecule into which it is incorporated. In preferred embodiments a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, compartment, receptor e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand. Ligands providing enhanced affinity for a selected target are also termed targeting ligands.
Some ligands can have endosomolytic properties. The endosomolytic ligands promote the lysis of the endosome and/or transport of the ition of the invention, or its components, from the endosome to the cytoplasm of the cell. The molytic ligand may be a pEmionic peptide or omimetic which shows pH—dependent membrane activity [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM and fusogenicity. In one embodiment, the molytic ligand assumes its active conformation at endosomal pH. The e" conformation is that conformation in which the endosomolytic ligand promotes lysis of the endosome and/or transport of the composition of the invention, or its components, from the endosome to the asm of the cell. Exemplary endosomolytic ligands include the GALA e (Subbarao et al., Biochemistry, 1987, 26: 2964-2972), the EALA peptide (Vogel et al., J. Am. Chem. 500., 1996, 118: 1581—1586), and their derivatives (Turk et al., Biochem. Biophys. Acta, 2002, 1559: 56-68). In one ment, the endosomolytic component may contain a chemical group (e.g., an amino acid) Which will undergo a change in charge or protonation in response to a change in pH.
The endosomolytic component may be linear or branched.
Ligands can improve transport, hybridization, and specificity properties and may also improve se resistance of the resultant natural or modified oligoribonucleotide, or a polymeric molecule comprising any combination of monomers described herein and/or natural or modified ribonucleotides.
Ligands in general can include therapeutic ers, 6.57., for enhancing uptake; diagnostic compounds or reporter groups e.g., for monitoring distribution; cross—linking agents; and nuclease—resistance conferring moieties. General examples include lipids, steroids, ns, sugars, proteins, peptides, polyamines, and peptide mimics.
Ligands can include a naturally occurring nce, such as a n (e.g., human 2O serum albumin (HSA), low—density lipoprotein (LDL), high-density lipoprotein (HDL), or globulin); a carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or onic acid); or a lipid. The ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid, an oligonucleotide (e.g., an aptamer). Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L—aspartic acid, poly L—glutamic acid, styrene—maleic acid anhydride copolymer, poly(L— lactide—co—glycolied) copolymer, divinyl ether—maleic anhydride copolymer, N—(2— hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), nyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine. e of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide—polyamine, omimetic polyamine, dendrimer polyamine, arginine, amidine, ine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide. s can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell. A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N- acetyl—galactosamine, N—acetyl—gulucosamine multivalent mannose, multivalent fucose, glycofited polyaminoacids, multivalent galactose, transferrin, bisphosphonate, ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM utamate, polyaspartate, a lipid, cholesterol, a d, bile acid, folate, vitamin B12, biotin, an RGD e, an RGD peptide mimetic or an aptamer.
Other examples of ligands e 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 or a chelator (e.g., EDTA), lipophilic molecules, e.g., cholesterol, cholic acid, tane acetic acid, l—pyrene butyric acid, dihydrotestosterone, l,3—Bis— O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, bomeol, menthol, 1,3— propanediol, heptadecyl group, palmitic acid, myristic acid,O3—(oleoyl)lithocholic acid, 03— (oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine)and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g., biotin), transport/absorption facilitators (e.g., aspirin, n E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine—imidazole conjugates, Eu3+ complexes of tetraazamacrocycles), dinitrophenyl, HRP, or AP.
Ligands can be 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. Ligands may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, yl— osamine, N—acetyl—gulucosamine multivalent mannose, multivalent fucose, or aptamers.
The ligand can be, for example, a lysaccharide, an activator of p38 MAP kinase, or an tor of NF—KB.
The ligand can be a nce, 6.57., a drug, Which can increase the uptake of the iRNA agent into the cell, for example, by disrupting the cell’s cytoskeleton, 6.5)., by disrupting the cell’s microtubules, microfilaments, and/or intermediate filaments. The drug can be, for example, taxon, vincristine, stine, cytochalasin, nocodazole, inolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.
The ligand can increase the uptake of the ucleotide into the cell by, for example, activating an inflammatory response. Exemplary ligands that would have such an effect include tumor necrosis factor alpha (TNFalpha), interleukin—1 beta, or gamma interferon.
In one aspect, the ligand is a lipid or lipid—based molecule. Such a lipid or lipid— based molecule preferably binds a serum protein, e.g., human serum albumin (HSA). An HSA binding ligand allows for bution of the conjugate to a target , e.g., a non- kidney target tissue of the body. For example, the target tissue can be the liver, including parenfinal cells of the liver. Other molecules that can bind HSA can also be used as ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM ligands. For example, naproxen or aspirin can be used. A lipid or lipid—based ligand can (a) increase ance to degradation of the conjugate, (b) se targeting or transport into a target cell or cell membrane, and/or (c) can be used to adjust binding to a serum protein, 6.5)., HSA.
A lipid based ligand can be used to modulate, e.g., control the binding of the conjugate to a target tissue. For example, a lipid or lipid—based ligand that binds to HSA more strongly will be less likely to be targeted to the kidney and therefore less likely to be cleared from the body. A lipid or lipid—based ligand that binds to HSA less strongly can be used to target the conjugate to the kidney.
In a preferred embodiment, the lipid based ligand binds HSA. Preferably, it binds HSA with a sufficient affinity such that the conjugate will be preferably distributed to a non— kidney tissue. However, it is preferred that the affinity not be so strong that the HSA—ligand binding cannot be reversed.
In another preferred ment, the lipid based ligand binds HSA weakly or not at all, such that the conjugate will be preferably distributed to the kidney. Other es that target to kidney cells can also be used in place of or in addition to the lipid based ligand.
In another , the ligand is a , e.g. a vitamin, which is taken up by a target cell, e.g., a proliferating cell. These are particularly useful for treating disorders characterized by unwanted cell proliferation, 6.57., of the malignant or non—malignant type, 2O e.g., cancer cells. Exemplary Vitamins include vitamin A, E, and K. Other ary vitamins include B vitamins, e.g., folic acid, B12, riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by cancer cells. Also included are HAS, low density lipoprotein (LDL) and ensity lipoprotein (HDL).
In another aspect, the ligand is a cell-permeation agent, preferably a helical cell— permeation agent. ably, the agent is amphipathic. An exemplary agent is a peptide such as tat or antennopedia. If the agent is a peptide, it can be modified, ing a peptidylmimetic, invertomers, non—peptide or pseudo—peptide linkages, and use of D—amino acids. The helical agent is preferably an alpha—helical agent, which preferably has a lipophilic and a lipophobic phase.
The ligand can be a peptide or peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptidomimetic) is a molecule capable of folding into a defined three— dimensional structure similar to a natural peptide. The e or peptidomimetic moiety can be about 5—50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long. A peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting ily of Tyr, Trp or Phe). The peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide. In another alternative, the peptide moiety can include a hydrophobic membfi translocation sequence (MTS). An exemplary hydrophobic MTS-containing [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP (SEQ ID NO: 11). An RFGF analogue (e.g., amino acid ce AALLPVLLAAP (SEQ ID NO: 12)) containing a hydrophobic MTS can also be a ing moiety. The peptide moiety can be a "delivery" peptide, which can carry large polar molecules including peptides, oligonucleotides, and protein across cell membranes. For example, sequences from the HIV Tat protein (GRKKRRQRRRPPQ) (SEQ ID NO: 13) and the Drosophila Antennapedia protein (RQIKIWFQNRRMKWKK) (SEQ ID NO: 14) have been found to be capable of functioning as delivery peptides. A peptide or peptidomimetic can be encoded by a random sequence of DNA, such as a peptide identified from a phage—display library, or one—bead—one— compound (OBOC) combinatorial library (Lam et al., Nature, 354:82—84, 1991). Preferably the peptide or peptidomimetic tethered to an iRNA agent via an incorporated monomer unit is a cell targeting peptide such as an arginine—glycine—aspartic acid (RGD)—peptide, or RGD mimic. A peptide moiety can range in length from about 5 amino acids to about 40 amino acids. The peptide es can have a structural modification, such as to increase stability or direct conformational properties. Any of the structural cations described below can be utilized.An RGD peptide moiety can be used to target a tumor cell, such as an endothelial tumor cell or a breast cancer tumor cell ann et al. Cancer Res. , , 9—43, 2002).
An RGD peptide can tate targeting of an iRNA agent to tumors of a variety of other tissues, including the lung, kidney, spleen, or liver (Aoki et al., Cancer Gene y 8:783— 787, 2001). Preferably, the RGD peptide will facilitate ing of an iRNA agent to the kidney. The RGD peptide can be linear or , and can be modified, e.g. , glycosylated or methylated to facilitate targeting to specific tissues. For example, a glycosylated RGD peptide can deliver an iRNA agent to a tumor cell expressing (xVB3 (Haubner et al., Jonr.
Nucl. Med., 42:326—336, 2001). Peptides that target markers enriched in proliferating cells can be used. For example, RGD ning peptides and peptidomimetics can target cancer cells, in particular cells that exhibit an integrin. Thus, one could use RGD peptides, cyclic peptides containing RGD, RGD peptides that include D—amino acids, as well as synthetic RGD . In addition to RGD, one can use other moieties that target the integrin ligand.
Generally, such ligands can be used to control erating cells and angiogeneis. Preferred conjugates of this type of ligand target PECAM—l, VEGF, or other cancer gene, e.g., a cancer gene described herein.
A "cell permeation peptide" is capable of permeating a cell, e.g., a microbial cell, such as a bacterial or fungal cell, or a mammalian cell, such as a human cell. A microbial cell—permeating peptide can be, for example, an a—helical linear e (e.g., LL—37 or Ceropin P1), a disulfide bond—containing e (e.g., or —defensin, B—defensin or ecin), or a peptide containing only one or two dominating amino acids (e.g., PR-39 or indolicidin).
A cell permeation peptide can also include a nuclear localization signal (NLS). For example, a cell fineation peptide can be a bipartite amphipathic peptide, such as MPG, which is ation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM derived from the fusion peptide domain of HIV—1 gp41 and the NLS of SV40 large T antigen (Simeoni et al., Nucl. Acids Res. 31:2717—2724, 2003).
In one embodiment, a targeting peptide can be an athic (x—helical peptide.
Exemplary amphipathic a—helical peptides include, but are not limited to, cecropins, lycotoxins, paradaxins, buforin, CPF, bombinin—like peptide (BLP), icidins, ceratotoxins, S. clava peptides, hagfish intestinal antimicrobial peptides (HFIAPs), magainines, brevinins—2, dermaseptins, melittins, pleurocidin, HzA peptides, Xenopus peptides, esculentinis—l, and caerins. A number of factors will ably be considered to maintain the integrity of helix stability. For example, a maximum number of helix stabilization residues will be ed (e.g., leu, ala, or lys), and a minimum number helix destabilization residues will be utilized (e.g., proline, or cyclic monomeric units. The g residue will be considered (for example Gly is an exemplary N—capping residue and/or C—terminal amidation can be used to provide an extra H—bond to stabilize the helix.
Formation of salt bridges between residues with opposite charges, separated by i i 3, or i i 4 ons can provide stability. For example, cationic es such as lysine, arginine, homo—arginine, omithine or histidine can form salt bridges with the anionic residues glutamate or aspartate.
Peptide and peptidomimetic ligands include those having naturally occurring or modified peptides, e.g., D or L peptides; (x, B, or y peptides; N—methyl es; azapeptides; peptides having one or more amide, i.e., peptide, linkages replaced with one or more urea, thiourea, carbamate, or sulfonyl urea linkages; or cyclic peptides.
The targeting ligand can be any ligand that is e of targeting a specific receptor.
Examples are: folate, , galactose, mannose, mannose—6P, clusters of sugars such as GalNAc cluster, mannose cluster, galactose cluster, or an er. A cluster is a combination of two or more sugar units. The targeting ligands also include integrin receptor ligands, Chemokine receptor ligands, transferrin, biotin, serotonin receptor s, PSMA, endothelin, GCPII, somatostatin, LDL and HDL ligands. The s can also be based on nucleic acid, 6.g. an aptamer. The aptamer can be unmodified or have any combination of modifications disclosed herein.
Endosomal release agents include imidazoles, poly or oligoimidazoles, PEIs, es, fusogenic peptides, polycaboxylates, polyacations, masked oligo or poly cations or anions, s, polyacetals, /polyketyals, orthoesters, polymers with masked or unmasked ic or anionic charges, dendrimers with masked or unmasked cationic or anionic charges.
PK modulator stands for pharmacokinetic modulator. PK modulators include iles, bile acids, steroids, phospholipid analogues, peptides, protein binding agents, PEG, vitamins etc. Examplary PK modulators e, but are not limited to, cholesterol, fatty afi, cholic acid, lithocholic acid, dialkylglycerides, diacylglyceride, phospholipids, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM ed set by KIM sphingolipids, naproxen, ibuprofen, vitamin E, biotin etc. Oligonucleotides that comprise a number of phosphorothioate linkages are also known to bind to serum protein, thus short oligonucleotides, e.g., oligonucleotides of about 5 bases, 10 bases, 15 bases or 20 bases, comprising multiple phosphorothioate es in the backbaone are also amenable to the present invention as ligands (e.g., as PK modulating ligands).
In addition, aptamers that bind serum components (e.g., serum proteins) are also amenable to the present invention as PK modulating ligands.
Other ligand conjugates le to the invention are described in US. Patent Applications USSN: 10/916,185, filed August 10, 2004; USSN: 10/946,873, filed September 21, 2004; USSN: 10/833,934, filed August 3, 2007; USSN: 11/115,989 filed April 27, 2005 and USSN: 11/944,227 filed November 21, 2007, which are incorporated by reference in their entireties for all purposes.
When two or more s are present, the ligands can all have same properties, all have different properties or some ligands have the same ties while others have ent properties. For example, a ligand can have targeting properties, have endosomolytic activity or have PK modulating properties. In a preferred embodiment, all the ligands have different properties.
Ligands can be coupled to the ucleotides at various places, for example, 3’—end, ’—end, and/or at an internal position. In preferred embodiments, the ligand is attached to the oligonucleotides via an intervening tether, e.g., a carrier described herein. The ligand or tethered ligand may be present on a monomer when the monomer is incorporated into the growing strand. In some embodiments, the ligand may be incorporated via coupling to a rsor" monomer after the "precursor" monomer has been incorporated into the growing . For e, a monomer having, e.g., an amino—terminated tether (i.e., having no associated ligand), e.g., TAP—(CH2)HNH2 may be incorporated into a growing oligonucelotide strand. In a subsequent ion, i.e., after incorporation of the sor monomer into the strand, a ligand having an electrophilic group, 6.57., a pentafluorophenyl ester or aldehyde group, can subsequently be attached to the precursor monomer by coupling the electrophilic group of the ligand with the terminal nucleophilic group of the precursor monomer’ s tether.
In another example, a monomer having a chemical group suitable for taking part in Click Chemistry reaction may be incorporated, e.g., an azide or alkyne terminated tether/linker. In a subsequent operation, i.e., after incorporation of the precursor monomer into the strand, a ligand having complementary al group, e.g. an alkyne or azide can be attached to the precursor monomer by ng the alkyne and the azide together.
For double— stranded oligonucleotides, ligands can be ed to one or both strands.
In some embodiments, a double-stranded iRNA agent contains a ligand conjugated to the sense strand. In other embodiments, a double—stranded iRNA agent contains a ligand conjufid to the nse strand.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In some embodiments, ligand can be conjugated to nucleobases, sugar moieties, or intemucleosidic linkages of nucleic acid molecules. ation to purine nucleobases or derivatives f can occur at any on including, endocyclic and exocyclic atoms. In some embodiments, the 2—, 6—, 7—, or 8—positions of a purine nucleobase are attached to a conjugate moiety. Conjugation to dine nucleobases or derivatives thereof can also occur at any position. In some embodiments, the 2—, 5—, and 6—positions of a pyrimidine nucleobase can be substituted With a ate moiety. Conjugation to sugar moieties of nucleosides can occur at any carbon atom. Example carbon atoms of a sugar moiety that can be attached to a conjugate moiety include the 2', 3', and 5' carbon atoms. The 1' position can also be attached to a conjugate moiety, such as in an abasic residue. Intemucleosidic linkages can also bear conjugate moieties. For phosphorus—containing linkages (e.g., phosphodiester, phosphorothioate, phosphorodithiotate, phosphoroamidate, and the like), the conjugate moiety can be attached directly to the phosphorus atom or to an O, N, or S atom bound to the phosphorus atom. For amine- or amide—containing intemucleosidic linkages (e.g., PNA), the conjugate moiety can be ed to the nitrogen atom of the amine or amide or to an adjacent carbon atom.
Any suitable ligand in the field of RNA interference may be used, although the ligand is typically a carbohydrate e. g. ccharide (such as GalNAc), disaccharide, trisaccharide, tetrasaccharide, polysaccharide. 2O Linkers that conjugate the ligand to the nucleic acid include those discussed above.
For example, the ligand can be one or more GalNAc (N—acetylglucosamine) derivatives attached through a bivalent or trivalent branched linker.
In one embodiment, the dsRNA of the invention is ated to a bivalent and trivalent branched linkers include the structures shown in any of formula (IV) — (VII): A—RZA, T2A_L2A 2A JP3A_Q3A_R3A| T3A_L3A q 3A P23-Q23-REB LIT-I—25-L28 \fP3B-Q3B-R3Bl_ T3B_L33 (1313 Formula (IV) Formula (V) P5A_Q5A_R5A I T5A_L5A A_R4A L4A qSA q PSB_Q513_R513 j—qSBT53_L53 P4B-Q4B-R4B jiT4B-L4B 4B PSC-Qso-Rso], T50_L50 q so Formula (VI) Formula (VII) wherein: [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM qZA, qZB, q3A, q3B, q4A, q4B, qSA, q513 and q5C represent independently for each occurrence 0—20 and wherein the repeating unit can be the same or different; PZA’ PZB, P3A’ P313, P4A’ P43, PSA, P53, Psc’ TZA’ TZB, T3A, T33, T4A’ T43, T4A’ TSB, Tsc are each independently for each occurrence absent, CO, NH, O, S, OC(O), NHC(O), CH2, CHzNH or CHZO; QZA, QZB, Q3A, Q3B, Q4A, Q43, QSA, Q53, Q5C are independently for each occurrence absent, ne, substituted alkylene wherin one or more methylenes can be interrupted or terminated by one or more of O, S, S(O), S02, N(RN), C(R’)=C(R"), CEC or C(O); RZA, RZB, R3A, R313, R", R43, RSA, RSB, R5C are each independently for each occurrence absent, NH, O, S, CH2, C(O)O, C(O)NH, NHCH(Ra)C(O), CH(Ra)-NH-, CO, CH=N-08, r":5, VW‘"N>< v" WW,S—WSWorheterocyclyl; LZA, L23, L3A, L3B, L4A, L4B, LSA, L513 and L5C represent the ligand; Le. each independently for each occurrence a monosaccharide (such as GalNAc), disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide; and Ra is H or amino acid side chain.
Trivalent conjugating GalNAc derivatives are particularly useful for use with RNAi agents for ting the expression of a target gene, such as those of formula (VII): PSA_QSA_RSA iSA T5A_L5A PSB-QSB-RSB i—SBT5B_LSB C_RSC ]?T5C_L5C a (VII) wherein LSA, L513 and L5C represent a monosaccharide, such as GalNAc derivative.
Examples of suitable bivalent and trivalent branched linker groups conjugating GalNAc derivatives include, but are not limited to, the following compounds: HOmofl AcHN O\/\/\[Ol/N\/\/N [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM O\/\O/\/O\/\N HO HOO H HOHofim‘' O\/\O/\/O\/\N,‘(\/O\gNW HO HO H O O HO ’0 O\/\O/\/O\/\H O HO HO O\/\O/\/O\/\N HO HOO H HOHfilfi- O\/\O/\/O\/\N,(\/O HO HO H O O 0%HO K O\/\O/\/O\/\N O HO HO E g O o\/\o O NHA HO \/o c NHAc OH \ HO NM OH HO O¢\O/\/0 "0&‘kflof NHAc NHAc HO OH HO OH HO OWYN O\/\/\__O HO OH NHAc HO OH NHAcW0 NHAcHO O OH HO O\/\/\n/NH Hoooé; NHAc O NHAc [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM o 0 AcHN H ,or HO OH o H HO N Tr AcHN H o HO OH flox/jk H HO HmN O ACHN xnz HO OH o O H O OWLNWJL HO N O AcHN H In other embodiments, the RNAi agent for use in the methods of the invention is AD— 59743. 111. Delivery of an iRNA of the Invention The delivery of an iRNA agent of the invention to a cell e.g., a cell Within a subject, such as a human subject (e.g., a subject in need thereof, such as a subject having a TMPRSS6 associated er, such as a hemochromatosis) can be achieved in a number of different ways. For example, delivery may be performed by ting a cell with an iRNA of the invention either in vitra or in viva. In viva delivery may also be performed directly by administering a composition sing an iRNA, e.g., a dsRNA, to a subject. Alternatively, in viva delivery may be performed indirectly by administering one or more s that encode and direct the expression of the iRNA. These alternatives are discussed further belowE [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM In general, any method of delivering a nucleic acid molecule (in vitra or in viva) can be adapted for use with an iRNA of the invention (see e.g., Akhtar S. and Julian RL. (1992) Trends Cell. Biol. 2(5): 139—144 and WO94/02595, which are incorporated herein by reference in their entireties). For in viva delivery, factors to consider in order to deliver an iRNA molecule include, for example, biological stability of the delivered molecule, prevention of non—specific s, and accumulation of the delivered molecule in the target tissue. The non—specific effects of an iRNA can be zed by local administration, for example, by direct injection or implantation into a tissue or topically administering the preparation. Local stration to a treatment site maximizes local concentration of the agent, limits the re of the agent to systemic tissues that can otherwise be harmed by the agent or that can degrade the agent, and permits a lower total dose of the iRNA molecule to be administered. Several studies have shown successful knockdown of gene ts when an iRNA is administered locally. For example, intraocular delivery of a VEGF dsRNA by intravitreal injection in cynomolgus s (Tolentino, MJ., et al (2004) Retina 24: 132— 138) and inal injections in mice (Reich, 8]., et al (2003) Mal. Vis. 9:210—216) were both shown to prevent neovascularization in an experimental model of age-related macular ration. In addition, direct intratumoral injection of a dsRNA in mice reduces tumor volume (Pille, J et al (2005) Mal. Ther.1 1:267—274) and can prolong survival of tumor— bearing mice (Kim, WJ., et al (2006) Mal. Ther. 142343—350; Li, S., et al (2007) Mal. Ther. 15:515—523). RNA interference has also shown s with local delivery to the CNS by direct injection (Dom, G., et al. (2004) Nucleic Acids 32:e49; Tan, PH., et al (2005) Gene Ther. 12:59—66; Makimura, H., et al (2002) BMC Neurasci. 3:18; Shishkina, GT., et al (2004) Neuroscience 129:521—528; Thakker, ER., et al (2004) Prac. Natl. Acad. Sci. USA. 101:17270—17275; Akaneya,Y., et al (2005) J. hysial. 93:594—602) and to the lungs by intranasal administration (Howard, KA., et al (2006) Mal. Ther. 14:476—484; Zhang, X., et al (2004) J. Biol. Chem. 279:10677—10684; Bitko, V., et al (2005) Nat. Med. 11:50—55). For stering an iRNA systemically for the treatment of a disease, the RNA can be modified or alternatively delivered using a drug delivery system; both methods act to prevent the rapid degradation of the dsRNA by endo— and exo—nucleases in viva. Modification of the RNA or the pharmaceutical carrier can also permit targeting of the iRNA composition to the target tissue and avoid undesirable off—target effects. iRNA molecules can be ed by al conjugation to lipophilic groups such as cholesterol to enhance cellular uptake and prevent degradation. For example, an iRNA directed against ApoB conjugated to a lipophilic cholesterol moiety was injected ically into mice and resulted in knockdown of apoB mRNA in both the liver and jejunum (Soutschek, J et al (2004) Nature 432: 173—178).
Conjugation of an iRNA to an aptamer has been shown to inhibit tumor growth and mediate tumor regression in a mouse model of prostate cancer (McNamara, J0., et al (2006) Nat.
Biatefil. 24:1005-1015). In an alternative embodiment, the iRNA can be delivered using [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM drug delivery systems such as a rticle, a dendrimer, a polymer, liposomes, or a cationic delivery system. Positively charged cationic delivery systems facilitate binding of an iRNA molecule (negatively charged) and also enhance interactions at the negatively charged cell membrane to permit efficient uptake of an iRNA by the cell. Cationic , dendrimers, or polymers can either be bound to an iRNA, or induced to form a vesicle or e (see e.g.
Kim SH., et al (2008) Journal of lled Release 129(2): 107—1 16) that encases an iRNA.
The formation of es or es further ts degradation of the iRNA when administered systemically. Methods for making and administering cationic— iRNA complexes are well within the abilities of one skilled in the art (see e.g., Sorensen, DR., et al (2003) J. Mol. Biol 327:761-766; Verma, UN., et al (2003) Clin. Cancer Res. —1300; Arnold, AS et al (2007) J. Hypertens. 25: 197—205, which are incorporated herein by reference in their entirety). Some non—limiting es of drug delivery systems useful for systemic delivery of iRNAs include DOTAP (Sorensen, DR., et al (2003), supra; Verma, UN., et al (2003), supra), Oligofectamine, "solid nucleic acid lipid particles" (Zimmermann, TS., et al (2006) Nature 441:111—114), cardiolipin (Chien, PY., et al (2005) Cancer Gene Ther. -328; Pal, A., et al (2005) Int J. Oncol. 26:1087—1091), polyethyleneimine (Bonnet ME., et al (2008) Pharm. Res. Aug 16 Epub ahead of print; Aigner, A. (2006) J.
Biomed. Biotechnol. , Arg—Gly—Asp (RGD) peptides (Liu, S. (2006) Mol. Pharm. 3:472—487), and polyamidoamines (Tomalia, DA., et al (2007) Biochem. Soc. Trans. 35:61— 67; Yoo, H., et al (1999) Pharm. Res. 16:1799—1804). In some embodiments, an iRNA forms a complex with cyclodextrin for ic administration. Methods for administration and pharmaceutical compositions of iRNAs and cyclodextrins can be found in US. Patent No. 7,427,605, which is herein incorporated by reference in its entirety.
A. Vector encoded iRNAs of the Invention iRNA targeting the TMPRSS6 gene can be expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., T10. (1996), 12:5—10; Skillern, A., et al., International PCT Publication No. WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, and Conrad, US. Pat. No. 6,054,299). sion can be transient (on the order of hours to weeks) or sustained (weeks to months or longer), ing upon the specific construct used and the target tissue or cell type. These transgenes can be introduced as a linear construct, a circular d, or a viral vector, which can be an integrating or non— ating vector. The transgene can also be ucted to permit it to be inherited as an hromosomal plasmid (Gassmann, et al., Proc. Natl. Acad. Sci. USA (1995) 92:1292).
The individual strand or strands of an iRNA can be transcribed from a promoter on an expression vector. Where two separate strands are to be expressed to generate, for example, a dsRNA, two separate expression vectors can be co-introduced (e.g., by transfection or infection) into a target cell. Alternatively each individual strand of a dsRNA can be transcfid by promoters both of which are located on the same expression plasmid. In one [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM embodiment, a dsRNA is expressed as inverted repeat cleotides joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure. iRNA expression vectors are generally DNA plasmids or viral vectors. Expression vectors compatible with eukaryotic cells, preferably those compatible with vertebrate cells, can be used to produce recombinant constructs for the expression of an iRNA as described . Eukaryotic cell expression vectors are well known in the art and are available from a number of commercial sources. lly, such vectors are provided containing convenient ction sites for insertion of the desired nucleic acid segment. Delivery of iRNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex—planted from the patient followed by reintroduction into the patient, or by any other means that allows for introduction into a desired target cell. iRNA expression plasmids can be transfected into target cells as a x with cationic lipid carriers (e. g., Oligofectamine) or non—cationic lipid—based rs (e. g., Transit— TKOTM). Multiple lipid transfections for iRNA—mediated knockdowns targeting different regions of a target RNA over a period of a week or more are also contemplated by the ion. Successful introduction of vectors into host cells can be monitored using various known methods. For example, transient ection can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP). Stable ection of cells ex vivo can be ensured using markers that provide the transfected cell with resistance to specific environmental factors (e. g., otics and drugs), such as hygromycin B resistance.
Viral vector systems which can be utilized with the methods and itions described herein include, but are not limited to, (a) adenovirus vectors; (b) retrovirus vectors, including but not limited to lentiviral vectors, moloney murine leukemia virus, eta; (c) adeno- associated virus vectors; (d) herpes simplex virus vectors; (e) SV 40 vectors; (f) polyoma virus vectors; (g) papilloma virus vectors; (h) picomavirus vectors; (i) pox virus vectors such as an orthopox, e.g., vaccinia virus vectors or avipox, e. g. canary pox or fowl pox; and (j) a helper—dependent or gutless adenovirus. Replication—defective viruses can also be advantageous. Different vectors will or will not become incorporated into the cells’ genome. The constructs can include viral sequences for transfection, if desired.
Alternatively, the construct can be incorporated into s capable of episomal replication, e. g. EPV and EBV s. Constructs for the recombinant expression of an iRNA will generally require regulatory elements, e.g., promoters, enhancers, etc., to ensure the expression of the iRNA in target cells. Other aspects to er for vectors and constructs are further described below.
Vectors useful for the delivery of an iRNA will include regulatory elements (promoter, er, etc.) sufficient for expression of the iRNA in the desired target cell or tissue. The regulatory elements can be chosen to provide either constitutive or regulafi/inducible expression.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Expression of the iRNA can be precisely regulated, for example, by using an inducible regulatory ce that is sensitive to certain logical regulators, e.g., circulating glucose levels, or hormones (Docherty et al., 1994, FASEB J. 8:20—24). Such ble expression systems, suitable for the control of dsRNA expression in cells or in mammals e, for example, regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of zation, and isopropyl—beta-Dl — thiogalactopyranoside (IPTG). A person skilled in the art would be able to choose the appropriate regulatory/promoter sequence based on the intended use of the iRNA transgene.
Viral vectors that contain nucleic acid sequences encoding an iRNA can be used. For example, a retroviral vector can be used (see Miller et al. , Meth. Enzymol. 217:581—599 (1993)). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA. The nucleic acid sequences encoding an iRNA are cloned into one or more vectors, which facilitate delivery of the nucleic acid into a patient. More detail about retroviral vectors can be found, for example, in Boesen et al., Biotherapy 6:291—302 (1994), which describes the use of a retroviral vector to deliver the mdrl gene to poietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene y are: Clowes et al., J. Clin. Invest. 93:644—651 (1994); Kiem et al., Blood 83: 1467— 1473 (1994); Salmons and rg, Human Gene Therapy 4:129—141 (1993); and an and Wilson, Curr. Opin. in Genetics and Devel. 3: 1 10—1 14 (1993). Lentiviral vectors contemplated for use include, for example, the HIV based vectors described in US.
Patent Nos. 6,143,520; 5,665,557; and 5,981,276, which are herein incorporated by nce.
Adenoviruses are also contemplated for use in delivery of iRNAs of the invention.
Adenoviruses are ally attractive vehicles, e. g., for delivering genes to atory epithelia. Adenoviruses naturally infect respiratory lia where they cause a mild disease.
Other targets for adenovirus-based ry systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non—dividing cells. Kozarsky and Wilson, t Opinion in Genetics and Development 3:499—503 (1993) present a review of adenovirus—based gene therapy. Bout et al., Human Gene Therapy 5:3—10 (1994) demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al., e 1—434 (1991); Rosenfeld et al., Cell 68:143—155 (1992); Mastrangeli et al., J. Clin. Invest. 91:225-234 ; PCT Publication WO94/12649; and Wang, et al., Gene y 2:775—783 (1995). A suitable AV vector for expressing an iRNA featured in the ion, a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia H et al. (2002), Nat. Biotech. 20: 1006—1010.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM Adeno—associated virus (AAV) vectors may also be used to delivery an iRNA of the invention (Walsh er al., Proc. Soc. Exp. Biol. Med. 204:289—300 (1993); US. Pat. No. ,436,146). In one embodiment, the iRNA can be expressed as two separate, complementary single—stranded RNA les from a recombinant AAV vector having, for e, either the U6 or H1 RNA promoters, or the cytomegalovirus (CMV) promoter. Suitable AAV vectors for expressing the dsRNA featured in the ion, methods for constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J. Virol. 61: 3096-3101; Fisher K J et al. (1996), J. Virol, 70: 520—532; Samulski R et al. (1989), J. Virol. 63: 3822—3826; US. Pat. No. 5,252,479; US.
Pat. No. 5,139,941; International Patent Application No. W0 88; and International Patent Application No. WO 93/24641, the entire disclosures of which are herein incorporated by reference.
Another viral vector suitable for delivery of an iRNA of the on is a pox virus such as a vaccinia virus, for example an attenuated ia such as Modified Virus Ankara (MVA) or NYVAC, an avipox such as fowl pox or canary pox.
The tropism of viral vectors can be modified by pseudotyping the vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as riate. For example, lentiviral vectors can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the like. 2O AAV vectors can be made to target different cells by engineering the vectors to express different capsid protein serotypes; see, e.g., Rabinowitz J E et al. (2002), J Viral —801, the entire disclosure of which is herein incorporated by reference.
The ceutical preparation of a vector can include the vector in an acceptable diluent, or can include a slow release matrix in which the gene delivery vehicle is imbedded.
Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene ry system.
IV. Pharmaceutical Compositions of the Invention The present invention also includes pharmaceutical compositions and formulations which include the iRNAs of the invention. In one embodiment, provided herein are pharmaceutical compositions containing an iRNA, as bed herein, and a pharmaceutically acceptable carrier. The ceutical compositions containing the iRNA are useful for treating a 6 associated disease or disorder, 6.g. romatosis.
Such pharmaceutical compositions are formulated based on the mode of delivery. One example is compositions that are formulated for systemic administration via parenteral delivery, e.g., by intravenous (IV) delivery. Another example is compositions that are [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM formulated for direct delivery into the brain hyma, e.g., by infusion into the brain, such as by continuous pump infusion.
The ceutical compositions comprising RNAi agents of the invention may be, for e, solutions with or without a buffer, or compositions containing ceutically acceptable carriers. Such compositions include, for example, aqueous or crystalline compositions, liposomal formulations, micellar formulations, emulsions, and gene therapy vectors.
In the s of the invention, the RNAi agent may be administered in a solution. A free RNAi agent may be administered in an unbuffered solution, e.g., in saline or in water.
Alternatively, the free siRNA may also be administred in a suitable buffer solution. The buffer solution may comprise acetate, citrate, prolamine, carbonate, or phosphate, or any combination thereof. In a preferred embodiment, the buffer solution is phosphate buffered saline (PBS). The pH and osmolarity of the buffer solution containing the RNAi agent can be adjusted such that it is suitable for administering to a subject.
In some embodiments, the buffer solution further comprises an agent for controlling the osmolarity of the solution, such that the osmolarity is kept at a desired value, e.g., at the physiologic values of the human plasma. Solutes which can be added to the buffer solution to control the osmolarity include, but are not limited to, proteins, peptides, amino acids, non— metabolized rs, ns, ions, sugars, metabolites, organic acids, , or salts. In some embodiments, the agent for controlling the osmolarity of the solution is a salt. In certain embodiments, the agent for controlling the osmolarity of the solution is sodium chloride or potassium chloride.
The pharmaceutical compositions of the invention may be administered in dosages sufficient to inhibit expression of a TMPRSS6 gene.
In general, a suitable dose of an iRNA of the invention will be in the range of about 0.001 to about 200.0 milligrams per kilogram body weight of the recipient per day, generally in the range of about 1 to 50 mg per kilogram body weight per day. For example, the dsRNA can be stered at about 0.01 mg/kg, about 0.05 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 1.5 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg about 10 mg/kg, about 20 mg/kg, about mg/kg, about 40 mg/kg, or about 50 mg/kg per single dose.
For example, the RNAi agent, e. g., dsRNA, may be administered at a dose of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4,1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, or about 10 mg/kg. Values and ranges interrrfiate to the d values are also intended to be part of this invention.
[Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In another embodiment, the RNAi agent, e.g., dsRNA, is administered at a dose of about 0.1 to about 50 mg/kg, about 0.25 to about 50 mg/kg, about 0.5 to about 50 mg/kg, about 0.75 to about 50 mg/kg, about 1 to about 50 mg/mg, about 1.5 to about 50 mg/kb, about 2 to about 50 mg/kg, about 2.5 to about 50 mg/kg, about 3 to about 50 mg/kg, about 3.5 to about 50 mg/kg, about 4 to about 50 mg/kg, about 4.5 to about 50 mg/kg, about 5 to about 50 mg/kg, about 7.5 to about 50 mg/kg, about 10 to about 50 mg/kg, about 15 to about 50 mg/kg, about 20 to about 50 mg/kg, about 20 to about 50 mg/kg, about 25 to about 50 mg/kg, about 25 to about 50 mg/kg, about 30 to about 50 mg/kg, about 35 to about 50 mg/kg, about 40 to about 50 mg/kg, about 45 to about 50 mg/kg, about 0.1 to about 45 mg/kg, about 0.25 to about 45 mg/kg, about 0.5 to about 45 mg/kg, about 0.75 to about 45 mg/kg, about 1 to about 45 mg/mg, about 1.5 to about 45 mg/kb, about 2 to about 45 mg/kg, about 2.5 to about 45 mg/kg, about 3 to about 45 mg/kg, about 3.5 to about 45 mg/kg, about 4 to about 45 mg/kg, about 4.5 to about 45 mg/kg, about 5 to about 45 mg/kg, about 7.5 to about 45 mg/kg, about to about 45 mg/kg, about 15 to about 45 mg/kg, about 20 to about 45 mg/kg, about 20 to about 45 mg/kg, about 25 to about 45 mg/kg, about 25 to about 45 mg/kg, about 30 to about 45 mg/kg, about 35 to about 45 mg/kg, about 40 to about 45 mg/kg, about 0.1 to about 40 mg/kg, about 0.25 to about 40 mg/kg, about 0.5 to about 40 mg/kg, about 0.75 to about 40 mg/kg, about 1 to about 40 mg/mg, about 1.5 to about 40 mg/kb, about 2 to about 40 mg/kg, about 2.5 to about 40 mg/kg, about 3 to about 40 mg/kg, about 3.5 to about 40 mg/kg, about 4 to about 40 mg/kg, about 4.5 to about 40 mg/kg, about 5 to about 40 mg/kg, about 7.5 to about 40 mg/kg, about 10 to about 40 mg/kg, about 15 to about 40 mg/kg, about 20 to about 40 mg/kg, about 20 to about 40 mg/kg, about 25 to about 40 mg/kg, about 25 to about 40 mg/kg, about 30 to about 40 mg/kg, about 35 to about 40 mg/kg, about 0.1 to about 30 mg/kg, about 0.25 to about 30 mg/kg, about 0.5 to about 30 mg/kg, about 0.75 to about 30 mg/kg, about 1 to about 30 mg/mg, about 1.5 to about 30 mg/kb, about 2 to about 30 mg/kg, about 2.5 to about 30 mg/kg, about 3 to about 30 mg/kg, about 3.5 to about 30 mg/kg, about 4 to about 30 mg/kg, about 4.5 to about 30 mg/kg, about 5 to about 30 mg/kg, about 7.5 to about 30 mg/kg, about 10 to about 30 mg/kg, about 15 to about 30 mg/kg, about 20 to about mg/kg, about 20 to about 30 mg/kg, about 25 to about 30 mg/kg, about 0.1 to about 20 mg/kg, about 0.25 to about 20 mg/kg, about 0.5 to about 20 mg/kg, about 0.75 to about 20 mg/kg, about 1 to about 20 mg/mg, about 1.5 to about 20 mg/kb, about 2 to about 20 mg/kg, about 2.5 to about 20 mg/kg, about 3 to about 20 mg/kg, about 3.5 to about 20 mg/kg, about 4 to about 20 mg/kg, about 4.5 to about 20 mg/kg, about 5 to about 20 mg/kg, about 7.5 to about 20 mg/kg, about 10 to about 20 mg/kg, or about 15 to about 20 mg/kg. Values and ranges intermediate to the recited values are also intended to be part of this invention.
For example, the RNAi agent, e. g., dsRNA, may be administered at a dose of about 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1H3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, 5.1, 5.2, 5.3, 5.4, .5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, or about 10 mg/kg. Values and ranges intermediate to the d values are also intended to be part of this invention.
In another embodiment, the RNAi agent, 6.g. ,dsRNA, is administered at a dose of about 0.5 to about 50 mg/kg, about 0.75 to about 50 mg/kg, about 1 to about 50 mg/mg, about 1.5 to about 50 mg/kg, about 2 to about 50 mg/kg, about 2.5 to about 50 mg/kg, about 3 to about 50 mg/kg, about 3.5 to about 50 mg/kg, about 4 to about 50 mg/kg, about 4.5 to about 50 mg/kg, about 5 to about 50 mg/kg, about 7.5 to about 50 mg/kg, about 10 to about 50 mg/kg, about 15 to about 50 mg/kg, about 20 to about 50 mg/kg, about 20 to about 50 mg/kg, about 25 to about 50 mg/kg, about 25 to about 50 mg/kg, about 30 to about 50 mg/kg, about to about 50 mg/kg, about 40 to about 50 mg/kg, about 45 to about 50 mg/kg, about 0.5 to about 45 mg/kg, about 0.75 to about 45 mg/kg, about 1 to about 45 mg/mg, about 1.5 to about 45 mg/kb, about 2 to about 45 mg/kg, about 2.5 to about 45 mg/kg, about 3 to about 45 mg/kg, about 3.5 to about 45 mg/kg, about 4 to about 45 mg/kg, about 4.5 to about 45 mg/kg, about 5 to about 45 mg/kg, about 7.5 to about 45 mg/kg, about 10 to about 45 mg/kg, about to about 45 mg/kg, about 20 to about 45 mg/kg, about 20 to about 45 mg/kg, about 25 to about 45 mg/kg, about 25 to about 45 mg/kg, about 30 to about 45 mg/kg, about 35 to about 2O 45 mg/kg, about 40 to about 45 mg/kg, about 0.5 to about 40 mg/kg, about 0.75 to about 40 mg/kg, about 1 to about 40 mg/mg, about 1.5 to about 40 mg/kb, about 2 to about 40 mg/kg, about 2.5 to about 40 mg/kg, about 3 to about 40 mg/kg, about 3.5 to about 40 mg/kg, about 4 to about 40 mg/kg, about 4.5 to about 40 mg/kg, about 5 to about 40 mg/kg, about 7.5 to about 40 mg/kg, about 10 to about 40 mg/kg, about 15 to about 40 mg/kg, about 20 to about 40 mg/kg, about 20 to about 40 mg/kg, about 25 to about 40 mg/kg, about 25 to about 40 mg/kg, about 30 to about 40 mg/kg, about 35 to about 40 mg/kg, about 0.5 to about 30 mg/kg, about 0.75 to about 30 mg/kg, about 1 to about 30 mg/mg, about 1.5 to about 30 mg/kb, about 2 to about 30 mg/kg, about 2.5 to about 30 mg/kg, about 3 to about 30 mg/kg, about 3.5 to about 30 mg/kg, about 4 to about 30 mg/kg, about 4.5 to about 30 mg/kg, about 5 to about 30 mg/kg, about 7.5 to about 30 mg/kg, about 10 to about 30 mg/kg, about 15 to about 30 mg/kg, about 20 to about 30 mg/kg, about 20 to about 30 mg/kg, about 25 to about mg/kg, about 0.5 to about 20 mg/kg, about 0.75 to about 20 mg/kg, about 1 to about 20 mg/mg, about 1.5 to about 20 mg/kb, about 2 to about 20 mg/kg, about 2.5 to about 20 mg/kg, about 3 to about 20 mg/kg, about 3.5 to about 20 mg/kg, about 4 to about 20 mg/kg, about 4.5 to about 20 mg/kg, about 5 to about 20 mg/kg, about 7.5 to about 20 mg/kg, about to about 20 mg/kg, or about 15 to about 20 mg/kg. In one embodiment, the dsRNA is stered at a dose of about 10mg/kg to about 30 mg/kg. Values and ranges intermediate to the fited values are also intended to be part of this invention. ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM For example, subjects can be administered a therapeutic amount of iRNA, such as about 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, 10, 10.5,11, 11.5, 12, 12.5, 13,135, 14, 14.5,15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 22.5, 23, 23.5, 24, 24.5, 25, 25.5, 26, 26.5, 27, 27.5, 28, 28.5, 29, 29.5, 30, 31, 32, 33, 34, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or about 50 mg/kg. Values and ranges intermediate to the recited values are also intended to be part of this invention.
In certain embodiments, for example, When a composition of the invention ses a dsRNA as described herein and a lipid, subjects can be administered a therapeutic amount of iRNA, such as about 0.01 mg/kg to about 5 mg/kg, about 0.01 mg/kg to about 10 mg/kg, about 0.05 mg/kg to about 5 mg/kg, about 0.05 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.2 mg/kg to about 5 mg/kg, about 0.2 mg/kg to about 10 mg/kg, about 0.3 mg/kg to about 5 mg/kg, about 0.3 mg/kg to about 10 mg/kg, about 0.4 mg/kg to about 5 mg/kg, about 0.4 mg/kg to about 10 mg/kg, about 0.5 mg/kg to about 5 mg/kg, about 0.5 mg/kg to about 10 mg/kg, about 1 mg/kg to about 5 mg/kg, about 1 mg/kg to about 10 mg/kg, about 1.5 mg/kg to about 5 mg/kg, about 1.5 mg/kg to about 10 mg/kg, about 2 mg/kg to about about 2.5 mg/kg, about 2 mg/kg to about 10 mg/kg, about 3 mg/kg to about 5 mg/kg, about 3 mg/kg to about 10 mg/kg, about 3.5 mg/kg to about 5 mg/kg, about 4 mg/kg to about 5 mg/kg, about 4.5 mg/kg to about 5 mg/kg, about 4 mg/kg to about 10 mg/kg, about 4.5 mg/kg to about 10 mg/kg, about 5 mg/kg to about 10 mg/kg, about 5.5 mg/kg to about 10 mg/kg, about 6 mg/kg to about 10 mg/kg, about 6.5 mg/kg to about 10 mg/kg, about 7 mg/kg to about 10 mg/kg, about 7.5 mg/kg to about 10 mg/kg, about 8 mg/kg to about 10 mg/kg, about 8.5 mg/kg to about 10 mg/kg, about 9 mg/kg to about 10 mg/kg, or about 9.5 mg/kg to about 10 mg/kg. Values and ranges intermediate to the recited values are also ed to be part of this invention.
For example, the dsRNA may be administered at a dose of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, .1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, or about 10 mg/kg. Values and ranges intermediate to the recited values are also intended to be part of this invention.
In certain embodiments of the invention, for example, when a double-stranded RNAi agent includes cations (6.5)., one or more motifs of three identical modifications on three fiecutive tides, including one such motif at or near the cleavage site of the [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM agent), six phosphorothioate linkages, and a ligand, such an agent is administered at a dose of about 0.01 to about 0.5 mg/kg, about 0.01 to about 0.4 mg/kg, about 0.01 to about 0.3 mg/kg, about 0.01 to about 0.2 mg/kg, about 0.01 to about 0.1 mg/kg, about 0.01 mg/kg to about 0.09 mg/kg, about 0.01 mg/kg to about 0.08 mg/kg, about 0.01 mg/kg to about 0.07 mg/kg, about 0.01 mg/kg to about 0.06 mg/kg, about 0.01 mg/kg to about 0.05 mg/kg, about 0.02 to about 0.5 mg/kg, about 0.02 to about 0.4 mg/kg, about 0.02 to about 0.3 mg/kg, about 0.02 to about 0.2 mg/kg, about 0.02 to about 0.1 mg/kg, about 0.02 mg/kg to about 0.09 mg/kg, about 0.02 mg/kg to about 0.08 mg/kg, about 0.02 mg/kg to about 0.07 mg/kg, about 0.02 mg/kg to about 0.06 mg/kg, about 0.02 mg/kg to about 0.05 mg/kg, about 0.03 to about 0.5 mg/kg, about 0.03 to about 0.4 mg/kg, about 0.03 to about 0.3 mg/kg, about 0.03 to about 0.2 mg/kg, about 0.03 to about 0.1 mg/kg, about 0.03 mg/kg to about 0.09 mg/kg, about 0.03 mg/kg to about 0.08 mg/kg, about 0.03 mg/kg to about 0.07 mg/kg, about 0.03 mg/kg to about 0.06 mg/kg, about 0.03 mg/kg to about 0.05 mg/kg, about 0.04 to about 0.5 mg/kg, about 0.04 to about 0.4 mg/kg, about 0.04 to about 0.3 mg/kg, about 0.04 to about 0.2 mg/kg, about 0.04 to about 0.1 mg/kg, about 0.04 mg/kg to about 0.09 mg/kg, about 0.04 mg/kg to about 0.08 mg/kg, about 0.04 mg/kg to about 0.07 mg/kg, about 0.04 mg/kg to about 0.06 mg/kg, about 0.05 to about 0.5 mg/kg, about 0.05 to about 0.4 mg/kg, about 0.05 to about 0.3 mg/kg, about 0.05 to about 0.2 mg/kg, about 0.05 to about 0.1 mg/kg, about 0.05 mg/kg to about 0.09 mg/kg, about 0.05 mg/kg to about 0.08 mg/kg, or about 0.05 mg/kg to about 0.07 mg/kg.
Values and ranges intermediate to the foregoing recited values are also intended to be part of this invention, e.g.,, the RNAi agent may be administered to the subject at a dose of about 0.015 mg/kg to about 0.45 mg/mg.
For example, the RNAi agent, 6.5)., RNAi agent in a pharmaceutical composition, may be administered at a dose of about 0.01 mg/kg, 0.0125 mg/kg, 0.015 mg/kg, 0.0175 mg/kg, 0.02 mg/kg, 0.0225 mg/kg, 0.025 mg/kg, 0.0275 mg/kg, 0.03 mg/kg, 0.0325 mg/kg, 0.035 mg/kg, 0.0375 mg/kg, 0.04 mg/kg, 0.0425 mg/kg, 0.045 mg/kg, 0.0475 mg/kg, 0.05 mg/kg, 0.0525 mg/kg, 0.055 mg/kg, 0.0575 mg/kg, 0.06 mg/kg, 0.0625 mg/kg, 0.065 mg/kg, 0.0675 mg/kg, 0.07 mg/kg, 0.0725 mg/kg, 0.075 mg/kg, 0.0775 mg/kg, 0.08 mg/kg, 0.0825 mg/kg, 0.085 mg/kg, 0.0875 mg/kg, 0.09 mg/kg, 0.0925 mg/kg, 0.095 mg/kg, 0.0975 mg/kg, 0.1 mg/kg, 0.125 mg/kg, 0.15 mg/kg, 0.175 mg/kg, 0.2 mg/kg, 0.225 mg/kg, 0.25 mg/kg, 0.275 mg/kg, 0.3 mg/kg, 0.325 mg/kg, 0.35 mg/kg, 0.375 mg/kg, 0.4 mg/kg, 0.425 mg/kg, 0.45 mg/kg, 0.475 mg/kg, or about 0.5 mg/kg. Values intermediate to the ing recited values are also intended to be part of this invention.
The pharmaceutical composition can be administered once daily, or the iRNA can be administered as two, three, or more sub—doses at riate intervals throughout the day or even using continuous infusion or delivery through a lled release ation. In that case, the iRNA contained in each sub—dose must be pondingly smaller in order to achiefie total daily dosage. The dosage unit can also be compounded for delivery over [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM l days, e.g., using a conventional sustained release formulation which es sustained release of the iRNA over a several day period. Sustained e formulations are well known in the art and are ularly useful for delivery of agents at a particular site, such as could be used with the agents of the present invention. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.
In other embodiments, a single dose of the pharmaceutical compositions can be long g, such that subsequent doses are administered at not more than 3, 4, or 5 day intervals, or at not more than 1, 2, 3, or 4 week intervals. In some embodiments of the invention, a single dose of the ceutical compositions of the invention is stered once per week. In other embodiments of the invention, a single dose of the pharmaceutical compositions of the invention is stered bi—monthly.
The skilled artisan will appreciate that certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments. Estimates of effective dosages and in viva half—lives for the dual iRNAs encompassed by the invention can be made using conventional methodologies or on the basis of in viva g using an appropriate animal model, as described elsewhere herein.
Advances in mouse genetics have generated a number of mouse models for the study of various human diseases, such as a disorder ated with iron overload that would benefit from reduction in the expression of TMPRSS6. Such models can be used for in viva testing of iRNA, as well as for determining a eutically effective dose. Suitable mouse models are known in the art and include, for example, the thalassemic Th3/+ mouse as a model of assemia (Douet et al., Am. J. Pathal. (2011), l78(2):774—83), the HFE knockout mouse as a model of hereditary hemochromatosis (Zhou et al. (1998) Prac. Natl Acad. Sci USA, 85:2492—2497); a Uros(mut248) mouse as a model of congenital erythropoietic porphyria (Ged er al. (2006) Genamics, 87(1):84-92).
The pharmaceutical compositions of the present invention can be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration can be topical (e.g., by a ermal patch), pulmonary, e. g., by inhalation or ation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal, oral or parenteral. Parenteral administration includes enous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; subdermal, e.g., via an implanted device; or intracranial, e. g., by intraparenchymal, hecal or intraventricular, administration The iRNA can be delivered in a manner to target a particular tissue, such as the liver (e. g., fiepatocytes of the liver).
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Pharmaceutical compositions and formulations for topical administration can include transdermal patches, ointments, lotions, creams, gels, drops, itories, sprays, liquids and powders. Conventional ceutical rs, aqueous, powder or oily bases, thickeners and the like can be necessary or ble. Coated condoms, gloves and the like can also be useful. Suitable l ations include those in which the iRNAs featured in the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Suitable lipids and liposomes e neutral (e. g., dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g., dimyristoylphosphatidyl ol DMPG) and cationic (e.g., yltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA). iRNAs featured in the ion can be encapsulated within liposomes or can form complexes thereto, in particular to cationic liposomes. Alternatively, iRNAs can be complexed to lipids, in particular to cationic lipids.
Suitable fatty acids and esters e but are not limited to arachidonic acid, oleic acid, eicosanoic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl l— monocaprate, l—dodecylazacycloheptan—2—one, an acylcarnitine, an acylcholine, or a C190 alkyl ester (e. g., isopropylmyristate 1PM), monoglyceride, diglyceride or pharmaceutically acceptable salt thereof). Topical formulations are described in detail in US. Patent No. 6,747,014, which is incorporated herein by reference.
A. iRNA Formulations Comprising Membranous Molecular Assemblies An iRNA for use in the compositions and methods of the invention can be formulated for delivery in a membranous molecular assembly, e. g., a liposome or a micelle. As used herein, the term ome" refers to a vesicle composed of amphiphilic lipids arranged in at least one bilayer, e.g., one bilayer or a plurality of bilayers. Liposomes include unilamellar and multilamellar vesicles that have a membrane formed from a ilic al and an aqueous interior. The aqueous portion contains the iRNA composition. The lipophilic material isolates the s interior from an aqueous exterior, which typically does not include the iRNA composition, although in some examples, it may. Liposomes are useful for the transfer and delivery of active ingredients to the site of . Because the liposomal membrane is structurally r to biological membranes, when liposomes are applied to a tissue, the liposomal bilayer fuses with bilayer of the ar membranes. As the merging of the liposome and cell progresses, the internal aqueous contents that include the iRNA are delivered into the cell where the iRNA can specifically bind to a target RNA and can mediate RNAi. In some cases the liposomes are also specifically targeted, e.g., to direct the iRNA to particular cell types.
A liposome containing a RNAi agent can be prepared by a variety of methods. In one exam the lipid component of a liposome is ved in a detergent so that micelles are [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM formed with the lipid component. For example, the lipid component can be an amphipathic cationic lipid or lipid conjugate. The detergent can have a high critical micelle concentration and may be nonionic. Exemplary detergents include cholate, CHAPS, octylglucoside, deoxycholate, and lauroyl sarcosine. The RNAi agent ation is then added to the micelles that include the lipid component. The cationic groups on the lipid interact with the RNAi agent and condense around the RNAi agent to form a liposome. After condensation, the detergent is removed, e.g., by dialysis, to yield a liposomal preparation of RNAi agent.
If necessary a carrier compound that assists in condensation can be added during the condensation on, e.g., by controlled addition. For e, the carrier compound can be a polymer other than a nucleic acid (e.g., spermine or spermidine). pH can also adjusted to favor condensation.
Methods for ing stable polynucleotide delivery vehicles, which incorporate a polynucleotide/cationic lipid complex as structural components of the delivery vehicle, are further described in, e.g., WO 96/37194, the entire ts of which are incorporated herein by reference. Liposome formation can also e one or more aspects of exemplary methods bed in Felgner, P. L. et al., Proc. Natl. Acad. Sci, USA 8:7413—7417, 1987; US. Pat. No. 4,897,355; US. Pat. No. 5,171,678; Bangham, et al. M. Mol. Biol. , 1965; Olson, et al. Biochim. Biophys. Acta 557:9, 1979; Szoka, et al. Proc. Natl. Acad. Sci. 75: 4194, 1978; Mayhew, et al. Biochim. s. Acta 775:169, 1984; Kim, et al. Biochim.
Biophys. Acta 728:339, 1983; and Fukunaga, et al. Endocrinol. 115:757, 1984. Commonly used techniques for preparing lipid aggregates of appropriate size for use as delivery vehicles include sonication and freeze—thaw plus extrusion (see, e.g., Mayer, et al. Biochim. Biophys.
Acta 858: 161, 1986). Microfluidization can be used when tently small (50 to 200 nm) and relatively uniform aggregates are desired (Mayhew, et al. Biochim. Biophys. Acta 775:169, 1984). These s are readily adapted to packaging RNAi agent preparations into liposomes.
Liposomes fall into two broad classes. Cationic liposomes are positively charged liposomes which interact with the negatively d nucleic acid molecules to form a stable complex. The positively charged nucleic acid/liposome complex binds to the negatively d cell surface and is internalized in an endosome. Due to the acidic pH within the endosome, the liposomes are ed, releasing their contents into the cell cytoplasm (Wang et al., Biochem. Biophys. Res. Commun., 1987, 147, 980—985).
Liposomes which are pH—sensitive or negatively—charged, entrap nucleic acids rather than complex with it. Since both the nucleic acid and the lipid are similarly charged, repulsion rather than complex formation . Nevertheless, some nucleic acid is entrapped within the aqueous interior of these mes. pH-sensitive liposomes have been used to deliver nucleic acids encoding the thymidine kinase gene to cell monolayers in culture. ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Expression of the ous gene was detected in the target cells (Zhou et al., Journal of Controlled Release, 1992, 19, 269—274).
One major type of liposomal composition includes phospholipids other than naturally— derived phosphatidylcholine. Neutral liposome compositions, for example, can be formed from dimyristoyl phosphatidylcholine (DMPC) or dipalmitoyl phosphatidylcholine (DPPC).
Anionic liposome compositions generally are formed from dimyristoyl phosphatidylglycerol, while anionic fusogenic liposomes are formed primarily from dioleoyl phosphatidylethanolamine (DOPE). Another type of liposomal composition is formed from phosphatidylcholine (PC) such as, for example, soybean PC, and egg PC. Another type is formed from mixtures of phospholipid and/or phosphatidylcholine and/or cholesterol.
Examples of other methods to introduce liposomes into cells in vitro and in vivo include US. Pat. No. 5,283,185; US. Pat. No. 5,171,678; WO 94/00569; WO 93/24640; WO 91/16024; Felgner, J. Biol. Chem. 269:2550, 1994; Nabel, Proc. Natl. Acad. Sci. 90211307, 1993; Nabel, Human Gene Ther. 3:649, 1992; Gershon, m. 3, 1993; and Strauss EMBO J. 112417, 1992.
Non—ionic liposomal systems have also been examined to determine their utility in the delivery of drugs to the skin, in particular systems comprising non—ionic surfactant and cholesterol. nic liposomal formulations comprising NovasomeTM I (glyceryl ate/cholesterol/polyoxyethylene—10—stearyl ether) and meTM II ryl distearate/cholesterol/polyoxyethylene—10—stearyl ether) were used to deliver cyclosporin—A into the dermis of mouse skin. Results indicated that such non—ionic liposomal systems were effective in facilitating the deposition of cyclosporine A into different layers of the skin (Hu et al. S.T.P.Pharma. Sci., 1994, 4(6) 466).
Liposomes also include "sterically stabilized" liposomes, a term which, as used herein, refers to liposomes sing one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are those in which part of the vesicle—forming lipid portion of the liposome (A) ses one or more glycolipids, such as monosialoganglioside GMl, or (B) is derivatized with one or more hydrophilic polymers, such as a hylene glycol (PEG) moiety. While not wishing to be bound by any particular theory, it is thought in the art that, at least for sterically stabilized liposomes containing osides, omyelin, or PEG-derivatized lipids, the enhanced circulation half—life of these sterically stabilized liposomes derives from a reduced uptake into cells of the loendothelial system (RES) (Allen et al., FEBS s, 1987, 223, 42; Wu et al., Cancer Research, 1993, 53, 3765).
Various liposomes comprising one or more glycolipids are known in the art.
Papahadjopoulos et al. (Ann. N. Y. Acad. Sci., 1987, 507, 64) reported the ability of monofiganglioside GMl, galactocerebroside sulfate and atidylinositol to improve [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM blood half-lives of liposomes. These findings were expounded upon by Gabizon et al. (Proc.
Natl. Acad. Sci. , 1988, 85, 6949). US. Pat. No. 4,837,028 and WO 24, both to Allen et al., disclose liposomes comprising (1) omyelin and (2) the ganglioside GM1 or a galactocerebroside sulfate ester. US. Pat. N0. 152 (Webb et al.) discloses liposomes comprising sphingomyelin. Liposomes comprising 1,2—sn—dimyristoylphosphatidylcholine are disclosed in W0 97/13499 (Lim et al).
In one embodiment, cationic liposomes are used. Cationic liposomes possess the advantage of being able to fuse to the cell membrane. Non-cationic liposomes, although not able to fuse as efficiently with the plasma membrane, are taken up by macrophages in vivo and can be used to deliver RNAi agents to macrophages. r ages of liposomes include: liposomes obtained from natural phospholipids are patible and biodegradable; liposomes can incorporate a wide range of water and lipid soluble drugs; liposomes can t encapsulated RNAi agents in their internal compartments from metabolism and ation (Rosoff, in "Pharmaceutical Dosage Forms," man, Rieger and Banker (Eds.), 1988, volume 1, p. 245). Important considerations in the preparation of liposome formulations are the lipid surface charge, vesicle size and the aqueous volume of the mes.
A positively charged synthetic cationic lipid, N—[1—(2,3—dioleyloxy)propyl]-N,N,N- trimethylammonium chloride (DOTMA) can be used to form small liposomes that interact 2O spontaneously with nucleic acid to form lipid—nucleic acid complexes which are capable of fusing with the negatively charged lipids of the cell membranes of tissue culture cells, resulting in delivery of RNAi agent (see, e.g., Felgner, P. L. et al., Proc. Natl. Acad. Sci., USA 8:7413—7417, 1987 and US. Pat. No. 355 for a description of DOTMA and its use with DNA).
A DOTMA analogue, 1,2—bis(oleoyloxy)—3—(trimethylammonia)propane (DOTAP) can be used in combination with a phospholipid to form mplexing vesicles.
LipofectinTM Bethesda Research Laboratories, Gaithersburg, Md.) is an effective agent for the delivery of highly c nucleic acids into living tissue culture cells that comprise positively d DOTMA liposomes which interact spontaneously with negatively charged polynucleotides to form complexes. When enough positively charged liposomes are used, the net charge on the resulting complexes is also positive. Positively charged complexes prepared in this way spontaneously attach to negatively charged cell surfaces, fuse with the plasma membrane, and efficiently deliver functional nucleic acids into, for example, tissue culture cells. Another commercially available cationic lipid, s(oleoyloxy)—3,3- (trimethylammonia)propane ("DOTAP") (Boehringer im, Indianapolis, Indiana) differs from DOTMA in that the oleoyl moieties are linked by ester, rather than ether linkages.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Other reported cationic lipid compounds include those that have been ated to a variety of moieties including, for example, carboxyspermine which has been conjugated to one of two types of lipids and includes compounds such as 5—carboxyspermylglycine dioctaoleoylamide ("DOGS") (TransfectamTM, Promega, Madison, Wisconsin) and dipalmitoylphosphatidylethanolamine 5—carboxyspermyl—amide ("DPPES") (see, e.g., U.S.
Pat. No. 5,171,678).
Another cationic lipid conjugate includes tization of the lipid with cholesterol ("DC—Chol") which has been formulated into liposomes in combination with DOPE (See, Gao, X. and Huang, L., Biochim. Biophys. Res. Commun. 179:280, 1991). Lipopolylysine, made by conjugating polylysine to DOPE, has been reported to be effective for transfection in the presence of serum (Zhou, X. et al., Biochim. Biophys. Acta 10658 1991). For certain cell lines, these liposomes containing conjugated cationic lipids, are said to exhibit lower toxicity and provide more efficient ection than the DOTMA—containing compositions.
Other commercially available cationic lipid products include DMRIE and DMRIE-HP (Vical, La Jolla, California) and ctamine (DOSPA) (Life Technology, Inc., Gaithersburg, Maryland). Other ic lipids suitable for the delivery of oligonucleotides are described in WO 59 and WO 94.
Liposomal formulations are particularly suited for l administration, liposomes present several advantages over other formulations. Such advantages include reduced side 2O effects related to high ic absorption of the administered drug, increased accumulation of the administered drug at the desired target, and the ability to administer RNAi agent into the skin. In some implementations, liposomes are used for delivering RNAi agent to epidermal cells and also to enhance the ation of RNAi agent into dermal tissues, e.g., into skin. For example, the liposomes can be applied topically. Topical ry of drugs formulated as liposomes to the skin has been documented (see, e.g., Weiner et al., Journal of Drug ing, 1992, vol. 2,405—410 and du Plessis et al., ral Research, 18, 1992, 259—265; Mannino, R. J. and Fould—Fogerite, S., Biotechniqnes 6:682—690, 1988; Itani, T. et al. Gene 56:267—276. 1987; Nicolau, C. et al. Meth. Enz. 149:157—176, 1987; Straubinger, R.
M. and Papahadjopoulos, D. Meth. Enz. 101:512—527, 1983; Wang, C. Y. and Huang, L., Proc. Natl. Acad. Sci. USA 84:7851-7855, 1987).
Non—ionic liposomal systems have also been examined to determine their utility in the delivery of drugs to the skin, in particular systems comprising non—ionic surfactant and cholesterol. Non—ionic liposomal formulations comprising Novasome I (glyceryl dilaurate/cholesterol/polyoxyethylene-10—stearyl ether) and Novasome II (glyceryl distearate/ terol/polyoxyethylene—10—stearyl ether) were used to deliver a drug into the dermis of mouse skin. Such formulations with RNAi agent are useful for treating a dermatological disorder.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM ed set by KIM Liposomes that include iRNA can be made highly deformable. Such deformability can enable the liposomes to penetrate through pore that are smaller than the average radius of the liposome. For example, transfersomes are a type of deformable liposomes.
Transferosomes can be made by adding surface edge activators, y surfactants, to a standard liposomal composition. Transfersomes that include RNAi agent can be delivered, for example, subcutaneously by ion in order to deliver RNAi agent to keratinocytes in the skin. In order to cross intact mammalian skin, lipid vesicles must pass through a series of fine pores, each with a diameter less than 50 nm, under the influence of a suitable transdermal gradient. In addition, due to the lipid properties, these transferosomes can be self—optimizing (adaptive to the shape of pores, 6.57., in the skin), self—repairing, and can ntly reach their targets without fragmenting, and often oading.
Other formulations amenable to the present invention are described in United States provisional application serial Nos. 61/018,616, filed January 2, 2008; 61/018,611, filed January 2, 2008; 61/039,748, filed March 26, 2008; ,087, filed April 22, 2008 and 61/051,528, filed May 8, 2008. PCT application no , filed October 3, 2007 also describes formulations that are amenable to the present ion.
Transfersomes are yet another type of liposomes, and are highly deformable lipid aggregates which are attractive candidates for drug delivery vehicles. Transfersomes can be described as lipid droplets which are so highly deformable that they are easily able to penetrate through pores which are smaller than the droplet. Transfersomes are adaptable to the environment in which they are used, e.g., they are self—optimizing (adaptive to the shape of pores in the skin), self—repairing, frequently reach their targets without fragmenting, and often self—loading. To make transfersomes it is possible to add surface edge—activators, y surfactants, to a standard liposomal composition. Transfersomes have been used to r serum albumin to the skin. The transfersome—mediated delivery of serum albumin has been shown to be as effective as subcutaneous injection of a solution containing serum albumin.
Surfactants find wide application in formulations such as emulsions (including microemulsions) and liposomes. The most common way of classifying and ranking the properties of the many different types of tants, both natural and synthetic, is by the use of the hydrophile/lipophile balance (HLB). The nature of the hydrophilic group (also known as the "head") es the most useful means for rizing the different surfactants used in formulations (Rieger, in ceutical Dosage Forms, Marcel Dekker, Inc., New York, N.Y., 1988, p. 285).
If the tant molecule is not ionized, it is classified as a nonionic surfactant.
Nonionic surfactants find wide ation in pharmaceutical and cosmetic products and are usable over a wide range of pH values. In general their HLB values range from 2 to about 18 depenfi on their structure. Nonionic surfactants include nonionic esters such as ethylene [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM glycol esters, propylene glycol esters, glyceryl esters, polyglyceryl esters, sorbitan esters, sucrose esters, and lated esters. ic alkanolamides and ethers such as fatty alcohol ethoxylates, propoxylated alcohols, and ethoxylated/propoxylated block polymers are also included in this class. The polyoxyethylene tants are the most popular members of the nonionic surfactant class.
If the surfactant molecule carries a negative charge when it is dissolved or dispersed in water, the surfactant is classified as anionic. Anionic surfactants include carboxylates such as soaps, acyl lactylates, acyl amides of amino acids, esters of sulfuric acid such as alkyl sulfates and ethoxylated alkyl sulfates, ates such as alkyl benzene sulfonates, acyl isethionates, acyl taurates and sulfosuccinates, and phosphates. The most important members of the anionic surfactant class are the alkyl sulfates and the soaps.
If the surfactant molecule carries a positive charge when it is dissolved or dispersed in water, the surfactant is classified as ic. ic surfactants include quaternary ammonium salts and ethoxylated amines. The quaternary ammonium salts are the most used members of this class.
If the surfactant molecule has the ability to carry either a positive or negative charge, the surfactant is classified as eric. Amphoteric tants include acrylic acid derivatives, substituted mides, N—alkylbetaines and phosphatides.
The use of surfactants in drug ts, formulations and in emulsions has been reviewed (Rieger, in Pharmaceutical Dosage Forms, Marcel , Inc., New York, N.Y., 1988, p. 285).
The iRNA for use in the methods of the ion can also be provided as micellar formulations. "Micelles" are d herein as a particular type of molecular ly in which amphipathic molecules are arranged in a spherical structure such that all the hydrophobic portions of the molecules are directed inward, leaving the hydrophilic portions in contact with the surrounding aqueous phase. The converse arrangement exists if the environment is hydrophobic.
A mixed micellar formulation suitable for delivery through transdermal membranes may be prepared by mixing an aqueous on of the siRNA composition, an alkali metal C8 to C22 alkyl sulphate, and a e forming compounds. Exemplary micelle forming compounds include in, hyaluronic acid, pharmaceutically acceptable salts of hyaluronic acid, glycolic acid, lactic acid, chamomile extract, cucumber extract, oleic acid, linoleic acid, linolenic acid, monoolein, monooleates, monolaurates, borage oil, evening of primrose oil, menthol, trihydroxy oxo cholanyl glycine and pharmaceutically acceptable salts thereof, glycerin, polyglycerin, lysine, polylysine, triolein, polyoxyethylene ethers and analogues thereof, polidocanol alkyl ethers and analogues thereof, chenodeoxycholate, deoxycholate, and mixtures thereof. The micelle forming compounds may be added at the same time or after fiition of the alkali metal alkyl sulphate. Mixed micelles will form with substantially [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM any kind of mixing of the ingredients but vigorous mixing in order to provide smaller size micelles.
In one method a first ar composition is ed which contains the siRNA composition and at least the alkali metal alkyl sulphate. The first micellar composition is then mixed with at least three micelle forming compounds to form a mixed micellar composition. In another method, the micellar composition is prepared by mixing the siRNA composition, the alkali metal alkyl sulphate and at least one of the micelle forming compounds, followed by addition of the remaining micelle forming compounds, with vigorous mixing.
Phenol and/or m—cresol may be added to the mixed micellar composition to stabilize the formulation and protect against bacterial growth. Alternatively, phenol and/or m—cresol may be added with the e g ingredients. An isotonic agent such as in may also be added after formation of the mixed micellar composition.
For delivery of the ar formulation as a spray, the formulation can be put into an aerosol dispenser and the dispenser is charged with a propellant. The propellant, which is under pressure, is in liquid form in the dispenser. The ratios of the ingredients are adjusted so that the s and propellant phases become one, i. 6., there is one phase. If there are two phases, it is necessary to shake the dispenser prior to dispensing a portion of the contents, e.g., through a metered valve. The dispensed dose of ceutical agent is propelled from the metered valve in a fine spray.
Propellants may include hydrogen—containing chlorofluorocarbons, hydrogen— containing fluorocarbons, dimethyl ether and diethyl ether. In certain ments, HFA 134a (l,l,l,2 tetrafluoroethane) may be used.
The specific concentrations of the ial ingredients can be determined by relatively straightforward experimentation. For absorption through the oral es, it is often desirable to increase, e.g., at least double or triple, the dosage for through injection or administration through the gastrointestinal tract.
B. Lipid particles iRNAs, e.g., dsRNAs of in the invention may be fully encapsulated in a lipid formulation, 6.57., a LNP, or other nucleic acid—lipid particle.
As used herein, the term "LNP" refers to a stable nucleic acid—lipid particle. LNPs contain a cationic lipid, a tionic lipid, and a lipid that prevents aggregation of the particle (e.g., a PEG—lipid conjugate). LNPs are extremely useful for systemic applications, as they exhibit extended circulation lifetimes following enous (i.v.) injection and accumulate at distal sites (e.g., sites physically separated from the administration site). LNPs include "pSPLP," which include an encapsulated condensing agent-nucleic acid complex as set forth in PCT Publication No. WO 00/03683. The particles of the present ion typicafimve a mean diameter of about 50 nm to about 150 nm, more typically about 60 nm [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM to about 130 nm, more typically about 70 nm to about 110 nm, most typically about 70 nm to about 90 nm, and are substantially nontoxic. In addition, the nucleic acids when present in the nucleic acid— lipid particles of the present invention are resistant in aqueous solution to degradation with a nuclease. Nucleic acid—lipid particles and their method of preparation are disclosed in, e.g., US. Patent Nos. 5,976,567; 5,981,501; 6,534,484; 6,586,410; 6,815,432; US. Publication No. 2010/0324120 and PCT Publication No. WO 96/40964.
In one embodiment, the lipid to drug ratio (mass/mass ratio) (e.g., lipid to dsRNA 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. Ranges intermediate to the above recited ranges are also contemplated to be part of the invention.
The cationic lipid can be, for e, N,N-dioleyl-N,N—dimethylammonium chloride (DODAC), N,N—distearyl—N,N—dimethylammonium e (DDAB), N—(I —(2,3— dioleoyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTAP), N—(I -(2,3— dioleyloxy)propyl)-N,N,N—trimethylammonium chloride (DOTMA), methyl—2,3— dioleyloxy)propylamine (DODMA), 1,2—DiLinoleyloxy—N,N—dimethylaminopropane (DLinDMA), l,2—Dilinolenyloxy—N,N—dimethylaminopropane (DLenDMA), 1,2— Dilinoleylcarbamoyloxy—3—dimethylaminopropane (DLin—C-DAP), 1,2—Dilinoleyoxy—3— hylamino)acetoxypropane (DLin—DAC), 1,2—Dilinoleyoxy—3—morpholinopropane (DLin—MA), 1,2—Dilinoleoyl-3—dimethylaminopropane (DLinDAP), 1,2—Dilinoleylthio—3— dimethylaminopropane S—DMA), 1—Linoleoyl—2—linoleyloxy—3—dimethylaminopropane (DLin—2—DMAP), 1,2—Dilinoleyloxy-3—trimethylaminopropane chloride salt (DLin-TMACl), 1,2—Dilinoleoyl—3—trimethylaminopropane chloride salt (DLin—TAP.Cl), 1,2—Dilinoleyloxy—3— (N—methylpiperazino)propane (DLin-MPZ), or 3—(N,N—Dilinoleylamino)—1,2—propanediol (DLinAP), 3—(N,N—Dioleylamino)—1,2—propanedio (DOAP), 1,2—Dilinoleyloxo—3—(2—N,N— dimethylamino)ethoxypropane (DLin—EG—DMA), l,2—Dilinolenyloxy—N,N- dimethylaminopropane (DLinDMA), 2,2—Dilinoleyl—4—dimethylaminomethyl—[1,3]—dioxolane K—DMA) or analogs thereof, (3aR,53,6aS)—N,N—dimethyl—2,2-di((9Z,12Z)—octadeca- 9,12—dienyl)tetrahydro—3aH—cyclopenta[d][1,3]dioxol—5—amine (ALN100), (6Z,9Z,28Z,3IZ)— heptatriaconta-6,9,28,31—tetraen—19-yl 4—(dimethylamino)butanoate (MC3), 1,1'—(2-(4—(2—((2— —hydroxydodecyl)amino)ethy1)(2—hydroxydodecyl)amino)ethyl)piperazin— 1— yl)ethylazanediyl)didodecan-2—ol (Tech GI), or a mixture thereof. The ic lipid can comprise from about 20 mol % to about 50 mol % or about 40 mol % of the total lipid present in the particle.
In r embodiment, the compound 2,2—Dilinoleyl—4—dimethylaminoethyl—[1,3]— dioxolane can be used to prepare lipid-siRNA nanoparticles. Synthesis of linoleyl dimethylaminoethyl—[1,3]—dioxolane is described in United States provisional patent [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM application number 61/107,998 filed on r 23, 2008, which is herein incorporated by reference.
In one embodiment, the lipid—siRNA particle es 40% 2, 2—Dilinoleyl—4— dimethylaminoethyl-[l,3]—dioxolane: 10% DSPC: 40% Cholesterol: 10% PEG—C—DOMG (mole percent) with a particle size of 63.0 i 20 nm and a 0.027 siRNA/Lipid Ratio.
The ionizable/non—cationic lipid can be an anionic lipid or a neutral lipid including, but not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl—phosphatidylethanolamine , palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoylphosphatidylethanolamine (POPE), yl— phosphatidylethanolamine 4—(N—maleimidomethyl)—cyclohexane—l— carboxylate (DOPE—mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl—phosphatidyl—ethanolamine (DSPE), 16—O-monomethyl PE, 16—O-dimethyl PE, 18—1 —trans PE, 1 oyl—2—oleoyl— phosphatidyethanolamine (SOPE), cholesterol, or a mixture thereof. The non—cationic lipid can be from about 5 mol % to about 90 mol %, about 10 mol %, or about 58 mol % if cholesterol is included, of the total lipid present in the particle.
The conjugated lipid that inhibits aggregation of particles can be, for example, a polyethyleneglycol (PEG)—lipid including, without limitation, a PEG—diacylglycerol (DAG), a PEG-dialkyloxypropyl (DAA), a PEG—phospholipid, a PEG-ceramide (Cer), or a mixture thereof. The PEG—DAA ate can be, for example, a PEG—dilauryloxypropyl (C12), a PEG-dimyristyloxypropyl (Ci4), a PEG—dipalmityloxypropyl (Ci6), or a PEG— distearyloxypropyl (C]g). The conjugated lipid that prevents aggregation of particles can be from 0 mol % to about 20 mol % or about 2 mol % of the total lipid present in the le.
In some embodiments, the nucleic acid—lipid le further includes cholesterol at, e.g., about 10 mol % to about 60 mol % or about 48 mol % of the total lipid present in the particle.
In one embodiment, the id ND98-4HCl (MW 1487) (see US. Patent Application No. 12/056,230, filed 3/26/2008, which is incorporated herein by reference), Cholesterol (Sigma—Aldrich), and PEG—Ceramide C16 (Avanti Polar Lipids) can be used to prepare lipid— dsRNA nanoparticles (i.e., LNP01 les). Stock solutions of each in l can be prepared as follows: ND98, 133 mg/ml; Cholesterol, 25 mg/ml, PEG—Ceramide C16, 100 mg/ml. The ND98, Cholesterol, and ramide C16 stock ons can then be combined in a, e.g., 42:48: 10 molar ratio. The combined lipid solution can be mixed with aqueous dsRNA (e.g., in sodium acetate pH 5) such that the final ethanol concentration is about 35-45% and the final sodium acetate concentration is about 100-300 mM. Lipid- dsRNA nanoparticles typically form spontaneously upon mixing. Depending on the desired particfize distribution, the resultant nanoparticle mixture can be extruded through a [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM polycarbonate ne (e.g., 100 nm cut—off) using, for example, a thermobarrel extruder, such as Lipex Extruder (Northern Lipids, Inc). In some cases, the extrusion step can be omitted. Ethanol removal and aneous buffer exchange can be accomplished by, for e, dialysis or tangential flow filtration. Buffer can be exchanged with, for example, phosphate buffered saline (PBS) at about pH 7, e.g., about pH 6.9, about pH 7.0, about pH 7.1, about pH 7.2, about pH 7.3, or about pH 7.4.
H H WJJ\/\ /\/N\/\ /\/N N\/\/\/\/\/\/ H 3 N: WO H H ND98|somer| Formulal LNPOl formulations are described, e.g., in International Application Publication No. , Which is hereby incorporated by reference.
Additional exemplary dsRNA formulations are described in Table A.
Table A. cationic lipid/non-cationic Ionizable/Cationic Lipid lipid/cholesterol/PEG-lipid conjugate Li RNA ratio DLinDMA/DPPC/CholesterolflDEG-CDMA l,2—Dilinolenyloxy-N,N-dimethylaminopropane 7.1/34.4/1.4) (DLinDMA) lipidzsiRNA ~ 7:1 XTC/DPPC/Cholesterol/PEG-cDMA 2,2-Dilinoleyldimethylaminoethyl-[ 1 , 3] - 57.1/7.1/34.4/1.4 dioxolane (XTC) 1ioid:siRNA ~ 7:1 XTC/DSPC/Cholesterol/PEG-DMG 2,2-Dilinoleyldimethylaminoethyl-[ l , 3] - 57.5/7.5/31.5/3.5 dioxolane (XTC) 1ioid:siRNA ~ 6:1 XTC/DSPC/Cholesterol/PEG-DMG 2,2-Dilinoleyldimethylaminoethyl-[ 1 , 3] - 57.5/7.5/31.5/3.5 dioxolane (XTC) siRNA ~ 11:1 linoleyldimethylaminoethyl-[ 1 , 3] - XTC/DSPC/Cholesterol/PEG-DMG dioxolane (XTC) 60/7513 1/1 .5, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 1ipid:siRNA ~ 6:1 XTC/DSPC/Cholesterol/PEG-DMG 2,2-Dilinoleyldimethylaminoethyl-[ 1 , 3] - /31/1.5, dioxolane (XTC) 1ioid:siRNA ~ 11:1 XTC/DSPC/Cholesterol/PEG-DMG 2,2-Dilinoley1dimethylaminoethy1-[1, 3] - 50/10/38.5/1.5 dioxolane (XTC) LioidzsiRNA 10:1 (3aR,5s,6aS)-N,N-dimethyl-2,2—di((9Z,122)- ALN]OO/DSPCiCholesterol/PEG-DMG 0ctadeca—9, 12-dieny1)tetrahydro-3aH- 50/10/38.5/1.5 cyclopenta[d] [1,3]dioxolamine (ALN 100) Lipid:siRNA 10:1 (6Z,9Z,282,3 1Z)-heptatriac0nta—6,9,28,3 1- MC-3fl)SPC/CholesteIOI/PEG-DMG n-19 —yl 4-(dimethylamin0)butanoate 50/10/38.5/1.5 (MC3) Li oidzsiRNA 10:1 1,1‘-(2-(4—(2-((2-(bis(2- Tech G1/DSPC/Cholester01/PEG-DMG hydroxydodecyl)amin0)ethy1)(2- 50/10/38.5/1.5 hydroxydodecyl)amin0)ethy1)piperazin Lipid:siRNA 10:1 l)eth lazanedi l)did0decan01 (Tech G 1) XTC/DSPC/Chol/PEG-DMG 50/10/38.5/1.5 LioidzsiRNA: 33:1 MC3/DSPC/Ch01fl3EG-DMG 40/15/40/5 Lioid:siRNA: 11:1 MC3/DSPC/Chol/PEG—DSG/GalNAc-PEG-DSG 50/10/35/4.5/0.5 siRNA: 11:1 MC3/DSPC/Cholfl3EG-DMG 50/10/38.5/1.5 Lioid:siRNA: 7:1 MC3/DSPC/Chol/PEG—DSG 50/10/38.5/1.5 Lipid:siRNA: 10:1 ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM MC3/DSPC/Ch0WEG-DMG MC3 50/10/38.5/1.5 Liid:siRNA: 12:1 MC3/DSPC/Chol/PEG-DMG MC3 50/10/35/5 Li oidzsiRNA: 8:1 MC3/DSPC/Ch0WEG-DPG MC3 50/10/38.5/1.5 Lipid:siRNA: 10:1 C12-200/DSPC/Chol/PEG-DSG C12 200 38.5/1.5 LipidzsiRNA: 7:1 XTC/DSPC/Chol/PEG-DSG XTC 50/10/38.5/1.5 Lipid:siRNA: 10:1 DSPC: distearoylphosphatidylcholine DPPC: dipalmitoylphosphatidylcholine PEG—DMG: PEG—didimyristoyl glycerol (Cl4—PEG, or PEG—C14) (PEG with avg mol wt of 2000) G: PEG—distyryl glycerol (Cl8—PEG, or PEG—C18) (PEG with avg mol wt of 2000) PEG—cDMA: PEG—carbamoyl—l,2—dimyristyloxypropylamine (PEG with avg mol wt of 2000) LNP ilinolenyloxy—N,N—dimethylaminopropane (DLinDMA)) comprising formulations are described in ational Publication No. WO2009/127060, filed April 15, 2009, which is hereby orated by reference.
XTC comprising formulations are described, e.g., in US. Provisional Serial No. 61/l48,366, filed January 29, 2009; US. Provisional Serial No. 61/156,851, filed March 2, 2009; US. Provisional Serial No. filed June 10, 2009; US. Provisional Serial No. 61/228,373, filed July 24, 2009; US. Provisional Serial No. 61/239,686, filed September 3, 2009, and International Application No. , filed January 29, 2010, which are hereby incorporated by reference.
MC3 comprising formulations are described, e.g., in US. Publication No. 2010/0324120, filed June 10, 2010, the entire contents of which are hereby incorporated by reference.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM ALNY—100 comprising formulations are described, e.g., International patent application number PCT/USO9/63933, filed on November 10, 2009, which is hereby incorporated by reference.
C12—200 comprising formulations are described in US. Provisional Serial No. 61/175,770, filed May 5, 2009 and International Application No. PCT/USlO/33777, filed May 5, 2010, which are hereby incorporated by reference.
Synthesis of ionizable/cationic lipids Any of the compounds, e.g., ic lipids and the like, used in the c acid—lipid particles of the invention can be prepared by known c synthesis techniques, including the methods described in more detail in the Examples. All tuents are as defined below unless indicated otherwise. " means a straight chain or branched, lic or cyclic, saturated aliphatic hydrocarbon containing from 1 to 24 carbon atoms. Representative saturated straight chain alkyls e methyl, ethyl, n—propyl, n—butyl, n—pentyl, n—hexyl, and the like; while saturated branched alkyls include isopropyl, sec—butyl, isobutyl, tert—butyl, isopentyl, and the like.
Representative saturated cyclic alkyls include cyclopropyl, utyl, cyclopentyl, cyclohexyl, and the like; while unsaturated cyclic alkyls include entenyl and cyclohexenyl, and the like.
"Alkenyl" means an alkyl, as defined above, containing at least one double bond between adjacent carbon atoms. Alkenyls include both cis and trans isomers. Representative straight chain and branched alkenyls include nyl, propylenyl, 1—butenyl, 2—butenyl, isobutylenyl, 1—pentenyl, 2—pentenyl, 3—methyl—1—butenyl, 2-methyl-2—butenyl, 2,3—dimethyl— 2—butenyl, and the like.
"Alkynyl" means any alkyl or alkenyl, as defined above, which additionally contains at least one triple bond between adjacent carbons. Representative straight chain and branched alkynyls include acetylenyl, propynyl, 1—butynyl, 2-butynyl, 1—pentynyl, 2-pentynyl, 3— methyl—l l, and the like.
"Acyl" means any alkyl, alkenyl, or alkynyl wherein the carbon at the point of attachment is substituted with an oxo group, as d below. For example, —C(=O)alkyl, — C(=O)alkenyl, and —C(=O)alkynyl are acyl groups.
"Heterocycle" means a 5— to 7—membered clic, or 7— to 10—membered bicyclic, heterocyclic ring which is either saturated, unsaturated, or aromatic, and which contains from 1 or 2 heteroatoms independently ed from nitrogen, oxygen and sulfur, and wherein the nitrogen and sulfur heteroatoms can be optionally oxidized, and the nitrogen heteroatom can be optionally nized, including bicyclic rings in which any of the above heterocycles are fused to a benzene ring. The heterocycle can be attached via any heteroatom or carbon atom.
Heterocycles include heteroaryls as defined below. Heterocycles include morpholinyl, pyrrolfionyl, pyrrolidinyl, piperidinyl, piperizynyl, hydantoinyl, lactamyl, oxiranyl, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
The terms nally substituted alkyl", "optionally substituted l", "optionally substituted alkyny ", "optionally substituted acy ", and "optionally substituted heterocycle" means that, when substituted, at least one en atom is replaced with a tuent. In the case of an oxo substituent (=0) two hydrogen atoms are replaced. In this regard, substituents include oxo, halogen, heterocycle, —CN, —ORX, —NRXRy, —NRXC(=O)Ry, —NRXSOZRy, -C(=O)RX, —C(=O)ORX, —C(=O)NRny, —SOnRX and —SOnNRXRy, wherein n is 0, 1 or 2, RX and Ry are the same or ent and ndently hydrogen, alkyl or heterocycle, and each of said alkyl and heterocycle substituents can be further substituted with one or more of oxo, halogen, —OH, —CN, alkyl, —ORX, heterocycle, —NRXRy, —NRXC(=O)Ry, —NRXSOZRy, -C(=O)RX, -C(=O)ORX, -C(=O)NRny, -SOnRX and —SOnNRXRy.
"Halogen" means fluoro, chloro, bromo and iodo.
In some ments, the methods of the ion can require the use of protecting groups. Protecting group methodology is well known to those skilled in the art (see, for example, tive Groups in Organic Synthesis, Green, T.W. er al., Wiley—Interscience, New York City, 1999). Briefly, protecting groups within the context of this invention are any group that reduces or eliminates unwanted reactivity of a functional group. A protecting group can be added to a functional group to mask its vity during certain reactions and then removed to reveal the original functional group. In some embodiments an "alcohol protecting group" is used. An "alcohol protecting group" is any group which decreases or eliminates unwanted reactivity of an alcohol functional group. Protecting groups can be added and removed using techniques well known in the art.
Synthesis ofFormula A In some embodiments, nucleic acid—lipid particles of the invention are formulated using a cationic lipid of a A: N—R4 3O where R1 and R2 are independently alkyl, alkenyl or alkynyl, each can be optionally substituted, and R3 and R4 are independently lower alkyl or R3 and R4 can be taken together to form an optionally substituted heterocyclic ring. In some ments, the cationic lipid is XTC (2,2—Dilinoleyl—4—dimethylaminoethyl—[1,3]—dioxolane). In general, the lipid of [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM formula A above can be made by the following Reaction Schemes 1 or 2, wherein all substituents are as defined above unless indicated otherwise.
Scheme 1 Br OH Formula A Lipid A, where R1 and R2 are independently alkyl, alkenyl or alkynyl, each can be optionally tuted, and R3 and R4 are independently lower alkyl or R3 and R4 can be taken together to form an optionally substituted heterocyclic ring, can be prepared according to Scheme 1. Ketone l and bromide 2 can be purchased or prepared according to methods known to those of ordinary skill in the art. Reaction of l and 2 yields ketal 3. Treatment of ketal 3 with amine 4 yields lipids of formula A. The lipids of a A can be ted to the corresponding ammonium salt with an organic salt of formula 5, where X is anion counter ion selected from n, hydroxide, phosphate, sulfate, or the like.
Scheme 2 _ R2 Bng R1 + R2‘CN —> O:< R2> Grignard t 6 and cyanide 7 can be purchased or prepared according to methods known to those of ordinary skill in the art. Reaction of 6 and 7 yields ketone 1. Conversion of ketone l to the corresponding lipids of formula A is as described in Scheme 1.
El [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Synthesis 0fMC3 ation of DLin-M—C3—DMA (i.e., ,28Z,31Z)—heptatriaconta—6,9,28,31— tetraen—l9—yl 4—(dimethylamino)butanoate) was as follows. A solution of ,28Z,31Z)— heptatriaconta—6,9,28,3l—tetraen—19-ol (0.53 g), 4—N,N—dimethylaminobutyric acid hydrochloride (0.51 g), 4—N,N—dimethylaminopyridine (0.61 g) and l—ethy1—3—(3— dimethylaminopropyl)carbodiimide hydrochloride (0.53 g) in dichloromethane (5 mL) was stirred at room temperature overnight. The solution was washed with dilute hydrochloric acid followed by dilute aqueous sodium bicarbonate. The organic fractions were dried over anhydrous magnesium sulphate, ed and the t removed on a p. The e was passed down a silica gel column (20 g) using a 1—5% methanol/dichloromethane elution gradient. Fractions containing the purified product were combined and the solvent removed, yielding a colorless oil (0.54 g). sis ofALNY-IOO Synthesis of ketal 519 [ALNY—100] was performed using the following scheme 3: NHBoc NHMe NCsze .sNCDZMe NCsze NMO' 0504 LAH CbZ-OSu, NEt3 + 514 515 515 517A 51713OH 0 J PTSA MezN"" Synthesis 0f516 To a stirred solution of compound 515 in 100 mL dry DCM in a 250 mL two neck RBF, was added NEt3 (37.2 mL, 0.2669 mol) and cooled to 0 0C under nitrogen atmosphere.
After a slow addition of N—(benzyloxy—carbonyloxy)—succinimide (20 g, 0.08007 mol) in 50 mL drECM, reaction mixture was allowed to warm to room temperature. After completion [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM of the on (2—3 h by TLC) mixture was washed successively with 1N HCl solution (1 x 100 mL) and saturated NaHCO3 solution (1 X 50 mL). The organic layer was then dried over anhyd. Na2SO4 and the solvent was evaporated to give crude material which was purified by silica gel column chromatography to get 516 as sticky mass. Yield: 11g (89%). 1H—NMR (CDCl3, 400MHz): 8 = 7.36—7.27(m, 5H), 5.69 (s, 2H), 5.12 (s, 2H), 4.96 (br., 1H) 2.74 (s, 3H), , 2H), 2.30-2.25(m, 2H). LC—MS [M+H] —232.3 (96.94%).
Synthesis 0f51 7A and 51 7B The entene 516 (5 g, 0.02164 mol) was dissolved in a solution of 220 mL acetone and water (10: 1) in a single neck 500 mL RBF and to it was added N—methyl morpholine—N—oxide (7.6 g, 0.06492 mol) followed by 4.2 mL of 7.6% solution of OsO4 (0.275 g, 0.00108 mol) in tert—butanol at room temperature. After completion of the reaction (~ 3 h), the mixture was quenched with on of solid Na2SO3 and resulting mixture was d for 1.5 h at room temperature. Reaction mixture was diluted with DCM (300 mL) and washed with water (2 x 100 mL) followed by saturated NaHCO3 (1 x 50 mL) solution, water (1 x 30 mL) and finally with brine (1x 50 mL). Organic phase was dried over an.Na2SO4 and t was removed in vacuum. Silica gel column chromatographic purification of the crude material was afforded a mixture of diastereomers, which were separated by prep HPLC.
Yield: — 6 g crude 517A - Peak—1 (white solid), 5.13 g (96%). 1H-NMR (DMSO, 400MHz): 5: 7.39—7.31(m, 5H), 5.04(s, 2H), .73 (m, 1H), 4.48—4.47(d, 2H), .93(m, 2H), 2.71(s, 3H), 1.72- , 4H). LC-MS - [M+H]-266.3, [M+NH4 +]—283.5 present, HPLC—97.86%.
Stereochemistry confirmed by X—ray. sis of5I 8 Using a procedure analogous to that described for the sis of compound 505, compound 518 (1.2 g, 41%) was obtained as a colorless oil. 1H—NMR (CDCl3, 400MHz): 8: 7.35-7.33(m, 4H), 7.30-7.27(m, 1H), 5.37—5.27(m, 8H), 5.12(s, 2H), 4.75(m,1H), 4.58- 4.57(m,2H), 2.78-2.74(m,7H), 2.06—2.00(m,8H), 1.96-1.91(m, 2H), 1.62(m, 4H), 1.48(m, 2H), 1.37—1.25(br m, 36H), 0.87(m, 6H). HPLC—98.65%.
General Procedurefor the Synthesis of Compound 519 A solution of compound 518 (1 eq) in hexane (15 mL) was added in a drop-wise fashion to an ice—cold solution of LAH in THF (1 M, 2 eq). After complete addition, the mixture was heated at 400C over 0.5 h then cooled again on an ice bath. The mixture was carefully hydrolyzed with saturated aqueous Na2SO4 then filtered through celite and reduced to an oil. Column chromatography provided the pure 519 (1.3 g, 68%) which was obtained as a colorless oil. 13C NMR 8 = 130.2, 130.1 (x2), 127.9 (x3), 112.3, 79.3, 64.4, 44.7, 38.3, .4, 31.5, 29.9 (x2), 29.7, 29.6 (x2), 29.5 (x3), 29.3 (x2), 27.2 (x3), 25.6, 24.5, 23.3, 226, 14.1; Electrospray MS (+ve): Molecular weight for C44H80NO2 (M + H)+ Calc. 654.6, Foundééw. ation] KIM None set by KIM ation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Formulations ed by either the standard or extrusion—free method can be terized in similar manners. For example, formulations are typically characterized by visual inspection. They should be whitish ucent solutions free from aggregates or sediment. Particle size and particle size distribution of lipid—nanoparticles can be measured by light scattering using, for example, a Malvern Zetasizer Nano ZS (Malvern, USA).
Particles should be about 20-300 nm, such as 40—100 nm in size. The particle size distribution should be unimodal. The total dsRNA concentration in the formulation, as well as the ped fraction, is estimated using a dye exclusion assay. A sample of the formulated dsRNA can be ted with an RNA—binding dye, such as Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, 6.57., 0.5% Triton—X100. The total dsRNA in the formulation can be determined by the signal from the sample ning the surfactant, relative to a standard curve. The entrapped fraction is determined by subtracting the "free" dsRNA content (as measured by the signal in the absence of surfactant) from the total dsRNA content. Percent entrapped dsRNA is lly >85%. For LNP formulation, the particle size is at least 30 nm, at least 40 nm, at least 50 nm, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, and at least 120 nm. The suitable range is typically about at least 50 nm to about at least 110 nm, about at least 60 nm to about at least 100 nm, or about at least 80 nm to about at least 90 Compositions and formulations for oral administration e powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non—aqueous media, capsules, gel capsules, sachets, tablets or minitablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders can be desirable. In some embodiments, oral formulations are those in which dsRNAs featured in the invention are administered in conjunction with one or more penetration enhancer surfactants and ors. Suitable surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof.
Suitable bile salts include chenodeoxycholic acid (CDCA) and ursodeoxychenodeoxycholic acid (UDCA), cholic acid, dehydrocholic acid, deoxycholic acid, glucholic acid, glycholic acid, glycodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, sodium tauro-24,25—dihydro-fusidate and sodium glycodihydrofusidate. Suitable fatty acids e arachidonic acid, undecanoic acid, oleic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl l—monocaprate, l— dodecylazacycloheptan—Z—one, an acylcarnitine, an acylcholine, or a yceride, a diglyceride or a pharmaceutically acceptable salt thereof (e.g., sodium). In some embodiments, combinations of penetration enhancers are used, for example, fatty acids/salts in combination with bile acids/salts. One exemplary ation is the sodium salt of lauric acid, fie acid and UDCA. Further penetration enhancers include polyoxyethylenelauryl ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ether, polyoxyethylene—20-cetyl ether. DsRNAs featured in the ion can be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or rticles. DsRNA compleXing agents e poly—amino acids; polyimines; polyacrylates; polyalkylacrylates, polyoxethanes, polyalkylcyanoacrylates; cationized gelatins, albumins, starches, acrylates, polyethyleneglycols (PEG) and starches; kylcyanoacrylates; DEAE—derivatized polyimines, pollulans, celluloses and starches.
Suitable compleXing agents include an, N—trimethylchitosan, poly—L—lysine, polyhistidine, polyornithine, ermines, protamine, polyvinylpyridine, polythiodiethylaminomethylethylene P(TDAE), polyaminostyrene (e.g., p—amino), poly(methylcyanoacrylate), poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(isohexylcynaoacrylate), DEAE—methacrylate, DEAE— hexylacrylate, DEAE—acrylamide, DEAE—albumin and DEAE—dextran, polymethylacrylate, polyhexylacrylate, poly(D,L—lactic acid), poly(DL—lactic—co—glycolic acid (PLGA), alginate, and polyethyleneglycol (PEG). Oral formulations for dsRNAs and their preparation are described in detail in US. Patent 6,887,906, US Publn. No. 27780, and US. Patent No. 014, each of which is incorporated herein by reference.
Compositions and formulations for parenteral, intraparenchymal (into the brain), intrathecal, entricular or intrahepatic administration can include sterile aqueous solutions which can also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.
Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome—containing formulations. These compositions can be generated from a variety of components that include, but are not limited to, preformed s, self—emulsifying solids and self—emulsifying semisolids. ularly preferred are formulations that target the liver when treating hepatic disorders such as hepatic carcinoma.
The pharmaceutical formulations of the present ion, which can conveniently be presented in unit dosage form, can be prepared according to conventional techniques well known in the pharmaceutical ry. Such ques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the t.
The compositions of the present invention can be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions of the present ion can also be formulated as suspensions in aqueous, non—aqueous or mixed media. Aqueous suspefins can further contain substances which increase the viscosity of the suspension [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The sion can also contain stabilizers.
C. Additional Formulations Emulsions The compositions of the present invention can be prepared and formulated as emulsions. Emulsions are typically heterogeneous systems of one liquid dispersed in another in the form of droplets usually ing 0.1 pm in diameter (see 6.g. s Pharmaceutical Dosage Forms and Drug ry Systems, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, NY, volume 1, p. 199; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker , 1988, Marcel Dekker, Inc., New York, NY, Volume 1, p. 245; Block in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, NY, volume 2, p. 335; i et al., in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 301). Emulsions are often biphasic systems comprising two immiscible liquid phases intimately mixed and dispersed with each other. In general, emulsions can be of either the water—in—oil (w/o) or the oil—in—water (o/w) variety. When an aqueous phase is finely divided into and sed as minute droplets into a bulk oily phase, the resulting composition is called a in—oil (w/o) emulsion.
Alternatively, when an oily phase is finely divided into and dispersed as minute ts into a bulk aqueous phase, the resulting composition is called an oil—in—water (o/w) emulsion.
Emulsions can contain additional components in addition to the dispersed , and the active drug which can be present as a solution in either the aqueous phase, oily phase or itself as a separate phase. Pharmaceutical excipients such as emulsifiers, stabilizers, dyes, and anti— oxidants can also be present in emulsions as needed. Pharmaceutical ons can also be multiple emulsions that are comprised of more than two phases such as, for example, in the case of oil—in—water—in—oil (o/w/o) and water—in—oil—in—water (w/o/w) emulsions. Such complex formulations often provide certain advantages that simple binary emulsions do not.
Multiple emulsions in which individual oil ts of an o/w emulsion enclose small water droplets constitute a w/o/w emulsion. Likewise a system of oil droplets enclosed in globules of water stabilized in an oily continuous phase provides an o/w/o emulsion.
Emulsions are characterized by little or no thermodynamic stability. Often, the dispersed or discontinuous phase of the on is well sed into the external or continuous phase and maintained in this form through the means of emulsifiers or the viscosity of the formulation. Either of the phases of the emulsion can be a lid or a solid, as is the case of emulsion-style ointment bases and creams. Other means of stabilizing emulsions entail the use of emulsifiers that can be incorporated into either phase of the emuls'fi Emulsifiers can broadly be classified into four categories: synthetic surfactants, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM naturally occurring emulsifiers, absorption bases, and finely dispersed solids (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug ry s, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). tic surfactants, also known as surface active agents, have found wide applicability in the formulation of ons and have been reviewed in the literature (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; , in ceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), Marcel Dekker, Inc., New York, N.Y., 1988, volume 1, p. 199). tants are typically amphiphilic and comprise a hydrophilic and a hydrophobic portion. The ratio of the hydrophilic to the hydrophobic nature of the surfactant has been termed the hydrophile/lipophile balance (HLB) and is a valuable tool in categorizing and ing surfactants in the ation of formulations. Surfactants can be classified into different classes based on the nature of the hydrophilic group: nonionic, c, cationic and amphoteric (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug ry Systems, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285).
Naturally occurring emulsifiers used in emulsion formulations include lanolin, beeswax, phosphatides, lecithin and acacia. Absorption bases possess hydrophilic ties such that they can soak up water to form w/o emulsions yet retain their semisolid consistencies, such as anhydrous lanolin and hydrophilic petrolatum. Finely divided solids have also been used as good emulsifiers especially in combination with tants and in viscous ations. These include polar inorganic solids, such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin, montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and nonpolar solids such as carbon or glyceryl tristearate.
A large variety of non—emulsifying materials are also included in emulsion formulations and contribute to the properties of emulsions. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hilic colloids, vatives and antioxidants (Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Hydrophilic ds or hydrocolloids include lly occurring gums and synthetic polymers such as polysaccharides (for example, acacia, agar, alginic acid, carrageenan, guar gum, karaya gum, and anth), cellulose derivatives (for e, carboxymethylcellulose and carboxypropylcellulose), and synthetic polymers (for example, carbomers, cellulose ethers, and carboxyvinyl polymers). These disperse or swell in water to form colloidal solutions that stabilize emulsions by forming strong interfacial films around the dispersed—phase droplets and by increasing the viscosity of the external phase.
Since ons often contain a number of ingredients such as carbohydrates, proteins, sterols and phosphatides that can readily support the growth of microbes, these formulations often incorporate preservatives. Commonly used preservatives included in emulsion ations include methyl paraben, propyl paraben, quaternary ammonium salts, benzalkonium chloride, esters of p—hydroxybenzoic acid, and boric acid. Antioxidants are also commonly added to emulsion formulations to t deterioration of the formulation.
Antioxidants used can be free radical scavengers such as tocopherols, alkyl gallates, butylated hydroxyanisole, butylated ytoluene, or reducing agents such as ic acid and sodium metabisulfite, and antioxidant synergists such as citric acid, tartaric acid, and lecithin.
The application of emulsion formulations via dermatological, oral and eral routes and methods for their manufacture have been reviewed in the literature (see 6.g.
Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich 2O NG., and Ansel HC., 2004, Lippincott ms & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Emulsion formulations for oral delivery have been very widely used because of ease of ation, as well as efficacy from an absorption and bioavailability standpoint (see e.g. Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Mineral—oil base laxatives, oil—soluble vitamins and high fat nutritive preparations are among the materials that have commonly been administered orally as o/w ons. ii. Microemulsions In one embodiment of the present ion, the compositions of iRNAs and nucleic acids are formulated as microemulsions. A microemulsion can be defined as a system of water, oil and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug ry Systems, Allen, LV., ch NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New Hg NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM (Eds), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Typically microemulsions are systems that are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component, generally an intermediate chain—length alcohol to form a transparent system. Therefore, mulsions have also been described as thermodynamically stable, isotropically clear dispersions of two immiscible liquids that are stabilized by interfacial films of surface-active molecules (Leung and Shah, in: Controlled Release of Drugs: rs and Aggregate Systems, Rosoff, M., Ed., 1989, VCH Publishers, New York, pages 185—215). Microemulsions commonly are prepared via a combination of three to five components that include oil, water, surfactant, cosurfactant and electrolyte. r the microemulsion is of the water—in-oil (w/o) or an oil— in—water (o/w) type is dependent on the properties of the oil and surfactant used and on the structure and geometric packing of the polar heads and hydrocarbon tails of the surfactant molecules t, in Remington's Pharmaceutical es, Mack Publishing Co., Easton, Pa., 1985, p. 271).
The phenomenological approach utilizing phase diagrams has been extensively studied and has yielded a comprehensive knowledge, to one skilled in the art, of how to formulate microemulsions (see e.g., s Pharmaceutical Dosage Forms and Drug Delivery s, Allen, LV., Popovich NG., and Ansel HC., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel , Inc., New York, N.Y., volume 1, p. 245; Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel , Inc., New York, N.Y., volume 1, p. 335). Compared to conventional emulsions, microemulsions offer the advantage of solubilizing water—insoluble drugs in a formulation of thermodynamically stable ts that are formed spontaneously. tants used in the preparation of microemulsions include, but are not limited to, ionic surfactants, non-ionic surfactants, Brij 96, polyoxyethylene oleyl , polyglycerol fatty acid esters, tetraglycerol monolaurate (ML310), tetraglycerol monooleate (M0310), hexaglycerol monooleate (P0310), hexaglycerol pentaoleate ), decaglycerol monocaprate (MCA750), ycerol monooleate (M0750), decaglycerol sequioleate (S0750), decaglycerol decaoleate (DA0750), alone or in combination with cosurfactants.
The cosurfactant, usually a short—chain l such as l, 1—propanol, and 1—butanol, serves to increase the interfacial fluidity by penetrating into the surfactant film and consequently creating a disordered film because of the void space generated among surfactant molecules. Microemulsions can, however, be prepared without the use of cosurfactants and alcohol—free self—emulsifying microemulsion systems are known in the art. The aqueous phase can lly be, but is not limited to, water, an aqueous solution of the drug, glycerol, PEG300, PEG400, polyglycerols, propylene glycols, and derivatives of ethylene glycol. The oil phfican include, but is not limited to, materials such as Captex 300, Captex 355, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Capmul MCM, fatty acid esters, medium chain 2) mono, di, and tri—glycerides, polyoxyethylated glyceryl fatty acid esters, fatty ls, polyglycolized glycerides, saturated ycolized C8—C10 glycerides, vegetable oils and silicone oil.
Microemulsions are particularly of interest from the standpoint of drug lization and the enhanced absorption of drugs. Lipid based microemulsions (both o/W and W/o) have been proposed to enhance the oral bioavailability of drugs, including peptides (see e. 57., US.
Patent Nos. 6,191,105; 7,063,860; 7,070,802; 7,157,099; Constantinides et al., Pharmaceutical Research, 1994, 11, 1385—1390; Ritschel, Meth. Find. Exp. Clin.
Pharmacol, 1993, 13, 205). Microemulsions afford advantages of improved drug solubilization, protection of drug from enzymatic hydrolysis, possible enhancement of drug absorption due to surfactant—induced alterations in membrane fluidity and permeability, ease of preparation, ease of oral stration over solid dosage forms, ed clinical potency, and decreased toxicity (see e.g., US. Patent Nos. 6,191,105; 7,063,860; 7,070,802; 7,157,099; Constantinides et al., Pharmaceutical Research, 1994, 11, 1385; Ho et al., J.
Pharm. Sci, 1996, 85, 138-143). Often microemulsions can form spontaneously when their components are brought er at ambient ature. This can be particularly advantageous When formulating thermolabile drugs, peptides or iRNAs. mulsions have also been effective in the transdermal delivery of active components in both cosmetic and pharmaceutical applications. It is expected that the microemulsion compositions and formulations of the present invention Will facilitate the increased systemic absorption of iRNAs and nucleic acids from the gastrointestinal tract, as well as improve the local cellular uptake of iRNAs and nucleic acids.
Microemulsions of the present ion can also contain onal components and additives such as sorbitan monostearate (Grill 3), Labrasol, and penetration enhancers to e the properties of the formulation and to e the absorption of the iRNAs and nucleic acids of the present invention. Penetration enhancers used in the microemulsions of the present invention can be classified as belonging to one of five broad categories—— surfactants, fatty acids, bile salts, chelating agents, and non-chelating non—surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier s, 1991, p. 92). Each of these classes has been discussed above. iii. Microparticles An RNAi agent of the invention may be incorporated into a particle, e. g., a microparticle. Microparticles can be produced by spray—drying, but may also be produced by other methods ing lyophilization, evaporation, fluid bed drying, vacuum drying, or a combination of these techniques. iv. Penetration Enhancers In one embodiment, the present invention employs various ation enhancers to effectfiefficient ry of nucleic acids, particularly iRNAs, to the skin of animals. Most [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has been discovered that even non—lipophilic drugs can cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of pophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs.
Penetration enhancers can be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non—chelating non—surfactants (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p.92). Each of the above mentioned classes of penetration enhancers are described below in greater detail.
Surfactants (or ce—active agents") are chemical entities which, when dissolved in an aqueous solution, reduce the surface tension of the solution or the interfacial tension between the aqueous solution and another , with the result that absorption of iRNAs through the mucosa is enhanced. In addition to bile salts and fatty acids, these penetration enhancers include, for example, sodium lauryl sulfate, polyoxyethylene—9—lauryl ether and polyoxyethylene—20—cetyl ether) (see 6.5)., Malmsten, M. Surfactants and rs in drug delivery, Informa Health Care, New York, NY, 2002; Lee et 511., al Reviews in eutic Drug Carrier Systems, 1991, p.92); and rochemical emulsions, such as FC—43. Takahashi er al., J. Pharm. Pharmacol., 1988, 40, 252). s fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid (n—decanoic acid), myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein (1—monooleoyl—rac— ol), dilaurin, caprylic acid, arachidonic acid, glycerol 1—monocaprate, 1— dodecylazacycloheptan—2—one, acylcamitines, acylcholines, C1_20 alkyl esters thereof (e.g., methyl, isopropyl and t—butyl), and mono— and di—glycerides f (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.) (see e.g., Touitou, E., et al.
Enhancement in Drug Delivery, CRC Press, Danvers, MA, 2006; Lee et a!., al s in Therapeutic Drug Carrier Systems, 1991, p.92; Muranishi, Critical Reviews in eutic Drug Carrier s, 1990, 7, 1—33; El Hariri et aL, J. Pharm. Pharmacol., 1992, 44, 651- 654).
The logical role of bile includes the facilitation of dispersion and tion of lipids and fat—soluble vitamins (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Brunton, Chapter 38 in: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al. Eds., McGraw— Hill, New York, 1996, pp. 934-935). Various natural bile salts, and their synthetic derivatives, act as ation enhancers. Thus the term "bile salts" includes any of the naturafisccurring components of bile as well as any of their synthetic derivatives. Suitable [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM bile salts include, for example, cholic acid (or its pharmaceutically acceptable sodium salt, sodium cholate), dehydrocholic acid (sodium dehydrocholate), deoxycholic acid (sodium holate), lic acid (sodium glucholate), glycholic acid (sodium glycocholate), glycodeoxycholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chenodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25—dihydro—fusidate (STDHF), sodium ihydrofusidate and yethylene—9—lauryl ether (POE) (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, a Health Care, New York, NY, 2002; Lee et 5123., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; rd, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., o, ed., Mack Publishing Co., Easton, Pa., 1990, pages 782—783; Muranishi, Critical Reviews in eutic Drug Carrier Systems, 1990, 7, 1—33; Yamamoto et al., J. Pharm. Exp. Ther., 1992, 263, 25; Yamashita et al., J. Pharm. Sci., 1990, 79, 579-583).
Chelating agents, as used in connection with the present invention, can be defined as compounds that remove metallic ions from solution by forming complexes therewith, with the result that absorption of iRNAs through the mucosa is enhanced. With regards to their use as penetration enhancers in the present invention, chelating agents have the added advantage of also serving as DNase inhibitors, as most characterized DNA ses require a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 618, 315—339). Suitable ing agents e but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (6.57., sodium salicylate, 5— methoxysalicylate and homovanilate), N—acyl derivatives of en, laureth—9 and N—amino acyl derivatives of beta—diketones (enamines)(see e.g., Katdare, A. et al., Excipient development for ceutical, biotechnology, and drug delivery, CRC Press, Danvers, MA, 2006; Lee et 51]., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug r Systems, 1990, 7, 1—33; Buur et (11., J. Control Rel., 1990, 14, 43-51).
As used herein, non—chelating non—surfactant penetration ing compounds can be defined as compounds that demonstrate insignificant activity as chelating agents or as surfactants but that nonetheless enhance absorption of iRNAs through the alimentary mucosa (see e.g., Muranishi, Critical s in Therapeutic Drug Carrier Systems, 1990, 7, 1—33).
This class of penetration enhancers includes, for example, rated cyclic ureas, 1—alkyl- and 1—alkenylazacyclo—alkanone derivatives (Lee et al., Critical Reviews in Therapeutic Drug Carrier s, 1991, page 92); and non—steroidal anti—inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone hita et al., J. Pharm.
Pharmacol., 1987, 39, 621-626).
Agents that enhance uptake of iRNAs at the cellular level can also be added to the pharrrfiutical and other compositions of the present invention. For example, cationic lipids, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM such as lipofectin (Junichi et al, US. Pat. No. 5,705,188), cationic glycerol derivatives, and polycationic molecules, such as polylysine (Lollo er (11., PCT ation WO 97/30731), are also known to enhance the cellular uptake of dsRNAs. Examples of cially available transfection reagents include, for example LipofectamineTM (Invitrogen; Carlsbad, CA), Lipofectamine 2000TM (Invitrogen; Carlsbad, CA), 293fectinTM (Invitrogen; Carlsbad, CA), CellfectinTM (Invitrogen; ad, CA), DMRIE—CTM (Invitrogen; Carlsbad, CA), FreeStyleTM MAX (Invitrogen; Carlsbad, CA), LipofectamineTM 2000 CD (Invitrogen; Carlsbad, CA), LipofectamineTM (Invitrogen; Carlsbad, CA), RNAiMAX rogen; Carlsbad, CA), OligofectamineTM (Invitrogen; Carlsbad, CA), OptifectTM (Invitrogen; Carlsbad, CA), X—tremeGENE Q2 Transfection Reagent (Roche; cherstrasse, Switzerland), DOTAP Liposomal Transfection Reagent acherstrasse, Switzerland), DOSPER Liposomal ection Reagent (Grenzacherstrasse, Switzerland), or Fugene (Grenzacherstrasse, Switzerland), Transfectam® Reagent (Promega; Madison, WI), TransFastTM ection Reagent (Promega; Madison, WI), foTM—20 Reagent (Promega; Madison, WI), foTM—50 Reagent ga; Madison, WI), DreamFectTM (OZ Biosciences; Marseille, France), EcoTransfect (OZ Biosciences; Marseille, France), TransPass" Dl Transfection t (New England Biolabs; Ipswich, MA, USA), LyoVecTM/LipoGenTM (Invitrogen; San Diego, CA, USA), PerFectin Transfection Reagent (Genlantis; San Diego, CA, USA), NeuroPORTER Transfection Reagent (Genlantis; San Diego, CA, USA), GenePORTER Transfection reagent (Genlantis; San Diego, CA, USA), GenePORTER 2 Transfection reagent (Genlantis; San Diego, CA, USA), Cytofectin Transfection Reagent (Genlantis; San Diego, CA, USA), BaculoPORTER Transfection Reagent (Genlantis; San Diego, CA, USA), TroganPORTERTM transfection Reagent (Genlantis; San Diego, CA, USA ), RiboFect ne; Taunton, MA, USA), PlasFect (Bioline; Taunton, MA, USA), UniFECTOR (B—Bridge International; Mountain View, CA, USA), SureFECTOR (B—Bridge International; Mountain View, CA, USA), or HiFectTM (B—Bridge International, Mountain View, CA, USA), among .
Other agents can be utilized to enhance the penetration of the administered nucleic acids, including glycols such as ethylene glycol and ene glycol, s such as 2— pyrrol, azones, and terpenes such as limonene and menthone. v. Carriers n compositions of the present invention also incorporate carrier nds in the formulation. As used herein, "carrier compound" or "carrier" can refer to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological ty per se) but is recognized as a nucleic acid by in viva processes that reduce the bioavailability of a nucleic acid having biological activity by, for e, degrading the biologically active nucleic acid or promoting its l from circulation. The coadministration of a nucleic acid and a carriefimpound, typically with an excess of the latter substance, can result in a substantial ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM reduction of the amount of nucleic acid recovered in the liver, kidney or other irculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor. For example, the recovery of a partially phosphorothioate dsRNA in hepatic tissue can be reduced when it is coadministered with polyinosinic acid, dextran sulfate, polycytidic acid or amido—4'isothiocyano—stilbene— 2,2'—disulfonic acid (Miyao et al., DsRNA Res. Dev., 1995, 5, 1; Takakura et al., DsRNA & Nucl. Acid Drug Dev., 1996, 6, 177—183. vi. Excipients In contrast to a carrier compound, a "pharmaceutical carrier" or "excipient" is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert e for delivering one or more nucleic acids to an animal. The excipient can be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical ition. Typical ceutical carriers include, but are not limited to, g agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, eta); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc.); lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, ic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium te, sodium acetate, eta); disintegrants (e.g., , sodium starch glycolate, etc.); and wetting agents (e.g., sodium lauryl sulphate, etc).
Pharmaceutically acceptable organic or inorganic excipients suitable for non— parenteral administration which do not deleteriously react with c acids can also be used to formulate the compositions of the present invention. Suitable pharmaceutically able carriers include, but are not limited to, water, salt solutions, alcohols, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, nylpyrrolidone and the like.
Formulations for topical administration of nucleic acids can include sterile and non- sterile aqueous solutions, non—aqueous solutions in common ts such as alcohols, or solutions of the nucleic acids in liquid or solid oil bases. The ons can also contain buffers, diluents and other suitable additives. Pharmaceutically able organic or nic excipients suitable for non-parenteral administration which do not deleteriously react with nucleic acids can be used.
Suitable pharmaceutically able excipients include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like. vii. Other Components [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM The compositions of the present invention can additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art—established usage levels. Thus, for example, the compositions can contain onal, compatible, pharmaceutically—active materials such as, for example, antipruritics, astringents, local anesthetics or anti—inflammatory agents, or can contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, iers, thickening agents and izers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if d, mixed with auxiliary , e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic re, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the nucleic acid(s) of the formulation. s suspensions can contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran.
The suspension can also contain stabilizers.
In some embodiments, pharmaceutical compositions featured in the invention include (a) one or more iRNA compounds and (b) one or more agents which function by a non—RNAi mechanism and which are useful in treating a bleeding disorder. Examples of such agents include, but are not lmited to an anti-inflammatory agent, anti—steatosis agent, anti-viral, and/or ibrosis agent. In addition, other substances commonly used to protect the liver, such as silymarin, can also be used in conjunction with the iRNAs described herein. Other agents useful for treating liver es include telbivudine, entecavir, and protease inhibitors such as telaprevir and other disclosed, for example, in Tung et al., US. Application Publication Nos. 2005/0148548, 2004/0167116, and 2003/0144217; and in Hale er al., US.
Application Publication No. 2004/0127488.
Toxicity and therapeutic efficacy of such compounds can be determined by standard ceutical procedures in cell cultures or mental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are red.
The data ed from cell e assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of compositions ed herein in the invention lies generally within a range of ating concentrations that include the ED50 with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the fihods featured in the ion, the eutically effective dose can be estimated [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM initially from cell culture assays. A dose can be formulated in animal models to e a circulating plasma concentration range of the compound or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a sed concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test compound which achieves a half—maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma can be measured, for e, by high performance liquid chromatography.
In addition to their administration, as discussed above, the iRNAs featured in the invention can be administered in combination with other known agents ive in treatment of pathological processes that are mediated by iron overload and that can be treated by inhibiting TMPRSS6 sion. In any event, the administering physician can adjust the amount and timing of iRNA administration on the basis of s observed using rd measures of cy known in the art or described herein.
V. Methods For Inhibiting TMPRSS6 Expression The present invention provides methods of inhibiting sion of TMPRSS6 (matriptase—Z) in a cell. The methods include contacting a cell with an RNAi agent, 6.57., a double stranded RNAi agent, in an amount effective to inhibit expression of the TMPRSS6 in the cell, thereby inhibiting expression of the TMPRSS6 in the cell.
Contacting of a cell with a double stranded RNAi agent may be done in vitra or in viva. Contacting a cell in viva with the RNAi agent includes contacting a cell or group of cells within a subject, e.g., a human subject, with the RNAi agent. Combinations of in vitra and in viva methods of contacting are also possible. Contacting may be direct or indirect, as discussed above. Furthermore, contacting a cell may be accomplished via a targeting ligand, including any ligand described herein or known in the art. In preferred embodiments, the targeting ligand is a carbohydrate , e.g., a GalNAC3 ligand, or any other ligand that directs the RNAi agent to a site of interest, e.g., the liver of a subject.
The term "inhibiting," as used herein, is used interchangeably with "reducing," "silencing," egulating" and other similar terms, and includes any level of tion.
The phrase "inhibiting expression of a TMPRSS6" is ed to refer to inhibition of expression of any TMPRSS6 gene (such as, e.g., a mouse TMPRSS6 gene, a rat 6 gene, a monkey TMPRSS6 gene, or a human TMPRSS6 gene) as well as variants or mutants of a TMPRSS6 gene. Thus, the TMPRSS6 gene may be a wild—type TMPRSS6 gene, a mutant TMPRSS6 gene, or a enic 6 gene in the context of a genetically manipulated cell, group of cells, or organism.
"Inhibiting expression of a TMPRSS6 gene" includes any level of inhibition of a TMPRSS6 gene, e.g., at least partial suppression of the expression of a TMPRSS6 gene. The exprefin of the TMPRSS6 gene may be assessed based on the level, or the change in the [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM ed set by KIM level, of any variable associated with TMPRSS6 gene expression, e.g., TMPRSS6 mRNA level, TMPRSS6 protein level, or lipid levels. This level may be assessed in an individual cell or in a group of cells, including, for example, a sample derived from a subject.
Inhibition may be assessed by a decrease in an absolute or relative level of one or more variables that are associated with TMPRSS6 expression compared with a control level.
The control level may be any type of control level that is ed in the art, e.g., a pre—dose baseline level, or a level determined from a similar subject, cell, or sample that is untreated or treated with a control (such as, 6.5)., buffer only control or inactive agent control).
In some embodiments of the methods of the invention, expression of a TMPRSS6 gene is inhibited by at least about 5%, at least about 10%, at least about 15%, at least about %, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%. at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%.
Inhibition of the expression of a TMPRSS6 gene may be manifested by a reduction of the amount of mRNA expressed by a first cell or group of cells (such cells may be present, for example, in a sample d from a subject) in which a TMPRSS6 gene is ribed and which has or have been treated (e.g., by ting the cell or cells with an RNAi agent of the invention, or by administering an RNAi agent of the invention to a subject in which the cells are or were present) such that the expression of a TMPRSS6 gene is inhibited, as compared to a second cell or group of cells ntially identical to the first cell or group of cells but which has not or have not been so treated (control cell(s)). In red embodiments, the inhibition is assessed by expressing the level of mRNA in treated cells as a percentage of the level of mRNA in l cells, using the following formula: (mRNA in control cells) - (mRNA in treated cells) —. 10070 (mRNA in control cells) Alternatively, tion of the expression of a TMPRSS6 gene may be ed in terms of a reduction of a parameter that is functionally linked to TMPRSS6 gene expression, e.g., TMPRSS6 n expression, hepcidin gene or protein expression, or iron levels in tissues or serum. TMPRSS6 gene silencing may be ined in any cell expressing TMPRSS6, either constitutively or by genomic engineering, and by any assay known in the art. The liver is the major site of TMPRSS6 expression. Other significant sites of expression include the kidneys and the uterus.
Inhibition of the expression of a TMPRSS6 protein may be manifested by a reduction in the level of the TMPRSS6 protein that is expressed by a cell or group of cells (e.g., the level of protein expressed in a sample derived from a subject). As explained above for the assessment of mRNA suppression, the ton of protein expression levels in a treated cell [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM or group of cells may similarly be sed as a percentage of the level of protein in a l cell or group of cells.
A control cell or group of cells that may be used to assess the tion of the expression of a TMPRSS6 gene includes a cell or group of cells that has not yet been contacted with an RNAi agent of the invention. For e, the control cell or group of cells may be derived from an individual subject (6.g. a human or animal subject) prior to treatment of the subject with an RNAi agent.
The level of TMPRSS6 mRNA that is expressed by a cell or group of cells may be determined using any method known in the art for ing mRNA expression. In one embodiment, the level of expression of TMPRSS6 in a sample is determined by detecting a transcribed cleotide, or portion thereof, e.g., mRNA of the TMPRSS6 gene. RNA may be extracted from cells using RNA extraction techniques including, for example, using acid phenol/guanidine isothiocyanate extraction (RNAzol B; Biogenesis), RNeasy RNA preparation kits (Qiagen) or PAXgene (PreAnalytix, Switzerland). Typical assay formats utilizing ribonucleic acid hybridization include nuclear run—on assays, RT—PCR, RNase protection assays (Melton er al., Nuc. Acids Res. 12:7035), Northern blotting, in situ hybridization, and microarray analysis.
In one embodiment, the level of expression of TMPRSS6 is determined using a nucleic acid probe. The term "probe", as used , refers to any molecule that is capable 2O of selectively binding to a specific TMPRSS6. Probes can be synthesized by one of skill in the art, or derived from appropriate biological preparations. Probes may be specifically designed to be d. es of molecules that can be utilized as probes include, but are not limited to, RNA, DNA, proteins, antibodies, and organic molecules.
Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, rase chain on (PCR) analyses and probe arrays. One method for the determination of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to 6 mRNA.
In one embodiment, the mRNA is immobilized on a solid surface and contacted with a probe, for example by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a ne, such as ellulose. In an alternative embodiment, the probe(s) are immobilized on a solid surface and the mRNA is contacted with the probe(s), for example, in an Affymetrix gene chip array. A skilled artisan can readily adapt known mRNA detection methods for use in determining the level of TMPRSS6 mRNA.
An alternative method for determining the level of expression of TMPRSS6 in a sample involves the process of nucleic acid amplification and/or reverse transcriptase (to e cDNA) of for example mRNA in the , e.g., by RT-PCR (the experimental ment set forth in Mullis, 1987, U.S. Pat. No. 4,683,202), ligase chain reaction (Barany (1991 00. Natl. Acad. Sci. USA 88:189-193), self sustained sequence replication (Guatelli [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM et al. (1990) Proc. Natl. Acad. Sci. USA 87:1874—1878), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 3—1177), Q—Beta Replicase (Lizardi et al. (1988) Bio/Fechnology 6: 1 197), rolling circle replication (Lizardi et al., US. Pat. No. ,854,033) or any other nucleic acid amplification , followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. In particular aspects of the invention, the level of expression of 6 is ined by quantitative fluorogenic RT—PCR (i.e., the TaqManTM System).
The expression levels of TMPRSS6 mRNA may be monitored using a membrane blot (such as used in hybridization analysis such as Northern, rn, dot, and the like), or ells, sample tubes, gels, beads or fibers (or any solid support comprising bound nucleic acids). See US Pat. Nos. 5,770,722, 5,874,219, 5,744,305, 5,677,195 and 5,445,934, which are incorporated herein by reference. The determination of TMPRSS6 expression level may also comprise using nucleic acid probes in solution.
In preferred embodiments, the level of mRNA expression is assessed using branched DNA (bDNA) assays or real time PCR (qPCR). The use of these methods is described and exemplified in the es presented herein.
The level of TMPRSS6 protein sion may be determined using any method known in the art for the measurement of protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, fluid or gel precipitin reactions, absorption spectroscopy, a colorimetric assays, ophotometric assays, flow cytometry, immunodiffusion (single or ), immunoelectrophoresis, Western blotting, radioimmunoassay (RIA), enzyme-linked sorbent assays (ELISAS), immunofluorescent assays, electrochemiluminescence assays, and the like.
The term "sample" as used herein refers to a tion of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject.
Examples of biological fluids include blood, serum and serosal fluids, plasma, lymph, urine, cerebrospinal fluid, saliva, ocular fluids, and the like. Tissue samples may include samples from tissues, organs or localized regions. For example, samples may be derived from particular organs, parts of , or fluids or cells within those organs. In certain embodiments, samples may be derived from the liver (e.g., whole liver or certain segments of liver or certain types of cells in the liver, such as, e.g., hepatocytes). In preferred embodiments, a e derived from a subject" refers to blood or plasma drawn from the subject. In further embodiments, a "sample derived from a t" refers to liver tissue derived from the subject.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM In some embodiments of the methods of the invention, the RNAi agent is administered to a subject such that the RNAi agent is delivered to a specific site within the subject. The tion of expression of TMPRSS6 may be assessed using measurements of the level or change in the level of TMPRSS6 mRNA or TMPRSS6 protein in a sample derived from fluid or tissue from the specific site within the subject. In preferred embodiments, the site is the liver. The site may also be a subsection or subgroup of cells from any one of the aforementioned sites. The site may also include cells that express a particular type of receptor.
VI. Methods for Treating or ting a TMPRSS6 Associated Disorder The present invention also provides methods for treating or ting diseases and conditions that can be modulated by TMPRSS6 gene sion. For example, the compositions bed herein can be used to treat any disorder associated with iron overload, e.g., a thalassemia (e.g., [3—thalassemia or a-thalassemia), primary hemochromatosis, secondary hemochromatosis, severe juvenile hemochromatosis, erythropoietic ria, sideroblastic anemia, hemolytic anemia, thropoietic anemia, or sickle—cell anemia. In one embodiment, a TMPRSS6 iRNA is used to treat a hemoglobinopathy. The TMPRSS6 iRNAs of the ion can also be used to treat elevated levels of iron due to other conditions, such as chronic alcoholism.
In thalassemias, the bone marrow sizes insufficient amounts of a hemoglobin chain; this in turn reduces the production of red blood cells and causes anemia. Either the ct or the [3 chain may be affected, but [3 thalassemias are more common. Newborn babies are healthy because their bodies still produce HbF, which does not have [3 chains; during the first few months of life, the bone marrow es to producing HbA, and symptoms start to appear. [3—thalassemias result from mutation with either non-expressing ([30) or low expressing ([3+) alleles of the HBB gene, [3—thalassemias vary in severity depending on the pe, and include minor/trait [3-thalassemia ([3/ [3° or [3/ [3+), intermedia [3—thalassemia ([3°/[3+), and major [3—thalassemia ([30 /[3° or [3"7 [3+).
Thalassemia intermedia (TI) typically presents with little hemolysis, while major [3- thalassemia (TM) is typically accompanied by abundant hemolysis which causes, e.g., anemia and splenomegaly; and highly ctive erythropoiesis, which causes bone marrow drive tal changes, oteopenia), increased erythropoietin synthesis, hepato—splenomegaly, ption of haematinics (megablastic anemia), and high uric acid in blood. The iRNAs of the invention, e.g., TMPRSS6 iRNAs, are better suited for treating the iron overload that typically accompanies thalassemia's that are more TI like (e.g., for ng individuals having a [3°/[3+, [3/ [30 or [3/ [3+ genotype). ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM Symptoms of B—thalassemias also include, 6.5)., complication due to therapy, e.g., iron overload, which causes endocrinopathy, liver fibrosis and cardiac fibrosis. Administration of an iRNA agent that targets TMPRSS6 can be effective to treat one or more of these symptoms. a—thalassemias result from mutation with either non—expressing (a°) or low expressing (a+) alleles of the HBAl or HBA2 genes, orthalassemias vary in severity depending on the genotype, and include trait thalassemia (—(x/(xa), Hb Bart and Hydrops fetalis (aO /a°), a— Thalaseemia minor (— km), (—al—a), and HbH disease (—/—a). Lower a—globin chains are produced, resulting in an excess of B chains in adults and excess 7 chains in newborns. The excess B chains form unstable tetramers (called Hemoglobin H or HbH of 4 beta chains), which have abnormal oxygen dissociation . Administration of an iRNA agent that targets TMPRSS6 can be effective to treat iron overload in a subject who has an a- semias.
Symptoms of hemochromatosis include, e.g., abdominal pain, joint pain, fatigue, lack of energy, weakness, ing of the skin (often ed to as "bronzing"), and loss of body hair. Administration of an iRNA agent that targets TMPRSS6 can be ive to treat one or more of these symptoms.
Other symptoms associated with iron overload include increased risk for liver e (cirrhosis, cancer), heart attack or heart failure, diabetes mellitus, osteoarthritis, orosis, metabolic syndrome, hypothyroidism, hypogonadism, and in some cases ure death.
Iron mismanagement resulting in overload can also accelerate such neurodegenerative diseases as Alzheimer's, early—onset Parkinson's, Huntington's, epilepsy and multiple sclerosis. Administration of an iRNA agent that targets TMPRSS6, e.g., an iRNA described in Tables 1 or 2 can treat one or more of these symptoms, or prevent the development or progression of a disease or disorder that is aggrevated by increased iron levels.
The methods of the invention further relate to the use of an iRNA agent or a pharmaceutical composition thereof, e.g., for treating a disorder associated with iron overload, in combination with other pharmaceuticals and/or other therapeutic methods, e.g., with known pharmaceuticals and/or known therapeutic methods, such as, for example, those which are currently employed for treating these disorders. For e, in certain embodiments, an iRNA agent targeting TMPRSS6 is administered in combination with, e.g., iron chelators (e.g., desferoxamine), folic acid, a blood transfusion, a phlebotomy, agents to manage ulcers, agents to increase fetal hemoglobin levels (e.g., hydroxyurea), agents to l infection (e.g., otics and antivirals), agents to treat thrombotic state, or a stem cell or bone marrow transplant. A stem cell lant can utilize stem cells from an umbilical cord, such as from a relative, e.g., a g. Exemplary iron chelators include desferoxamine, Deferasirox (Exjade), deferiprone, vitamin E, wheat germ oil, tocophersolan, and indicafithin.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM The iRNA agent and an additional therapeutic agent can be administered in the same composition, e.g., parenterally, or the additional therapeutic agent can be administered as part of a separate composition or by another method described herein. Administration of the iRNA agent and the additional therapeutic agent can be at the same time, or at different times and, in any order.
Administration of the iRNA agent of the invention can lower iron levels, lower ferritin levels, and/or lower transferrin tion levels. For example, administration of the dsRNA can lower serum iron levels and/or lower serum ferritin levels. Transferrin saturation levels can be d by 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or more. In another embodiment, the transferrin saturation levels remain lower for 7 days, 10 days, 20 days, 30 days, or more following administration.
Transferrin tion levels can be lowered to below 50%, below 45%, below 40%, below 35%, below 35%, below 30%, below 25%, below 20%, below 15%, or lower. In another embodiment, the lower transferrin saturation levels are ined for 7 days, 10 days, 20 days, 30 days, or more following administration. Transferrin saturation is a measure of the amount of iron bound to serum transferrin, and ponds to the ratio of serum iron and total iron—binding capacity.
Serum iron levels can be lowered by 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more. In another embodiment, the serum iron levels remain lower for 7 days, 10 days, 20 days, 30 days, or more following administration.
Administration of the iRNA agent of the invention preferably results in d iron levels in the blood, and more particularly in the serum, or in one or more tissues of the . In some embodiments, iron levels are sed by at least 10%, 15%, 20%, 25%, %, 40%, 50%, 60%, 70%, 80%, 90% or more, as compared to pretreatment levels.
By "lower" in this context is meant a statistically significant decrease in such level.
The decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without such disorder.
Administration of the iRNA agent of the ion can increase serum hepcidin levels, and/or increase hepcidin gene expression. For example, administration of the dsRNA can increase serum in by at least about 10%, 25%, 50%, 100%, 150%, 200%, 250%, 300%, or more. In a further example, administration of the dsRNA can increase hepcidin mRNA levels by at least about 1.5—fold, 2—fold, 3—fold, 4—fold, 5—fold, or greater.
Efficacy of treatment or prevention of disease can be assessed, for example by measuring disease progression, e remission, symptom severity, reduction in pain, quality of life, dose of a tion required to sustain a treatment effect, level of a disease marker or any other measurable ter appropriate for a given disease being treated or targetfior prevention. It is well within the ability of one skilled in the art to monitor [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM efficacy of treatment or prevention by measuring any one of such parameters, or any combination of parameters. For example, the levels of transferrin saturation or serum ferritin can be red for efficacy of a given treatment regime.
Iron level tests are typically performed on a sample of a pateint's blood. An iron level test measure the amount of iron in the blood serum that is being d by the proteins transferrin. A TIBC (Total inding capacity) test measures the amount of iron that the blood would carry if the transferrin were fully saturated. Since transferrin is ed by the liver, the TIBC can be used to monitor liver function and nutrition. The transferrin test is a direct measure of errin (also called siderophilin) levels in the blood. The saturation level of transferrin can be ated by dividing the serum iron level by the TIBC. The ferritin test measures the level of a protein in the blood that stores iron for later use by the body.
The iRNA treatments bed herein can be used to treat individuals afflicted with a 6 associated disorder, e.g., elevated iron levels, as may be indicated by iron levels in serum e.g., iron levels measuring greater than 350 ug/dL, greater than 500 ug/dL, greater than 1000 ug/dL, or more. In an embodiment, ed levels of iron in serum, e.g., greater than , 20, 25, or 30 mg/g dry weight.
The iRNA treatments described herein can also be used to treat individuals having elevated iron levels, as may be indicated by elevated ferritin levels in serum, 6.g. ferritin levels measuring greater than 300 ug/L, greater than 500 ug/L, greater than 1000 ug/L, greater than 1500 ug/L, greater than 2000 ug/L, greater than 2500 ug/L, or 3000 ug/L, or more.
The iRNA ents described herein can further be used to treat individuals having elevated iron levels, as may be indicated by elevated transferrin levels in serum, 6.g. transferrin levels measuring greater than 400 mg/dL, greater than 500 mg/L, greater than 1000 mg/dL, or more.
The iRNA treatments described herein can also be used to treat individuals having moderately elevated iron levels, as may be indicated by moderately elevated transferrin saturation levels, e.g., saturation levels of 40%, 45%, or 50% or more. In addition, the treatment described herein may also be used to prevent elevated iron levels in individuals with only minor elevations in transferrin saturation. One of skill in the art can easily monitor the transferrin saturation levels in subjects ing treatment with iRNA as described herein and assay for a ion in transferrin saturation levels of at least 5% or 10%.
The iRNA treatments described herein can be used to treat individuals having elevated iron levels, as may be indicated by a TIBC value greater than 400 ug/dL, greater than 500 ug/dL, or greater than 1000 ug/dL, or more.
In some embodiments, individuals in need of treatment with an iRNA agent of the inven'fihave sed hematocrit , decreased hemoglobin levels, increased red blood [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM cell distribution width, increased number of reticulocytes, decreased number of mature red blood cells, sed unsaturated iron binding ty, decreased ineffective erythropoiesis, decreased extradedullary hematopoiesis, and/or decreased HAMPl expression levels.
A patient can be r monitored by assay of blood sugar (glucose) level or a fetoprotein level, by echocardiogram (e.g., to examine the s function), electrocardiogram (ECG) (e.g., to look at the electrical activity of the heart), imaging tests (such as CT scans, MRI and ultrasound), and liver function tests. Excess iron staining or iron concentrations can be ed on liver biopsy samples, or to confirm the extent of liver damage, e.g., the stage of liver disease.
A treatment or preventive effect is evident when there is a statistically significant improvement in one or more ters of disease status, or by a failure to worsen or to p ms where they would otherwise be anticipated. As an example, a favorable change of at least 10% in a able parameter of disease, and preferably at least 20%, %, 40%, 50% or more can be indicative of effective treatment. Efficacy for a given iRNA drug or formulation of that drug can also be judged using an experimental animal model for the given disease as known in the art. When using an experimental animal model, efficacy of treatment is evidenced when a statistically significant reduction in a marker or symptom is observed.
Alternatively, the efficacy can be measured by a reduction in the severity of disease as determined by one skilled in the art of diagnosis based on a clinically accepted disease severity grading scale.
As used herein, a "subject" includes a human or man animal, preferably a vertebrate, and more ably a mammal. A subject may include a transgenic organism.
Most preferably, the t is a human, such as a human suffering from or predisposed to developing a TMPRSS6 associated disorder.
In some embodiments of the methods of the ion, TMPRSS6 expression is decreased for an extended duration, e.g., at least one week, two weeks, three weeks, or four weeks or longer. For example, in certain instances, expression of the TMPRSS6 gene is suppressed by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 100% by administration of an iRNA agent described herein. In some embodiments, the TMPRSS6 gene is suppressed by at least about 60%, 70%, or 80% by administration of the iRNA agent. In some embodiments, the TMPRSS6 gene is suppressed by at least about 85%, 90%, or 95% by stration of the -stranded oligonucleotide. In another embodiment, the TMPRSS6 gene remains suppressed for 7 days, 10 days, 20 days, 30 days, or more following administration.
The RNAi agents of the invention may be administered to a subject using any mode of administration known in the art, including, but not limited to aneous, intravenous, intrarrfiular, intraocular, intrabronchial, intrapleural, intraperitoneal, intraarterial, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM lymphatic, cerebrospinal, and any combinations thereof. In preferred embodiments, the agents are administered subcutaneously.
In some ments, the administration is via a depot injection. A depot injection may release the RNAi agent in a consistent way over a prolonged time period. Thus, a depot injection may reduce the frequency of dosing needed to obtain a desired effect, e.g. , a desired inhibition of TMPRSS6, or a therapeutic or prophylactic effect. A depot ion may also provide more consistent serum concentrations. Depot ions may e subcutaneous ions or intramuscular injections. In preferred embodiments, the depot ion is a subcutaneous injection.
In some embodiments, the administration is via a pump. The pump may be an external pump or a surgically implanted pump. In certain embodiments, the pump is a subcutaneously implanted osmotic pump. In other embodiments, the pump is an infusion pump. An infusion pump may be used for intravenous, subcutaneous, arterial, or epidural infusions. In preferred embodiments, the infusion pump is a subcutaneous infusion pump. In other ments, the pump is a surgically ted pump that delivers the RNAi agent to the liver.
Other modes of administration include epidural, intracerebral, intracerebroventricular, nasal administration, rterial, intracardiac, intraosseous infusion, intrathecal, and intravitreal, and pulmonary. The mode of administration may be chosen based upon whether local or systemic treatment is desired and based upon the area to be treated. The route and site of administration may be chosen to enhance targeting.
The method includes administering an iRNA agent, e.g., a dose sufficient to depress levels of TMPRSS6 mRNA for at least 5, more ably 7, 10, 14, 21, 25, 30 or 40 days; and optionally, administering a second single dose of dsRNA, wherein the second single dose is administered at least 5, more preferably 7, 10, 14, 21, 25, 30 or 40 days after the first single dose is administered, thereby inhibiting the expression of the TMPRSS6 gene in a subject.
In one embodiment, doses of iRNA agent of the invention are administered not more than once every four weeks, not more than once every three weeks, not more than once every two weeks, or not more than once every week. In another ment, the administrations can be maintained for one, two, three, or six months, or one year or longer. In another embodiment, doses of iRNA agent of the ion are stered once a week for three weeks.
In general, the iRNA agent does not activate the immune system, e. g., it does not increase cytokine levels, such as TNF-alpha or IFN-alpha levels. For example, when measured by an assay, such as an in vitro PBMC assay, such as described herein, the increase in levels of TNF-alpha or IFN-alpha, is less than 30%, 20%, or 10% of control cells treated with a l dsRNA, such as a dsRNA that does not target TMPRSS6.
[Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM For example, a subject can be administered a therapeutic amount of an iRNA agent, such as 0.3 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, or 3 mg/kg of dsRNA. The iRNA agent can be administered by intravenous infusion over a period of time, such as over a 5 minute, 10 minute, 15 minute, 20 minute, or 25 minute period. The administration is repeated, for example, on a regular basis, such as biweekly (i.e., every two weeks) for one month, two months, three months, four months or longer. After an l treatment regimen, the treatments can be administered on a less frequent basis. For example, after stration biweekly for three , administration can be repeated once per month, for six months or a year or longer. stration of the iRNA agent can reduce TMPRSS6 levels, 6.g. in a cell, tissue, blood, urine or other compartment of the patient by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80 % or at least 90% or more.
Before stration of a full dose of the iRNA agent, patients can be administered a smaller dose, such as a dose resulting in less than 5% infusion reaction, and monitored for adverse effects, such as an allergic reaction, or for elevated lipid levels or blood pressure. In another e, the patient can be monitored for unwanted immunostimulatory effects, such as increased cytokine (e.g., TNF—alpha or INF—alpha) levels.
Many disorders associated with elevated iron levels are tary. ore, a patient in need of a TMPRSS6 iRNA may be identified by taking a family history. A healthcare provider, such as a doctor, nurse, or family member, can take a family history before prescribing or administering a TMPRSS6 dsRNA. A DNA test may also be performed on the patient to identify a mutation in the TMPRSS6 gene, before a TMPRSS6 dsRNA is administered to the patient. For example, diagnosis of hereditary hemochromatosis can be confirmed by identifying the two HFE (Hemochromatosis) gene mutations C282Y and H63D, according to GenBank Accession No. CAB07442.1 (GI: 1890180, record dated October 23, 2008).
A treatment or preventive effect is evident when there is a statistically significant improvement in one or more parameters of disease status, or by a failure to worsen or to develop symptoms where they would otherwise be anticipated. As an example, a favorable change of at least 10% in a measurable parameter of disease, and preferably at least 20%, %, 40%, 50% or more can be indicative of effective ent. cy for a given iRNA agent of the invention or formulation of that iRNA agent can also be judged using an experimental animal model for the given disease as known in the art. When using an mental animal model, efficacy of treatment is evidenced when a statistically significant reduction in a marker or symptom is observed.
In one embodiment, the RNAi agent is administered at a dose of n about 0.25 mg/kg to about 50 mg/kg, e.g., between about 0.25 mg/kg to about 0.5 mg/kg, between about 0.25 fig to about 1 mg/kg, between about 0.25 mg/kg to about 5 mg/kg, between about ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 0.25 mg/kg to about 10 mg/kg, between about 1 mg/kg to about 10 mg/kg, between about 5 mg/kg to about 15 mg/kg, between about 10 mg/kg to about 20 mg/kg, between about 15 mg/kg to about 25 mg/kg, between about 20 mg/kg to about 30 mg/kg, n about 25 mg/kg to about 35 mg/kg, or between about 40 mg/kg to about 50 mg/kg.
In some embodiments, the RNAi agent is administered at a dose of about 0.25 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, about 20 mg/kg, about 21 mg/kg, about 22 mg/kg, about 23 mg/kg, about 24 mg/kg, about 25 mg/kg, about 26 mg/kg, about 27 mg/kg, about 28 mg/kg, about 29 mg/kg, 30 mg/kg, about 31 mg/kg, about 32 mg/kg, about 33 mg/kg, about 34 mg/kg, about 35 mg/kg, about 36 mg/kg, about 37 mg/kg, about 38 mg/kg, about 39 mg/kg, about 40 mg/kg, about 41 mg/kg, about 42 mg/kg, about 43 mg/kg, about 44 mg/kg, about 45 mg/kg, about 46 mg/kg, about 47 mg/kg, about 48 mg/kg, about 49 mg/kg or about 50 mg/kg.
In certain embodiments, for example, when a composition of the invention comprises a dsRNA as described herein and a lipid, subjects can be administered a therapeutic amount of iRNA, such as about 0.01 mg/kg to about 5 mg/kg, about 0.01 mg/kg to about 10 mg/kg, about 0.05 mg/kg to about 5 mg/kg, about 0.05 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.2 mg/kg to about 5 mg/kg, about 0.2 mg/kg to about 10 mg/kg, about 0.3 mg/kg to about 5 mg/kg, about 0.3 mg/kg to about 10 mg/kg, about 0.4 mg/kg to about 5 mg/kg, about 0.4 mg/kg to about 10 mg/kg, about 0.5 mg/kg to about 5 mg/kg, about 0.5 mg/kg to about 10 mg/kg, about 1 mg/kg to about 5 mg/kg, about 1 mg/kg to about 10 mg/kg, about 1.5 mg/kg to about 5 mg/kg, about 1.5 mg/kg to about 10 mg/kg, about 2 mg/kg to about about 2.5 mg/kg, about 2 mg/kg to about 10 mg/kg, about 3 mg/kg to about 5 mg/kg, about 3 mg/kg to about 10 mg/kg, about 3.5 mg/kg to about 5 mg/kg, about 4 mg/kg to about 5 mg/kg, about 4.5 mg/kg to about 5 mg/kg, about 4 mg/kg to about 10 mg/kg, about 4.5 mg/kg to about 10 mg/kg, about 5 mg/kg to about 10 mg/kg, about 5.5 mg/kg to about 10 mg/kg, about 6 mg/kg to about 10 mg/kg, about 6.5 mg/kg to about 10 mg/kg, about 7 mg/kg to about 10 mg/kg, about 7.5 mg/kg to about 10 mg/kg, about 8 mg/kg to about 10 mg/kg, about 8.5 mg/kg to about 10 mg/kg, about 9 mg/kg to about 10 mg/kg, or about 9.5 mg/kg to about 10 mg/kg. Values and ranges intermediate to the recited values are also intended to be part of this invention.
For example, the dsRNA may be administered at a dose of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5, .1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7, 7.1, 7.2, 7.3, 7H5, 7.6, 7.7, 7.8, 7.9, 8, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9, 9.1, 9.2, 9.3, 9.4, [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 9.5, 9.6, 9.7, 9.8, 9.9, or about 10 mg/kg. Values and ranges intermediate to the d values are also intended to be part of this invention.
In certain embodiments of the invention, for example, When a double—stranded RNAi agent includes one or more modifications (e.g., motifs of three identical modifications on three consecutive nucleotides, including one such motif at or near the cleavage site of the agent), six phosphorothioate linkages, and a ligand, such an agent is administered at a dose of about 0.01 to about 0.5 mg/kg, about 0.01 to about 0.4 mg/kg, about 0.01 to about 0.3 mg/kg, about 0.01 to about 0.2 mg/kg, about 0.01 to about 0.1 mg/kg, about 0.01 mg/kg to about 0.09 mg/kg, about 0.01 mg/kg to about 0.08 mg/kg, about 0.01 mg/kg to about 0.07 mg/kg, about 0.01 mg/kg to about 0.06 mg/kg, about 0.01 mg/kg to about 0.05 mg/kg, about 0.02 to about 0.5 mg/kg, about 0.02 to about 0.4 mg/kg, about 0.02 to about 0.3 mg/kg, about 0.02 to about 0.2 mg/kg, about 0.02 to about 0.1 mg/kg, about 0.02 mg/kg to about 0.09 mg/kg, about 0.02 mg/kg to about 0.08 mg/kg, about 0.02 mg/kg to about 0.07 mg/kg, about 0.02 mg/kg to about 0.06 mg/kg, about 0.02 mg/kg to about 0.05 mg/kg, about 0.03 to about 0.5 mg/kg, about 0.03 to about 0.4 mg/kg, about 0.03 to about 0.3 mg/kg, about 0.03 to about 0.2 mg/kg, about 0.03 to about 0.1 mg/kg, about 0.03 mg/kg to about 0.09 mg/kg, about 0.03 mg/kg to about 0.08 mg/kg, about 0.03 mg/kg to about 0.07 mg/kg, about 0.03 mg/kg to about 0.06 mg/kg, about 0.03 mg/kg to about 0.05 mg/kg, about 0.04 to about 0.5 mg/kg, about 0.04 to about 0.4 mg/kg, about 0.04 to about 0.3 mg/kg, about 0.04 to about 0.2 mg/kg, about 0.04 to about 0.1 mg/kg, about 0.04 mg/kg to about 0.09 mg/kg, about 0.04 mg/kg to about 0.08 mg/kg, about 0.04 mg/kg to about 0.07 mg/kg, about 0.04 mg/kg to about 0.06 mg/kg, about 0.05 to about 0.5 mg/kg, about 0.05 to about 0.4 mg/kg, about 0.05 to about 0.3 mg/kg, about 0.05 to about 0.2 mg/kg, about 0.05 to about 0.1 mg/kg, about 0.05 mg/kg to about 0.09 mg/kg, about 0.05 mg/kg to about 0.08 mg/kg, or about 0.05 mg/kg to about 0.07 mg/kg.
Values and ranges ediate to the foregoing recited values are also intended to be part of this invention, e.g.,, the RNAi agent may be administered to the subject at a dose of about 0.015 mg/kg to about 0.45 mg/mg.
For e, the RNAi agent, e.g., RNAi agent in a pharmaceutical composition, may be administered at a dose of about 0.01 mg/kg, 0.0125 mg/kg, 0.015 mg/kg, 0.0175 mg/kg, 0.02 mg/kg, 0.0225 mg/kg, 0.025 mg/kg, 0.0275 mg/kg, 0.03 mg/kg, 0.0325 mg/kg, 0.035 mg/kg, 0.0375 mg/kg, 0.04 mg/kg, 0.0425 mg/kg, 0.045 mg/kg, 0.0475 mg/kg, 0.05 mg/kg, 0.0525 mg/kg, 0.055 mg/kg, 0.0575 mg/kg, 0.06 mg/kg, 0.0625 mg/kg, 0.065 mg/kg, 0.0675 mg/kg, 0.07 mg/kg, 0.0725 mg/kg, 0.075 mg/kg, 0.0775 mg/kg, 0.08 mg/kg, 0.0825 mg/kg, 0.085 mg/kg, 0.0875 mg/kg, 0.09 mg/kg, 0.0925 mg/kg, 0.095 mg/kg, 0.0975 mg/kg, 0.1 mg/kg, 0.125 mg/kg, 0.15 mg/kg, 0.175 mg/kg, 0.2 mg/kg, 0.225 mg/kg, 0.25 mg/kg, 0.275 mg/kg, 0.3 mg/kg, 0.325 mg/kg, 0.35 mg/kg, 0.375 mg/kg, 0.4 mg/kg, 0.425 mg/kg, 0.45 mg/kg, 0.475 mg/kg, or about 0.5 mg/kg. Values intermediate to the foregoing recited values are alfitended to be part of this invention.
[Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM The dose of an RNAi agent that is administered to a subject may be tailored to balance the risks and benefits of a particular dose, for example, to achieve a desired level of TMPRSS6 gene suppression (as assessed, e.g., based on TMPRSS6 mRNA suppression, TMPRSS6 protein expression, or a ion in lipid levels) or a desired therapeutic or prophylactic effect, while at the same time avoiding rable side effects.
In some embodiments, the RNAi agent is administered in two or more doses. If desired to facilitate repeated or frequent infusions, implantation of a delivery device, e.g., a pump, semi—permanent stent (e.g., intravenous, intraperitoneal, intracisternal or intracapsular), or reservoir may be ble. In some embodiments, the number or amount of subsequent doses is dependent on the achievement of a desired effect, e.g., the suppression of a TMPRSS6 gene, or the achievement of a therapeutic or prophylactic effect, e.g., reducing iron overload. In some embodiments, the RNAi agent is administered according to a schedule. For example, the RNAi agent may be administered once per week, twice per week, three times per week, four times per week, or five times per week. In some ments, the schedule involves regularly spaced administrations, e.g., hourly, every four hours, every six hours, every eight hours, every twelve hours, daily, every 2 days, every 3 days, every 4 days, every 5 days, weekly, biweekly, or y. In other embodiments, the schedule involves closely spaced administrations followed by a longer period of time during which the agent is not administered. For example, the schedule may e an initial set of 2O doses that are stered in a vely short period of time (e.g., about every 6 hours, about every 12 hours, about every 24 hours, about every 48 hours, or about every 72 hours) followed by a longer time period (e.g., about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, or about 8 weeks) during which the RNAi agent is not administered. In one ment, the RNAi agent is initially administered hourly and is later administered at a longer interval (e.g., daily, weekly, biweekly, or monthly). In another embodiment, the RNAi agent is initially administered daily and is later administered at a longer interval (e.g., weekly, biweekly, or monthly). In certain embodiments, the longer interval increases over time or is determined based on the achievement of a d effect. In a specific embodiment, the RNAi agent is administered once daily during a first week, followed by weekly dosing starting on the eighth day of administration. In another specific embodiment, the RNAi agent is administered every other day during a first week ed by weekly dosing starting on the eighth day of administration.
In some embodiments, the RNAi agent is administered in a dosing regimen that includes a "loading phase" of closely spaced administrations that may be followed by a "maintenance phase", in which the RNAi agent is administred at longer spaced intervals. In one embodiment, the loading phase ses five daily administrations of the RNAi agent during first week. In another embodiment, the maintenance phase comprises one or two [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM weekly administrations of the RNAi agent. In a further embodiment, the maintenance phase lasts for 5 weeks.
Any of these schedules may optionally be repeated for one or more iterations. The number of iterations may depend on the achievement of a desired effect, e.g., the suppression of a TMPRSS6 gene, and/or the achievement of a therapeutic or prophylactic effect, e.g., ng iron levels or reducing a symptom of thalassemia, e.g., B—thalassemia, or hemotochromatosis.
In another aspect, the invention features, a method of instructing an end user, e.g., a caregiver or a subject, on how to administer an iRNA agent described herein. The method es, optionally, providing the end user with one or more doses of the iRNA agent, and instructing the end user to administer the iRNA agent on a regimen described herein, thereby cting the end user.
VII. Kits The present invention also provides kits for using any of the iRNA agents and/or ming any of the methods of the invention. Such kits include one or more RNAi agent(s) and instructions for use, e.g., instructions for inhibiting expression of a TMPRSS6 in a cell by contacting the cell with the RNAi agent(s) in an amount effective to inhibit expression of the TMPRSS6. The kits may optionally further comprise means for contacting the cell with the RNAi agent (e.g., an injection device), or means for measuring the inhibition of TMPRSS6 (e.g., means for measuring the inhibition of TMPRSS6 mRNA or TTR protein). Such means for measuring the inhibition of TMPRSS6 may comprise a means for obtaining a sample from a subject, such as, e.g., a plasma sample. The kits of the invention may optionally further comprise means for administering the RNAi s) to a subject or means for determining the therapeutically effective or prophylactically effective amount.
Unless ise defined, all cal and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the iRNAs and methods featured in the ion, suitable methods and materials are described below. All publications, patent applications, s, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including tions, will l. In addition, the als, methods, and examples are rative only and not intended to be limiting.
EXAMPLES Materials and Methods fie following materials and methods were used in the Examples.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM cDNA synthesis using ABI High ty cDNA reverse transcription kit (Applied Biosystems, Foster City, CA, Cat #4368813) A master mix of 2p] 10X , 0.8 ul 25X dNTPs, 2ul Random primers, lul Reverse Transcriptase, lul RNase inhibitor and 3.2m of H20 per reaction was added into "] total RNA. cDNA was generated using a Bio—Rad C—1000 or S—1000 thermal cycler (Hercules, CA) through the following steps: 25°C 10 min, 37°C 120 min, 85°C 5 sec, 4°C hold.
Cell culture and transfections Hep3B cells (ATCC, as, VA) were grown to near confluence at 37 °C in an atmosphere of 5% CO2 in EMEM (ATCC) supplemented with 10% FBS, omycin, and glutamine (ATCC) before being released from the plate by trypsinization. ection was carried out by adding 14.8ul of Opti-MEM plus 0.2ul of Lipofectamine RNAiMax per well (Invitrogen, Carlsbad CA. cat # 13778—150) to 5p] of siRNA duplexes per well into a 96—well plate and incubated at room temperature for 15 minutes. Subsequently, 80p] of complete growth media without antibiotic containing ~2 x104 Hep3B cells were then added to the siRNA mixture. Cells were incubated for 24 hours prior to RNA purification. Single dose experiments were performed at 10nM and 0.1nM final duplex concentration.
Total RNA isolation using DYNABEADS mRNA Isolation Kit (Invitrogen, part #.' ) Cells were harvested and lysed in lSOul of Lysis/Binding Buffer then mixed for 5 minute at 850 rpm using a platform shaker (the mixing speed was the same throughout the process). Ten microliters of ic beads and 80ul Lysis/Binding Buffer mixture were added to a round bottom plate and mixed for 1 minute. Magnetic beads were captured using magnetic stand and the supernatant was removed without disturbing the beads. After the supernatant was removed, the lysed cells were added to the remaining beads and mixed for 5 s. After the supernatant was removed, magnetic beads were washed 2 times with 150ul Wash Buffer A and mixed for 1 minute. Beads were capture again and supernatant removed. Beads were then washed with 150ul Wash Buffer B, captured and supernatant was removed. Beads were next washed with 150ul Elution Buffer, captured and atant removed. Beads were allowed to dry for 2 s. After drying, 50ul of Elution Buffer was added and mixed for 5 minutes at 75°C. Beads were captured on magnet for 5 s, and 50p] of supernatant containing the purified RNA was removed and added to a new 96 well plate.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Real time PCR Two ul of cDNA was added to a master mix containing 0.5ul human GAPDH TaqMan Probe ed tems Cat 17E), 0.5ul human TMPRSS6 TaqMan probe (Applied Biosystems cat # Hs00542184_m1) and Sul Lightcycler 480 probe master miX (Roche Cat #04887301001) per well in a 384 well plate (Roche cat # 04887301001).
Real time PCR was performed in a Roche LC480 Real Time PCR system (Roche) using the AACt(RQ) assay. Each duplex was tested in two independent transfections and each ection was assayed in duplicate, unless otherwise noted.
To calculate relative fold change, real time data were analyzed using the AACt method and normalized to assays performed with cells transfected with 10nM AD—1955, or mock transfected cells.
The sense and antisense sequences of AD—1955 are: SENSE: 5’— cuuAchuGAGuAcuchAdTsdT-3’ (SEQ ID NO: 15); and NSE: 5’— UCGAAGuACUcAGCGuAAGdedT-3’ (SEQ ID NO: 16).
Table B: Abbreviations of nucleotide monomers used in nucleic acid sequence re resentation.
Abbreviation __'-’ _beta—L—uanos1ne—3— hos hate beta—L— _uanosine—3‘— hos nhorothioate 2’ —fluoroguanosine—3’—phosphate 2’ —fluoroguanosine—3’—phosphorothioate 2’ —fluoro—5—methyluridine—3 ’ hate 2’ —fluoro—5—methyluridine—3 ’ —phosphorothioate [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM 2’ —flu0r0uridine—3’— hos nhate 2’ —fluorouridine —3 ’ —phosph0rothioate _2'—O—methy1aden0sine—3 ’ te 2'—O—methy1adenosine—3’ — phosphorothioate 2'—O—meth lc tidine—3’—hos hate 2'—O—meth 1c tidine—3’— hos ioate ; 2'—O—meth 1_--uanosine—3’—hoshate s 2'—O—meth 1 uanosine—3’— nuhoshorothioate 2’ —O—meth 1—5—meth luridine-3’ — uhos hate ts 2’ —O—meth 1—5—meth luridine—3 ’ - nhos horothioate 2'—O—methy1uridine—3’—hoshate 2'—O—methy1uridine—3 ’ —ph0sph0r0thi0ate m phosphorothioate linkage L96 N— [tris(Ga1NAC—a1ky1)—amidodecanoyl)] —4—hydroxyprolinol Hyp— (GalNAc-alk D3 EAAAAAAAAAAAAAA U)Aeo) 2’ —O—methox eth ladeno sine-3’— hos hate Aeos) 2’ —O—meth0X eth 1aden0sine—3’— nhos horothioate Geo) 2’—O—methox eth luanosine-3’—hos hate Geos) 2’—O—meth0X eth 1 uanosine—3’— nohoshorothioate Teo) Teos) mSCeo) mSCeos) A3m) A3mx) 63m) 3 O methylguanosme 2 -phosphate G3mx) 3'—O—meth l—X lofuranos 1 C3m) 3‘-O-meth lc tidine-2‘-hos hate C3mx) eth l—X lofuranos 1c tidine-2'—h0s hate 3‘ -O-meth 1uridine-2‘ - - hos hate [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 3'—O—meth lx louridine—2'— hos hate 2'—O—hexadecyl—cytidine—3'— hos nhate 2'—O—hexadecyl—uridine—3'— nhos-hate 2'—O—hexadecyl—cytidine—3'—phosphate (m5Cam) 2‘—O—(I\'—meth lacetamide)—5—meth lc tidine—3‘—hos hate (m5Cams) 2‘—O—(N—meth lacetamide)—5—meth lc -3‘—--hoshorothioate 2‘—O—(I\'—meth lacetamide)th midine—3‘—hos hate - '—methylacetamide)thymidine—3‘—hoshorothioate 2‘—O—(N—methylacetamide)adenosine—3‘—phosphate 2‘—O—(N—methylacetamide)adenosine—3‘—phosphorothi0ate N—methylacetamide)guanosine—3‘—phosphate Y44 2—hydroxymethyl—tetrahydrofurane—S—phosphate Example 1. Design, Specificity and Efficacy Prediction of Oligonucleotides Transcripts siRNA design was carried out to fy siRNAs targeting human, rhesus (Macaca mulatta), mouse, and rat TMPRSS6 ripts annotated in the NCBI Gene database (http://www.ncbi.nlm.nih.gov/gene/). Design used the following transcripts from the NCBI RefSeq collection: Human —NM_153609.2; Rhesus — XM_001085203.2 and 085319. 1; Mouse — NM_027902.2; Rat -NM_001130556.1. Due to high primate/rodent sequence divergence, siRNA duplexes were designed in several separate batches, including but not limited to batches containing duplexes matching human and rhesus transcripts only; human, rhesus, and mouse transcripts only; human, rhesus, mouse, and rat transcripts only; and mouse and rat ripts only. All siRNA duplexes were ed that shared 100% identity with the listed human transcript and other species transcripts considered in each design batch (above).
The specificity of all le 19mers was predicted from each sequence. Candidate 19mers that lacked repeats longer than 7 nucleotides were then selected. These 1259 canditE human/rhesus, 91 human/rhesus/mouse, 37 human/rhesus/mouse/rat, and 810 ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM mouse/rat siRNAs were used in comprehensive searches against the appropriate transcriptomes (defined as the set of NM_ and XM_ records within the human, rhesus, mouse, or rat NCBI Refseq sets) using an exhaustive "brute—force" algorithm implemented in the python script ‘BruteForcepy’. The script next parsed the transcript—oligo alignments to generate a score based on the position and number of ches between the siRNA and any potential 'off—target' transcript. The off—target score is weighted to emphasize differences in the 'seed' region of siRNAs, in ons 2—9 from the 5' end of the molecule. Each oligo— transcript pair from the brute—force search was given a mismatch score by g the individual mismatch scores; mismatches in the position 2—9 were counted as 2.8, mismatches in the ge site positions 10—11 were d as 1.2, and mismatches in region 12—19 counted as 1.0. An additional off—target prediction was carried out by comparing the frequency of heptamers and octomers derived from 3 distinct, seed-derived hexamers of each oligo. The hexamers from positions 2—7 relative to the 5’ start were used to create 2 heptamers and one octomer. Heptamerl was created by adding a 3’ A to the hexamer; heptamer2 was d by adding a 5’ A to the hexamer; the octomer was created by adding an A to both 5’ and 3’ ends of the r. The frequency of octomers and ers in the human, rhesus, mouse, or rat me (defined as the subsequence of the transcriptome from NCBI’s Refseq database where the end of the coding region, the ‘CDS’, is clearly defined) was pre—calculated. The octomer frequency was ized to the heptamer 2O frequency using the median value from the range of octomer frequencies. A ‘mirSeedScore’ was then calculated by calculating the sum of ( (3 X normalized octomer count ) + ( 2 X heptamer2 count) + (l X heptamerl count)).
Both siRNA strands were assigned to a ry of specificity according to the calculated scores: a score above 3 qualified as highly specific, equal to 3 as specific and between 2.2 and 2.8 qualified as moderately specific. The siRNAs were sorted by the specificity of the nse strand. Duplexes from the human/rhesus and mouse/rat sets whose antisense oligos lacked GC at the first position, lacked G at both positions 13 and 14, and had 3 or more Us or As in the seed region (characteristics of duplexes with high predicted efficacy) were then selected. Similarly, duplexes from the human/rhesus/mouse and human/rhesus/mouse/rat sets that had had 3 or more Us or As in the seed region were selected.
Candidate —conjugated duplexes, 21 and 23 nucleotides long on the sense and antisense strands respectively, were designed by extending antisense l9mers 4 additional tides in the 3’ direction (preserving t complementarity with the target transcript).
The sense strand was specified as the reverse complement of the first 21 nucleotides of the antisense 23mer. Duplexes were selected that maintained perfect matches to all selected species transcripts across all 23 nucleotides.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM siRNA sequence ion A total of 39 sense and 39 antisense derived human/rhesus, 6 sense and 6 antisense derived human/rhesus/mouse, 3 sense and 3 antisense derived human/rhesus/mouse/rat, and 16 sense and 16 antisense derived rat siRNA 21/23mer oligos were synthesized and formed into —conjugated duplexes.
The sequences of the sense and antisense strands of the modified duplexes are shown in Table l, and the sequences of the sense and antisense strands of the unmodified duplexes are shown in Table 2.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Ln O \—| The modified and conjugated TMPRSS6 siRNA duplexes were also evaluated for efficacy by transfection assays in human cell line Hep3B. TMPRSS6 siRNAs were transfected at two doses, 10nM and 0.1nM. The results of these assays are shown in Table 3 and the data are expressed as a fraction of the message remaining in cells transfected with siRNAs targeting TMPRSS6, relative to cells transfected with a negative control siRNA, AD— 1955 i the standard deviation (SD).
Table 3. TMPRSS6 sin ' le dose screen.
-Duplex ID Avg 10nM SD 0.1nM -AD-58686. 1 74.50 34.00 -AD-58687. l 89.33 20.06 -AD-58688. 1 34.43 0.43 689. 1 73.04 3.00 -AD-58690. 1 33.40 44.04 -AD-58692. 1 73.44 0.04 -AD-58693. 1 77.43 47.37 -AD-58694. l 50.54 3.04 -AD-58695. 1 54.37 43.53 -AD-58696. 1 33.43 m- 3.04 697. 1 74.35 40.43 -AD-58698. 1 44.34 43.70 699. 1 44.33 44.55 -AD-58700. l 74.40 7.00 -AD-58701. 1 79.10 090 -AD-58702. l 33.43 40.35 -AD-58703. 1 73.45 079 -AD-58704. l 37.34 4.44 -AD-58705. 1 59.50 3.20 -AD-58706. l 74.37 44.00 -AD-58707. 1 77.03 1.34 -AD-58708. l 73.07 4.04 -AD-58709. 1 34.34 m- 43.40 -AD-58710. l 53.43 44.33 -AD-587 l 1.1 33.34 3.33 -AD-58712. l 35.73 m- 4.50 I:-AD-S l 04.44 5.30 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Duplex ID 470 101.11 am- AD-587l4.l 04.70 "-1.00 AD-58715.1 01.05 11.00 AD-58716.l 70.07 2.00 AD-58717.1 14.27 4.32 AD-587l8.l 02.10 15.00 AD-58719.1 72.94 9.97 .l 00.51 5.00 AD-58721.1 17.70 2.57 AD-58722.l 51.05 0.50 AD-58723.1 50.27 10.20 AD-58724.l 50.00 4.00 AD-58725.1 54.50 1.50 AD-58726.l 10.00 7.07 AD-58727.1 02.00 10.10 AD-58728.l 40.00 1.74 AD-58729.1 40.00 10.00 AD-58730.l 50.00 1.00 AD-58731.1 05.20 02.14 AD-58734.l 05.71 0.11 37.l 70.07 7.40 Example 3. In vivo single dose screen using AD-59743 The ability of AD—59743 to suppress expression of TMPRSS6 n was assessed by measuring levels of TMPRSS6 and hepcidin mRNA in the liver of wild-type C57BL/6 mice following administration of AD—59743. A single dose of 1, 3 or 10 mg/kg of AD—59743 was administered aneously, and the mice were sacrified on day 3 or day 7. Levels of TMPRSS6 and in mRNA in the liver were measured by qPCR using the methods bed above. A control group received injections with PBS.
The levels of TMPRSS6 mRNA following administration of AD—59743 are shown in Figure 1, and the levels of hepcidin mRNA following administration of AD-59743 are shown in Figure 2. The results demonstrate a dose—dependent decrease in the levels of TMPRSS6 transcripts that is ned through day 7.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM Example 4. In vivo effect of TMPRSS6 iRNA agents in combination with an iron chelator The purpose of this study was to test the effect of co—administered TMPRSS6 specific siRNA and iron chelators on iron levels. In the study, 6—week old wild—type 6 and thalassemic Th3/+ mice (Douet et al., Am. J. Pathol. (2011), 178(2):774—83) were fed low— iron diets containing 3—5 ppm iron. The mice were stered intravevously the formulation —46273 containing deferiprone, an iron chelator at a dose of 250 mg/kg/day and an iRNA agent with the following structure: oligoSeq—sense — uGGuAuuuccuAGGGuAcAdedT (SEQ ID NO: 209); oligoSeq—antisense — UGuACCCuAGGAAAuACcAdedT (SEQ ID NO: 210). The formulation also contained MC—3/DSPC/Cholesterol/PEG—DMG 50/10/38.5/ 1.5. Liver and spleen tissues were collected and tissue nonheme iron concentrations were ined as described previously (see, e.g., Schmidt et al. (2013) Blood 121(7):1200—8; Cook, JD, et al.. Tissue iron stores. In: Cook JD . Methods in Hematology. Vol 1. New York, NY: Churchill Livingstone Press; 1980. p. 104—109).
The results of these experiments demonstrate an additive effect of AD—46273 and deferiprone in Th3/+ mice, with the decreased iron levels relative to the negative controls.
Example 5. Design, icity and Efficacy Prediction of Oligonucleotides Transcripts siRNA design was carried out to identify siRNAs targeting human, cynomolgus monkey (Macacafascicularis; henceforth ), mouse, and rat TMPRSS6 transcripts ted in the NCBI Gene database (http://www.ncbi.nlm.nih.gov/gene/). Design used the following transcripts from the NCBI RefSeq collection: Human 3609.2; Mouse - NM_027902.2; Rat -NM_001130556.1. For cyno, a transcript sequence was obtained via ent with human TMPRSS6 of sequence assembled from two ions: "ENSP00000384964 [mRNA] locus=chr10:82446450z8248540 :-" and FR874253. 1, available from the M. fascicularis genome project and NCBI Nucleotide databases, respectively (http ://macaque.genomics.org.cn/page/species/download.j sp and /www.ncbi.nlm.nih.gov/nucleotide/). Due to high primate/rodent sequence divergence, siRNA duplexes were designed in several te batches, including but not limited to batches containing duplexes matching human and cyno transcripts only; human, cyno, and mouse transcripts only; and human, cyno, mouse, and rat transcripts only. Most siRNA duplexes were designed that shared 100% identity in the designated region with the listed human transcript and other species transcripts considered in each design batch (above).
In some instances, mismatches between duplex and mRNA target were allowed at the first antiseEGast sense) position when the antisense strandztarget mRNA complementary [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM basepair was a GC or CG pair. In these cases, duplexes were designed with UA or AU pairs at the first antisense:last sense pair. Thus the duplexes maintained complementarity but were mismatched with respect to target (U:C, U:G, AC. or AG).
The specificity of all possible 19mers was predicted from each ce. Candidate 19mers that lacked repeats longer than 7 nucleotides were then selected. These 1128 candidate human/cyno, 69 human/cyno/mouse, and 23 cyno/mouse/rat siRNAs were used in hensive searches against the appropriate transcriptomes (defined as the set of NM_ and XM_ records within the human, mouse, or rat NCBI Refseq sets, and the cyno transcriptome set in NCBI nucleotide) using an exhaustive "brute—force" algorithm implemented in the python script ‘BruteForcepy’. The script next parsed the transcript—oligo alignments to generate a score based on the position and number of mismatches between the siRNA and any potential 'off—target' transcript. The off—target score is weighted to emphasize differences in the 'seed' region of siRNAs, in positions 2—9 from the 5' end of the molecule.
Each oligo—transcript pair from the force search was given a mismatch score by summing the individual mismatch scores; mismatches in the position 2—9 were counted as 2.8, mismatches in the cleavage site positions 10—11 were counted as 1.2, and mismatches in region 12—19 counted as 1.0. An additional off—target prediction was carried out by comparing the frequency of ers and octomers d from 3 distinct, seed-derived hexamers of each oligo. The hexamers from positions 2—7 ve to the 5’ start were used to create 2 heptamers and one octomer. Heptamerl was created by adding a 3’ A to the hexamer; heptamer2 was created by adding a 5’ A to the hexamer; the octomer was created by adding an A to both 5’ and 3’ ends of the hexamer. The frequency of octomers and heptamers in the human, cyno, mouse, or rat 3’UTRome (defined as the subsequence of the transcriptome from NCBI’s Refseq database where the end of the coding region, the ‘CDS’, is clearly defined) was lculated. The octomer frequency was normalized to the heptamer frequency using the median value from the range of octomer frequencies. A ‘mirSeedScore’ was then ated by calculating the sum of ( (3 X normalized octomer count ) + ( 2 X er2 count) + (1 X heptamerl count)).
Both siRNAs strands were assigned to a category of specificity according to the 3O calculated scores: a score above 3 qualifies as highly ic, equal to 3 as ic and between 2 and 2.8 as moderately specific. We sorted by the specificity of the antisense strand. We then selected tely (or higher) specific duplexes whose antisense oligos possessed characteristics of duplexes with high predicted efficacy, ing maximal UA content in the seed region and low l GC content.
For GalNaC—conjugated duplexes, sense 21mer and antisense 23mer oligos were designed by extending antisense 19mers (described above) to 23 nucleotides of target— complementary sequence. All species transcripts included in the design batch were checked [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM for mentarity. For each duplex, the sense 21mer was specified as the e complement of the first 21 nucleotides of the antisense strand. siRNA sequence selection A total of 5 sense and 5 antisense human, 32 sense and 32 antisense derived human/cyno, 4 sense and 4 antisense derived human/cyno/mouse, 8 sense and 8 antisense derived human/cyno/mouse/rat, l9 sense and 19 antisense derived human/cyno/rat, 2 sense and 2 antisense derived human/mouse, and 1 sense and l antisense d mouse/rat siRNA 21/23mer oligos were synthesized and formed into GalNAc—conjugated duplexes.
The sequences of the sense and antisense strands of the unmodified es are shown in Table 4, and the sequences of the sense and antisense strands of the modified duplexes are shown in Table 5.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 52:81 moommfi .22 II 8». mom HE 8533 wmcmmzc< UUUU Cells were incubated for 24 hours prior to RNA purification. Experiments were performed at 10nM and 0.1nM final duplex concentration.
Total RNA ion using DYNABEADS mRNA Isolation Kit (Invitrogen, part #: 610-12) Cells were harvested and lysed in 150u1 of Lysis/Binding Buffer then mixed for 5 minutes at 850rpm using an Eppendorf Thermomixer (the mixing speed was the same throughout the process). Ten microliters of magnetic beads and 80u1 Lysis/Binding Buffer mixture were added to a round bottom plate and mixed for 1 minute. ic beads were captured using magnetic stand and the supernatant was removed without bing the beads.
After removing supernatant, the lysed cells were added to the remaining beads and mixed for minutes. After removing supernatant, magnetic beads were washed 2 times with 150p] Wash Buffer A and mixed for 1 minute. Beads were capture again and atant removed.
Beads were then washed with 150ul Wash Buffer B, captured and supernatant was removed.
Beads were next washed with 150ul Elution Buffer, captured and supernatant removed.
Beads were allowed to dry for 2 minutes. After drying, 50u1 of Elution Buffer was added and mixed for 5 minutes at 70°C. Beads were captured on magnet for 5 minutes. 40ul of supernatant was removed and added to another 96 well plate. cDNA sis using ABI High capacity cDNA reverse transcription kit (Applied 3O Biosystems, Foster City, CA, Cat #4368813) A master mix of 2p] 10X Buffer, 0.8u1 25X dNTPs, 2u1 Random primers, lul Reverse riptase, lul RNase inhibitor and 3.2ul of H20 per reaction were added into 10p] total RNA. cDNA was generated using a Bio—Rad C—1000 or S—1000 l cycler (Hercules, CA) through the following steps: 25°C 10 min, 37°C 120 min, 85°C 5 sec, 4°C hold.
Real time PCR [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM 2n] of cDNA were added to a master mix containing 0.5ul GAPDH TaqMan Probe (Applied Biosystems Cat #4326317E), 0.5ul TMPRSS6 TaqMan probe (Applied Biosystems cat # Hs00542184_m1) and Sul Lightcycler 480 probe master mix (Roche Cat #04887301001) per well in a 384 well 50 plates (Roche cat # 01001). Real time PCR was done in an Roche Lightcycler Real Time PCR system (Roche) using the AACt(RQ) assay. Each duplex was tested in two independent transfections and each transfection was assayed in duplicate, unless otherwise noted in the summary tables.
To calculate relative fold change, real time data were analyzed using the AACt method and normalized to assays performed with cells transfected with 10nM AD—l955, or mock transfected cells.
Data are sed as a fraction of TMPRSS6 message remaining in cells transfected with siRNAs targeting TMPRSS6, relative to naive cells. All siRNAs were transfected at least two times and qPCR reactions were performed in ate. Data are show in Table 6.
Table 6. TMPRSS6 single dose screen.
U) \l (D I—‘ —_——m ————m Imam" [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-60963 117.4 138.1 2.6 16.8 AD-60964 80.7 AD-60965 30.1 AD-60966 54.1 AD-60967 122.2 AD-60968 86.9 AD-60969 106.2 AD-60970 54.6 AD-60971 50.5 AD-60972 55.6 5’" h AD-60973 126.1 74 82.6 75 88.2 AD-60976 46.3 AD-60977 13.5 ED N AD-60978 72.7 AD-60979 103.8 AD-60980 28.4 AD-60981 56.0 AD-60982 102.4 AD-60983 60.8 AD-60984 53.6 AD-60985 72.6 86 90.1 AD-60987 83.1 AD-60988 69.4 AD-60989 112.4 AD-60990 90.4 AD-60991 97.6 AD-60992 104.0 AD-60993 118.6 AD-60994 25.9 9m AD-60996 77.3 97 60.1 AD-60998 32.6 AD-60999 133.6 AD-61000 55.8 AD-61001 57.9 AD-61002 15.4 03 82.3 AD-61004 106.4 AD-61005 138.0 AD-61006 31.7 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM Example 7. In vivo effect of single dose administration of TMPRSS6 iRNA agent Female C57BL/6 mice were administered a single aneous injection of AD— 60940 at a dose of 0.3 mg/kg, 1.0 mg/kg or 3.0 mg/kg, or PBS alone as a l. Three mice were evaluated per dose for hepatic 6 mRNA, hepatic hepcidin mRNA, serum hepcidin, total serum iron, and percent transferrin saturation at various time . Mice receiving 1.0 mg/kg or 3.0 mg/kg of AD—60940 or PBS were evaluated at day 0 (pre— treatment) and 7, 11, 14 and 21 days after treatment. Mice receiving 0.3 mg/kg AD—60940 were evaluated at day 0 (pre—treatment) and at 7 and 11 days after treatment. Hepatic TMPRSS6 mRNA and hepatic hepcidin mRNA levels were determined by qPCR, normalized to GAPDH mRNA levels and sed relative to the mRNA levels in mice administered PBS alone. Serum hepcidin was measured by ELISA (Intrinsic Life es). Total serum iron and percent transferrin saturation (% TfSat) were ed using an Olympus AU400 Serum Chemistry Analyzer. Each data point represents the mean value from three mice. The standard deviation of the mean is represented by error bars.
Single dose stration of AD—60940 resulted in robust and durable suppression of c TMPRSS6 mRNA relative to the control. TMPRSS6 mRNA concentration was suppressed by greater than 90% for up to three weeks following administration of the 3.0 mg/kg dose (Figure 3A). As a result of the suppression of hepatic TMPRSS6 mRNA concentration, hepcidin mRNA levels, increased two—fold relative to the control e 3B), and serum hepcidin concentration increased greater than 2—fold relative to the control (Figure 3C). In addition, total serum iron e 3D) decreased and percent transferrin tion decreased by greater than 50% relative to the control (Figure 3E). The decreases in total serum iron and percent transferring saturation were durable for up to three weeks following administration of AD—60940. Figure 3F demonstrates the relative hepatic TMPRSS6 mRNA concentration as a function of 40 dose at 11 days following administration. Each data point represents the maximum suppression of TMPRSS6 mRNA concentration observed at each dose level. The data were fit to the Hill equation.
The degree to which AD—60940 modulates hepcidin and serum iron mobilization is h3/+ nearly identical to that observed in the previous Hlabt mouse studies dt et al., Blood (2013), 121(7), 1200—1208) and indicates that AD—60940 is a potent RNAi therapeutic for producing disease modifying s in B—Thalassemia.
Example 8. In vivo effect of multi-dose administration of TMPRSS6 iRNA agent Female C57BL/6 mice were administered a subcutaneous injection of AD—60940 at a dose of 0.3 mg/kg, 1.0 mg/kg, or PBS alone (as a control) once per week for three weeks then sacrificed 7 days after the final dose (Figure 4A). Three mice per dose were evaluated for hepatiEVlPRSS6 mRNA, hepatic hepcidin mRNA, and percent transferrin saturation.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM c TMPRSS6 mRNA and hepatic hepcidin mRNA levels were determined by qPCR, normalized to GAPDH mRNA levels and expressed relative to the mRNA levels in mice administered PBS alone. Percent transferrin saturation (% TfSat) was measured using an Olympus AU400 Serum Chemistry Analyzer. Each data point represents the mean value from three mice. The standard deviation of the mean is represented by error bars.
Multi—dose administration of 1.0 mg/kg AD—60940 resulted in greater than 90% ssion of TMPRSS6 mRNA concentration (Figure 4B). Hepcidin mRNA tration increased two—fold and percent errin saturation decreased by greater than 50% relative to the control e 4B). Figure 4C demonstrates the relative hepatic TMPRSS6 mRNA concentration as a function of AD—60940 dose. The data were fit to the Hill equation. These data indicate that the multi-dose ED80 is less than 1.0 mg/kg.
This study demonstrates that AD—60940 exhibits robust and durable ssion of TMPRSS6, resulting in hepcidin induction and systemic iron ction and tes that AD—60940 is a potent RNAi therapeutic for producing disease modifying effects in B— semia.
Example 9. Relationship between liver TMPRSS6 mRNA levels and serum hepcidin concentration and percent transferrin saturation Data generated using AD—59743, AD—61002, AD—60940, and other TMPRSS6 iRNA agents were further analyzed to evaluate the relationship between liver TMPRSS6 mRNA levels and serum hepcidin levels and percent transferrin saturation. Serum hepcidin concentration demonstrated a non—linear relationship to TMPRSS6 mRNA levels using the Hill equation (Figure 5A). The percent transferrin saturation demonstrated a linear relationship to TMPRSS6 mRNA levels when fit to a simple linear regression equation (Figure 5B). The linear relationship between TMPRSS6 mRNA levels and percent transferrin saturation indicate that iron restriction can be precisely and predictably modulated by AD- 60940. Serum hepcidin tration and relative hepcidin mRNA levels also trated a linear relationship when fit to a simple linear regression equation (Figure 5C). In contrast, the relationship between percent transferrin saturation and serum hepcidin concentration was 3O near and fit to the Hill equation (Figure 5D).
Example 10. In vivo single dose screen TMPRSS6 siRNA duplexes as indicated in Figure 6 were evaluated for cy by their ability to suppress levels of TMPRSS6 mRNA in the liver of female C57BL/6 mice following stration of the siRNA . A single subcutaneous dose of 3 mg/kg of TMPRSS6 siRNA duplex was administered, and the mice were sacrificed 7 days later. The level of TMPRSS6 mRNA in the liver was measured by qPCR using the s described aboveEice in a control group received an injection of PBS.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM The levels of TMPRSS6 mRNA following administration of a TMPRSS6 siRNA duplex are shown in Figure 6. The results demonstrate that administration of AD—60940, AD—59743 and AD—61002 resulted in substantial suppression of liver TMPRSS6 mRNA with AD60940 producing the greatest silencing. Specifically, TMPRSS6 siRNA duplex AD— 60940 reduced 6 mRNA by greater than 80% ve to the control. The data also trate that treatment with AD—59743, AD—60940, AD—61002, AD-60994, AD—60998 and AD—61001 result in a decrease in the level of TMPRSS6 ript that is maintained through day 7.
Example 11. In vivo multi-dose screen TMPRSS6 siRNA duplexes as indicated in Figure 7 were evaluated for efficacy by their ability to suppress levels of TMPRSS6 mRNA in the liver of ype 6 mice following stration of the siRNA duplex. A subcutaneous dose of either 0.3 mg/kg or 1.0 mg/kg of TMPRSS6 siRNA duplex was administered once a week for three weeks. The mice were sacrificed 7 days after the last dose. The level of TMPRSS6 mRNA in the liver was measured by qPCR using the methods described above. Mice in a control group received an injection of PBS.
The levels of TMPRSS6 mRNA following administration of a TMPRSS6 siRNA duplex are shown in Figure 7. The results demonstrate that the 1.0 mg/kg dosing regimen of TMPRSS6 siRNA duplex 40 reduces TMPRSS6 mRNA by greater than 80% relative to the control.
Example 12. Optimization of 40 Based on the observation that administration of AD—60940 durably reduced TMPRSS6 mRNA by greater than 80% relative to the control, additional siRNAs based on the parent sequence of AD—60940 with a variety of chemical modifications were evaluated for efficacy in single dose screens at 10nM and 0.1nM by transfection in Hep3B cells. The sequences of the sense and nse strands of these agents are shown in Table 8 and the 3O results of this screen are shown in Table 9. The data in Table 9 are expressed as the average fraction message remaining relative to control.
In on, a subset of siRNA described in Tables 4 and 5, above, were modified to replace a 2’F with a 2’OMe modification at the 5’-end of the sense strand and to add a 5’— phosphate on the antisense strand. These siRNA agents were also evaluated for in vitro efficacy in single dose screens at 10nM and 0.1nM by transfection in Hep3B cells. The sequences of the sense and antisense s of these agents are shown in Table 10 and the results of this screen are shown in Table 11. The data in Table 11 are expressed as the averaEaction e remaining ve to control.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM #mowoo fid‘d‘d‘ mmmmm $83563:gammfiaueumsmanmé mmmmmweowo $33563:£<~§<=§U~Smm5m€ $33368:"Emuflficfigmaam: umwmmEoU mmmmmzflu mm mm mm wm mm mm mm mmmmuu ~86 awn—Bu mama—89 m». amazon wmwmuu mmmmmzuwu «.5 mmeuU m5 m5 m5 33 866323080 mm9.34.69£33<~mm~3u8~5mm5m3 m5 mm835635:32:65«3952. $33368:£<~§<=§U~Smm5m= mmmwuu $3956m5£<~§<=§U§Gm5m= 8: 33388331338386 m5 =22ammEeueuEaumSm: m5 wmwmmzuumwnszwmmzsuuuwmemem: mmmmouw<3 "EEEEDEDED . . nmEEuEEDmsn "$355539th "3:555 £ NM_153609.3. Three thousand one hundred and eighty duplexes were initially identified that did not contain repeats longer than 7 tides, spanning substantially the entire 3209 nucleotide transcript. All 3180 duplexes were then scored for predicted efficacy according to a linear model that tes the nucleotide pair at each duplex position, and the dose and cell line used for screening. The duplexes were also matched against all transcripts in the human RefSeq collection using a custom brute force algorithm, and scored for lowest numbers of mismatches (per strand) to transcripts other than TMPRSS6. Duplexes to be synthesized and ed were then selected from the 3180, according to the following scheme: Beginning at the 5’ end of the transcript, a duplex was selected within a "window" of every 10 i 2 nucleotides that had the highest predicted efficacy, had at least one ch in both strands to all transcripts other than TMPRSS6, and had not already been synthesized and ed as part of other duplex sets.
If no duplex is identified within a given window that satisfied all criteria, that window was skipped. Three hundred and three duplexes were selected according to the above criteria.
An additional 31 duplexes were also selected.
A detailed list of the 334 TMPRSS6 sense and nse strand sequences is shown in Table 12.
Cell culture and transfeetions Hep3B2.1—7 cells were obtained from American Type Culture Collection (Rockville, Md., cat. No. HB—8064) and cultured in EMEM (ATCC #30—2003), mented to contain % fetal calf serum (FCS) (Biochrom AG, Berlin, y, cat. No. 80115) and Penicillin 100 U/ml, Streptomycin 100 mg/ml (Biochrom AG, Berlin, Germany, cat. No. A2213), at 37°C in an atmosphere with 5% C0211] a humidified incubator (Heraeus HERAcell, Kendro Laboratory Products, selbold, Germany).
Transfection of dsRNA was performed directly after g 15,000 cells / well on a 96—well plate, and was carried out with Lipofectamine 2000 (Invitrogen GmbH, Karlsruhe, Germany, cat.No. 11668—019) as described by the manufacturer. Transfections were performed in quadruplicates and dsRNAs were transfected at a concentration of 10 nM.
[Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM ation] KIM Unmarked set by KIM Branched DNA assa s- unatiGene 2.0 Panomics cat #.' $001 I For measurement of TMPRSS6 mRNA cells were harvested 24 hours after transfection and lysed at 53°C following procedures recommended by the cturer of the Quantigene II Kit for TMPRSS6 and Quantigene I Explore Kit for bDNA (Panomics, Fremont, Calif, USA, cat. No. 15735 or QG0004, respectively). Subsequently, 50 ul of the lysates were incubated with probesets specific to human TMPRSS6 and 10 ul of the lysates for human GAPDH and processed according to the manufacturer’s protocol for QuantiGene.
Chemoluminescence was measured in a 2—Light (Perkin Elmer, Wiesbaden, Germany) as RLUs (relative light units) and values obtained with the human TMPRSS6 probeset were normalized to the respective human GAPDH values for each well and then related to the mean of three unrelated control dsRNAs.
The in vitro efficacy of the compounds is shown in Table 13.
[Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM wmcmmzc< HdmmoNHLq H.HowoNH-< H.mowoNH-< H.momoNH-< HKomoNHLQ H.momoNH-< HLq figmnwoNHLq HLq HKnmoNHLQ H.mnmoNH-< NH-< H.mwwoNH-< H.mmmoNH-< HfiwmoNHLq H.mmmmNH-< H.HmwoNH-< H.mmwoNH-< H.mmmoNH-< HfimmoNHLq Hdmwomfii H.HomoNH-< 0mm HOZ mfiOH OmOH HmoH NmOH mmOH «mofi mmOH omOH wmofi meH mmofi Ooofi HooH NooH moofi #oofi moofi ooOH ._._o._.—U mucosuwm ._.E._Q AD1 AD1 AD-63338.1 28.0 AID-633441 AD1 AD1 AD1 AD1 AD1 AD1—n AID-633391 AD1 AD1 AD-63357.1 35.8 " AD1 AD1 AD-63375.1 2.4 AD1 40-1"— AD-63346.1 3.5 AD-63352.1 3.9 AD-63358.1 8.1 AD-63364.1 AD-63370.1 2.8 AD-63376.1 4.2 AD-63382.1 7.5 WEE—— [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-63365.1 27.3 3.4 AID-63371.1 AD1 AD-63383.1 AD-63342.1 7.3 AD1 AD1 AD1 AD1—m- AD1—m AD-63378.1 64.1 m AD1 AD1 AD1 AD1—n AD1 AD-63414.1 6.2 AD1 AD1 AD1 AD1 AD1 AD1 AD1 AD1 21-1"— AD1 86-1 AD1 AD1 AD-63404.1 8.1 AD1 AD1—m AD-63422.1 2.7 28.1 9 7 AD-63387.1 AD-63393.1 4.4 AD-63399.1 4054 . 9 3 AD-63411.1 14.9 ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-63394.1 31.5 4.6 400.1"— AD1 AD1 AD-63418.1 4.3 AD1 AD1 AD1 AD1 AD1 AD-63407.1 4.9 AD1—m AD1 AD-63425.1 78.8 n AD1—n AD1 AD-63443.1 56.4 " AID-634491 AD1 AD1 AD1 AD1 AD1 38-1 AID-634441 AD1 AD1 AD1 AD1 AD1 33-1 AD1 AD1 AD-63451.1 49.6 " AD-63457.1 4.9 AD-63463.1 3.8 AID-63469.1 14-0 AD-63475.1 14 AD-63434.1 AD-63440.1 4.7 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-6347o.1 44.8 7.8 AID-63476.1 AD1 AD1 AD-63447.1 46.5 m AD-63453.1 59-1—n 65.1 27.3 n 71-1 AD1 AD-63436.1 4.7 AD1 AD1—m- AD1 AD1 AD1 AD-63472.1 4.1 AD1 AD1 AD1 AD1 AD1"— AD1 14-1 AD1"— AD1 AD1 AD1"— AD1 03-1 AD-63509.1 29.7 " AD-63515.1 AD1 AD-63480.1 0.7 AD-63486.1 12.1 AD-63492.1 3.7 AD-63498.1 20.3 AD-63504.1 1.9 AD-63510.1 4.0 AD-63516.1 "Egan—— [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-63499.1 44.2 17.7 05.1"— AD1 AD1 AD1 AD-63482.1 AD1 AD1 00-1 AD-63506.1 AD1 AD1 AD1 AD1 AD1 AD1 AD-63501.1 2.1 AD1 AD1 19~1—n AD1 AD1 37-1 AD1 AD1 AD1 AD1 AD-63567.1 39.2 _ AD1"— A01—_ AD-63538.1 50.8 AD-63544.1 AD1 AD-63556.1 31.0 m AD-63562.1 2.4 AD-63568.1 2.7 AD-63527.1 2.4 AD-63533.1 AD-63539.1 3.5 AD-63545.1 8.5 mum—— [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD-63528.1 52.9 9.0 AD-63534.1 42.5 "- AD1 AD1 AD-63552.1 5.0 58.1 64-1 AD-63570.1 50.6 " 29-1 AD-63535.1 AD1 AD1—n AD-63553~1—m- AD1 AD1 AD1 AD1 AD1 42-1—-!- AD1 AD1 AD1 AD1—m- AD1 AD1 AD1 AD1 AD-63596.1 63.5 m AD1 AD1 AD-63614.1 7.4 73.1 AD1 AD-63585.1 2.1 AD-63591.1 2 5 AD-63597.1 5.8 AD-63603.1 4.0 AD-63609.1 2.7 AD-63615.1 9.7 AD-63574.1 10.3 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM AD—63604.1 98.7 7.1 AID-63610.1 AD1 AD1 AD-63581.1 3.2 AD1 AD1 AD1 AD1 AD1 AD-63617.1 45.6 m AD1 AD1 AD-63588.1 6.7 AD-63594.1 18.0 AD-63600.1 11.9 06.1 2.7 12.1 5.3 AD-63618.1 AD1 AD1—m AD-63589.1 61.9 _ _m-_ The claims ng the invention are as follows: 1. A double stranded RNAi agent for inhibiting expression of TMPRSS6 in a cell, comprising a sense strand and an antisense strand forming a double-stranded region, n the antisense strand comprises at least 15 contiguous nucleotides which differ by no more than three nucleotides from the nucleotide sequence AGAAUGAACCAGAAGAAGCAGGU (SEQ ID NO:187), wherein each strand is independently 15 to 30 nucleotides in length, wherein all of the nucleotides of the sense strand and all of the nucleotides of the antisense strand are modified nucleotides, and wherein at least one strand is conjugated to a ligand. 2. The double stranded RNAi agent of claim 1, wherein the nse strand comprises at least 16 contiguous nucleotides which differ by no more than three nucleotides from the nucleotide sequence AGAAUGAACCAGAAGAAGCAGGU (SEQ ID NO:187). 3. The double stranded RNAi agent of claim 1, wherein the antisense strand comprises at least 17 contiguous nucleotides which differ by no more than three nucleotides from the nucleotide sequence AGAAUGAACCAGAAGAAGCAGGU (SEQ ID NO:187). 4. The double stranded RNAi agent of any one of claims 1-3, wherein each strand is independently 17-25 nucleotides in .
. The double stranded RNAi agent of any one of claims 1-3, wherein each strand is independently 19-25 tides in length. 6. The double stranded RNAi agent of any one of claims 1-3, n each strand is independently 19-23 nucleotides in length. 7. The double stranded RNAi agent of any one of claims 1-3, wherein at least one of the modified nucleotides is selected from the group consisting of a 3’-terminal deoxythymine (dT) nucleotide, a 2'-O-methyl modified tide, a 2'-fluoro modified tide, a xy-modified nucleotide, a locked nucleotide, an abasic nucleotide, a 2’-amino- modified nucleotide, a yl-modified tide, a morpholino tide, a phosphoramidate, a non-natural base comprising nucleotide, a nucleotide comprising a 5'- phosphorothioate group, a nucleotide comprising a 5’ phosphate or 5’ phosphate mimic, and a terminal nucleotide linked to a cholesteryl derivative or a dodecanoic acid bisdecylamide group. 8. The double stranded RNAi agent of any one of claims 1-3, wherein at least one of the modified nucleotides is ed from the group ting of a 2'-O-methyl modified nucleotide, a oro modified nucleotide, a nucleotide comprising a 5'- phosphorothioate group, an abasic nucleotide, and a 2’-amino modified nucleotide. 9. The double stranded RNAi agent of any one of claims 1-3, wherein at least one strand comprises an overhang of at least 1 nucleotide. 10. The double stranded RNAi agent of any one of claims 1-9, wherein the agent further comprises at least one phosphorothioate or methylphosphonate internucleotide linkage. 11 The double stranded RNAi agent of claim 10, wherein the phosphorothioate or methylphosphonate internucleotide linkage is at the 3’-terminus of one strand. 12. The double stranded RNAi agent of claim 11, n the strand is the antisense strand. 13. The double stranded RNAi agent of claim 11, wherein the strand is the sense strand. 14. The double stranded RNAi agent of claim 10, wherein the phosphorothioate or phosphonate internucleotide linkage is at the 5’-terminus of one strand.
. The double stranded RNAi agent of claim 14, wherein the strand is the antisense strand. 16. The double stranded RNAi agent of claim 10, wherein the phosphorothioate or methylphosphonate internucleotide linkage is at both the 5’- and 3’-terminus of one . 17. The double ed RNAi agent of claim 16, wherein the strand is the antisense . 18. The double stranded RNAi agent of claim 10, wherein the double stranded RNAi agent comprises 6-8 phosphorothioate internucleotide linkages. 19. The double stranded RNAi agent of any one of claims 1-3, wherein the ligand comprises an N-acetylgalactosamine (GalNAc) derivative.
. The double stranded RNAi agent of claim 19, wherein the N- acetylgalactosamine (GalNAc) derivative ligand is attached through a trivalent ed linker. 21. The double stranded RNAi agent of claim 20, wherein the trivalent branched linker comprises the structure shown in Formula (VII): wherein: q5A, q5B and q5C represent ndently for each occurrence 0-20 and wherein the repeating unit can be the same or different; P5A, P5B, P5C, T5A, T5B, and T5C are each independently for each occurrence absent, CO, NH, O, S, OC(O), NHC(O), CH2, CH2NH or CH2O; Q5A, Q5B, and Q5C are independently for each occurrence absent, alkylene, substituted alkylene wherin one or more methylenes can be interrupted or ated by one or more of O, S, S(O), SO2, N(RN), C(R’)=C(R’’), C≡C or C(O); R5A, R5B, R5C are each independently for each occurrence absent, NH, O, S, CH2, C(O)O, C(O)NH, NHCH(Ra)C(O), -C(O)-CH(Ra)-NH-, CO, , N , S S S S N S S H , , , or heterocyclyl; L5A, L5B and L5C are independently for each occurrence a monosaccharide, a GalNAc derivative, a disaccharide, a trisaccharide, a tetrasaccharide, an oligosaccharide, or a polysaccharide 22. A pharmaceutical composition for inhibiting expression of a TMPRSS6 gene comprising the double stranded RNAi agent of any one of claims 1-21. 23. An in vitro method of inhibiting TMPRSS6 expression in a cell, the method comprising: (a) ting the cell in vitro with the double stranded RNAi agent of any one of claims 1-21, or a pharmaceutical composition of claim 22; and (b) maintaining the cell produced in step (a) in vitro for a time sufficient to obtain degradation of the mRNA transcript of a TMPRSS6 gene, y inhibiting expression of the TMPRSS6 gene in the cell. 24. The method of claim 23, or the double stranded RNAi agent of any one of claims 1 to 21, or the pharmaceutical composition of claim 22, wherein the TMPRSS6 expression is inhibited by at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 100%; serum hepcidin tration is increased by at least 10%, 25%, 50%, 100%, 150%, 200%, 250%, or 300%; serum iron concentration is decreased by at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 100%; or percent transferrin saturation is decreased by at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 100%.
. Use of a double stranded RNAi agent of any one of claims 1-21, or the pharmaceutical composition of claim 22 in the manufacture of a medicament for treating a TMPRSS6 associated disorder in a subject. 26. The use of claim 25, wherein the TMPRSS6 associated disorder is hereditary hemochromatosis, β-thalassemia, opoietic porphyria, or a disorder associated with iron overload. 27. The use of claim 25, wherein the double stranded RNAi agent is ated for administration at a dose of 0.01 mg/kg to 10 mg/kg or 1 mg/kg to 10 mg/kg. 28. An isolated cell containing the double stranded RNAi agent of any one of claims 1-21. 29. The use of any one of claims 25-27, wherein the double ed RNAi agent is for subcutaneous administration.
. The use of any one of claims 25-27, wherein the double stranded RNAi agent is for intravenous administration.
[Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 1/19 TMPRSSS mRNA 3 ..............
Day 3 {)3}: 3" Figure 1 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 2/19 Hepcidin mRNA ........................................................................................................................................................................................ ............................................................................................................................................................................................. m\\\\\\\\\m\\\\\\\\\\\\\\\\\\ Gay 3 Say 3? ...............................................................................................................................................................................................................................................
Figure 2 [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 3/19 w"PBS "\VWO 3 mg/kg Concentration 1.2 3‘ .......1 :cce¢ J_.() [11 £; mRNA vsse§_\q \\\fi\L \va§ \\\\\‘\\\\\\\ \T TmprssG .0 m \ .. \ .0 4:.
Relative u\mm»»»»»‘u\m»m»»»»\ummmmm"\ummmm»mum‘mmww‘xfiémm»»»»mumm»we"cs¢vx wxs$¢yxs¢ \\ \zS:fao5xxxxxxxxvaeeeccceeaeafig§onxxogx fissfifleas \\\\\\\"\\\ \ Viae~‘5535‘¢g\\\\\\\S‘ \\\\\\\\\\\\\\\ ‘anaaagiiuwvis ‘ «classlag! ‘gausaaaxuias a ‘fillinnneeeaaabssaaeeeaan O U‘I 10 15 Time (days post dose) Figure 3A 2.5 ........,............¢ .\\\ flit-"V PBS Concentration »\\\&w0.3 mg/kg .\\.\\\\\\\‘ 1.0 mg/kg .\\\\}’\\‘3.0 mg/kg H U'I mRNA .I.
‘E sxewrrrrtccvqgk fi§§\ 2 5?M/\ I l \"\\\"\\W\\\§ -.\' §"N\\\m\\\\m\mxx\\\\\\\\x‘ \\,4 " V in . & \\"{Q\\\\\\\\ \Six "\ \w" s~"\ \ "43‘ - 'r l 0 $"\\\\\\W\\\\Km\\\\\\\m"\wm \\§J\'§‘QQQ§$\\\\\\M""wwwww"m" «WWWmmwwww.""«www.‘agg§:ce:Q:{t§§¥§$$i$§§§§3eel§§§? Hepci .
Relative .0 u-I .0O ..4,..................,.;................,.
O \l 14 21 28 Time (days post dose) Figure 3B [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 4/19 . ‘\\ . \"PBS ...'.....l.....}.....1:"...1.....4..... ‘ \\\\\\\\\\\‘0.3 mg/kg I \\\\\\§\\\\l_0 mg/kg Ln0O - \93‘1)T‘rTg7k?""""""""""""w (fig/ml)o0 ......,, pcidin WOO ...1'.....4’.....«w""""w‘wsl‘w‘"w"www«"""ww«ww" -[ W‘w‘ww‘w‘ .\rww"""""««««‘«""""«««ww" Serum \\ ¢\‘ . ‘3, \\\\\\\ \\\\\\\\\ \ \."w?" & \K\\\\\\\\\\\\\\\\,\,\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\ \\‘ \ \\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\L® \ \\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\ ‘\\\\" 200 '17 |_\OO .¢,....;.....:.....""‘\.""":"""1" ‘‘‘‘""?"""\."""‘~‘""‘"""""""‘ ""«k «"‘""«\« > ‘: ? ««\f««\f«««=«"«1‘"‘"\« > ‘ > ""‘t"""}"""\"""""""‘. ~ \ O \I 14 21 28 Time (days post dose).
Figure 3C \\\\\\\\\\\\\\\\\\\\\\\\\\\\\\ (pg/dL) \m\\\\\\\\\\\\\\\\\\%\\\\\\\\\\\\\\\\\\\\\\ '\\\\\\\\\\m\\\\\\\\\\\\\\\ 150 ""4," Iron 1,,,,L.,,i,"/’,,,,$,,,L,.J,,, \\\ | \\ l \\\\\\\\\\\\ Serum m‘mm":":\\""""mwww""mwwwm"""""""‘EX}. x\\\V<:caer.\\\\\\m\\\*\\\ \ Total \\ Ngl‘gomQ"\\\\\\\\w\\\\\\\\\\\\\\m‘nb\\\\\\\1 50 A, "riflflbfliafl/L," 4 ~ . V‘""\‘"""§"""\"""‘r"""r""‘\‘""‘\"""\"""\ 0 7 14 21 28 Time (days post dose).
Figure 3D [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM /19 \\\\\§N\ P BS ;\L «\\\\\\\~0. 3 mg/kg f""WWW""""""IQ;;;;;;;\;;;;;;\:{{c&ssax\\\\\"\‘"""m*3""""m"bl\ \ \\\\\\\\\\\\\ """"""x<<§ccs1‘;o"rfi§7k"g‘"""" "New 3.0 mg/kg '\ "um\\\\\\x \\\\\\\\\\\\\\ ‘ \\ "I \\\\\\\\\\\\\\\\ $\\\\\\\\\\\\\\\\\\‘~\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\N \\\\ x wwwmfi W\%mxuxxxx\\\\\m\\xmmwmwmwmx‘. ""fl‘t""«\‘"""V‘"‘"r‘fi‘wv"‘"3"""‘:"‘"‘V"""1"""Yv""\"‘""‘~"""‘r""""«"":«"‘\‘"""\_‘"""§«‘"‘\?"""\"""‘:"""‘;«""\‘«""‘:‘"«‘\‘"««\ 0 7 14 21 28 Time (days post dose)_ Figure 3E Q "LE Tmprssfi Cancentrat {TE .8 {3 _3 xi, fiaiafiwe $33 mfihfifi 33.2 a «g 2 :3 4 5 ALNWTMP {m gmg} Figure 3F [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM 6/19 Dose 1 Sac (start) Dose 2 Dose 3 (end) fime waW) 0 7 1 4 21 FEUm 4A 2 s. g. T m mRm 2 a. HEe d.m mRNA cams w: s.
Essa gumgcmuneo «I n. «mmmzfi a a.
El .m. u m 4 B a... 2 as a. mmflmefisnmmmm csmfiefifim U a.
V «Ems mu2 a. nu. 5. 1 a. m m ALNu—TMP (mgikgj Figure 4C [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 7/19 Serum in vs. Tmprssfi mRNA sec: infirm}; 500 11:35 Figure 5A Hepcidin 3:35 Serum 286 0.8 02 {LA (3.6 0.3 1 .0 1 .2 Reiative Tmprssa mRNA Concentration W831: vs Tmprssfi mRNA {9/05 Figure SB TfSat 0.6 {3.2 0.4 {3.6 0.8 1.4:) 1.2 Reiative Tmprssfi mRNA Concentration Serum Hepcidin vs. Hepcidin mRNA 633a inglmij Figure 5C Hepcidin 38mm .5 1.6 1.5 Efl 2.5 Reiative Hepcidin mRNA Concentration TfSat we Serum Hepcidin Figure 5D 109 280 am} am} 5m} 69!} Serum Hepcidin {ngs‘mi} [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM ation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 8/19 1.20 §\\\\\\\\\\\\\\\\\\\\\\\ "‘‘ mmmmmoS. 1.00 "‘ \\\\\ mmflacc. $2é 80 «>333. 60 Z0 . 0 ‘ ’ \§AD-59743 \ AD-60940 AD-61002§ AD-60994 98 AD-61006 AD-60944 AD-61001 Figure 6 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 9/19 100 . .‘" . "...¥\¥\¥. in; 80 ‘ ‘‘« ‘ «‘‘‘‘‘‘N «mamas. 60 ‘‘ " ‘‘ ‘‘ ‘‘‘‘‘‘‘‘‘‘ @358... \ 40 "\\ 3:23. 20 ‘‘‘‘‘" \\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\\ PBS 43 AD-60940 AD-61002 Figure 7 [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM /19 SEQ ID NO:L >gi | 56682967 | ref | NM_153609 .2 | Homo s transmembrane protease, serine 6 (TMPRSS6) mRNA CTTGAGCCAGACCCAGTCCAGCTCTGGTGCCTGCCCTCTGGTGCGAGCTGACCTGAGATGCACTTCCCTC CTCTGTGAGC"GTCTCGGCACCCACTTGCAGTCACTGCCGCC"GATGTTG""ACTCTTCCACTCCAAAAG GA GCCCG GGCCGAGGCCCCCCAGGTGGC"GGCGGGCAGGGGGACGGAGG"GATGGCGAGGAAGCGGAG GGGATGTTCAAGGCCLG_GAGGAC_CCAAGAGAAAAGCCCGGGGC_ACC_CCGCCLGGLGCCCC TG"TTGTGCTGCTGGCCCTGCTCGMGCTGGCTTCGGCGGGGGHGCTACTCHGGTAHTTCCTAGGGTACAA GGCGGAGG GATGG CAGCCAGG"GTACTCAGGCAGiCiGCG G ACTCAA CGCCAC AGGAT C ACCCGCCGGGAATCTAGTGCCTTCCGCAGTGAAACCGCCAAAGCCCAGAAGA-GC-CAAGGAGCTCA TCACCAGCACCCGCCTGGGAACTHACTACAACTCCAGCTCCGHCHATTCCHHTGGGGAGGGACCCCTCAC C GCTTC C"GGT CCAAATCCCCGAGCACCGCCGGC GATGC CGAGGTGG"GCAG GCACLGC GG-GGAGGAGCTGCTGTCCACAGTCAACAGCTCGGC GCCG CCCCTACAGGGCCGAGTACG AAGTGGACCCCGAGGGCC_AGLGA_CCLGGAAGCCAGTGTGAAAGACALAGCiGCAL_GAALLCCACGC_ GGGTTGTHACCGCTACAGCTACGTGGGCCAGGGCCAGGTCCTCCGGCTGAAGGGGCC"GACCACCHGGCC TCCAGCTGCC"GTGGCACCTGCAGGGCCCCAAGGACC CATGC CAAAC CCGGCLGGAGTGGACGCTGG CAGAGTGCCGGGACCGAC-GGCCA-GTAiGACGTGGCCGGGCCCCTGGAGAAGAGGCTCATCACC-CGG- GTACGGC"GCAGCCGCCAGGAGCCCGTGGTGGAGGTTCTGGCG'CGGGGGCCATCATGGCGGTCGHCTGG AAGAAGGGCC"GCACAGCTACTACGACCCC TCGTGC CTCCG GCAGCCGGTGGTC CTGTG AAGTGAACC"GACGCTGGACAACAGGCTCGACTCCCAGGGCG CC-CAGCACCCCGTACTTCCCCAGCTA CTACTCGCCCCAAACCCACTGCTCCTGGCACCTCACGGTGCCC"C"CTGGACTACGGCHTGGCCCHCTGG Ti GATGCC ATGCACLGAGGAGGCAGAAG ATGA GCCG GCACCCAGGGCCAG GGACGA CCAGA ACAGGAGGC GTG-GGCLTGCGCA CC-GCAGCCC"ACGCCGAGAGGATCCCCGTGG"GGCCACGGCCGG GA-CACCALCAAC__CACCTCCCAGATCTCCCTCACCGGGCCCGGTGTGCGGGTGCAC_ATGGC__GTAC AACCAGTCGGACCCCTGCCCTGGAGAGHHCCTCTG""CTGTGAA"GGAC"CMGTGTCCCTGCCTGHGATG GGGTCAAGGACTGCCCCAACGGCC GGA GAGAGAAACTGCGTT"GCAGAGCCACAT"CCAGTGCAAAGA GGACAGCACALGCA-C-CACiGCCCAAGGTCTGTGA"GGGCAGCC-GAL-G-CTCAACGGCAGCGACGAA GAGCAGTGCCAGGAAGGGGTGCCAHGTGGGACATTCACCTTCCAGHGTGAGGACCGGAGCTGCGTGAAGA AGCCCAACCCGCAG G GATGGGCGGCCCGACTGCAGGGACGGC CGGA GAGGAGCACTGTGACTGTGG CCTCCAGGGCCCCTCCAGCCGCA -GT-GGiGGAGC GLGTCC CCGAGGG"GAGTGGCCATGGCAGGCC AGCCTCCAGGTTCGGGGTCGACACA_C_G_GGGGGGGCCCTCATCGCTGACCGC_GGG_GALAACAGCLG CCCACTGC"TCCAGGAGGACAGCAHGGCC CCACGG GCTGTGGACCGTG""CC"GGGCAAGGTGTGGCA GAACTCGCGC"GGCCTGGAGAGG GTCC CAAGGLGAGCCGCCTGCTCC"GCACCCG"ACCACGAAGAG GACAGCCATGACTACGACGTGGCGC_GC_GCAGCLCGACCACCCGGTGGTGCGCTCGGCCGCCGTGCGCC CCGTCTGCCTGCCCGCGCGCTCCCACTTC"TCGAGCCCGGCCTGCACTGCHGGAHTACGGGCTGGGGCGC OD’OD’HO GCGCGAGGGCGGCCCCATCAGCAACGCTCTGCAGAAAGTGGATGTGCAGTTGATCCCACAGGACCTG GCAGCGAGGTCTATCGCTACCAGGTGACGCCACGCATGCTGTGTGCCGGCTACCGCAAGGGCAAGAAGG "GCCTGTCAGGGTGACTCAGGHGGHCCGCTGGTG"GCAAGGCACTCAGTGGCCGCTGGTTCCTGGCGGG C"GGTCAGC"GGGGCCTGGGC"GTGGCCGGCCTAAC"ACTTCGGCGTCTACACCCGCATCACAGGTGTG CAGCLGGA CCAGCAAGTGG"GACCTGAGGAAC"GCCCCCCTGCAAAGCAGGGCCCACCTCC"GGACT AGAGAGCCCAGGGCAACTGCCAAGCAGGGGGACAAGTATTCTGGCGGGGGGTGGGGGAGAGAGCAGGCC C"GTGGTGGCAGGAGGTGGCATCTTGTCTCGMCCC"GATGTCTGCTCCAGTGATGGCAGGAGGA"GGAGA AGTGCCAGCAGCTGGGGGTCAAGACGTCCCC"GAGGACCCAGGCCCACACCCAGCCCTTCTGCC"CCCAA i-CLCTCLCCLCCGLCCCCT-CCLCCACiGC-GCC-AALGCAAGGCAGTGGCTCAGCAGCAAGAATGCTG G"TCTACATCCCGAGGAGTGHCTGAGGTGCGCCCCACTCTGTACAGAGGCTGHTTGGGCAGCC""GCCTC CAGAGAGCAGATTCCAGCTTCGGAAGCCCCTGGTC"AACTTGGGATCTGGGAATGGAAGGTGC"CCCATC GGAGGGGACCCTCAGAGCCCTGGAGACTGCCAGGTGGGCCTGCTGCCACTGTAAGCCAAAAGG"GGGGAA GTCCTGACTCCAGGGTCCTTGCCCCACCCCTGCCTGCCACCTGGGCCCTCACAGCCCAGACCCTCACTGG GAGGTGAGCTCAGCTGCCCTHTGGAATAAAGCTGCCTGATCAAAAAAAAAAAAAAAAAAAAA Figure 8 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 11/19 SEQ ID NO:2 >gi | 125656151 | ref | 902 . 2| Mus musculus transmembrane serine protease 6 (Tmprss6) mRNA AGTTTCATTGTCGCCCTGGACCTGACAGGAGAGGCCCATGGAACTTGGGGCCACAGGCCACAAGGGACAA GGGCCAGACACCCCAGCCATGGC"CCAGGCCATTGA"CCAACCTAAGCTGGCCAGT"GGGGGTGGAAAGA CCTTGGCC"GGATAAACAGAGGCCTCCAGGCCTGTG"GCAGGCCCGGCACC ACCL CCACTC GAAGA GA-G__TCCAGC_CCCC_GlTC_ACCAGGATGCCCACCACCGAGGTCCCCCAAGCGGCTGATGG TCAGGGCGATGCGGGTGAHGGAGAGGAAGCTGCTGAGCCAGAGGGGAAGTHCAAGCCCCCAAAAAACACC AAGAGAAAAAACCGGGACTACGTCCGCT"CACGCCACTGTTGCTGGTC"TGGCTGCGCTGGTC"CAGCAG LGCL--GGLA-iTCCiAGGGTACAAAGCGGAAGTGACCGTAAGCCAGGTGTACTCTGGCAGCCT CCGGGTGCTCAACCGTCAHTTCTCCCAGGACCTGGGCCGACGGGAGTCHAHTGCTTTCCGCAGHGAATCT GCCAAAGCCCAGAAGA"GCTCCAAGAAC"GGTTGCCAGCACCCGCCTGGG ACTTAC ACAAC C AGTT C G CTACiCCTTiGGGGAGGGACCCCTCACCTGC iC-TC-GG -TA-CCLLGACA CCCTGAG"ACCA GCGACTGACCC_GAGCCC_GAAGLAGTGCGCGAGC_CC_GG_GGA_GAGC-ACTGLCCAACAGCTCAACC TCCTAHAAGACCGAATATGAGG"GGACCCGGAAGGCC"GGTGATCCTGGAAGCCAG"G"GAACG ACA AGTCGLACTGAALLCCACGC GGGCiG TALCGC ACAGC ATG GAACCCAGGCCAGG"CCTCCC ATTGAAGGGGCCTGACCAGCAGACCACAAGC-GCC-GLGGCALC-GCAAGGGCCCGAAGACC-CALGA-C CGGCTGGAGTGGACCCGGGTCGATTGCAGAGACAGGG'GGCGATGTACGACGCAGC'GGGCCCC "GGAGAAGAGACTTATCACC"CGG"CTATGGGTGCAGCCGCCAGGAACCTGTGATGGAGGTGC"GGCA"C GGGCiCCGiCAiGGCCGLGG-G GGAAAAAGGGCA"GCATAGC ACTALGACCCT-iCCiGC-CiCAG-G AAGTCTGTGGCCTTCCAGGACTGCCAGGTGAACCHGACACTGGAGGGCCGGCTGGACACACAGGGCTTCC "CCGTACACCCTACTACCCCAG"TACTACTCTCCCAGiACCCAC GCTCCiGGCAHCiCACGG ACCC"C C GGACiACGGCiiGGCGC-C GGTTCGATGCCTACGCACTGAGGAGGCAGAAG"ACAACCGAC GTG ACTCAGGGCCAGTGGATGATCCAGAACAGGAGGC_G_GlGGCLiCCGTACCCTGCAGCCATATGCTGAGA GGATCCCCATGGTGGCCTCAGA"GGTGTCACCATCAACTTCACC"CCCAGATCTCCCTCACAGGCCCGGG G GCAAG G ACTACAGCT"G"ACAACCAATCAGACCCCTGCCCTGGTGAGTTCC C GCTC G GAA GGAC-G-G-G-CCCiGCGiG-GACGGGATCAAGGAC"GCCCCAA-GGCC-GGATGAGAGAAAC-G-GTC- GCAGAGCCATGTTCCAGTGCCAAGAGGACAGCACGTGCATTTCACTGCC"AGAGTCHGHGACCGGCAGCC CGACHGHC"CAATGGCAGTGACGAAGAACAGTGCCAAGAAGGAG GCCC G GGGACA TCAC i CCAG TG"GAGGACCGGAGCTGTGTGAAGAAGCCCAACCCAGAGTGTGACGGCCAG CAGA iGCAGAGACGGCT CAGAT AGCAACACTGTGACTGTGGCCTCCAGGGCCTCTCCAGCCGTA__G_GGGCGGGACCG_G_CCLC CGAGGG"GAGTGGCCA"GGCAGGCCAGCC CCAGAT CGGGGTCGACACA"CTGTGGGGGGGC"C"CATC GC GACCGC GGGiCA AACGGCCGCCCAC"GCTTCCAGGAGGACAGCA"GGCCTCCCCGAAGCTGTGGA CCGlGT_CC_GGGAAAGATGCGGCAGAACTCGCGCTGGCCAGGCGAGG_G_CCTLCAAGGTGAGCCGTCT GT"CCTGCACCCGTACCACGAGGAGGACAGCCATGACTACGACGTGGCCCHGCTGCAGCTCGACCACCCC GTGGTGHAC"CGGCCAC"GTGCGCCCCGTC"GCCTGCCTGCCCGCTCCCACTTCT"TGAGCCAGGCCAGC AC GCTGGA CACAGGCTGGGGAGCCCAGCGAGAGGGTGGTCCGGTGAGCAACACCCTGCAGAAGGTGGA CGHACAGCTGGTCCCHCAGGACCTCTGCAG"GAGGCCTACCGCTACCAGGHGTCCCCACGCAHGCTCTGT GC"GGCHACCGCAAGGGCAAGAAAGATGCC"GCCAGGGTGAC C CCACHGG"TTGCAGGGAGC CCAGTGGCCGCTGG CCTGGCAGGGTLGG -AGC GGGGCC GGGCTGiGGCCGACCCAAT iCi-TGG CGTCTACACCCGTG_CACACG_G1GATCAAC_GGA_CCAGCAGGTGCTGACClGAGGGClG-_C_ACAGA CCTGCCTCCAGGCCAAGTTCAGGG"GTCCACCCAGCCAGGACACAAGTA"TCHGGGGCAAGTGA CCC"GCTAAGGCCTG iiCCC CAGGCCiACCCCAGTGACAG"ACAGAGAAGGATGTCAGCTGG"GGTTA GGA-GCCLCCLGAGG-CCAGGGGCCAGCC-CGGCLAGGLTTCAC-iCTAACCCTT-C--AT-CiAGiCCi TTCCCCTCCCTGCTCCTACCACHGTTTTGGAGTGGGGTCTGGCGGCCATGACCTTGGCCTCCGGGTCTCT GTAGGAAAGAAAGAA"CCTTCCCCTTGCAAAAGCCTCTTGGGGGAACTGCACAGAGAAAGAAGG"GCCTC TA"CAAGGCTCTATCAGAGCCC iGAGiC GCCAAGiGGGCiGiACTCiAAGCCAAA"CACCGGGCAGCC TCAGCTGCAGATGCCTGCTGAAGCTCTGCCTGCTACAGGGGCCTCCCTGCCATTCACTGGAGGCCCACTG TC"GTTCTGGGAATAAAGCACT"GACCAAGCCCTGACACTGAAAAAAAAAAAAAAA Figure 9 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 12/19 SEQ ID NO:3 >gi | 194474097 Ire leM_OOll30556 . l] Rattus norvegicus transmembrane se, serine 6 (Tmprss6) mRNA ATTGTCCGTCCTGGACCTGACAGGAGGCCCATGGAACTTGGGGCCACAGGCCACGAGGGACAAGGGCCAG CAGLCATGGL LATTGA iAAGCTGGCCAGT"GTGGGTGGAGAGACCTTGGC C"GGATAAACAGAGGCCTCCAGGCCTGiG CAGGCCCAGCACC ACCL CCACTC GAAGA GCCAAG A_Gl_TCCAGC_CCCC_GlTC_ACCAGGATGCCCACCGCTGAGGTTCCCCAAGCAGC_GGTGG_CAGGG_ GATGGAGGTGAHGGAGAGGAAGCTGCAGAGCCAGAGGGGGTGTHCAAGGCCCCCAGAAACGCCAAGAGAA AAGACAGGGACTACGTCCGCT"CACACCAC GTTGCLGGTC iGGCTGCG iGGCi CGGCAGGAGTCA" GCiC-GGiA i-CCiAGGGTACAAGGCGGAAGTGACCATAAGCCAGGTGTACTCTGGCAGCCTCCGGGTG CTCAACCGCCAHTTTTCACAGGACTTGGCCCGACGGGAGTCHAHTGCTTTCCGCACHGAAACTGCCAAAG CCCAGAAGA"G"TCCAAGAGC"GGTTGCCAGCACCCGCHTGGG ACTTAC ACAAC CCAGTTCCATC"A CGCCTTTGGGGAGGGACCCCL ALCTGC iC-TC-GG -CA-CCLLGACA CCCCGAGTACCAGCGACTG ACCC_GAGCCC_GAGGlGGTGCGCGAGC_CC_GG_GGG_GAGC-ACTGLCCAACAGC_CAGCCL_GGC_L CC"A"AGGACCGAATATGAGGHGGACCCGGAAGGCC"GGTGATACTAGAAGCCAGCG"GAACGACATAGT CG ACTGAALLCCACGC GGGCiG TACCGC ACAGC ACG GAACCCGGGCCAGG CCiCCGG iGAGG GGGCCCGACCAGCAGACCACLAGC-GCC-GiGGCACC-GCAGGGGCCCGAGGACCTCATGCTCAAAGTGC AGCTAGAGTGGAC'CGGGTTGATTGCAGAGACAGGGHGGCGATGTACGACGCAGC'GGGCCCCTGGAGAA GAGACTTATCACC"CGG"CTATGGGTGCAGCCGCCAGGAACCCGTGATGGAGGTGC"GGCGTCGGGCTCT GiCAiGGCCGLGG-G GGAAGAAGGGC- GCATAGC CTALGACCCT-1 CLGC-C CAGTGAAGTCTG TGGCCTTCCAGGACTGCCAGGTGAACC"GACCCTGGAAGGCCGGC"GGATCCACAGGGCTTCCTCCGTAC ACCCTACTACCCCAG"TACTACTCGCCCAGiACCCAC GCTCCiGGCA"C CACGG CCCTCLCTGGAC iAiGGCiiGGCAC-C GGTTLGACGCC-A GCACiGAGGAGGCAGAAG-ACAACC AC ATG ACLCAGG GCCAGTGGATGATCCAGAACAGGAGGC_A_GlGGCLiCCGTACCCTGCAGCCATA-GC_GAGAGGATCCC CGTGGTGGCC"CGGA"GGTATCACCATCAACTTCACC"CCCAGATCTCCC"CACAGGCCCGGGTGTGCAA G G GCT"G"ACAACCAATCAGACCCCTGCCCiGGAGAG TCCiC GCTC G GAA GGATTG G-G-CCCiGC-1G-GACGGAATCAAGGAC"GCCCCAACGGCCTGGATGAGAGGAAC-G-GTC-GCAGAGC CATGTTCCAGHGCCAAGAGGACAGCACGTGCATCTCACTGCCGAGAGTCTGHGACCGGCAGCCCGACTG" C CAATGG AGCGACGAAGAGCAGTGCCAAGAAGGAGiGCCC G GGGACA TCAC i CCAGLG GAGG GC G-GTGAAGAAGCCCAACCCCGAGTGTGACGGGCAGGCAGACTGCAGGGA GGC-CGGATGA GGAGCACTGTGACTGTGGCCTCCAGGGCCCCTCCAGCCGCA__G_GGGCGGGGCCA_G_CCLCGGAGGGT GAGTGGCCC"GGCAGGCCAGTC"CCAGATTCGGGGTCGACACA"CTGTGGGGGGGC"CHCATCGCHGACC GC GGGTCA AACAGCCGCTCAC"GCTTCCAGGAGGACAGCA"GGCCTCCCCGAGGCTGTGGACCGTGT" TCTGGGAAAGATGCGGCAGAA-_CACGCLGGCCGGGCGAGG_G_CCTTCAAGGTGAGCCGCCTGTTCCTG CACCCGTATCATGAGGAGGACAGCCATGACTACGACGTGGCCCHGCTGCAGCTGGACCACCCTGTGGTG" AC"CGGCCACCGTGCGCCCCG C GCCTGCCCGCACGCTCTCACTTCTTTGAGCCAGGCCAGCAC"GCTG AGGCTGGGGAGCCCAGCGAGAGGGTGGTCCTGGTAGCAGCACCCTTCAGAAGGTGGATGTGCAA CTGATCCCTCAGGACCTGTGCAAHGAGGCCTACCG"TACCAGGHGACCCCACGCATGCHCTGTGCHGGT" ATCGCAAGGGCAAGAAAGATGCC"GCCAGGGCGAC C GGAGGCCCACTGG"TTGCAAGGAGCCCAGGTG ACCACCCAGCCAGGGCACAAG A CTGGGGCGAGCGACCC-GC AAGGCC GTCCCC CATGCC ACCC CAGGGACAGTACAGAGAAGGA-G_CAGC_GGlGGL_AGGATGCCTCCAGGGGCTAGCC_CAGCLCGGCL_ CACTTCCAACCCTTTCTTATTC"AGTCC"T"CCCC"C"CCCCTCCTACTGCHGTTTTGGGGTGGGGTCTG G GGCAALGA GCTGGiiCCAAGG CTG GGGAAAGLAAGA iCCiTCCCC iGCAAAAGCCTC"AGGGG GATCCGAGAAAGAAGG-GCCTC-A-CAAGGC-CTG-CAGAGCCC--GAGACLGCCAAG-AGGGC CATACCGTAAGCCAAATCATGGGGCAGCCTCAGCTGCGGGTGCCTGCTGTGCTCTGCCTGCTACAGGGCC C"CCCTGCCA"TCACTGGAGGCCCACTG C GTTCCGGAAA"AAAGCAGT"GGCCAAGC Figure 10 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 13/19 SEQ ID NO:4 >gi | 297260989 | ref | XM_001085203 . 2] PREDICTED: Macaca mulatta transmembrane protease, serine 6, transcript variant 3 (TMPRSS6) mRNA GCCTGTGGCCAAGGCCCCCCAGGTGGCTGGTGGGCAGGGGGACGGAGGTGATGGCGAGGAAGCG GAGCCAGAGGGGATG"TCGAGGCCCGTGAGGACTCCAAGAGAAAAGCCCGGGGCTACCTCCGCCTGGCGC CCCiGTGGCiGACCC GGTTG GCiGACi CAGTGGGGGTGCTACTCTGGTATTTCCTAGGGTACAAGGC GGAGGTGACGGTCAGCCAGGTGTACTCAGGCAGCC_GCGCG_GC_CAA_CGCCACLLClCCCAGGATCTT CGGGAATCCAGTGCCHHCCGCAGHGAAACCGCCAAAGCCCAGAAGATGCTCAAGGAGCTCATCG CCAGCACCCGCCTGGGAACTTA ACAAC CCAGC CCGTC Al CCT GGGGAGGGACCGCTCACCTG C iC-TC-GG--CAi CiCCAAA-CCCCGAGCACCGCCGGC-GA-GCTGAGCCCCGAGGTGGTGCAGGCA C"GCHGG"GGAGGAGC"GCTGHCCACAGTCAACAGCTCGGCGGC"GTCCCCTACAGGGCCGAGTACGAAG "GGACCCCGAGGGCC AGTGA CCTAGAAGCCAGHGTGAAAGACATAGC"GCACHGAATTCCACGCTGGG G -ACCGC-ACAGC ACGLGGGCCAGGGLCAGG GC GAAGGGACCCGACCACCTGGCCTCC C_GlGGCACC_GCAGGGCCCCGAAGACCTCATGCTGAAACTCCGGC_GGAG_GGACGC_GGCCG AGTGCCGGGACCGAC"GGCCAHGTATGACGTGGCHGGGCCCC"GGAGAAGAGGC"CA"CACC"CGGTGTA CAGCCGCCAGGAGCC"GTGGTGGAAGTCC"GGCATCGGGGGCCATCATGGCGGiGG C GGAAG AAGGGCCTGCACAGCTACTACGACCCC-i-ALGC-CLCCGTGCAGiCGG-GG-CTLCCAGGCCTGCGAGG 'AAACCTGACGCTGGAHGACAGGCTGGAC'CCCAGGGCGTCCHCAGCACCCCGTACTHCCCCAGCTACTA CTCGCCCCGAACCCAC GCTCCiGGCACC CACGG GCCCTC C GGAC ACGGCTLGGCCC C GGTTL GACGCCTACGCACTGCGGAGGCAGAAG-A GATT-GCCGTGCACCCAGGGCCAGTGGACGATCCAGAACA GGAGGCTGTGTGGCCTGCGCAHCCTGCAGCCTTACGCCGAGAGGATCCCCGTGGTGGCCACGGCCGGCAT CACCATCAALTTCACC CCCAGALCTCCC CACAGGGCCTGG G GCGGGiGCACTAiGGCT"G ACAAC CAG"CGGACCCCTGCCCiGGAGAGiTCCiC GCTC G GAACGGACTC-GCGLCCCLGCCTG GALGGGG TCAAGGACTGCCCCAACGGCCTGGATGAGAGAAAC_GCGTT_GCAGAGCCACATTCCAGTGCCAAGAGGA CAGCACGTGCATCTCACTGCTHAAGGTCTG"GACGGGCAGCCHGACTG"CTCAACGGCAGCGATGAAGAG CGG"GCCAGGAAGGGG GCCC GCGGGACA iCACCi CCAG G CAGAGCTGCGTGAAGAAGC CCAACCCACAGTGTGATGGGCGGCCCGACTGCAGGGACGGCTCAGACGAGCAGCACiG-GAC-GiGGCCi CCAGGGCCCCTCCAGTCGCATHGTHGGTGGGGCCGTG"CCTCCGAGGG"GAGTGGCCAHGGCAGGCCAGC C CCAGG iCGGGGiCGACACA"C"GTGGGGGCGCCC CATCGC GACCGC GGG"GA AACAGC GCCC A GCTiCCAGGAGGACAGCA"GGCCTCCCCGGCGCiGLGGACGGLGT-CC GGGCAAGGTGTGGCAGAA C_CGCGC_GGCCTGGAGAGGTGTCCTTCAAGGTGAGCCGCCLAC_CCTGCA-CCG-AlCACGAAGAGGAC AGCCACGACTACGACGTGGCGC"G"TGCAGCTCGACCACCCGGTGGTGCGC"CGGCCGCCGTGCG"CCAG TC"GCCTGCCCGCGCGCLCCCACL CT CGAACCCGGCCTGCAC GCTGGA CAC GGCLGGGGCGCCC" GCGCGAAGGCGGCCCCACCAGCAA_GC_CLGCAGAAAG_GGACG_GCAGLiGATCCCACAGGACCTGTGC AGCGAGGCCTATCGCTACCAGGHGACGCCACGCATGCTGTGTGCCGGCTACCGCAAGGGCAAGAAGGATG CC"GCCAGGGTGAC"CGGGTGG"CCGC"GG"ATGCAAGGCACTCAGTGGCCGCTGG"TCCTGGCAGGGC" GG CAGCiGGGGCC-GGGCTGiGGCCGGCC AACLACL-CGGCG-CTACACCCGCA CACAGGLG-GALC GGCTGGATCCAGCAAGTGGTGACCTGAGGAACTGCCCCCCTGCAGAGCAGGTCCCACCTC Figure 11 [Annotation] KIM None set by KIM [Annotation] KIM ionNone set by KIM [Annotation] KIM ed set by KIM [Annotation] KIM None set by KIM ation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 14/19 SEQ ID NO:5 >gi | 109094061 | ref | XM_001085319 . l] PREDICTED: Macaca mulatta similar to transmembrane protease, serine 6, transcript variant 4 (LOC696094) mRNA CTTGAGCCACACCCAGTCCAGCTCTGGTGCCTGCCCTCTGGGGTGAGCTGCCTTGAGATGCACTTCGCTC CTCTGTGAACTGTCTCGGCACCCACTTCCGGTCACTGCCGCCTGATGTTGT"ACTCTTCCACTCTGAAAG GA GCCiG GGCCCCCCAGGTGGC"GGTGGGCAGGGGGACGGAGG"GATGGCGAGGAAGCGGAG CCAGAGGGGATGTTCGAGGCCCGTGAGGACTCCAAGAGAAAAGCCCGGGGC_ACC_CCGCCLGGCGCCCC TG"GGCTGACCCTGGTTGHGCTGACTTCAG"GGGGGTGCTACTCTGGTATT"CCTAGGGTACAAGGCGGA GG GACGG CAGCCAGGTGTACTCAGGCAGCCTGCGCGTGC"CAATCGCCACTTC"CCCAGGATCTTACC GAATCCAGTGCCTTCCGCAGTGAAACCGCCAAAGCCCAGAAGATGCTCAAGGAGCTCATCGCCA GCACCCGCCTGGGAACTTATTACAACTCCAGCTCCGTCTAT"CCTTTGGGGAGGGACCGCTCACCTGCTT C CTGGL CATTC CCAAATCCCCGAGCACCGCCGGCTGAHGCTGAGCCCCGAGG"GGHGCAGGCACTG C"GGTGGAGGAGCTGCTGTCCACAGTCAACAGCTCGGCGGC-G CCCCiACAGGGCCGAGTACGAAGTGG ACCCCGAGGGCCTAGTGATCCTAGAAGCCAGTGTGAAAGACATAGCTGCAC_GAAL_CCACGC_GGGTLG T"ACCGCTACAGCTACGTGGGCCAGGGTCAGGTCCTCCGGCHGAAGGGACCCGACCACCHGGCCTCCAGC TGCCTGTGGCACCTGCAGGGCCCCGAAGACC CALGC GAAAC CCGGC GGAGTGGACGCTGGCCGAGT GCCGGGACCGACTGGCCA-GLALGACGLGGC-GGGCCCCTGGAGAAGAGGCTCATCACC-CGG-G-ATGG C"GCAGCCGCCAGGAGCCHGTGGTGGAAGTCCTGGCA'CGGGGGCCATCATGGCGGTGGHCTGGAAGAAG GGCCTGCACAGCTACTACGACCCCTTTATGC"CTCCG GCAGLCGGTGG C TCCAGGCCTGCGAGGTAA ACC-GACGCiGGA-GACAGGCTGGACTCCCAGGGCG"CCTCAGCACCCCGTACTTCCCCAGCTACTACTC GCCCCGAACCCACHGCTCCTGGCACCTCACGGTGCCC"CTCTGGACTACGGCTTGGCCCHCTGGTHTGAC GCACTGCGGAGGCAGAAGTATGA iHGCCG GCACCCAGGGCCAG GGACGA CCAGAACAGGA GGC GTG GGCCTGCGCATCC GCAGCCL ACGCCGAGAGGATCCCCGTGG"GGCCACGGCCGGCATCAC CALCAAL__CACC_CCCAGALC_CCCTCACAGGGCCTGGTGTGCGGGTGCAClALGGC-_GLACAACCAG TCGGACCCCTGCCCTGGAGAG""CCTCTGCTCTGHGAACGGAC"C"GCGTCCCTGCCTGMGATGGGGTCA AGGACTGCCCCAACGGCCTGGA"GAGAGAAACTGCG"TTGCAGAGCCACAT"CCAGTGCCAAGAGGACAG CACGLGCA-CLCACLGC-iAAGGLCTGLGACGGGCAGCCTGAC G-CTCAACGGCAGCGATGAAGAGCGG TGCCAGGAAGGGGTGCCCTGCGGGACATTCACCT"CCAGTGHGAGGACCAGAGCTGCGTGAAGAAGCCCA ACCCACAG"GTGATGGGCGGCCCGACTGCAGGGACGGCTCAGACGAGCAGCACTG"GACTGTGGCCTCCA GGGCCCCTCCAGTCGCA-TGL GGLGGGGCCGTG-CC CCGAGGG-GAGiGGCCA-GGCAGGCCAGCCTC CAGGTTCGGGGTCGACACATC_G_GGGGGCGCCC_CA_CGC_GACCGC_GGG_GA_AACAGC_GCCCAL_ GCTTCCAGGAGGACAGCATGGCC"CCCCGGCGCTGTGGACGGTGT"CC"GGGCAAGGTGTGGCAGAACTC GCGCTGGCCTGGAGAGG GTCC CAAGGLGAGCCGCCLAC CCiGCA CCG ATCACGAAGAGGACAGC CACGACTACGACGTGGCGCTG-_GCAGCLCGACCACCCGGTGGTGCGCTCGGCCGCCGlGCG_CCAGTC_ GCCTGCCCGCGCGCTCCCACTHCMTCGAACCCGGCCTGCACHGCTGGA"CACMGGCTGGGGCGCCCTGCG CGAAGGCGGCCCCACCAGCAA"GCTCTGCAGAAAG"GGACG"GCAGTTGATCCCACAGGACC"GTGCAGC GAGGCCTATCGCTACCAGGTGACGCCACGCA-GC-GLGLGCCGGCLACCGCAAGGGCAAGAAGGA-GCC- GCCAGGGTGACHCGGGHGGTCCGCTGGTATGCAAGGCACTCAGTGGCCGCTGGTTCCTGGCAGGGCTGG" CAGCTGGGGCC"GGGC"GTGGCCGGCCTAAC"ACH"CGGCGHCTACACCCGCATCACAGGTG"GA"CGGC TGGATCCAGCAAGTGG"GACCTGAGGAACTGCCCCCCTGCAGAGCAGGTCCCACCTCTTGGAC"CAGAGA GCCCAGGGCAATTGCCAAGCAGGGGGACAAG_AT_CiGGGGGGAGGGGGGCGCGAGCAGGCCC_G_GGLG GCAGGAGGTGGCATC""GTCTTGMCCCTGATGTCHGCTCCAGTGATGGCAGGAGGATGGAGGAG"GCCAG GGGG"CAAGACGTCCCC"AGGGACCCAGGCCCACACCCAGCCCiiCiGCCiCCCGAi C CTCi CCLCLGLCCCC-TCC-CCACLGC-GCCLALiGCAAGGAAGTGGCTCAGCAGCAAGAA-GCTGGC-CLACG TCCCCAGGAGHGTCTGAGCTGTGCCCCACTCTGTACAGAGGCTGCTTGGGCAGCCTTGCCTCTAGAGAGC AGA GCCAGC "CGGAAGCCCCTGGTCTAACTTGGGATCTGGGAATGGAAGGTGCCCCCATAGGAGGGGA CCCTCACAGCCCCGGGGACTGCCAGGTGGGCCGGCTGCCACCGTAAGCCAAAAAAGG"GGGGAAGCCCTG ACTCCAAGGTCCTTGCCCCACCCCTGCCTGCCACCTGGCCCCTCACAGCCCAGACCCTCACCGGCAGGTG AGCHCAGCTGCCCTTTGGAATAAAGCTGCCTGATCCAA Figure 12 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM /19 SEQ ID NO:6 Reverse Complement of >gi|56682967 releM_153609.2| Homo sapiens transmembrane protease, serine 6 (TMPRSS6), mRNA TTTTTTTTTTTTTTTTTTTTTGATCAGGCAGCTTTATTCCAAAGGGCAGCTGAGCTCACCTCCCAGTGAGGG TCTGGGCTG"GAGGGCCCAGGTGGCAGGCAGGGGHGGGGCAAGGACCCTGGAGTCAGGACTTCCCCACCT TGGCTTACAG"GGCAGCAGGCCCACCTGGCAGTC"CCAGGGCTCTGAGGG"CCCCTCCGATGGGAGCACC CCATTCCCAGATCCCAAGTTAGACCAGGGGCTTCCGAAGCTGGAAlCTGC_C_CLGGAGGCAAGGCTGCCCA AACAGCCTC"GTACAGAGTGGGGCGCACCHCAGACACTCCTCGGGATGTAGAACCAGCATTCTTGCTGCTGA GCCACTGCC GCALiAGGCAGCAGTGGAGGAAGGGGACGGAGGAGAGAGAA"TGGGAGGCAGAAGGGCTGG GiG-GGGCC GGGLCCiCAGGGGACGTCT"GACCCCCAGCTGC GGCACL C-CCALCCLCCLGCCATCACT GGAGCAGACAHCAGGGACGAGACAAGATGCCACC"CCTGCCACCACAGGGCC"GCTCHCTCCCCCACCCCCC GCCAGAA"AC"TGTCCCCCTGCLiGGCAG GGCTC CLGAGTCCAGGAGG GGGCCC GCTTTGC GCAGTTCCTCAGGTCACCACTiGC GGA-CCAGCTGA CACACC G-GAiGCGGGTG"AGACGCCG AAGTAGTTAGGCCGGCCACAGCCCAGGCCCCAGCTGACCAGCCCCGCCAGGAACCAGCGGCCACTGAGTGCC H"GCACACCAGCGGACCACCTGAGHCACCC"GACAGGCATCC""CTTGCCCT"GCGG"AGCCGGCACACAGC GGCG CACC GGTAGCGA AGACC CGC GCACAGG CCiGTGGGA CAAC GCACA CCACT C TGCAGAGCG--GCiGALGGGGCCGCCCTCGCGCAAGGCGCCCCAGCCCGTAATCCAGCAGTGCAGGCCGGGC HCGAAGAAG"GGGAGCGCGCGGGCAGGCAGACGGGGCGCACGGCGGCCGAGCGCACCACCGGGHGGTCGAGC "GCAGCAGCGCCACG CA GGC G CCTC iCGTGG ACGGGTGCAGGAGCAGGCGGCiCACC G AAGGACACC"CTCCAGGCCAGCGCGAG- C GCCACACCTTGCCCAGGAACACGGTCCACAGCACCGTGGAG GCCATGCTG"CCTCC"GGAAGCAG"GGGCAGCTGHHATCACCCAGCGGTCAGCGAHGAGGGCCCCCCCACAG A"GTGTCGACCCCGAACCTGGAGGCTGGCC GCCA GGCCAC CACCCTCGGAGGACACAGCTCCACCAACA A"GCGGCTGGAGGGGCCCTGGAGGCCACAG CACAGLGCTCC AGCCGTCCCTGCAGTCGGGCCGC CCAlCACAC_GCGGG_lGGGC__C_iCACGCAGCTCCGGTCC_CACACLGGAAGG_GAA1GLCCCACALGGC ACCCCTTCC"GGCAC"GCTCT"CG"CGCTGCCGT"GAGACAAHCAGGCTGCCCATCACAGACCTTGGGCAGT GAGATGCALG GCTG CCTCT GCAC"GGAATG GGC ACGCAG TTC CAGGCCGTTG GGGCAGLCC--GACCCCATCACAGGCAGGGACACAGAG-CCA-iCACAGAACAGAGGAACTCTCCAGGGCAG GGGHCCGAC"GGTTGHACAAGCCAHAG"GCACCCGCACACCGGGCCCGGTGAGGGAGA"CTGGGAGGTGAAG TTGATGG"GA"CCCGGCCGTGGCCACCACGGGGA"CCTCTCGGCGTAGGGC GCAGGA GCGCAAGCCACAC AGCCLCC G -CTGGALCGTCCAC-GGCCCiGGG-GCACGGCAAALCA-AC LCTGCC CCTCAG GCA-AG GCATCAAACCAGAGGGCCAAGCCG_AG_CCAGAGAGGGCACCGTGAGGTGCCAGGAGCAGTGGG__lGGGGC GAGMAGTAGC"GGGGAAGTACGGGGTGCTGAGGACGCCCTGGGAGTCGAGCCTGTTG CCAGCG CAGG" C ACT"CACAGGCCTGGAAGACCACCGGC"GCACGGAGAGCACGAAGGGG CG AGTAGC GTGCAGGCCC C TTCCAGACGACCGCCATGATGGCCCCCGACGCCAGAACCTCCACCACGGGCTCCTGGCGGCTGCAGCCGTAC ACCGAGGTGAMGAGCC"CTTCTCCAGGGGCCCGGCCACGTCATACATGGCCAGTCGG"CCCGGCACTCTGCC AGCGTCCACTCCAGCCGGAGTL GAGCA GAGGTCC"TGGGGCCCTGCAGG"GCCACAGGCAGCTGGAGGCC AGGTGGTCAGGCCCCT"CAGCCGGAGGACCTGGCCC"GGCCCACGLAGCiG-AGCGG AACAACCCAGCGTG GAATTCAATGCAGCTA"GTCTTHCACAC"GGCTTCCAGGATCACTAGGCCCHCGGGGHCCACTHCGTACTCG GCCCTGTAGGGGACGGCAGCCGAGCTG""GACTGTGGACAGCAGCTCCTCCACCAGCAGTGCC"GCACCACC "CGGGGCTCAGCATCAGCCGGCGGiGC-CGGGGAii GGAGAA GAACCAGAAGAAGCAGGTGAGGGGTCCC TCCCCAAAGGAATAGACGGAGC_GGAG__GlAGTAAG_lCCCAGGCGGGLGClGG_GAlGAGC_CClTGAGC ATC"TCTGGGCTTTGGCGGTTTCACTGCGGAAGGCACMAGATTCCCGGCGGGTAAGA"CCTGGGAGAAG"GG CGA"TGAGTACACGCAGACTGCCTGAG"ACACCTGGC GACCA CACCLCCGCCT G GGAAA AC CAGAGTAGCACCCCCGCCGAAGCCAGCACGAGCAGGGCCAGCAGCACAAACAGGGGCACCAGGCGGAGGTAG CCCCGGGCTTTTCTCTTGGAGTCCTCACAGGCCTTGAACATCCCCTCCGGCTCCGCTHCCTCGCCATCACCT CCCTGCCCGCCAGCCACCTGGGGGGCCTCGGCCACGGGCATCCTTTTGGAG"GGAAGAG"AACAAC ATCAGGCGGCAGTGACTGCAAGTGGGTGCCGAGACAGCTCACAGAGGAGGGAAGTGCATCTCAGGTCAGCTC GCACCAGAGGGCAGGCACCAGAGCTGGACTGGGTCTGGCTCAAG Figure 13 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 16/19 SEQ ID NO:7 Reverse Complement of >gi | 125656151 | releM_02 7902 . 2| Mus musculus transmembrane serine protease 6 (Tmprss6) mRNA TTTTTTTTTTTTTTTCAGTGTCAGGGCTTGGTCAAGTGCTTTATTCCCAGAACAGACAGTGGGCCTCCAGTG AA"GGCAGGGAGGCCCCTGTAGCAGGCAGAGCTTCAGCAGGCA"CTGCAGCTGAGGCTGCCCGG"GATT"GG CT"AGAGTACAGCCCACTTGGCAGACTCAAGGGCTC"GATAGAGCCTTGATAGAGGCACCTTCi CTC G1 CCCCCAAGAGGCTTTTGCAAGGGGAAGGA__ClTTCL__CCTACAGAGACCCGGAGGCCAAGGTCA TGGCCGCCAGACCCCACTCCAAAACAGTGGTAGGAGCAGGGAGGGGAAAGGACTAGAATAAGAAAGGGTHAG AAG GAAACC AGCCGAGGC"GGCCCCTGGACCTCAGGAGGCA"CCTAACCACCAGCiGACA CC TCTC G iAC-GTCACiGGGG-AGGCC-GAGGGAAACAGGCC- AGCAGGG-CAC1-GCCCCAGAALAC-1G-GTCC G TGGACACCCTGAACHTGGCCTGGAGGCAGGHCCAGCTC"GTAGAACAGCCCTCAGGHCAGCACCHG C"GGATCCAG"TGA CACACGiG GACACGGGTGHAGACGCCAAAGAAA GGGTCGGCCACAGCCCAGGCC CCAGCTAACCAACCCTGCCAGGAACCAGCGGCCAC"GGGCTCCC"GCAAACCAGTGGGCCTCCAGAGTCACC CTGGCAGGCA-CT-_CllGCCC-_GCGGLAGCCAGCACAGAGCATGCGTGGGGACACC_GG-AGCGGTAGGC C"CACTGCAGAGG"CCTGAGGGACCAGCTGTACGHCCACCTTC GCAGGG GHTGCTCACCGGACCACCCTC TCGCTGGGCTCCCCAGCCTGTGA"CCAGCAGTGC"GGCCTGGC"CAAAGAAG"GGGAGCGGGCAGGCAGGCA GACGGGGCGCACAGTGGCCGAGTACACCACGGGG-GGLCGAGC-GCAGCAGGGCCACG-CG-AGLCATGGCL G"CCTCCTCGHGG'ACGGGTGCAGGAACAGACGGCTCACCTTGAAGGACACC"CGCCTGGCCAGCGCGAGTT C GCCGCA C TTCCCAGGAACACGGTCCACAGC"TCGGGGAGGCCATGCTG"CCTCC"GGAAGCAGTGGGC TA"GACCCAGCGGTCAGCGATGAGAGCCCCCCCACAGATGTGTCGACCCCGAATCTGGAGGCTGGC C"GCCATGGCCACHCACCCTCGGAGGACACGGTCCCGCCCACAATACGGCTGGAGAGGCCCHGGAGGCCACA G"CACAGTG""GC"CATCTGAGCCGTCTCTGCAA"CTGACTGGCCGTCACACMCTGGG TGGGC TCTTCAC ACAGCTCCGG CC-CACACTGGAAAGTGAATGTCCCACAGGGCACLCC-1C -GGCAC GTTC CGTCACL GCCATTGAGACAGTCGGGCTGCCGGTCACAGACTCTAGGCAGTGAAATGCACGTGC_G_CC_C__GGCAC_G GAACATGGC"CTGCAGACACAG"TTCTCTCAMCCAGGCCATTGGGGCAGTCC"TGAHCCCGMCACACGCAGG GACACACAG"CCA"TCACAGAGCAGAGGAAC"CACCAGGGCAGGGGTC GA "GG G ACAAGCLGTAG A CACACCCGGGCCTGTGAGGGAGA C-GGGAGG-GAAG1iGATGGLGACACCATCTGAGGCCACCAT GGGGATCCTC"CAGCAHATGGC"GCAGGGHACGGAAGCCACACAGCCTCCTGHTCHGGATCATCCACTGGCC CTGAGTACACAGTCGG TGTAC iCTGCC CCTCAGiGCGTAGGCATCGAACCAGAGCGCCAAGCCGTAG"C CAGAGAGGGTACCG"GAGATGCCAGGAGCAG-GGG AC-GGGAGAGTAGTAACTGGGG"AGTAGGGTGTACG GAGGAAGCCC_GTG_G_CCAGCCGGCCC_CCAGTG_CAGGTLCACCTGGCAG_CC_GGAAGGCCACAGAC__ CACTGAGAGCAGGAAAGGGTCA AGTAGC AMGCA"GCCCTTTTTCCACACCACGGCCATGACGGAGCCCGA TGCCAGCACC CCA CACAGGi CCTGGCGGCTGCACCCATAGACCGAGGiGATAAG CLCTLC CCAGGGG CCCAGCTGCGTCGTACATCGCCACCCTG_ClCTGCAA-CGACCCGGGTCCACTCCAGCCGCAC___GA_CA_ TTCGGGCCCTTGCAGAMGCCACAGGCAGC"TGTGGTCTGCTGGHCAGGCCCCTTCAAHGGGAGGAC C"GGCCTGGG TCACAiAGCTG AGCGA AACAGCCCAGCGTGGAATTCAGiACGAC ATGTCG CACAC GGCTTCCAGGATCACCAGGCCL CCGGG-CCACCiCA-ATTCGGiCTTA-AGGAAGCCAGGG- GAGC-G TAGC"CATCCACCAGGAGCTCGCGCACTACTHCAGGGCTCAGGGHCAGTCGCTGGTACHCAGGGA" G CAAGGA GAAGAAGCAGGTGAGGGGTCCC"CCCCAAAGGAG"AGACAGAACTAGAG GTAG A AG"ACCCAGGCGGGTGC"GGCAACCAGi C1-GGAGCA-CT-C1GGGC1 GGCAGA iCAC GCGGAAAGC CTCCCGTCGGCCCAGGTCCTGGGAGAAATGACGGTTGAGCACCCGGAGGCTGCCAGAG_ACACC_G GC"TACGG"CACTTCCGCTTTG"ACCCTAGGAAATACCAAAGCA"GACCCCTGCTGAGACCAGCGCAGCCAA GACCAGCAACAGTGGCG"GAAGCGGACG AG CCCGG TT C CTTGG GiTTi GGGGGC iGAAC CCCCTCTGGCTCAGCAGCiTCC-CLCCA-CACCCGCA-CGCCC-GACCALCAGCCGC--GGGGGACCTCGGT GG"GGGCA"CCTGGTAGAACAGGGGAGC"GGAAACATCHCGGCA"CTTCAAGAGTGGAAGGTAGGTGCCGGG CC"GCACACAGGCCTGGAGGCC CTGTT A CCAGGCCAAGG C TTCCACCCCCAAC"GGCCAGCTTAGG" AA GGCCiGGAGCCATGGCTGGGG GTCiGGCCCTiG CCCTiGiGGCCiG GGCCCCAAGTTCCA TGGGCCTCTCCTGTCAGGTCCAGGGCGACAATGAAACT Figure 14 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 17/19 SEQ ID NO:8 Reverse Complement of 4474097|ref|NM_OOll30556.ll Rattus norvegicus transm mbran prot as s rim 6 mRNA , (Tmprss6), GCTTGGCCAACTGCTTTATTTCCGGAACAGACAGTGGGCCTCCAGTGAATGGCAGGGAGGGCCCTGTAGCAG GCAGAGCACAGCAGGCACCCGCAGCTGAGGCMGCCCCAHGAT GGCTTACGGTATGGCCCTACTTGGCAGT C"CAAGGGC"CTGACAGAGCCTTGATAGAGGCACCi C TTC CGGATCCAGTTCCCCCTAGAGGCTTTTGC AAGGGGAAGGAATCTTACTTTCCCACAGACCTTGGAACCAGCATCATTGCCACCAGACCCCACCCCAAAACA GCAGTAGGAGGGGAGAGGGGAAAGGACTAGAATAAGAAAGGG"HGGAAGTGAAGCCGAGCTGAGGCTAGCCC C"GGAGGCA"CCTAACCACCAGCLGACA CC TCTC G ACLG CCCTGGGGTAGGCATGAGGGGACAGGCC - AGCAGGG"CGCTCGCCCCAGAALACL-G-GCCCiGGCTGGG"GGTCACCLGGGC-CC-TGCAAACCAGTG CAGAGHCGCCCHGGCAGGCATC"H"CTTGCCC"TGCGATAACCAGCACAGAGCATGCGTGGGGTCA CC"GGTAACGGHAGGCC"CATTGCACAGG"CCTGAGGGATCAG iGCACA CCACC CMGAAGGGTGCTGC "ACCAGGACCACCC"CTCGCTGGGCTCCCCAGCCTGTGATCCAGCAGTGC GC-CAAAGAAGTGAG AGCGTGCGGGCAGGCAGACGGGGCGCACGGTGGCCGAGTACACCACAGGGTGGTCCAGCTGCAGCAGGGCCA CG CGTAGLCAHGGC G"CCTCCTCATGA"ACGGG"GCAGGAACAGGCGGCTCACC""GAAGGACACC"CGC CCGGCCAGCGLGAA C GCCGCA CTiiCCCAGAAACACGG"CCACAGCCTCGGGGAGGCCATGCTGTCCT CCTGGAAGCAGTGAGCGGCTG-LA-GACCCAGCGG-CAGCGA-GAGAGCCCCCCCACAGATG-G-CGACCCC GAATCTGGAGACTGGCC"GCCAGGGCCAC'CACCC'CCGAGGACATGGCCCCGCCCACAATGCGGCTGGAGG GGCCCTGGAGGCCACAG CACAGLGCTCC CATCCGAGCCATCCCTGCAGiCTGCC GCCCG CACACHCGG GG LGGGC iC-TCACACAGCTCCGGTCC CACAC GGAAAG GAATGLCCCACAGGGCACTCC LCT-GGC CT"CGHCGCTACCATHGAGACAGHCGGGCHGCCGGTCACAGACTCTCGGCAGTGAGATGCACGHGC "G CCTCT GGCAC"GGAACA"GGCTCTGCAGACACAG"TCC"CTCATCCAGGCCG"TGGGGCAGTCC""GA "TCCGTCACAAGCAGGGACACACAATCCA"TCACAGAGCAGAGGAACTCTCCAGGGCAGGGG C GATTGGL _G_ACAAGClG_AG_ACACTLGCACACCCGGGCCTGTGAGGGAGATCTGGGAGGTGAAGTTGATGGTGATAC CA"CCGAGGCCACCACGGGGAHCCTCTCAGCATATGGC"GCAGGGMACGGAAGCCACATAGCCTCCTGHHCT TCCACTGGCCCLGAG ACATAGLAGGTTG AC TCLGCC CCLCAGTGCA AGGCG CAAACCAGA GTGCCAAGCCA-AG-CCAGAGAGGGAACCG-GAGA-GCCAGGAGCAGTGGGTACTGGGCGAG-AGLAAC-GG GGHAGTAGGGTGTACGGAGGAAGCCCTGTGGATCCAGCCGGCCT"CCAGGGTCAGGHHCACCHGGCAGHCCT GGAAGGCCACAGAC GAGCAGAAAAGGG"CA"AGAAGC"ATGCAAGCCC CTTCCACACCACGG CCATGACAGAGCCCGACGCCAGCACCTCCA CACGGGL-CCiGGCGGC-GCACCCA"AGACCGAGGTGATAA G_C_CTLC_CCAGGGGCCCAGCTGCGTCGTACATCGCCACCCLG-CTC_GCAATCAACCCGAG_CCAC_CLA GCiGCACT GAGCA GAGGTCCTCGGGCCCCTGCAGG"GCCACAGGCAGCTAGTGG"CTGCTGGHCGGGCC CCC"CAACCGGAGGACC"GGCCCGGGT"CACGTAGC1G AGCGG AACAGCCCAGCG GGAAL CAGTACGA C_A_GTCG__CACGC_GGCTLC_AG1A_CACCAGGCCL_CCGGG_CCACCLCATAL_CGGTCC_A_AGGAAG CCAAGGCTGAGCTG""GGACAGHAGCTCACCCACCAGGAGCTCGCGCACCACCTCAGGGCTCAGGGTCAGTC GCLGGTAC CGGGGA G CAAGGA GAACCAGAAGAAGCAGATAAGGGGTCCCTCCCCAAAGGCG"AGATGG AAC-GGAG AGTACCCAAGCGGGTGCTGGCAACCAGCTCTTGGAACATC CLGGGC -LGGCAG TGCGGAAAGCAATAGACTCCCGHCGGGCCAAG"CCTGTGAAAAATGGCGG""GAGCACCCGGAGGC TGCCAGAG ACACC GGCTTALGGHCAC iCCGCC iGHACCCiAGGAAAiACCAGAGCATGAC"CCTGCCG AAGCCAACGCAGCCAAGACCAGCAACAG"GGTGTGAAGCGGACG AGTCCCLGTCL CTC GGCGTL C TGGGGGCC-_GAACACCCCC_CLGGCTC_GCAGCL_CC_CTCCA-CACCLCCATCACCCTGACCACCAGCTG CTTGGGGAACCTCAGCGGTGGGCAMCCTGGTAGAACAGGGGAGC"GGAAACATCTTGGCATC""CAAGAG"G GAAGGTAGG"GCTGGGCCTGAACACAGGCCTGGAGGCC CTGii ATCCAGGCCAAGG CTC CCACCCACA ACTGGCCAGCLLAGGLLGGA-CAA-AGCCiGGAACCALGACLGGGGTGLCLGGCCCLLG-CCC-CGLGGCCL GTGGCCCCAAGTTCCATGGGCCTCCTGTCAGGTCCAGGACGGACAAT Figure 15 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 18/19 SEQ ID NO:9 Reverse Complement of >gi|297260989|ref|XM_001085203.2| PR4DIC «D: Macaca mulatta transmembrane se, serine 6, transcrip: variant 3 (TMPRSS6), mRNA GAGGTGGGACCTGCTCTGCAGGGGGGCAG"TCCTCAGGTCACCACTTGC"GGATCCAGCCGAHCACACCTG" GATGCGGGTG"AGACGCCGAAGTAGTTAGGCCGGCCACAGCCCAGGCCCCAGCTGACCAGCCCTGCCAGGAA CCAGCGGCCACTGAGTGCCTTGCATACCAGCGGACCACCCGAG_CACCC_GGCAGGCA1CC__CllGCCCL_ GCCGGCACACAGCATGCGTGGCGHCACCHGGMAGCGAHAGGCC"CGCTGCACAGGHCCTGTGGGA" CAACTGCACG CCAC iCTGCAGAGCA GCTGGiGGGGCCGCCTTCGCGCAGGGCGCCCCAGCCAGTGA" CCAGCAGTGCAGGCCGGGTTCGAAGAAGTGGGAGCGCGCGGGCAGGCAGACTGGACGCACGGCGGCCGAGCG CACCACCGGGHGGTCGAGCTGCAACAGCGCCACG"CGHAGTCGHGGCTGHCCTCTTCGHGAHACGGATGCAG GAGTAGGCGGCTCACC"TGAAGGACACC"CTCCAGGCCAGCGCGAGTTC"GCCACACC"TGCCCAGGAACAC CG"CCACAGCGCCGGGGAGGCCALGCTG CCLCC-GGAAGCAA"GGGCAGC"GTTATCACCCAGCGGTCAGC GATGAGGGCGCCCCCACAGALG_G_CGACCCCGAACC_GGAGGC_GGCC_GCCALGGCCAC_CACCCTCGGA GGACACGGCCCCACCAACAATGCGACTGGAGGGGCCC"GGAGGCCACAG"CACAGTGC"GCHCGTCTGAGCC GTCCCTGCAGTCGGGCCGCCCA CACAC GiGGG iGGGCTiC GCTCTGG"CCTCACACTGGAA GGTGAATGTCCCGCAGGGCACCCC-TCC-GGCACCGC-CTTCA-CGCTGCCGTTGAGACAGTCAGGCTGCCC GTCACAGACCTTAAGCAGTGAGATGCACG'GCTG'CC'CTTGGCACTGGAATGTGGCTCTGCAAACGCAG"T iC CTCA CCAGGCCGTTGGGGCAGTCC GACCCCA"CACAGGCAGGGACGCAGAGTCCGTTCACAGAGCA GAGGAACTCTCCAGGGCAGGGG CCGAC ACAAGCCA"AGTGCACCCGCACACCAGGCCCTGTGAG GGAGATCHGGGAGGTGAAATTGATGGTGAHGCCGGCCGTGGCCACCACGGGGATCCTCHCGGCGHAAGGCHG CAGGATGCGCAGGCCACACAGCCiCCTG CTGGALCGTCCAC"GGCCCTGGGTGCACGGCAAA CATAC C"GCCTCCGCAGTGCG AGGCG AGAGGGCCAAGCCG"AGTCCAGAGAGGGCACCGTGAGGTGCCA GGAGCAGTGGGTTCGGGGCGAG_AGlAGC_GGGGAAGTACGGGGTGCTGAGGACGCCC_GGGAG_CCAGCC_ G"CA"CCAGCGTCAGG"TTACC"CGCAGGCCTGGAAGACCACCGACTGCACGGAGAGCATAAAGGGGTCG"A G AGCTG GCAGGCCC"TCTTCCAGACCACCGCCA GA GGCCCCCGATGCCAGGACT"CCACCACAGGC"C CTGGCGGCTGCAGCCATACACCGAGGTGA GAGCC-C--CTCCAGGGGCCCAGCCACGTCATACATGGCCAG TCGGHCCCGGCACHCGGCCAGCG"CCACTCCAGCCGGAGTT"CAGCATGAGGHCTTCGGGGCCCHGCAGGHG CCACAGGCAGCTGGAGGCCAGG"GGTCGGGTCCCT"CAGCCGGAGGACCTGACCCTGGCCCACG AGCTG A GCGGTAACAACCCAGCGLGGAA CAGLGCAGCTA G CTT-CACACTGGC -CTAGGALCACLAGGCCC C GGGG_CCACl_CG_AClCGGCCC_G_AGGGGACAGCCGCCGAGC_GTTGAC_GTGGACAGCAGCTCCTCCAC CAGCAGTGCC"GCACCACCTCGGGGCTCAGCATCAGCCGGCGGTGCTCGGGGATTTGGAGAATGAACCAGAA GAAGCAGGTGAGCGGTCCCTCCCCAAAGGAA"AGACGGAGC"GGAGTTG1AA AAG iCCCAGGCGGGTGC" GGCGATGAGC_CC_LGAGCALC__C_GGGCL_TGGCGGlTTCAC_GCGGAAGGCACTGGATTCCCGGCGGGT AAGATCCTGGGAGAAGTGGCGAHHGAGCACGCGCAGGCTGCCTGAGTACACC"GGCHGACCGTCACCTCCGC CTTGTACCCTAGGAAATACCAGAG"AGCACCCCCACTGAAG"CAGCACAACCAGGG"CAGCCACAGGGGCGC CAGGCGGAGGTAGCCCCGGGCL -CTCLiGGAGiCCiCACGGGCCTCGAACATCCCCTCTGGCTCCGCTTC CTCGCCATCACCTCCGTCCCCCHGCCCACCAGCCACCTGGGGGGCCTTGGCCACAGGCATCCTG Figure 16 [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM [Annotation] KIM None set by KIM [Annotation] KIM MigrationNone set by KIM [Annotation] KIM Unmarked set by KIM 19/19 SEQ ID NO:lO Reverse Complement of >gi | 109094061 | releM_0010853l9 PR4 DICL 4 D: . 1| Macaca mulatta similar to embrane protease, serine 6, transcript variant 4 (LOC696094) , mRNA TTGGATCAGGCAGCTT"ATTCCAAAGGGCAGCTGAGCTCACC"GCCGGTGAGGGTC"GGGCTGTGAGGGGCC AGGTGGCAGGCAGGGG"GGGGCAAGGACCiiGGAGiCAGGGC CCCCACCLLTT GGCTTACGGTGGCAG CCGGCCCACCTGGCAGTCCCCGGGGCTGTGAGGGTCCCCTCC_A_GGGGGCACCL_CCA_TCCCAGATCCCA AGTTAGACCAGGGGCTMCCGAAGC"GGCATCTGCTC"CTAGAGGCAAGGC"GCCCAAGCAGCCTCMGTACAG AGTGGGGCACAGCTCAGACAC CC GGGGACGTAGAGCCAGCA CTTGC GCTGAGCCACTTCC"TGCAAT AGGCAGCAGTGGAGGAAGGGGACAGAGGAGAGAGAA"CGGGAGGCAGAAGGGCTGGGiG-GGGCC-GGGiCC C"AGGGGACGTCTTGACCCCCAGC"GCTGGCACTCC"CCATCCHCCTGCCATCACTGGAGCAGACATCAGGG ACAAGACAAGATGCCACCTCC"GCCACCACAGGGCC"GCTCGCGCCCCCC"CCCCCCAGAATAC GTCCCC C"GCTTGGCAATTGCCCTGGGCTCTCTGAGTCCAAGAGGTGGGACCTGCTCTGCAGGGGGGCAG CCTCAG G_CACCACLlGCTGGA-CCAGCCGA1CACACClG_GAiGCGGG_G_AGACGCCGAAGTAGTTAGGCCGGCCA CAGCCCAGGCCCCAGC"GACCAGCCCTGCCAGGAACCAGCGGCCACTGAG"GCCTTGCATACCAGCGGACCA CCCGAGTCACCCTGGCAGGCALCC TC iGCCCT GCGGTAGCCGGCACACAGCALGCGiGGCG CACC GG -AGCGA-AGGCCTCGCTGCACAGG-CC-GLGGGA-CAACTGCACG-CCAC---CTGCAGAGCA--GCTGGLG GGGCCGCCTTCGCGCAGGGCGCCCCAGCCAGTGA'CCAGCAGTGCAGGCCGGGTTCGAAGAAGHGGGAGCGC AGGCAGACTGGACGCACGGCGGCCGAGCGCACCACCGGG"GGTCGAGCTGCAACAGCGCCACG"CG -GGCLGTCCLC-TCG-GA-ACGGA GCAGGAGTAGGCGGCTCACC -GAAGGACACC"CTCCAGGC CAGCGCGAGTTCTGCCACACCHTGCCCAGGAACACCG"CCACAGCGCCGGGGAGGCCATGCTGHCCTCCHGG AAGCAAHGGGCAGCTG""ATCACCCAGCGG CAGCGA GAGGGCGCCCCCACAGATG"GTCGACCCCGAACC "GGAGGCTGGCCTGCCA GGCCAC CACCC CGGAGGACACGGCCCCACCAACAATGCGACTGGAGGGGCCC TGGAGGCCACAGTCACAGlGC_GC_CGLCiGAGCCG_CCCTGCAGTCGGGCCGCCCA_CACAC_GlGGG_LG HTCACGCAGCMCTGGHCC"CACAC"GGAAGGHGAATGTCCCGCAGGGCACCCCTTCCHGGCACCGC C CA"CGCLGCCGL GAGACAG CAGGC GCCCG CACAGACC"TAAGCAGTGAGA GCACG GCTG CC -C--GGCAC-GGAALG-GGCLCLGCAAACGCAGTiiC-CTCA-CCAGGCCGTTGGGGCAGTCC- GACCCCA HCACAGGCAGGGACGCAGAGTCCGHTCACAGAGCAGAGGAACHCTCCAGGGCAGGGG"CCGACHGGHTGHAC AAGCCAHAG"GCACCCGCACACCAGGCCCTGTGAGGGAGATC"GGGAGGTGAAATTGA GGTGA GCCGGCC G"GGCCACCACGGGGA CCTC CGGCG-AAGGCTGCAGGATGCGCAGGCCACACAGCC CCTG C-GGAiC G_CCAC_GGCCCTGGGTGCACGGCAAA_CA-ACT_C_GCCTCCGCAGTGCG_AGGCG_CAAACCAGAGGGCC HAG"CCAGAGAGGGCACCGTGAGG"GCCAGGAGCAG"GGGTTCGGGGCGAG AGTAGC GGGGAAG "ACGGGGTGCTGAGGACGCCC GGGAG CCAGCC G GCGTCAGG TACC CCTGGAAG GACTGCACGGAGAGCATAAAGGGG_CGTAG_AGCTG_GCAGGCCC__CLLCCAGACCACCGCCATG ATGGCCCCCGATGCCAGGACT"CCACCACAGGCTCC"GGCGGCTGCAGCCAMACACCGAGGTGAHGAGCCHC iC CCAGGGGCCCAGCCACG"CATACATGGCCAGTCGGTCCCGGCACTCGGCCAGCG"CCACTCCAGCCGG AG -iCAGCA-GAGG C-TCGGGGCCCTGCAGGTGCCACAGGCAGCTGGAGGCCAGGTGGTCGGGTCCCT"C AGCCGGAGGACCTGACCCTGGCCCACGHAGCTGTAGCGGTAACAACCCAGCGTGGAAT"CAGTGCAGCTA"G "C""TCACAC"GGC CHAGGALCACTAGGCCCTCGGGGTCCACTTCGTACTCGGCCC"GTAGGGGACAGCC GCCGAGC G GAC G GGACAGCAGCTCCTCCACCAGCAGTGCCTGCACCACCTCGGGGCTCAGCATCAGC CGGCGGLGC_CGGGGA__lGGAGAAiGAACCAGAAGAAGCAGGTGAGCGGTCCCTCCCCAAAGGAATAGACG GAGCTGGAG""GTAA"AAGTTCCCAGGCGGGTGCTGGCGATGAGCTCCTTGAGCATC""CTGGGCTTTGGCG G iiCAC GCGGAAGGCACTGGA"TCCCGGCGGGTAAGATCC"GGGAGAAG GGCGA GAGCACGCGCAGG CTGCCTGAG-ACACC-GGCTGACCGTCACCLCCGCC-LGTACCCLAGGAAA-ACCAGAGTAGCACCCCCACT GAAGTCAGCACAACCAGGGTCAGCCACAGGGGCGCCAGGCGGAGGTAGCCCCGGGCT""TCTCTTGGAGTCC "CACGGGCC CGAACALCCCCLC GGCTCCGCTTCC CGCCA CACCTCCG CCCCC GCCCACCAGCCACC TGGGGGGCC"TGGCCACAGGCATCCTTTCAGAGTGGAAGAGTAACAACATCAGGCGGCAGTGACCGGAAGTG GGTGCCGAGACAGTTCACAGAGGAGCGAAGTGCATCTCAAGGCAGCTCACCCCAGAGGGCAGGCACCAGAGC "GGACTGGG"GTGGCTCAAG Figure 17
NZ714577A 2013-05-22 2014-05-22 Tmprss6 irna compositions and methods of use thereof NZ714577B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361826178P 2013-05-22 2013-05-22
US61/826,178 2013-05-22
US201361912988P 2013-12-06 2013-12-06
US61/912,988 2013-12-06
PCT/US2014/039149 WO2014190157A1 (en) 2013-05-22 2014-05-22 Tmprss6 compositions and methods of use thereof

Publications (2)

Publication Number Publication Date
NZ714577A NZ714577A (en) 2021-11-26
NZ714577B2 true NZ714577B2 (en) 2022-03-01

Family

ID=

Similar Documents

Publication Publication Date Title
JP7437891B2 (en) TMPRSS6 IRNA composition and method of use thereof
TWI727963B (en) TRANSTHYRETIN (TTR) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES
US20180362978A1 (en) Compositions and Methods for Inhibition of Expression of Apolipoprotein C-III (APOC3) Genes
AU2021269372A1 (en) Compositions and methods for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
TW201831685A (en) Methods for treating or preventing ttr-associated diseases using transthyretin (ttr) irna compositions
TW201936624A (en) Serpina1 iRNA compositions and methods of use thereof
US20220380762A1 (en) HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
NZ714577B2 (en) Tmprss6 irna compositions and methods of use thereof
CN117561334A (en) Human chromosome 9 open reading frame 72 (C9 ORF 72) iRNA pharmaceutical compositions and methods of use thereof
NZ795574A (en) Methods for treating or preventing TTR-associated diseases using transthyretin (TTR) iRNA compositions
NZ714530B2 (en) SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EA042137B1 (en) COMPOSITIONS BASED ON iRNA TMPRSS6 AND METHODS FOR THEIR APPLICATION
NZ620151B2 (en) Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof