WO2016079197A1 - Activateurs d'administration d'arnsi conjugué et de nanoparticules lipidiques - Google Patents

Activateurs d'administration d'arnsi conjugué et de nanoparticules lipidiques Download PDF

Info

Publication number
WO2016079197A1
WO2016079197A1 PCT/EP2015/076993 EP2015076993W WO2016079197A1 WO 2016079197 A1 WO2016079197 A1 WO 2016079197A1 EP 2015076993 W EP2015076993 W EP 2015076993W WO 2016079197 A1 WO2016079197 A1 WO 2016079197A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
sirna
lipid nanoparticles
compound
formulation
Prior art date
Application number
PCT/EP2015/076993
Other languages
English (en)
Inventor
Marino Zerial
Marc Bickle
Jerome GILLERON
William Querbes
Martin Maier
Akin Akinc
Muthiah Manoharan
Original Assignee
MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
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 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. filed Critical MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
Publication of WO2016079197A1 publication Critical patent/WO2016079197A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5123Organic compounds, e.g. fats, sugars

Definitions

  • the present invention relates to delivery enhancers for facilitating the cellular delivery of (i) an active pharmaceutical ingredient (e.g., an siRNA) in a lipid nanoparticle or (ii) a conjugated siRNA.
  • an active pharmaceutical ingredient e.g., an siRNA
  • the present invention also relates to pharmaceutical compositions including (a) the delivery enhancer an (b) (i) a lipid nanoparticle containing an active pharmaceutical ingredient or (ii) a conjugated siRNA.
  • the invention further relates to processes for preparing such pharmaceutical compositions and their use.
  • RNAi therapeutics Interfering with gene expression has long been proposed as a potential therapeutic strategy.
  • the combination of potent RNAi therapeutics and innovative delivery strategies has opened new opportunities to efficiently silence disease-associated genes at therapeutically relevant doses.
  • Numerous delivery platforms such as viruses (Silva et al, Virology Journal, 7, 248, 2010, liposomes (Buyens et al, J. Controlled Release, 158(3), 362-370, 2012), polycationic polymers (Castillo et al, J. Drug Delivery, 218940, 2012), conjugates (Lopez et al., Curr. Opin.
  • LNPs lipid nanoparticles
  • siRNAs short interfering RNA sequences
  • LNPs lipid nanoparticles
  • RISC RNA-induced silencing complex
  • the present inventors have discovered compounds ("delivery enhancers") that enhance cellular delivery of (i) a siRNA conjugated to a ligand (such as a targeting ligand) (hereinafter referred to as a “conjugated siRNA”) and (ii) an active pharmaceutical ingredient (such as siRNA) when combined with a lipid nanoparticle (LNP).
  • a siRNA conjugated to a ligand such as a targeting ligand
  • an active pharmaceutical ingredient such as siRNA
  • LNP lipid nanoparticle
  • the invention relates to a composition (such as a pharmaceutical formulation) comprising (i) a conjugated siRNA and (ii) a delivery enhancer compound which enhances the cellular uptake of the conjugate siRNA and/or the intracellular release of the siRNA (for example, from endosomes).
  • a composition such as a pharmaceutical formulation
  • the composition is suitable for parenteral administration.
  • the delivery enhancer compound which enhances the cellular uptake of the conjugate siRNA and/or the intracellular release of the siRNA is selected from compounds of Formula (I):
  • Ri is, independently, alkyl (e.g., C]-C 8 alkyl, Ci-C 6 alkyl or C]-C 4 alkyl, such as methyl);
  • R4 is hydrogen or OH
  • alkyl e.g., Cj-C 8 alkyl, Ci-C 6 alkyl or Ci-C 4 alkyl, such as methyl
  • R 6 is hydrogen or alkyl (e.g., Ci-C 8 alkyl, Ci-C 6 alkyl or C 1 -C4 alkyl, such as methyl);
  • R 7 is hydrogen, hydroxy or alkyl (e.g., Q-Q alkyl, Q-C6 alkyl or Q-C4 alkyl, such as methyl);
  • n is 1 or 2; each occurrence of is an optional carbon carbon double bond;
  • Z is OH or -N(Ri) 2 .
  • the compounds of Formula (I) are preferably used with conjugated siRNA.
  • the compound of formula (I) does not contain two carbon-carbon double bonds. In one embodiment, the compound of formula (I) contains zero or one carbon- carbon double bond.
  • a preferred compound of formula (I) is a compound of formula (IA):
  • each occurrence of Ri is C1-C4 alkyl, such as methyl; hydrogen and R 3 is hydroxyl; and
  • each occurrence of R] is Q-C4 alkyl, such as methyl;
  • R 2 is hydrogen and R 3 is hydroxyl;
  • R5 is hydrogen or C1-C4 alkyl, such as methyl;
  • R 6 is hydroxyl or Q-C4 alkyl, such as methyl;
  • R 7 is hydrogen or C 1 -C 4 alkyl, such as methyl; and
  • Z is OH.
  • the compound of formula (I) is a compound of formula (IB):
  • R R 7 and Z are as defined above for the compound of formula (I).
  • each occurrence of R] is Q-C 4 alkyl, such as methyl;
  • each occurrence of Ri is C1-C4 alkyl, such as methyl
  • R5 is hydrogen
  • R 6 and R 7 are both C1-C4 alkyl, such as methyl
  • Z is OH.
  • the compound of formula (I) is a compound of formula (IC):
  • each occurrence of Ri is C1-C4 alkyl, such as methyl;
  • R 2 is hydrogen and R 3 is hydroxyl
  • each occurrence of Ri is Q-C4 alkyl, such as methyl; R 2 is hydrogen and R 3 is hydroxyl;
  • R 5 is alkenyl (e.g., C 2 -C 8 alkenyl, C 2 -C 6 alkenyl or C 2 -C 4 alkenyl, such as -
  • R and R 7 are hydrogen; and Z is OH.
  • the compound of formula (I) has the following stereochemistry:
  • delivery enhancer compounds which enhance the cellular uptake of the conjugate siRNA and/or the intracellular release of the siRNA (for example, from endosomes) include, but are not limited to, pentacyclic triterpenes and pentacyclic steroids, such as ursolic acid, oleanolic acid, moronic acid, betulinic acid, corosolic acid, and combinations thereof.
  • delivery enhancer compounds which enhance the cellular uptake of the conjugate siRNA and/or the intracellular release of the siRNA include, but are not limited to, those listed in Table A below.
  • the pharmaceutical formulation may include one or more pharmaceutically acceptable excipients, such as isotonicity agents.
  • the concentration of delivery enhancer compound in the pharmaceutical formulation ranges from about 0.1 ⁇ to about 100 ⁇ , such as from about 0.5 to about 50 ⁇ , from about 1 ⁇ to about 16 ⁇ , or from about 5 ⁇ to about 15 ⁇ (e.g., about 5 ⁇ or about 10 ⁇ ).
  • the invention relates to a process for preparing a composition by mixing conjugated siRNA with one or more delivery enhancer compounds selected from those described above, such as a delivery enhancer compound selected from formula I, IA, IB, IC or Table A.
  • a delivery enhancer compound selected from formula I, IA, IB, IC or Table A is another embodiment.
  • Yet another embodiment is a method of administering (e.g., parenterally such as intravenously) an siRNA by administering a pharmaceutical formulation containing a conjugated siRNA of the present invention.
  • the invention relates to lipid nanoparticles comprising an active pharmaceutical ingredient where the lipid nanoparticles are treated with one or more delivery enhancer compounds (e.g., those selected from Table B or of Formula II, IIA, III, IIIA, or IIIB) that enhance the cellular uptake of the active pharmaceutical ingredient and/or the intracellular release of the active pharmaceutical ingredient (for example, from endosomes).
  • the active pharmaceutical ingredient can be a nucleic acid, such as a siRNA.
  • the delivery enhancer compound which enhances cellular uptake of LNPs and/or the intracellular release of the siRNA (for example, from endosomes) is of formula
  • Ring A and Ring B are each, independently, selected from substituted and unsubstituted aryl (e.g., phenyl or naphthyl) and substituted and unsubstituted heteroaryl (e.g., pyridinyl or pyrimidinyl); each occurrence of Ri is, independently, selected from alkyl (e.g., C 1 -C 4 alkyl), aryl, heteroaryl, arylalkyl and heteroarylalkyl; each occurrence of L is, independently, selected from -0-, -S-, -NRg-, -C(0)0-, -OC(O)-, -NRg(CO)- and -C(0)NR 8 -; each occurrence of R 8 is hydrogen or alkyl (e.g., C1-C4 alkyl); each occurrence of X is, independently, selected from -O- and -S-; and each occurrence of n is independently 1, 2, 3, 4, 5 or 6.
  • aryl
  • each of rings A and B is bound to group L at the para position relative to the C(R 2 group.
  • both Ring A and Ring B are, independently, selected from substituted and unsubstituted aryl (e.g., phenyl).
  • the compound of formula (II) is a compound of formula (IIA)
  • R 1 , L, X and n are as defined above for formula (II).
  • Preferred embodiments of the compounds of formula (II) and (IIA) include one or more of the following:
  • each occurrence of Ri is, independently, alkyl (e.g., -Q alkyl, C ⁇ -C alkyl or C1-C4 alkyl);
  • each occurrence of L is, independently, selected from -O- and -S-; (iv) each occurrence of L is -0-;
  • the delivery enhancer compound which enhances cellular uptake of LNPs and/or the intracellular release of the siR A (for example, from endosomes) is of formula III:
  • Xi is, independently, CH or N;
  • Y is -0-, -NH-, or -S-;
  • R" is -CH 3 , -CH(CH 3 ) 2 , -(CH 2 ) 2 C0 2 H, -CH 2 C 6 H4 or each occurrence of R] is, independently, H, alkyl (e.g., C alkyl) or aryl (e.g., phenyl);
  • R 2 is H, alkyl (e.g., C ⁇ A alkyl) or aryl (e.g., phenyl); and each bond identified by * can independently be in the R or S configuration.
  • the compound of formula (III) may be a compound of formula (IIIA) or (IIIB):
  • Preferred embodiments of the compounds of formula (III), (IIIA) and (IIIB) include one or more of the following:
  • R" is -CH 2 C 6 H 4 ;
  • R 2 is H.
  • delivery enhancer compounds which enhance cellular uptake of LNPs and/or the intracellular release of the siRNA (for example, from endosomes) include, but are not limited to, those in Table B below.
  • the invention relates to lipid nanoparticles comprising an active pharmaceutical ingredient where the lipid nanoparticles are treated with one or more delivery enhancer compounds selected from a compound of Formula II, IIA, III, IIIA, or IIIB, or Table B that enhances delivery of the active pharmaceutical ingredient.
  • the active pharmaceutical ingredient can be a nucleic acid, such as a siRNA.
  • Another embodiment is compacted lipid nanoparticles prepared by treating lipid nanoparticles with one or more delivery enhancer compounds selected from a compound of Formula II, IIA, III, IIIA, or IIIB, or Table B, such as BADGE.
  • the lipid nanoparticles comprise an active pharmaceutical ingredient.
  • the compacted lipid nanoparticles may be incorporated in a pharmaceutical formulation as described herein.
  • the invention relates to lipid nanoparticles comprising (a) an active pharmaceutical ingredient, such as a nucleic acid (e.g., a siRNA), and (b) one or more delivery enhancer compounds selected from a compound of formula II, IIA, III, IIIA, or IIIB, or Table B.
  • an active pharmaceutical ingredient such as a nucleic acid (e.g., a siRNA)
  • one or more delivery enhancer compounds selected from a compound of formula II, IIA, III, IIIA, or IIIB, or Table B.
  • lipid nanoparticles comprising lipid nanoparticles, where the lipid nanoparticles comprise an active pharmaceutical ingredient, and one or more delivery enhancer compounds that enhance delivery of an active pharmaceutical ingredient such as siRNA.
  • the delivery enhancer compounds can be selected from a compound of Formula II, IIA, HI, IIIA, or IIIB, and Table B.
  • each nanoparticle comprises a cationic lipid and an active pharmaceutical ingredient (such as a nucleic acid).
  • the lipid nanoparticles have a d 5 o ranging from about 5 to about 500 nm, such as from about 5 to about 200 nm, from about 10 to about 50 nm, from about 25 to about 50 nm or from about 35 to about 45 nm.
  • the pharmaceutical formulation is suitable for parenteral administration.
  • the delivery enhancer compound is selected from BADGE and CP WOOl J18 and mixtures thereof:
  • Another embodiment is a pharmaceutical formulation comprising lipid nanoparticles and one or more delivery enhancer compounds selected from a compound of formula II, IIA, III, IIIA, or IIIB, and Table B, where each lipid nanoparticle comprises a cationic lipid and an active pharmaceutical ingredient (such as a nucleic acid), and the lipid nanoparticles have a d 50 ranging from about 10 to about 50 nm, such as from about 25 to about 50 nm or from about 35 to about 45 nm.
  • the concentration of delivery enhancer compound in the pharmaceutical formulation ranges from about 0.1 ⁇ to about 100 ⁇ , such as from about 0.5 to about 50 ⁇ , from about 1 ⁇ to about 16 ⁇ , or from about 5 ⁇ to about 15 ⁇ (e.g., about 5 ⁇ or about 10 ⁇ ).
  • the lipid nanoparticles comprise:
  • an active pharmaceutical ingredient e.g., a nucleic acid
  • a cationic lipid (b) a cationic lipid, (c) a non-cationic lipid (such as a neutral lipid),
  • an aggregation reducing agent such as polyethylene glycol (PEG) or PEG-modified lipid
  • the cationic lipid has a pKa ranging from about 4 to about 11, and preferably from about 5 to about 7.
  • the formulation includes less than about 3, about 2, about 1.5, about 1, or about 0.5 mole percent of the aggregation reducing agent (such as PEG or PEG-modified lipid), based upon the total moles of lipid (e.g., total moles of cationic lipid, non-cationic lipid, sterol, and aggregation reducing agent) in the lipid nanoparticle.
  • the aggregation reducing agent such as PEG or PEG-modified lipid
  • the formulation further comprises one or more isotonicity agents.
  • the formulation includes a sufficient amount of the isotonicity agent(s) to render the formulation physiologically isotonic (i.e., have a pharmaceutically acceptable osmolality) in order to avoid cell distortion or lysis.
  • the active pharmaceutical ingredient in the lipid nanoparticles is a nucleic acid, such as a siRNA.
  • the nucleic acid-lipid particle preferably has an encapsulation efficiency of greater than about 90, 92, 95, or 98%, after storage of the formulation for 1 month at about 4°C.
  • compositions containing conjugated siR As or LNPs described herein may be solutions or suspensions.
  • the present invention relates to a process for preparing lipid nanoparticles.
  • the process includes the step of incubating lipid nanoparticles in the presence of one or more delivery enhancer compounds selected from a compound of formula II, IIA, III, IIIA, or IIIB, and Table B.
  • the lipid nanoparticles comprise an active pharmaceutical ingredient.
  • Another embodiment is a process for preparing a pharmaceutical formulation containing LNPs according to any of the embodiments described herein.
  • the process includes adding one or more delivery enhancer compounds to one or more lipid nanoparticles.
  • the process may include, for example, mixing and/or incubating the delivery enhancer compound with the lipid nanoparticles.
  • the process comprises incubating the lipid nanoparticles and the delivery enhancer compound.
  • the incubation can be for, for example, from about 1 to about 48 hours. In one embodiment, the process involves incubating overnight (e.g., for about 12 or 16 hours). In another embodiment, the process involves incubating at a temperature between about 0 ° C and about 10° C, such as between about 2° C to about 5° C, e.g., at about 4° C. In one embodiment, the process involves incubating overnight (e.g., for about 12 or 16 hours) at about 4° C. In one embodiment, the incubation is conducted in an Eppendorf tube.
  • Yet another embodiment is a method of preparing a pharmaceutical formulation of the present invention comprising:
  • lipid nanoparticles comprising a cationic lipid and an active pharmaceutical ingredient (such as a nucleic acid);
  • a delivery enhancer of the present invention such as a compound of formula II, IIA, III, IIIA, or IIIB, or one from Table B) and mixtures thereof;
  • step (iii) optionally incubating (e.g., incubating overnight at about 4° C, e.g., in Eppendorf tubes) the mixture of step (ii).
  • Yet another embodiment is a method of reducing the size of lipid nanoparticles comprising incubating the lipid nanoparticles in the presence of one or more delivery enhancer compounds selected from a compound of Formula II, IIA, III, IIIA, or IIIB or Table B.
  • the incubation can be for, for example, from about 1 to about 48 hours.
  • the process involves incubating overnight (e.g., for about 12 or 16 hours).
  • the process involves incubating at a temperature between about 0 ° C and about 10° C, such as between about 2° C to about 5° C, e.g., at about 4° C.
  • the process involves incubating overnight (e.g., for about 12 or 16 hours) at about 4° C.
  • the incubation is conducted in an Eppendorf tube.
  • Yet another embodiment is a pharmaceutical formulation prepared by any of the processes described herein.
  • Yet another embodiment is a method of administering (e.g., parenterally such as intravenously) an active pharmaceutical ingredient, such as an siRNA, by administering a pharmaceutical formulation of the present invention.
  • an active pharmaceutical ingredient such as an siRNA
  • Figure la shows images of LNP-siRNA-gold detected in HeLa cells in vitro by electron microscopy after treatment with DMSO, Compound 1, or Compound 2.
  • siRNA-gold were detected in the cytosol (arrows) or within several endocytic compartments. Magnified images (insets) permit appreciation of the cytosolic localization of siRNA-gold.
  • Figure 2a shows images of LNP-siRNA-gold by electron microscopy after incubation with DMSO or Compound 1.
  • Figure 2b is a graph showing the fluorescence intensity (uptake kinetics) of LNP-siR A- alexa647 (40nM) treated with DMSO (black curve) or Compound 1 (red curve, ⁇ ) over time.
  • subject refers to a mammal, such as a human, domestic animal, such as a feline or canine subject, farm animal (e.g., bovine, equine, caprine, ovine, and porcine subject), wild animal (whether in the wild or in a zoological garden), research animal, such as mouse, rat, rabbit, goat, sheep, pig, dog, and cat, avian species, such as chicken, turkey, and songbird.
  • farm animal e.g., bovine, equine, caprine, ovine, and porcine subject
  • wild animal whether in the wild or in a zoological garden
  • research animal such as mouse, rat, rabbit, goat, sheep, pig, dog, and cat, avian species, such as chicken, turkey, and songbird.
  • the "subject” or “patient” can also be a plant.
  • treat and “treatment” refer to (a) relief from or alleviation of at least one symptom of a disorder in a subject, (b) relieving or alleviating the intensity and/or duration of a manifestation of a disorder experienced by a subject, (c) slowing or reversing the progression of such condition, and (d) arresting, delaying the onset (i.e., the period prior to clinical manifestation of a disorder) and/or reducing the risk of developing or worsening a disorder.
  • IV infusion refers to a method of administration of a composition directly into the vein of a patient.
  • IV infusion allows for direct administration of a pharmaceutical formulation to the bloodstream of a patient. This can be performed, for example, via subcutaneous or intradermal infusion.
  • IV infusion can be performed in many ways, including through the use of an injection needle, or with an infusion pump. It can be provided as, for example, a continuous infusion, an intermittent infusion, a patient-controlled infusion, or a circadian infusion.
  • An "isotonicity agent” generally refers to a compound that is physiologically tolerated and imparts a suitable tonicity to a formulation to prevent the net flow of water across cell membranes that are in contact with the formulation.
  • alkyl refers to a straight or branched chain saturated hydrocarbon moiety.
  • the alkyl group is a straight chain saturated hydrocarbon.
  • the "alkyl” group contains from 1 to 24 carbon atoms, such as from 1 to 12 carbon atoms, from 1 to 8 carbon atoms, from 1 to 6 carbon atoms or from 1 to 4 carbon atoms.
  • saturated straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl; while saturated branched alkyl groups include isopropyl, sec-butyl, isobutyl, tert-butyl, and isopentyl.
  • alkenyl refers to a straight or branched chain hydrocarbon moiety having one or more carbon-carbon double bonds. In one embodiment, the alkenyl group contains 1, 2, or 3 double bonds and is otherwise saturated. Unless otherwise specified, the "alkenyl” group contains from 2 to 24 carbon atoms, such as from 2 to 12 carbon atoms, from 2 to 8 carbon atoms, from 2 to 6 carbon atoms or from 2 to 4 carbon atoms. Alkenyl groups include both cis and trans isomers.
  • Representative straight chain and branched alkenyl groups include ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-l-butenyl, 2-methyl-2-butenyl, and 2,3-dimethyl-2-butenyl.
  • aryl refers to an aromatic monocyclic, bicyclic, or tricyclic hydrocarbon ring system.
  • aryl moieties include, but are not limited to, phenyl, naphthyl, anthracenyl, and pyrenyl.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 7-12 membered bicyclic, or 1 1-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, where the heteroatoms are selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively).
  • the heteroaryl groups herein described may also contain fused rings that share a common carbon-carbon bond.
  • halogen refers to fluoro, chloro, bromo and iodo.
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: halo, alkyl, alkenyl, aryl, heteroaryl, and any combination thereof.
  • Conjugated siRNA refers to an siRNA molecule conjugated to at least one ligand (preferably, at least one targeting ligand).
  • the siRNA may have a duplex region that is 17-21 or 19-21 nucleotides in length.
  • Suitable conjugated siRNA are described, for example, in U.S. Patent Publication Nos. 2013/0184328, 2013/0203836, 2013/0202652, 2013/0317080, 2013/0211063, and 2013/0184324, which are hereby incorporated by reference.
  • Other RNA conjugates are described in U.S. Patent Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465;
  • SiRNAs are generally RNA duplexes normally 16-30 nucleotides long that can associate with a cytoplasmic multi-protein complex known as RNAi-induced silencing complex (RISC). RISC loaded with siRNA mediates the degradation of homologous mRN A transcripts, therefore siRNA can be designed to knock down protein expression with high specificity.
  • RISC RNAi-induced silencing complex
  • siRNA molecules were RNA:RNA hybrids comprising both an RNA sense and an RNA antisense strand
  • DNA sense:RNA antisense hybrids and RNA sense'.DNA antisense hybrids are capable of mediating RNA interference (Lamberton, J. S. and Christian, A. T., (2003) Molecular Biotechnology 24: H il l 9).
  • siRNA includes the use of small interfering RNA molecules comprising any of these different types of double-stranded molecules.
  • RNAi molecules may be used and introduced to cells in a variety of forms.
  • siRNA molecules encompasses any and all molecules capable of inducing an RNA interference response in cells, including, but not limited to, (i) a double-stranded oligonucleotide comprising two separate strands, i.e.
  • a sense strand and an antisense strand (ii) a double-stranded oligonucleotide comprising two separate strands that are linked together by a non-nucleotidyl linker, (iii) a oligonucleotide comprising a hairpin loop of complementary sequences, which forms a double-stranded region, e.g., shKNAi molecules, and (iv) expression vectors that express one or more polynucleotides capable of forming a double- stranded polynucleotide alone or in combination with another polynucleotide.
  • the siRNA may be a single strand siRNA compound or a double stranded siRNA compound.
  • a "single strand siRNA compound” as used herein, is an siRNA compound which is made up of a single molecule. It may include a duplexed region, formed by intra-strand pairing, e.g., it may be, or include, a hairpin or pan-handle structure. Single strand siRNA compounds may be antisense with regard to the target molecule.
  • a single strand siRNA compound may be sufficiently long that it can enter the RISC and participate in RISC mediated cleavage of a target mRNA.
  • a single strand siRNA compound is at least 14, and in other embodiments at least 15, 20, 25, 29, 35, 40, or 50 nucleotides in length. In certain embodiments, it is less than 80 or 60 nucleotides in length.
  • Hairpin siRNA compounds will have a duplex region equal to or at least 17, 18, 19, 29, 21, 22, 23, 24, or 25 nucleotide pairs.
  • the duplex region will may be equal to or less than 30, 25, 23, or 22 nucleotide pairs in length. In certain embodiments, ranges for the duplex region are 15- 30, 17 to 23, 19 to 23, and 19 to 21 nucleotide pairs in length.
  • the hairpin may have a single strand overhang or terminal unpaired region. In certain embodiments, the overhangs are 2-3 nucleotides in length. In some embodiments, the overhang is at the sense side of the hairpin and in some embodiments on the antisense side of the hairpin.
  • a "double stranded siRNA compound” as used herein, is an siRNA compound which includes more than one, and typically two, strands in which interchain hybridization can form a region of duplex structure.
  • the antisense strand of a double stranded siRNA compound may be equal to or at least, 14, 15, 16, 17, 18, 19, 25, or 29 nucleotides in length. It may be equal to or less than 30, 25, 23, or 21 nucleotides in length. Ranges may be 17 to 25, 19 to 23, and 19 to 21 nucleotides in length.
  • antisense strand means the strand of an siRNA compound that is sufficiently complementary to a target molecule, e.g. a target RNA.
  • the sense strand of a double stranded siRNA compound may be equal to or at least 14, 15, 16, 17, 18, 19, 25, or 29 nucleotides in length. It may be equal to or less than 30, 25, 23, or 21 nucleotides in length. Ranges may be 17 to 25, 19 to 23, and 19 to 21 nucleotides in length.
  • the double strand portion of a double stranded siRNA compound may be equal to or at least 14, 15, 16 17, 18, 19, 20, 21, 22, 23, 24, 25, 29, or 30 nucleotide pairs in length. It may be equal to or less than 30, 25, 23, or 21 nucleotides pairs in length. Ranges may be 15 to 30, 17 to 23, 19 to 23, and 19 to 21 nucleotide pairs in length.
  • the siRNA compound is sufficiently large that it can be cleaved by an endogenous molecule, e.g., by Dicer, to produce smaller siRNA compounds, e.g., siRNAs agents.
  • the sense and antisense strands may be chosen such that the double-stranded siRNA compound includes a single strand or unpaired region at one or both ends of the molecule.
  • a double- stranded siRNA compound may contain sense and antisense strands, paired to contain an overhang, e.g., one or two 5' or 3' overhangs, or a 3' overhang of 1-3 nucleotides.
  • 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.
  • Some embodiments will have at least one 3' overhang.
  • both ends of an siRNA molecule will have a 3' overhang.
  • the overhang is 2 nucleotides.
  • the siRNA compounds described herein, including double-stranded siRNA compounds and single-stranded siRNA compounds can mediate silencing of a target RNA, e.g., mRNA, e.g. a transcript of a gene that encodes a protein (a target gene).
  • a target RNA e.g., mRNA, e.g. a transcript of a gene that encodes a protein (a target gene).
  • the RNA to be silenced is an endogenous gene or a pathogen gene.
  • RNAs other than mRNA, e.g., tRNAs, and viral RNAs can also be targeted.
  • an siRNA compound is "sufficiently complementary" to a target RNA, e.g., a target mRNA, such that the siRNA compound silences production of protein encoded by the target mRNA.
  • the siRNA compound is "exactly complementary" to a target RNA, e.g., the target RNA and the siRNA compound anneal, for example to form a hybrid made exclusively of Watson-Crick base pairs in the region of exact complementarity.
  • a "sufficiently complementary" target RNA can include an internal region (e.g., of at least 10 nucleotides) that is exactly complementary to a target RNA.
  • the siRNA compound specifically discriminates a single-nucleotide difference. In this case, the siRNA compound only mediates RNA interference if exact complementary is found in the region (e.g., within 7 nucleotides of) the single-nucleotide difference.
  • the ligand may be conjugated to any monomer in the siRNA, either directly or indirectly (such as through an intervening tether).
  • the ligand may be conjugated to a monomer at the 5' or 3' terminus of the siRNA molecule or may be attached to an internal monomer.
  • the ligand can be attached to the 3' end of the sense strand.
  • the ligand can facilitate targeting and/or delivery of the siRNA.
  • the ligand cholesterol for example, promotes entry into a cell.
  • the ligand includes a lipophilic moiety. While not wishing to be bound by theory, it is believed the attachment of a lipohilic agent increases the lipophilicity of the siRNA.
  • a ligand alters the distribution, targeting or lifetime of an siRNA into which it is incorporated.
  • a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, compartment, 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.
  • the ligand may be a liver targeting ligand.
  • Preferred ligands can improve transport, hybridization, and specificity properties and may also improve nuclease resistance of the resultant natural or modified siRNA.
  • Ligands in general can include therapeutic modifiers, e.g., for enhancing uptake; cross- linking agents; and nuclease-resistance conferring moieties.
  • therapeutic modifiers e.g., for enhancing uptake; cross- linking agents; and nuclease-resistance conferring moieties.
  • General examples include lipophiles, lipids, steroids (e.g., uvaol, hecigenin, and diosgenin), terpenes (e.g., triterpenes, e.g., sarsasapogenin, Friedelin, and epifriedelanol derivatized lithocholic acid), vitamins (e.g., folic acid, vitamin A, biotin, and pyridoxal), carbohydrates, proteins, protein binding agents, integrin targeting molecules, polycationics, peptides, polyamines, and peptide mimics.
  • the ligand can have endosomolytic properties.
  • the endosomolytic ligands promote the lysis of the endosome and/or transport of the siRNA from the endosome to the cytoplasm of the cell.
  • the endosomolytic ligand may be a polyanionic peptide or peptidomimetic which shows pH-dependent membrane activity and fusogenicity.
  • the endosomolytic ligand assumes its active conformation at endosomal pH.
  • the "active" conformation is that conformation in which the endosomolytic ligand promotes lysis of the endosome and/or transport of the siRNA from the endosome to the cytoplasm of the cell.
  • Exemplary endosomolytic ligands include the GALA peptide (Subbarao et al, Biochemistry, 1987, 26: 2964-2972), the EALA peptide (Vogel et al, J. Am. Chem. Soc, 1996, 118: 1581-1586), and their derivatives (Turk et al., Biochem. Biophys. Acta, 2002, 1559: 56-68).
  • 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.
  • Endosomal release agents include, but are not limited to, imidazoles, poly- or oligoimidazoles, PEIs, peptides, fusogenic peptides, polycarboxylates, polyacations, masked oligo or poly cations or anions, acetals, polyacetals, ketals/polyketyals, orthoesters, polymers with masked or unmasked cationic or anionic charges, and dendrimers with masked or unmasked cationic or anionic charges.
  • Ligands can include a naturally occurring substance, (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulin); carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); amino acid, or a lipid.
  • HSA human serum albumin
  • LDL low-density lipoprotein
  • globulin carbohydrate
  • carbohydrate e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid
  • amino 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.
  • polyamino acids include, but are not limited to, 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), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine.
  • PLL polylysine
  • poly L- aspartic acid poly L-glutamic acid
  • styrene-maleic acid anhydride copolymer poly(L-lactide-co- glycolied) copolymer
  • divinyl ether-maleic anhydride copolymer divinyl ether-maleic anhydride
  • polyamines include, but are not limited to, polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, and cationic moieties, e.g., cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
  • PLL polylysine
  • spermine spermine
  • spermidine polyamine
  • polyamine pseudopeptide-polyamine
  • peptidomimetic polyamine dendrimer polyamine
  • arginine amidine
  • protamine protamine
  • cationic moieties e.g., cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
  • Ligands 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 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, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, biotin, or an RGD peptide or RGD peptide mimetic.
  • a preferred targeting ligand is GalNAc. Exemplary targeting ligands are described in Table 2 of US 2013/0203836, which is hereby incorporated by reference.
  • ligands include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, and Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine and dihydrophenazine), artificial endonucleases (e.g.
  • intercalating agents e.g. acridines
  • cross-linkers e.g. psoralene, mitomycin C
  • porphyrins TPPC4, texaphyrin, and Sapphyrin
  • polycyclic aromatic hydrocarbons e.g., phenazine and dihydrophenazine
  • artificial endonucleases e.g.
  • lipophilic molecules e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, glycerol (e.g., esters and ethers thereof, e.g., Ci 0 -C 2 o alkyl, e.g., l,3-bis-0(hexadecyl)glycerol, l ,3-bis-0(octaadecyl)glycerol), geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3 -propanediol, heptadecyl group, palmitic acid, myristic acid, 03-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine), peptide conjug
  • biotin e.g., aspirin, vitamin E, and folic acid
  • transport/absorption facilitators e.g., aspirin, vitamin E, and folic acid
  • synthetic ribonucleases e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, and 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, N-acetyl-galactosamine, N-acetyl- gulucosamine multivalent mannose, or multivalent fucose.
  • the ligand can be, for example, a lipopolysaccharide, an activator of p38 MAP kinase, or an activator of NF-KB.
  • the ligand can be a substance, e.g., a drug, which can increase the uptake of the siRNA into the cell, for example, by disrupting the cell's cytoskeleton, such as by disrupting the cell's microtubules, microfilaments, and/or intermediate filaments.
  • the drug can be, for example, taxon, vincristine, vinblastine, cytochalasin, nocodazole, japlakinolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.
  • the ligand can increase the uptake of the siRNA into the cell, for example, by activating an inflammatory response.
  • exemplary ligands that have such an effect include tumor necrosis factor alpha (TNF-ot), interleukin-1 beta, or gamma interferon.
  • the ligand is a lipid or lipid-based molecule.
  • a lipid or lipid-based molecule may bind a serum protein, e.g., human serum albumin (HSA).
  • HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., a non-kidney target tissue of the body.
  • the target tissue can be the liver, including parenchymal cells of the liver.
  • Other molecules that can bind HSA can also be used as ligands. For example, neproxin or aspirin can be used.
  • a lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, and/or (c) can be used to adjust binding to a serum protein, e.g., HSA.
  • a serum protein e.g., HSA.
  • a lipid based ligand can be used to modulate, e.g., control the binding of the conjugate to a target tissue.
  • 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.
  • the lipid based ligand binds HSA.
  • it may bind HSA with a sufficient affinity such that the conjugate will be preferably distributed to a non-kidney tissue.
  • the affinity it is preferred that the affinity not be so strong that the HSA-ligand binding cannot be reversed.
  • the lipid based ligand binds HSA weakly or not at all, such that the conjugate will be preferably distributed to the kidney.
  • Other moieties that target to kidney cells can also be used in place of or in addition to the lipid based ligand.
  • the ligand is a moiety, e.g., a vitamin, which is taken up by a target cell, e.g., a proliferating cell.
  • a target cell e.g., a proliferating cell.
  • vitamins include vitamins A, E, and K.
  • Other exemplary vitamins include B vitamins, e.g., folic acid, vitamin B 12 , riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by cancer cells.
  • HSA and low density lipoprotein (LDL) are also included.
  • the ligand is a cell-permeation agent, such as a helical cell- permeation agent.
  • the agent may be amphipathic.
  • An exemplary agent is a peptide such as tat or antennopedia.
  • the helical agent may be an alpha-helical agent, for example, which has a lipophilic and a lipophobic phase.
  • the ligand can be a peptide or peptidomimetic.
  • a peptidomimetic also referred to as an oligopeptidomimetic is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide.
  • the peptide 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 (see, for example, Table 3 of US 2013/0203836, which is hereby incorporated by reference).
  • a peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting primarily of Tyr, Trp or Phe).
  • the peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide.
  • the peptide moiety can include a hydrophobic membrane translocation sequence (MTS).
  • An exemplary hydrophobic MTS-containing peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP.
  • An RFGF analogue e.g., amino acid sequence AALLPVLLAAP
  • a hydrophobic MTS can also be a targeting moiety.
  • the peptide moiety can be a "delivery" peptide, which can carry large polar molecules including peptides, oligonucleotides, and protein across cell membranes.
  • sequences from the HIV Tat protein GRKKRRQRRRPPQ
  • the Drosophila Antennapedia protein RQIKIWFQNRRMKWKK
  • 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).
  • the peptide or peptidomimetic tethered to an siRNA 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 moieties can have a structural modification, such as to increase stability or direct conformational properties.
  • An RGD peptide moiety can be used to target a tumor cell, such as an endothelial tumor cell or a breast cancer tumor cell (Zitzmann et al., Cancer Res., 62:5139-43, 2002).
  • An RGD peptide can facilitate targeting of an siRNA to tumors of a variety of other tissues, including the lung, kidney, spleen, or liver (Aoki et al., Cancer Gene Therapy 8:783-787, 2001).
  • the RGD peptide will facilitate targeting of an siRNA to the kidney.
  • the RGD peptide can be linear or cyclic, and can be modified, e.g., glycosylated or methylated to facilitate targeting to specific tissues.
  • Peptides that target markers enriched in proliferating cells can be used.
  • RGD containing peptides and petomimetics can target cancer cells, in particular cells that exhibit an integrin.
  • ligands can be used to control proliferating cells and angiogeneis.
  • the ligands can target PECAM-1, VEGF, or other cancer gene, e.g., a cancer gene described herein.
  • the ligand can target the liver.
  • a liver-targeting agent can be a lipophilic moiety.
  • lipophilic moieties include, but are not limited to, lipids, cholesterols, oleyl, retinyl, and cholesteryl residues.
  • liver- targeting agents include cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, l,3-bis-0(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, 03- (oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, and phenoxazine.
  • siRNA can also be targeted to the liver by association with a low-density lipoprotein (LDL), such as lactosylated LDL.
  • LDL low-density lipoprotein
  • Polymeric carriers complexed with sugar residues can also function to target siRNA to the liver.
  • a targeting agent that incorporates a sugar, e.g., galactose and/or analogues thereof, is particularly useful. These agents target, in particular, the parenchymal cells of the liver.
  • a targeting moiety can include more than one or preferably two or three galactose moieties, spaced about 15 angstroms from each other.
  • the targeting moiety can alternatively be lactose (e.g., three lactose moieties), which is glucose coupled to a galactose.
  • the targeting moiety can also be N-acetyl-galactosamine, N-Ac-glucosamine.
  • a mannose or mannose-6- phosphate targeting moiety can be used for macrophage targeting.
  • 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 alpha-helical linear peptide (e.g., LL-37 or Ceropin PI), a disulfide bond-containing peptide (e.g., alpha-defensin, beta-defensin or bactenecin), 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).
  • NLS nuclear localization signal
  • a cell permeation peptide can be a bipartite amphipathic peptide, such as MPG, which is derived from the fusion peptide domain of HIV- 1 gp41 and the NLS of SV40 large T antigen (Simeoni et ah, Nucl. Acids Res. 31:2717-2724, 2003).
  • a targeting peptide can be an amphipathic alpha-helical peptide.
  • amphipathic alpha-helical peptides include, but are not limited to, cecropins, lycotoxins, paradaxins, buforin, CPF, bombinin-like peptide (BLP), cathelicidins, ceratotoxins, S. clava peptides, hagfish intestinal antimicrobial peptides (HFIAPs), magainines, brevinins-2, dermaseptins, melittins, pleurocidin, H 2A peptides, Xenopus peptides, esculentinis-1, and caerins.
  • a number of factors may be considered to maintain the integrity of helix stability.
  • a maximum number of helix stabilization residues may be utilized (e.g., leu, ala, or lys), and a minimum number of helix destabilization residues nay be utilized (e.g., proline or cyclic monomeric units).
  • the capping residue may be considered (for example Gly is an exemplary recapping 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 ⁇ 3, or i ⁇ 4 positions can provide stability.
  • cationic residues such as lysine, arginine, homo-arginine, ornithine 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; alpha, beta, or gamma peptides; N-methyl peptides; 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.
  • D or L peptides alpha, beta, or gamma peptides
  • N-methyl peptides azapeptides
  • cyclic peptides include those having naturally occurring or modified peptides, e.g.,
  • the targeting ligand can be any ligand that is capable of targeting a specific receptor. Examples include, but are not limited to, folate, GalNAc, galactose, mannose, mannose-6P, clusters of sugars such as a GalNAc cluster, mannose cluster, galactose cluster, or an apatamer. A cluster is a combination of two or more sugar units.
  • the targeting ligand can also be an integrin receptor ligand, Chemokine receptor ligand, transferrin, biotin, serotonin receptor ligand, PSMA, endothelin, GCPII, somatostatin, or LDL or HDL ligand.
  • the ligand can also be based on a nucleic acid, e.g., an aptamer. The aptamer can be unmodified or have any combination of modifications disclosed herein.
  • the active pharmaceutical ingredient can be any compound suitable for incorporation into a lipid nanoparticle.
  • the active pharmaceutical ingredient is encapsulated within an aqueous interior of the lipid nanoparticle.
  • the active pharmaceutical ingredient is present within one or more lipid layers of the lipid nanoparticle.
  • the active pharmaceutical ingredient is bound to the exterior or interior of the lipid surface of the lipid nanoparticle.
  • the active pharmaceutical ingredient can be any compound capable of exerting a desired effect on a cell, tissue, organ, or subject. Such effects may be biological, physiological, or cosmetic, for example.
  • the active pharmaceutical ingredient can be a nucleic acid, peptide, polypeptide (e.g., an antibody), cytokine, growth factor, apoptotic factor, differentiation-inducing factor, cell surface receptor or a corresponding ligand, or hormone.
  • Suitable active pharmaceutical ingredient include, but are not limited to, anti-inflammatory compounds, anti-depressants, stimulants, analgesics, antibiotics, birth control medication, antipyretics, vasodilators, anti-angiogenics, cytovascular agents, signal transduction inhibitors, cardiovascular drugs (e.g., anti-arrhythmic agents), vasoconstrictors, hormones, steroids, and oncology drugs (e.g., an anti-tumor agent, an anti-cancer drug, or anti-neoplatic agent).
  • anti-inflammatory compounds include, but are not limited to, anti-inflammatory compounds, anti-depressants, stimulants, analgesics, antibiotics, birth control medication, antipyretics, vasodilators, anti-angiogenics, cytovascular agents, signal transduction inhibitors, cardiovascular drugs (e.g., anti-arrhythmic agents), vasoconstrictors, hormones, steroids, and oncology drugs (e.g., an anti-tumor agent, an anti-can
  • the active pharmaceutical ingredient is a nucleic acid.
  • the nucleic acid can be an interfering RNA (such as a siRNA, including conjugated siRNA as described herein), an antisense oligonucleotide, a DNAi oligonucleotide, a ribozyme, an aptamer, a plasmid, or any combination of any of the foregoing.
  • the nucleic acid can be encoded with a product of interest including, but not limited to, RNA, antisense oligonucleotide, an antagomir, a DNA, a plasmid, a ribosomal RNA (rRNA), a micro RNA (miRNA) (e.g., a miR A which is single stranded and 17-25 nucleotides in length), transfer RNA (tRNA), a small interfering RNA (siRNA), small nuclear RNA (snRNA), antigens, fragments thereof, proteins, peptides, and vaccines or mixtures thereof.
  • RNA antisense oligonucleotide
  • an antagomir e.g., a DNA, a plasmid, a ribosomal RNA (rRNA), a micro RNA (miRNA) (e.g., a miR A which is single stranded and 17-25 nucleotides in length), transfer RNA (t
  • the nucleic acid is an oligonucleotide (e.g., 15-50 nucleotides in length (or 15-30 or 20-30 nucleotides in length)).
  • An siRNA can have, for instance, a duplex region that is 16-30 nucleotides long (e.g., 17-21 or 19- 21 nucleotides long).
  • the nucleic acid is an immunostimulatory oligonucleotide, decoy oligonucleotide, supermir, miRNA mimic, or miRNA inhibitor.
  • a supermir refers to a single stranded, double stranded or partially double stranded oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or both or modifications thereof, which has a nucleotide sequence that is substantially identical to an miRNA and that is antisense with respect to its target.
  • miRNA mimics represent a class of molecules that can be used to imitate the gene silencing ability of one or more miRNAs.
  • the term "microRNA mimic” refers to synthetic non-coding RNAs (i.e. the miRNA is not obtained by purification from a source of the endogenous miRNA) that are capable of entering the RNAi pathway and regulating gene expression.
  • the nucleic acid that is present in a lipid nanoparticle can be in any form.
  • the nucleic acid can, for example, be single- stranded DNA or RNA, or double-stranded DNA or RNA, or DNA-RNA hybrids.
  • double-stranded RNA include siRNA.
  • Single-stranded nucleic acids include, e.g., antisense oligonucleotides, ribozymes, microRNA, and triplex-forming oligonucleotides.
  • the nucleic acid can be conjugated to one or more ligands (e.g., a targeting ligand).
  • the nucleic acid is selected from an interfering RNA, an antisense oligonucleotide, a DNAi oligonucleotide, a ribozyme, an aptamer, a plasmid, and any combination of any of the foregoing.
  • the RNA is selected from siRNA, aiRNA, miRNA, Dicer-substrate dsRNA, shRNA, ssRNAi oligonucleotides, and any combination of any of the foregoing.
  • the active pharmaceutical ingredient is an siRNA (e.g., an siRNA having a duplex region that is 17-21 or 19-21 nucleotides long).
  • siRNA e.g., an siRNA having a duplex region that is 17-21 or 19-21 nucleotides long.
  • Formulations containing siRNA are useful in down-regulating the protein levels and/or mRNA levels of target proteins.
  • the siRNA may be unmodified oligonucleotides or modified, and may be conjugated with lipophilic moieties such as cholesterol.
  • the active pharmaceutical ingredient is a micro RNA.
  • the active pharmaceutical ingredient e.g., a nucleic acid
  • the active pharmaceutical ingredient is fully encapsulated in the lipid nanoparticle.
  • the lipid nanoparticle may include any cationic lipid suitable for forming a lipid nanoparticle.
  • the cationic lipid carries a net positive charge at about physiological pH.
  • the cationic lipid may be an amino lipid.
  • amino lipid is meant to include those lipids having one or two fatty acid or fatty alkyl chains and an amino head group (including an alkylamino or dialkylamino group) that may be protonated to form a cationic lipid at physiological pH.
  • the cationic lipid may be, for example, N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N-dimethylammonium bromide (DDAB), 1,2- dioleoyltrimethyl ammonium propane chloride (DOTAP) (also known as N-(2,3-dioleoyloxy)propyl)-N,N,N- trimethylammonium chloride and l,2-Dioleyloxy-3-trimethylaminopropane chloride salt), N-(l- (2,3- dioleyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTMA), N,N-dimethyl-2,3- dioleyloxy)propylamine (DODMA), 1 ,2-DiLinoleyloxy-N,N-dimethylaminopropane (DLinDMA), l,2-Dilinolen
  • cationic lipids include, but are not limited to, N,N-distearyl-N,N- dimethylammonium bromide (DDAB), 3P-(N-(N',N'-dimethylaminoethane)- carbamoyl)cholesterol (DC-Choi), N-(l-(2,3-dioleyloxy)propyl)-N-2-
  • DDAB N,N-distearyl-N,N- dimethylammonium bromide
  • DC-Choi 3P-(N-(N',N'-dimethylaminoethane)- carbamoyl)cholesterol
  • DC-Choi N-(l-(2,3-dioleyloxy)propyl)-N-2-
  • sperminecarboxamido)ethyl)-N,N-dimethylammonium trifluoracetate DOSPA
  • DOGS dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidoglycyl carboxyspermine
  • DOPE dioctadecylamidog
  • cationic lipids can be used, such as, e.g., LIPOFECTIN (including DOTMA and DOPE, available from GIBCO/BRL), and LIPOFECTAMINE (comprising DOSPA and DOPE, available from GIBCO/BRL).
  • LIPOFECTIN including DOTMA and DOPE, available from GIBCO/BRL
  • LIPOFECTAMINE comprising DOSPA and DOPE, available from GIBCO/BRL
  • amino lipids include those having alternative fatty acid groups and other dialkylamino groups, including those in which the alkyl substituents are different ⁇ e.g., N-ethyl- N-methylamino-, and N-propyl-N-ethylamino-).
  • amino lipids having less saturated acyl chains are more easily sized, particularly when the complexes must be sized below about 0.3 microns, for purposes of filter sterilization.
  • Amino lipids containing unsaturated fatty acids with carbon chain lengths in the range of C 14 to C 22 may be used.
  • Other scaffolds can also be used to separate the amino group and the fatty acid or fatty alkyl portion of the amino lipid.
  • amino or cationic lipids of the invention have at least one protonatable or deprotonatable group, such that the lipid is positively charged at a pH at or below physiological pH (e.g. pH 7.4), and neutral at a second pH, preferably at or above physiological pH.
  • physiological pH e.g. pH 7.4
  • second pH preferably at or above physiological pH.
  • the protonatable lipids have a pKa of the protonatable group in the range of about 4 to about 11 , e.g., a pKa of about 5 to about 7.
  • Lipid particles can include two or more cationic lipids.
  • the cationic lipids can be selected to contribute different advantageous properties.
  • cationic lipids that differ in properties such as amine pK a , chemical stability, half-life in circulation, half-life in tissue, net accumulation in tissue, or toxicity can be used in the lipid nanoparticle.
  • the cationic lipids can be chosen so that the properties of the mixed-lipid particle are more desireable than the properties of a single-lipid particle of individual lipids.
  • Net tissue accumulation and long term toxicity (if any) from the cationic lipids can be modulated in a favorable way by choosing mixtures of cationic lipids instead of selecting a single cationic lipid in a given formulation. Such mixtures can also provide better encapsulation and/or release of the active pharmaceutical ingredient.
  • a series of structurally similar compounds can have varying pK a values that span a range, e.g. of less than 1 pKg unit, from 1 to 2 pKa units, or a range of more than 2 pK a units.
  • a pK a in the middle of the range is associated with an enhancement of advantageous properties (greater effectiveness) or a decrease in disadvantageous properties (e.g., reduced toxicity), compared to compounds having pK a values toward the ends of the range.
  • two (or more) different compounds having pK a values toward opposing ends of the range can be selected for use together in a lipid nanoparticle.
  • the net properties of the lipid nanoparticle (for instance, charge as a function of local pH) can be closer to that of a particle including a single lipid from the middle of the range.
  • Cationic lipids that are structurally dissimilar (for example, not part of the series of structurally similar compounds mentioned above) can also be used in a mixed-lipid nanoparticle.
  • two or more different cationic lipids may have widely differing pK a values, e.g., differing by 3 or more pKa units.
  • the net behavior of a mixed lipid nanoparticle will not necessarily mimic that of a single-lipid particle having an intermediate pK a . Rather, the net behavior may be that of a particle having two distinct protonatable (or deprotonatable, as the case may be) site with different pKa values.
  • the fraction of protonatable sites that are in fact protonated varies sharply as the pH moves from below the pKa to above the pKa (when the pH is equal to the pKa value, 50% of the sites are protonated).
  • the lipid nanoparticle can show a more gradual transition from non-protonated to protonated as the pH is varied.
  • the cationic lipid can comprise from about 20 mol % to about 70 or 75 mol % or from about 45 to about 65 mol % or about 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, or about 70 mol % of the total lipid present in the particle.
  • the lipid nanoparticles include from about 25% to about 75% on a molar basis of cationic lipid, e.g., from about 20 to about 70%, from about 35 to about 65%, from about 45 to about 65%, about 60%, about 57.5%, about 57.1%, about 50% or about 40% on a molar basis (based upon 100% total moles of lipid in the lipid nanoparticle).
  • the ratio of cationic lipid to nucleic acid is from about 3 to about 15, such as from about 5 to about 13 or from about 7 to about 11.
  • the non-cationic lipid incorporated in the LNP can be a neutral lipid, an anionic lipid, or an amphipathic lipid.
  • Neutral lipids when present, can be any of a number of lipid species which exist either in an uncharged or neutral zwitterionic form at physiological pH. Such lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, dihydrosphingomyelin, cephalin, and cerebrosides.
  • the selection of neutral lipids for use in the particles described herein is generally guided by consideration of, e.g., lipid particle size and stability of the lipid particle in the bloodstream.
  • the neutral lipid is a lipid having two acyl groups (e.g., diacylphosphatidylcholine and diacylphosphatidylethanolamine).
  • the neutral lipids contain saturated fatty acids with carbon chain lengths in the range of Cio to C 20 .
  • neutral lipids with mono or diunsaturated fatty acids with carbon chain lengths in the range of Cio to C 2 o are used.
  • neutral lipids having mixtures of saturated and unsaturated fatty acid chains can be used.
  • Suitable neutral lipids include, but are not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl- phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoylphosphatidylethanolamine (POPE), dioleoyl- phosphatidylethanolamine 4-(N- maleimidomethyl)-cyclohexane-l- carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), dimyristoyl phosphatidylcholine (
  • Anionic lipids suitable for use in lipid particles of the invention include, but are not limited to, phosphatidylglycerol, cardiolipin, diacylphosphatidylserine, diacylphosphatidic acid, N-dodecanoyl phosphatidylethanoloamine, N-succinyl phosphatidylethanolamine, N-glutaryl phosphatidylethanolamine, lysylphosphatidylglycerol, and other anionic modifying groups joined to neutral lipids.
  • Amphipathic lipids refer to any suitable material, wherein the hydrophobic portion of the lipid material orients into a hydrophobic phase, while the hydrophilic portion orients toward the aqueous phase.
  • Such compounds include, but are not limited to, phospholipids, aminolipids, and sphingolipids.
  • Representative phospholipids include sphingomyelin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatdylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, distearoylphosphatidylcholine, or dilinoleoylphosphatidylcholine.
  • Other phosphorus-lacking compounds such as sphingolipids, glycosphingolipid families, diacylglycerols, and ⁇ -acyloxyacids, can also be used.
  • the non-cationic lipid can be from about 5 mol % to about 90 mol %, about 5 mol % to about 10 mol %, about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, or about 90 mol % of the total lipid present in the particle.
  • the lipid nanoparticles include from about 0% to about 15 or 45% on a molar basis of neutral lipid, e.g., from about 3 to about 12%) or from about 5 to about 10%.
  • the lipid nanoparticles may include about 15%, about 10%, about 7.5%, or about 7.1% of neutral lipid on a molar basis (based upon 100% total moles of lipid in the lipid nanoparticle).
  • a preferred sterol for incorporation in the LNP is cholesterol.
  • the sterol can be about 10 to about 60 mol % or about 25 to about 40 mol % of the lipid particle. In one embodiment, the sterol is about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, or about 60 mol % of the total lipid present in the lipid particle.
  • the lipid nanoparticles include from about 5% to about 50% on a molar basis of the sterol, e.g., about 15 to about 45%, about 20 to about 40%, about 48%, about 40%, about 38.5%, about 35%, about 34.4%, about 31.5% or about 31% on a molar basis (based upon 100% total moles of lipid in the lipid nanoparticle).
  • the aggregation reducing agent in the LNP can be a lipid capable of reducing aggregation.
  • lipids include, but are not limited to, polyethylene glycol (PEG)-modified lipids, monosialoganglioside Gml, and polyamide oligomers (PAO) such as those described in U.S. Patent No. 6,320,017, which is incorporated by reference in its entirety.
  • PEG polyethylene glycol
  • PAO polyamide oligomers
  • ATTA-lipids are described, e.g., in U.S. Patent No. 6,320,017
  • PEG-lipid conjugates are described, e.g., in U.S. Patent Nos. 5,820,873, 5,534,499 and 5,885,613, each of which is incorporated
  • the aggregation reducing agent may be, for example, a polyethyleneglycol (PEG)-lipid including, without limitation, a PEG-diacylglycerol (DAG), a PEG-dialkylglycerol, a PEG- dialkyloxypropyl (DAA), a PEG-phospholipid, a PEG-ceramide (Cer), or a mixture thereof (such as PEG-Cerl4 or PEG-Cer20).
  • PEG polyethyleneglycol
  • the PEG-DAA conjugate may be, for example, a PEG- dilauryloxypropyl (Ci 2 ), a PEG-dimyristyloxypropyl (C 14 ), a PEG-dipalmityloxypropyl (Ci 6 ), or a PEG- distearyloxypropyl (C] 8 ).
  • pegylated-lipids include, but are not limited to, polyethylene glycol-didimyristoyl glycerol (C14-PEG or PEG-C14, where PEG has an average molecular weight of 2000 Da) (PEG-DMG); (R)-2,3-bis(octadecyloxy)propyl-l-(methoxy polyethylene glycol)2000)propylcarbamate) (PEG-DSG); PEG-carbamoyl-1,2- dimyristyloxypropylamine, in which PEG has an average molecular weight of 2000 Da (PEG- cDMA); N-Acetylgalactosamine-((R)-2,3-bis(octadecyloxy)propyl- 1 -(methoxy poly(ethylene glycol)2000)propylcarbamate)) (GalNAc-PEG-DSG); mPEG
  • the aggregation reducing agent is PEG- DMG. In another embodiment, the aggregation reducing agent is PEG-c-DMA.
  • the average molecular weight of the PEG moiety in the PEG-modified lipids can range from about 500 to about 8,000 Daltons (e.g., from about 1,000 to about 4,000 Daltons). In one preferred embodiment, the average molecular weight of the PEG moiety is about 2,000 Daltons.
  • the concentration of the aggregation reducing agent may range from about 0.1 to about 15 mol %, based upon the 100% total moles of lipid in the lipid particle. In one embodiment, the formulation includes less than about 3, 2, or 1 mole percent of PEG or PEG-modified lipid, based upon the total moles of lipid in the lipid particle.
  • the lipid nanoparticles include from about 0.1% to about 20% on a molar basis of the PEG-modified lipid, e.g., about 0.5 to about 10%, about 0.5 to about 5%, about 10%, about 5%, about 3.5%, about 1.5%, about 0.5%, or about 0.3% on a molar basis (based on 100% total moles of lipids in the lipid nanoparticle).
  • LNPs Lipid Nanoparticles
  • the lipid nanoparticles may have the structure of a liposome.
  • a liposome is a structure having lipid-containing membranes enclosing an aqueous interior. Liposomes may have one or more lipid membranes. Liposomes can be single-layered, referred to as unilamellar, or multi-layered, referred to as multilamellar. When complexed with nucleic acids, lipid particles may also be lipoplexes, which are composed of cationic lipid bilayers sandwiched between DNA layers.
  • the formulation is preferably substantially free of aggregates of lipid nanoparticles.
  • the formulation may have a deo (i.e., 90% of the particles have a particle size) less than about 1, about 0.9, about 0.8, about 0.7, or about 0.6 ⁇ .
  • the formulation includes less than about 5, about 4, about 3, about 2, or about 1% by volume of aggregates greater than about 2, about 1.5, about 1 , or about 0.8 ⁇ ..
  • the lipid nanoparticles in the formulation have a d9 8 of less than 1 micron, such as less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm or less than about 100 nm.
  • the lipid nanoparticles have a d 99 of less than 1 micron, such as less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm or less than about 100 nm.
  • the particle has a d 50 of less than about 100 nm, such as less than about 75nm, less than about 50 nm, less than about 40 nm, less than about 30 nm, less than about 20 nm or less than about 10 nm.
  • the lipid nanoparticles may have a d 99 ranging from about 50 to about 200 nm, or from about 75 to about 150 nm.
  • the lipid nanoparticles may have a d 50 ranging from about 5 to about 50 nm, such as from about 10 to about 50 nm, from about 25 to about 50 nm or from about 35 to about 45 nm.
  • the lipid nanoparticles have a d 50 ranging from about 10 to about 40 nm or from about 20 to about 30 nm.
  • lipid nanoparticles treated with Compound 1 have a reduced particle size.
  • the lipid nanoparticles may have the previously mentioned particle sizes pre- or post-treatment with Compound 1.
  • the lipid nanoparticles have a median diameter size of from about 50 nm to about 300 nm, such as from about 50 nm to about 250 nm, for example, from about 50 nm to about 200 nm.
  • the d 50 , d 98 or d 9 of the lipid nanoparticles in the formulation does not vary by more than 40, 30, 20, 10, or 5% after 1, 3, 6, 9, 12, and 24 months of storage at 4° C. In one embodiment, after 1 month of storage at 4° C, the lipid nanoparticles in the formulation have d 5 o, d 98 and/or d 99 values as set forth above.
  • the lipid nanoparticles in the formulation have d 98 or d 99 of less than 1 micron, such as less than about 500 nm, less than about 400 nm, less than about 300 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm or less than about 100 nm.
  • the lipid nanoparticles in the formulation of the present invention have a single mode particle size distribution (i.e., they are not bi- or poly-modal).
  • the lipid nanoparticles may further comprise one or more lipids and/or other components in addition to those mentioned above.
  • Other lipids may be included in the liposome compositions for a variety of purposes, such as to prevent lipid oxidation or to attach ligands onto the liposome surface. Any of a number of lipids may be present in lipid particles, including amphipathic, neutral, cationic, and anionic lipids. Such lipids can be used alone or in combination.
  • Additional components that may be present in a lipid particle include bilayer stabilizing components such as polyamide oligomers ⁇ see, e.g., U.S. Patent No. 6,320,017, which is incorporated by reference in its entirety), peptides, proteins, and detergents.
  • lipid nanoparticles having varying molar ratios of cationic lipid, non-cationic (or neutral) lipid, sterol (e.g., cholesterol), and aggregation reducing agent (such as a PEG- modified lipid) on a molar basis (based upon the total moles of lipid in the lipid nanoparticles) are provided in Table 1 below.
  • the weight ratio of lipid to siRNA is at least about 0.5: 1, at least about 1 :1, at least about 2:1, at least about 3:1, at least about 4:1, at least about 5:1, at least about 6: 1, at least about 7: 1, at least about 11: 1 or at least about 33:1. In one embodiment, the weight ratio of lipid to siRNA is from about 1:1 to about 35:1, about 3: 1 to about 15: 1, about 4:1 to about 15:1, or about 5:1 to about 13:1. In one embodiment, the weight ratio of lipid to siRNA is from about 0.5:1 to about 12:1.
  • the lipid nanoparticles have an in vivo half life (ti /2 ) (e.g., in the liver, spleen or plasma) of less than about 3 hours, such as less than about 2.5 hours, less than about 2 hours, less than about 1.5 hours, less than about 1 hour, less than about 0.5 hour or less than about 0.25 hours.
  • ti /2 in vivo half life
  • the medium containing the lipid nanoparticles is substantially free of negative counter-ions (i.e., anions).
  • the medium comprises a non-ionic or substantially non-ionic diluent, and preferably includes a non-ionic or substantially non-ionic diluent that does not destabilize the formulation.
  • the non-ionic or substantially non-ionic diluent increases the stability of the lipid nanoparticles, such as against mechanical disturbances, and/or inhibits the aggregation of the lipid nanoparticles.
  • the medium may comprise water.
  • the medium is deionized (e.g., deionized water).
  • the water in the medium may have been purified, for example, by reverse osmosis.
  • the medium (such as water) contains less than about 50 ppm of mineral acid(s), such as less than about 40 ppm, less than about 30 ppm, less than about 20 ppm, less than about 10 ppm, less than about 5 ppm or less than about 1 ppm of mineral acid(s).
  • the medium may include an acid so long as it is not predominantly in its dissociated form.
  • the formulation further comprises an acid, wherein the ratio of (a) the concentration of the anions formed from the acid to (b) the concentration of the acid is less than about 0.5, such as less than about 0.4, less than about 0.3, less than about 0.2 or less than about 0.1. In a particular embodiment, the ratio of anion concentration to acid concentration is less than about 0.2 to 0.5.
  • the anions present in the formulation may be derived from the acid in the medium.
  • the anion is a monovalent anion (such as an anion derived from acetic acid).
  • the isotonicity agent(s) included in the formulation are preferably substantially free of anions (e.g., substantially non-ionic), and more preferably are non-ionic.
  • Suitable non-ionic isotonicity agents include, but are not limited to, polyols (e.g., a sugar alcohol such as a C 3 -C 6 sugar alcohol), sugars (such as sucrose, fructose, dextrose, trehalose, or glucose), amino acids (such as glycine), and albumin.
  • Suitable sugar alcohols include, but are not limited to, glycerol, erythritol, threitol, arabitol, xylitol, ribitol, sorbitol, mannitol, dulcitol and iditol.
  • the isotonicity agent is a sugar such as a glucose.
  • the concentration of sugar (e.g., glucose) in the medium is at most about 300 mM, such as at most about 200, 100, 75, or 50 mM.
  • the amount of the isotonicity agent is preferably sufficient for the formulation to obtain an isotonic level.
  • the formulation is free or substantially free of isotonicity agents.
  • the concentration of conjugated siRNA in the formulation may range from about 0.01 to about 50 mg/mL. In one embodiment, the concentration of conjugated siRNA in the formulation ranges from about 0.1 to about 10 mg mL, such as 0.5 to about 5 mg/mL. In another embodiment, the concentration of conjugated siRNA in the formulation is about 0.5, about 0.75, about 1, about 1.5, about 2, about 2.5, about 3, about 4, or about 5 mg/mL.
  • the concentration of lipid nanoparticles in the formulation may range from about 0.01 to about 50 mg/mL. In one embodiment, the concentration of lipid nanoparticles in the formulation ranges from about 0.1 to about 10 mg/mL, such as 0.5 to about 5 mg/mL. In another embodiment, the concentration of lipid nanoparticles in the formulation is about 0.5, about 0.75, about 1, about 1.5, about 2, about 2.5, about 3, about 4, or about 5 mg/mL.
  • formulations may be administered parenterally, for example, intradermally, subcutaneously, intramuscularly, intravenously, or intraperitoneally.
  • the formulation is directly injected into a subject (e.g., by intravenous infusion).
  • the formulation is added to an intravenous fluid which is intravenously administered. Because many intravenous fluids contain significant quantities of anions which may over time cause aggregation of LNPs, the LNP-containing formulation of the invention is preferably added to the intravenous fluid shortly before (e.g., within 5, 10 or 15 minutes of) or simultaneously with the intravenous administration to the subject.
  • formulations may further include additional pharmaceutically acceptable diluents, excipients, and/or carriers.
  • excipients include, but are not limited to, isotonicity agents, pH adjusting and buffering agents.
  • the formulations may also include lipid-protective agents which protect lipids against free-radical and lipid-peroxidative damages on storage. Such agents include, but are not limited to, lipophilic free-radical quenchers, such as a-tocopherol and water-soluble iron-specific chelators, such as ferrioxamine.
  • the formulation can be sterilized by known sterilization techniques.
  • the aqueous solutions can then be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • the concentration of conjugated siRNA in the formulation can range, for example, from less than about 0.01% (e.g., at or at least about 0.05-5%) to as much as 10 to 30% by weight.
  • the dose of conjugated siRNA is dependent on many factors, including the disorder and active pharmaceutical ingredient.
  • the dose of conjugated siRNA administered may range from about 0.01 and about 50 mg per kilogram of body weight (e.g., from about 0.1 and about 5 mg/kg of body weight).
  • the concentration of lipid nanoparticles in the formulation can range, for example, from less than about 0.01% (e.g., at or at least about 0.05-5%) to as much as 10 to 30% by weight.
  • the dose of lipid nanoparticles is dependent on many factors, including the disorder and active pharmaceutical ingredient. In one embodiment, the dose of lipid nanoparticles administered may range from about 0.01 and about 50 mg per kilogram of body weight (e.g., from about 0.1 and about 5 mg/kg of body weight).
  • the formulation can be provided in kit form.
  • the kit will typically be comprised of a container that is compartmentalized for holding the various elements of the kit.
  • the kit may contain the lipid nanoparticles, the LNP-containing formulation (such as in dehydrated or concentrated form), or the conjugated siR A formulation with instructions for their rehydration or dilution and administration.
  • the conjugated siRNA can be prepared by methods known in the art, such as those described in U.S. Patent Publication Nos. 2013/0184328, 2013/0203836, 2013/0202652, 2013/0317080, 2013/0211063, and 2013/0184324, which are hereby incorporated by reference.
  • the lipid nanoparticles may be prepared by an in-line mixing method as follows.
  • both the lipids e.g., the cationic lipid, non-cationic lipid, sterol, and aggregation reducing agent
  • the nucleic acid are added in parallel into a mixing chamber.
  • the mixing chamber can be a simple T-connector. This method is disclosed, for example, in International Publication No. WO 2010/088537, U.S. Patent Nos. 6,534,018 and US 6,855,277, U.S. Patent Publication No. 2007/0042031 and Pharmaceuticals Research, Vol. 22, No. 3, Mar. 2005, p. 362-372, which are hereby incorporated by reference in their entirety.
  • individual and separate stock solutions are prepared - one containing lipid (e.g., the cationic lipid, non-cationic lipid, sterol, and aggregation reducing agent) and the other an active pharmaceutical ingredient, such as a nucleic acid (e.g., siRNA).
  • lipid e.g., the cationic lipid, non-cationic lipid, sterol, and aggregation reducing agent
  • an active pharmaceutical ingredient such as a nucleic acid (e.g., siRNA).
  • a lipid stock solution containing a cationic lipid, non-cationic lipid, sterol, and an aggregation reducing agent e.g., a PEG-modified lipid
  • an alcohol e.g., ethanol
  • the nucleic acid e.g., siRNA
  • acetate buffer for example, at a concentration of 0.8 mg/mL.
  • 5 mL of each stock solution may be prepared.
  • the stock solutions are completely clear, and the lipids are completely solubilized before combining them with the nucleic acid.
  • the stock solutions may be heated to completely solubilize the lipids.
  • the individual stock solutions i.e., the lipid stock solution and the nucleic acid stock solution
  • the lipid nanoparticles may be contacted with the delivery enhancer (e.g., Compound 1, Compound 2, or a mixture thereof) by any method known in the art.
  • the delivery enhancer may be added to and/or mixed and/or incubated with the lipid nanoparticles.
  • the delivery enhancer compound is added during the formation of the lipid nanoparticles.
  • the delivery enhancer compound may be added to and/or mixed and/or incubated with the lipid stock solution and the nucleic acid stock solution.
  • the method includes incubating the lipid nanoparticle with the delivery enhancer.
  • the incubation can be for from about 1 to about 48 hours.
  • the process involves incubating overnight (e.g., for about 12 or 16 hours).
  • the process involves incubating at a temperature between about 0 ° C and about 10° C, such as between about 2° C to about 5° C, e.g., at about 4° C.
  • the process involves incubating overnight (e.g., for about 12 or 16 hours) at about 4° C.
  • the incubation is conducted in an Eppendorf tube.
  • the medium of the lipid nanoparticles may be exchanged to one which is (a) non-ionic or substantially non-ionic and/or (b) free of or substantially free of anions.
  • This exchange can be performed by dialysis or tangential flow filtration.
  • the lipid nanoparticles may be dialyzed into reverse osmosis / deionized (RO/DI) water, and then concentrated (e.g., using centrifuge tubes).
  • the dispersion medium can then be changed to, for example, 300 mM glucose by adding an appropriate stock solution, for example, to give final lipid nanoparticles at ⁇ lmg/n L (based on siRNA).
  • the medium may be exchanged as follows.
  • the lipid nanoparticles are diluted into RO/DI water.
  • the diluted lipid nanoparticles are then concentrated using tangential flow filtration.
  • the concentration step includes washing with lOx larger volume-compared to concentrated formulation volume-of RO/DI water.
  • the dispersion medium can then be changed to, for example, 300 mM glucose by adding an appropriate stock solution, for example, to give final lipid nanoparticles at ⁇ lmg mL (based on siRNA).
  • the siRNAs used target GFP (eGFP plasmid, Clonetech, Mountain Via, CA).
  • the siRNAs were labelled with Alexa Fluor 647 (alexa637) and formulated into LNPs or conjugated to cholesterol.
  • Alexa Fluor 647 alexa637
  • the procedure used to produce LNP-siRNA, LNP- siRNA-alexa647 and LNP-siRNA-gold are as described in Gilleron et al, Nature Biotech., 31(7), 638-646, 2013.
  • GFP-HeLa cells (Bramsen et al, Nucleic Acid Res., 37(9), 2867-2881, 2009) were cultured in DMEM media complemented with 10% FBS and 1% penicillin-streptomycin at 37° C and 5% C0 2 .
  • Primary human fibroblasts (GM00041, Coriell Institute, Camden, NJ) were cultured and infected with Rab5-GFP as previously described (Pal et al, J. Cell Biol, 172(4), 605-618).
  • a high throughput screen was performed to identify chemical compounds that improve delivery of (i) cholesterol-conjugated siRNA and (ii) lipid nanoparticles containing siRNA.
  • the primary screen was performed in HeLa cells, a cell line which exhibits the key general features of the mammalian endocytic pathway. The results were validated by performing a secondary screen in primary cells, fibroblasts and hepatocytes.
  • the assay consisted of GFP-expressing HeLa cells transfected with suboptimal doses of LNPs containing an anti-GFP siRNA. The assay was optimized to obtain 20% GFP expression reduction as measured by quantitative microscopy.
  • silencing could be boosted to higher than 80% by adding a known transfection reagent. This demonstrated that the amount of siRNA per se was not limiting, but rather cellular uptake and/or escape from the endolysosomal system was suboptimal, allowing us to carry out a screen to improve these steps.
  • a pilot screen was initially performed on a small set of compounds, with or without overnight pre-incubation of the compounds with the delivery system prior to adding the mixtures to the cells.
  • This pilot screen revealed that an overnight preincubation increased the number of hits for LNPs. Therefore, an overnight compound preincubation step was incorporated for the LNP screen.
  • the cells were washed, and further incubated with fresh media for 72 hours. The cells were fixed, nuclei were stained with Hoechst and the GFP expression was quantified on an automated fluorescence microscope. All transfections were performed in serum containing media to mimic blood flow conditions. More than 50,000 compounds were tested. 25 and 27 compounds improved the silencing activity of LNPs and cholesterol-conjugated siRNAs, respectively.
  • Rab5-GFP human primary fibroblasts were transfected with LNP-siRNA formulation preincubated or not with the compounds (following a similar procedure as that in Example 1). After 72 hours, the cells were fixed with PFA 4% (pH 7.2 in phosphate buffer) for 20 minutes at room temperature. After washing, cells nuclei were labelled with Dapi and cytosol with SytoBlue. Acquisition and analysis of images (at least 25 fields per conditions) were done on an ArrayscanVTI with Twisterll automated wide field microscope (TDS, MPI-CBG, Dresden).
  • Example 2 The procedure in Example 2 was repeated with primary mouse hepatocytes expressing Lifeact-GFP instead of human primary fibroblasts.
  • the results for the lipid nanoparticles and cholesterol-conjugated siRNA are provided in Tables 4 and 5 below, respectively.
  • LNPs treated with BADGE or CP WOO 1 J18 or DMSO (control) were analyzed by electron microscopy. Morphological experiments were analysed in a blind fashion using a code that was not broken until the quantitation was completed.
  • For electron microscopy analysis on HeLa cells cells were transfected with LNP-siRNA-gold and fixed with 2.5% glutaraldehyde (in phosphate buffer) overnight. Then, cells were post-fixed in ferrocyanide reduced osmium as described in Karnovsky, Proceedings of the Eleventh Annual Meeting of the American Society or Cell Biology, 284, 186, 1971.
  • image equalization was performed to the interval [0;1] and thresholding with a threshold set at 0.3.
  • the binarized images were then analyzed by the watershed transform to split contiguous gold particles.
  • a last postprocessing step was performed to remove uncertain gold particles (particles having the average intensity value less than 5 standard deviations of the median intensity value in the whole image). Then, for a set of images, the number of particles were automatically counted and manually counted and an error rate ( ⁇ 1%) was determined, confirming that the procedure succeeded in correctly identifying gold particles. Finally, the procedure to determine the total number of gold particles in the images was applied. Then, the number of gold particles within the cytoplasm were counted manually based on morphological recognition.
  • BADGE increased the uptake of LNPs as indicated by the -15 fold increase in the amount of siRNA-gold internalized by the cells shown in Figure lb.
  • the ratio of siRNA-gold in the cytosol versus the total amount internalized was not increased ( Figure lc).
  • Figure lc the ratio of siRNA-gold in the cytosol versus the total amount internalized
  • the silencing enhancers were either pre-incubated with the cells or were added to the cells concomitantly with the delivery systems. If a compound were acting primarily on the cell, its silencing enhancer effect would be expected to occur also when added to the cells prior to the siRNAs. By this criterion, we found that 28% of the enhancer compounds were active only when incubated with the LNPs whereas the remaining 72% were most probably acting on the cells. Interestingly, almost all compounds that improved silencing activity by facilitating endo- lysosomal escape were active when pre-incubated with the cells, suggesting that they act upon the cells. The results are provided for the lipid nanoparticles and cholesterol conjugated siRNAs in tables 6 and 7, respectively.
  • Electron micrograph negative staining was performed on LNPs treated with BADGE and DMSO (control) to determine the differences in morphological properties, such as size, of the LNPs.
  • BADGE had an effect on the nanoparticles themselves.
  • BADGE reduced the size of LNPs by ⁇ 2 fold as shown in Figure 2a. This reduction in size was associated with a dramatic acceleration of uptake kinetics as shown in Figure 2b.
  • the uptake of LNPs exposed to BADGE is much less sensitive to the knockdown of clathrin, ARF-1 and RAC-1 compared to the control ( Figure 2c), suggesting that the smaller LNPs are captured through a broader set of endocytic mechanisms.
  • the present inventors theorize that the reduction in size of the LNPs is due to particle compaction, not fission.
  • the invention furthermore comprises the following items:
  • lipid nanoparticles of item 1 wherein the lipid nanoparticles are treated with a sufficient amount of the delivery enhancer to reduce the size of the lipid nanoparticles.
  • Lipid nanoparticles comprising (a) an active pharmaceutical ingredient and (b) a delivery enhancer described herein and mixtures thereof.
  • lipid nanoparticles of any one of items 1-3 wherein the lipid nanoparticles further comprise a cationic lipid, a non-cationic lipid, an aggregation reducing agent, and optionally a sterol.
  • a pharmaceutical formulation comprising the lipid nanoparticles of any one of items 1-6.
  • a pharmaceutical formulation comprising:
  • nucleic acid is selected from an interfering RNA, an antisense oligonucleotide, a DNAi oligonucleotide, a ribozyme, an aptamer, a plasmid, and any combination of any of the foregoing.
  • interfering RNA is selected from siRNA, aiRNA, miRNA, Dicer-substrate dsRNA, shR A, ssRNAi oligonucleotides, and any combination of any of the foregoing.
  • lipid nanoparticles further comprise a cationic lipid, a non-cationic lipid (such as a neutral lipid), an aggregation reducing agent (such as polyethylene glycol (PEG) or PEG-modified lipid), and optionally, a sterol.
  • a cationic lipid such as a neutral lipid
  • a non-cationic lipid such as a neutral lipid
  • an aggregation reducing agent such as polyethylene glycol (PEG) or PEG-modified lipid
  • PEG polyethylene glycol
  • sterol optionally, a sterol
  • a process for preparing lipid nanoparticles comprising the step of incubating lipid nanoparticles in the presence of a delivery enhancer described herein and mixtures thereof, wherein the lipid nanoparticles comprise an active pharmaceutical ingredient.
  • a process for preparing a pharmaceutical formulation comprising adding a delivery enhancer described herein and mixtures thereof to lipid nanoparticles, wherein the lipid nanoparticles comprise an active pharmaceutical ingredient.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dispersion Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des nanoparticules lipidiques et des formulations pharmaceutiques les contenant qui présentent une meilleure administration cellulaire de principes actifs pharmaceutiques tels que l'ARNsi. La présente invention concerne également un procédé de préparation de ces nanoparticules lipidiques et des formulations pharmaceutiques ainsi que leur utilisation.
PCT/EP2015/076993 2014-11-18 2015-11-18 Activateurs d'administration d'arnsi conjugué et de nanoparticules lipidiques WO2016079197A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462081296P 2014-11-18 2014-11-18
US62/081,296 2014-11-18

Publications (1)

Publication Number Publication Date
WO2016079197A1 true WO2016079197A1 (fr) 2016-05-26

Family

ID=54557425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/076993 WO2016079197A1 (fr) 2014-11-18 2015-11-18 Activateurs d'administration d'arnsi conjugué et de nanoparticules lipidiques

Country Status (1)

Country Link
WO (1) WO2016079197A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110548006A (zh) * 2018-05-30 2019-12-10 复旦大学 一种科罗索酸脂质体及其制备方法和用途

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120195957A1 (en) * 2009-04-30 2012-08-02 Mandip Singh Sachdeva Novel nanoparticle formulations for skin delivery
WO2012170889A1 (fr) * 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Lipides clivables
US20130072543A1 (en) * 2011-07-13 2013-03-21 Arrowhead Research Corporation Compounds and Compositions for Nucleic Acid Formulation and Delivery
US20130079383A1 (en) * 2011-07-13 2013-03-28 Arrowhead Research Corporation Lipid Compounds Targeting VLA-4
WO2013149140A1 (fr) * 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Lipides cationiques ionisables

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120195957A1 (en) * 2009-04-30 2012-08-02 Mandip Singh Sachdeva Novel nanoparticle formulations for skin delivery
WO2012170889A1 (fr) * 2011-06-08 2012-12-13 Shire Human Genetic Therapies, Inc. Lipides clivables
US20130072543A1 (en) * 2011-07-13 2013-03-21 Arrowhead Research Corporation Compounds and Compositions for Nucleic Acid Formulation and Delivery
US20130079383A1 (en) * 2011-07-13 2013-03-28 Arrowhead Research Corporation Lipid Compounds Targeting VLA-4
WO2013149140A1 (fr) * 2012-03-29 2013-10-03 Shire Human Genetic Therapies, Inc. Lipides cationiques ionisables

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110548006A (zh) * 2018-05-30 2019-12-10 复旦大学 一种科罗索酸脂质体及其制备方法和用途
CN110548006B (zh) * 2018-05-30 2022-07-29 复旦大学 一种科罗索酸脂质体及其制备方法和用途

Similar Documents

Publication Publication Date Title
US9415109B2 (en) Stable non-aggregating nucleic acid lipid particle formulations
US20210220274A1 (en) Novel lipid formulations for nucleic acid delivery
US10415037B2 (en) Compositions and methods for silencing hepatitis B virus gene expression
US20230212578A1 (en) Compositions and methods for treating hypertriglyceridemia
EP2281041B1 (fr) Réduction au silence de l'expression du gène csn5 au moyen d'arn interférant
US20110071208A1 (en) Lipid encapsulated dicer-substrate interfering rna
JP2022511977A (ja) KRASバリアントmRNA分子
EP3329003A2 (fr) Compositions et méthodes de silençage de l'expression du gène du virus de l'hépatite b
WO2016183366A2 (fr) Compositions et méthodes permettant l'extinction de l'expression de l'arn du virus de l'hépatite d
WO2016079197A1 (fr) Activateurs d'administration d'arnsi conjugué et de nanoparticules lipidiques
WO2023196188A1 (fr) Amélioration immunitaire de traitement du cancer
AU2013202932A1 (en) Novel lipid formulations for nucleic acid delivery

Legal Events

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

Ref document number: 15797092

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15797092

Country of ref document: EP

Kind code of ref document: A1