Peptide-Based In Vivo siRNA Delivery System

Information

  • Patent Application
  • 20170056472
  • Publication Number
    20170056472
  • Date Filed
    July 01, 2016
    7 years ago
  • Date Published
    March 02, 2017
    7 years ago
Abstract
The present invention is directed compositions for targeted delivery of RNA interference (RNAi) polynucleotides to hepatocytes in vivo. Targeted RNAi polynucleotides are administered together with co-targeted melittin delivery peptides. Delivery peptides provide membrane penetration function for movement of the RNAi polynucleotides from outside the cell to inside the cell. Reversible modification provides physiological responsiveness to the delivery peptides.
Description
BACKGROUND OF THE INVENTION

The delivery of polynucleotide and other substantially cell membrane impermeable compounds into a living cell is highly restricted by the complex membrane system of the cell. Drugs used in antisense, RNAi, and gene therapies are relatively large hydrophilic polymers and are frequently highly negatively charged. Both of these physical characteristics severely restrict their direct diffusion across the cell membrane. For this reason, the major barrier to polynucleotide delivery is the delivery of the polynucleotide across a cell membrane to the cell cytoplasm or nucleus.


One means that has been used to deliver small nucleic acid in vivo has been to attach the nucleic acid to either a small targeting molecule or a lipid or sterol. While some delivery and activity has been observed with these conjugates, the very large nucleic acid dose required with these methods is impractical.


Numerous transfection reagents have also been developed that achieve reasonably efficient delivery of polynucleotides to cells in vitro. However, in vivo delivery of polynucleotides using these same transfection reagents is complicated and rendered ineffective by in vivo toxicity, adverse serum interactions, or poor targeting. Transfection reagents that work well in vitro, cationic polymers and lipids, typically form large cationic electrostatic particles and destabilize cell membranes. The positive charge of in vitro transfection reagents facilitates association with nucleic acid via charge-charge (electrostatic) interactions thus forming the nucleic acid/transfection reagent complex. Positive charge is also beneficial for nonspecific binding of the vehicle to the cell and for membrane fusion, destabilization, or disruption. Destabilization of membranes facilitates delivery of the substantially cell membrane impermeable polynucleotide across a cell membrane. While these properties facilitate nucleic acid transfer in vitro, they cause toxicity and ineffective targeting in vivo. Cationic charge results in interaction with serum components, which causes destabilization of the polynucleotide-transfection reagent interaction, poor bioavailability, and poor targeting. Membrane activity of transfection reagents, which can be effective in vitro, often leads to toxicity in vivo.


For in vivo delivery, the vehicle (nucleic acid and associated delivery agent) should be small, less than 100 nm in diameter, and preferably less than 50 nm. Even smaller complexes, less that 20 nm or less than 10 nm would be more useful yet. Delivery vehicles larger than 100 nm have very little access to cells other than blood vessel cells in vivo. Complexes formed by electrostatic interactions tend to aggregate or fall apart when exposed to physiological salt concentrations or serum components. Further, cationic charge on in vivo delivery vehicles leads to adverse serum interactions and therefore poor bioavailability. Interestingly, high negative charge can also inhibit targeted in vivo delivery by interfering with interactions necessary for targeting, i.e. binding of targeting ligands to cellular receptors. Thus, near neutral vehicles are desired for in vivo distribution and targeting. Without careful regulation, membrane disruption or destabilization activities are toxic when used in vivo. Balancing vehicle toxicity with nucleic acid delivery is more easily attained in vitro than in vivo.


Rozema et al., in U.S. Patent Publication 20040162260 demonstrated a means to reversibly regulate membrane disruptive activity of a membrane active polyamine. The membrane active polyamine provided a means of disrupting cell membranes. pH-dependent reversible regulation provided a means to limit activity to the endosomes of target cells, thus limiting toxicity. Their method relied on modification of amines on a polyamine with 2-propionic-3-methylmaleic anhydride.




embedded image


This modification converted the polycation to a polyanion via conversion of primary amines to pairs of carboxyl groups (β carboxyl and γ carboxyl) and reversibly inhibited membrane activity of the polyamine. Rozema et al. (Bioconjugate Chem. 2003, 14, 51-57) reported that the β carboxyl did not exhibit a full apparent negative charge and by itself was not able to inhibit membrane activity. The addition of the γ carboxyl group was reported to be necessary for effective membrane activity inhibition. To enable co-delivery of the nucleic acid with the delivery vehicle, the nucleic acid was covalently linked to the delivery polymer. They were able to show delivery of polynucleotides to cells in vitro using their biologically labile conjugate delivery system. However, because the vehicle was highly negatively charged, with both the nucleic acid and the modified polymer having high negative charge density, this system was not efficient for in vivo delivery. The negative charge likely inhibited cell-specific targeting and enhanced non-specific uptake by the reticuloentothelial system (RES).


Rozema et al., in U.S. Patent Publication 20080152661, improved on the method of U.S. Patent Publication 20040162260 by eliminating the high negative charge density of the modified membrane active polymer. By substituting neutral hydrophilic targeting (galactose) and steric stabilizing (PEG) groups for the γ carboxyl of 2-propionic-3-methylmaleic anhydride, Rozema et al. were able to retain overall water solubility and reversible inhibition of membrane activity while incorporating effective in vivo hepatocyte cell targeting. As before, the polynucleotide was covalently linked to the transfection polymer. Covalent attachment of the polynucleotide to the transfection polymer was maintained to ensure co-delivery of the polynucleotide with the transfection polymer to the target cell during in vivo administration by preventing dissociation of the polynucleotide from the transfection polymer. Co-delivery of the polynucleotide and transfection polymer was required because the transfection polymer provided for transport of the polynucleotide across a cell membrane, either from outside the cell or from inside an endocytic compartment, to the cell cytoplasm. U.S. Patent Publication 20080152661 demonstrated highly efficient delivery of polynucleotides, specifically RNAi oligonucleotides, to liver cells in vivo using this new improved physiologically responsive polyconjugate.


However, covalent attachment of the nucleic acid to the polyamine carried inherent limitations. Modification of the transfection polymers, to attach both the nucleic acid and the masking agents was complicated by charge interactions. Attachment of a negatively charged nucleic acid to a positively charged polymer is prone to aggregation thereby limiting the concentration of the mixture. Aggregation could be overcome by the presence of an excess of the polycation or polyanion. However, this solution limited the ratios at which the nucleic acid and the polymer may be formulated. Also, attachment of the negatively charged nucleic acid onto the unmodified cationic polymer caused condensation and aggregation of the complex and inhibited polymer modification. Modification of the polymer, forming a negative polymer, impaired attachment of the nucleic acid.


Rozema et al. further improved upon the technology described in U.S. Patent Publication 20080152661, in U.S. Provisional Application 61/307,490. In U.S. Provisional Application 61/307,490, Rozema et al. demonstrated that, by carefully selecting targeting molecules, and attaching appropriate targeting molecules independently to both an siRNA and a delivery polymer, the siRNA and the delivery polymer could be uncoupled yet retain effective targeting of both elements to cells in vivo and achieve efficient functional targeted delivery of the siRNA. The delivery polymers used in both U.S. Patent Publication 20080152661 and U.S. Provisional Application 61/307,490 were relatively large synthetic polymers, poly(vinyl ether)s and poly(acrylate)s. The larger polymers enabled modification with both targeting ligands for cell-specific binding and PEG for increased shielding. Larger polymers were necessary for effective delivery, possibly through increased membrane activity and improved protection of the nucleic acid within the cell endosome. Larger polycations interact more strongly with both membranes and with anionic RNAs.


We have now developed an improved siRNA delivery system using a much smaller delivery peptide. The improved system provides for efficient siRNA delivery with decreased toxicity and therefore a wider therapeutic window.


SUMMARY OF THE INVENTION

In a preferred embodiment, the invention features a composition for delivering an RNA interference polynucleotide to a liver cell in vivo comprising: a) an asialoglycoprotein receptor (ASGPr)-targeted reversibly masked melittin peptide (delivery peptide) and b) an RNA interference polynucleotide conjugated to a hydrophobic group containing at least 20 carbon atoms (RNA-conjugate). The delivery peptide and the siRNA-conjugate are synthesized separately and may be supplied in separate containers or a single container. The RNA interference polynucleotide is not conjugated to the delivery peptide.


In another preferred embodiment, the invention features a composition for delivering an RNA interference polynucleotide to a liver cell in vivo comprising: a) an ASGPr-targeted reversibly masked melittin peptide (delivery peptide) and b) an RNA interference polynucleotide conjugated to a galactose cluster (RNA conjugate). The delivery peptide and the siRNA-conjugate are synthesized separately and may be supplied in separate containers or a single container. The RNA interference polynucleotide is not conjugated to the polymer.


In a preferred embodiment, an ASGPr-targeted reversibly masked melittin peptide comprises a melittin peptide reversibly modified by reaction of primary amines on the peptide with ASGPr ligand-containing masking agents. An amine is reversibly modified if cleavage of the modifying group restores the amine. Reversible modification of the melittin peptide with the masking agents disclosed herein reversibly inhibits membrane activity of the melittin peptide. In the masked state, the reversibly masked melittin peptide does not exhibit membrane disruptive activity. Reversible modification of more than 80%, or more than 90%, of the amines on the melittin peptide is required to inhibit membrane activity and provide cell targeting function, i.e. form a reversibly masked melittin peptide.


A preferred ASGPr ligand-containing masking agent has a neutral charge and comprises a galactosamine or galactosamine derivative having a disubstituted maleic anhydride amine-reactive group. Another preferred ASGPr ligand-containing masking agent comprises a galactosamine or galactosamine derivative having a peptidase cleavable dipeptide-p-amidobenzyl amine reactive carbonate derivative. Reaction of the amine reactive carbonate with an amine reversibly modifies the amine to form an amidobenzyl carbamate linkage.


In a preferred embodiment, a melittin peptide comprises an Apis florea (little or dwarf honey bee) melittin, Apis mellifera (western or European or big honey bee), Apis dorsata (giant honey bee), Apis cerana (oriental honey bee) or derivatives thereof. A preferred melittin peptide comprises the sequence: Xaa1-Xaa2-Xaa3-Ala-Xaa5-Leu-Xaa7-Val-Leu-Xaa10-Xaa11-Xaa12-Leu-Pro-Xaa15-Leu-Xaa17-Xaa18-Trp-Xaa20-Xaa21-Xaa22-Xaa23-Xaa24-Xaa25-Xaa26 wherein:

    • Xaa1 is leucine, D-leucine, isoleucine, norleucine, tyrosine, tryptophan, valine, alanine, dimethylglycine, glycine, histidine, phenylalanine, or cysteine,
    • Xaa2 is isoleucine, leucine, norleucine, or valine,
    • Xaa3 is glycine, leucine, or valine,
    • Xaa5 is isoleucine, leucine, norleucine, or valine,
    • Xaa7 is lysine, serine, asparagine, alanine, arginine, or histidine,
    • Xaa10 is alanine, threonine, or leucine,
    • Xaa11 is threonine or cysteine,
    • Xaa12 is glycine, leucine, or tryptophan,
    • Xaa15 is threonine or alanine,
    • Xaa17 is isoleucine, leucine, norleucine, or valine,
    • Xaa18 is serine or cysteine,
    • Xaa20 is isoleucine, leucine, norleucine, or valine,
    • Xaa21 is lysine or alanine,
    • Xaa22 is asparagine or arginine,
    • Xaa23 is lysine or alanine,
    • Xaa24 is arginine or lysine,
    • Xaa25 is lysine, alanine, or glutamine,
    • Xaa26 is optional and if present is glutamine, cysteine, glutamine-NH2, or cysteine-NH2; and,
    • and at least two of Xaa11, Xaa23, and Xaa25 are lysine.


A more preferred melittin comprises the sequence: Xaa1-Xaa2-Xaa3-Ala-Xaa5-Leu-Xaa7-Val-Leu-Xaa10-Xaa11-Xaa12-Leu-Pro-Xaa15-Leu-Xaa17-Ser-Trp-Xaa20-Lys-Xaa22-Lys-Arg-Lys-Xaa26 wherein:

    • Xaa1 is leucine, D-leucine, norleucine, or tyrosine,
    • Xaa2 is isoleucine, leucine, norleucine, or valine,
    • Xaa3 is glycine, leucine, or valine,
    • Xaa5 is isoleucine, valine, leucine, or norleucine,
    • Xaa7 is lysine, serine, asparagine, alanine, arginine, or histidine,
    • Xaa10 is alanine, threonine, or leucine,
    • Xaa11 is threonine, or cysteine,
    • Xaa12 is glycine, leucine, or tryptophan,
    • Xaa15 is threonine, or alanine,
    • Xaa17 is isoleucine, leucine, or norleucine,
    • Xaa10 is isoleucine, leucine, or norleucine,
    • Xaa22 is asparagine or arginine, and
    • Xaa26 is glutamine or cysteine.


A most preferred melittin comprises the sequence: Xaa1-Xaa2-Gly-Ala-Xaa5-Leu-Lys-Val-Leu-Ala-Xaa11-Gly-Leu-Pro-Thr-Leu-Xaa17-Ser-Trp-Xaa20-Lys-Xaa22-Lys-Arg-Lys-Xaa26 wherein:

    • Xaa1, Xaa2, Xaa5, Xaa17 and Xaa20 are independently isoleucine, leucine, or norleucine,
    • Xaa11 is threonine or cysteine,
    • Xaa22 is Asparagine or arginine, and
    • Xaa26 is glutamine or cysteine.


A preferred masking agent comprises a neutral hydrophilic disubstituted alkylmaleic anhydride:




embedded image


wherein R1 comprises a cell targeting group. A preferred alkyl group is a methyl or ethyl group. A preferred targeting group comprises an asialoglycoprotein receptor ligand. An example of a substituted alkylmaleic anhydride consists of a 2-propionic-3-alkylmaleic anhydride derivative. A neutral hydrophilic 2-propionic-3-alkylmaleic anhydride derivative is formed by attachment of a neutral hydrophilic group to a 2-propionic-3-alkylmaleic anhydride through the 2-propionic-3-alkylmaleic anhydride γ-carboxyl group:




embedded image


wherein R1 comprises a neutral ASGPr ligand and n=0 or 1. In one embodiment, the ASGPr ligand is linked to the anhydride via a short PEG linker.


A preferred masking agent comprises a hydrophilic peptidase (protease) cleavable dipeptide-p-amidobenzyl amine reactive carbonate derivative. Enzyme cleavable linkers of the invention employ a dipeptide connected to an amidobenzyl activated carbonate moiety. The ASGPr ligand is attached to the amino terminus of a dipeptide. The amidobenzyl activated carbonate moiety is at the carboxy terminus of the dipeptide. Peptidease cleavable linkers suitable for use with the invention have the structure:




embedded image


wherein R4 comprises an ASGPr ligand and R3 comprises an amine reactive carbonate moiety, and R1 and R2 are amino acid R groups. A preferred activated carbonate is a para-nitrophenol. However, other amine reactive carbonates known in the art are readily substituted for the para-nitrophenol. Reaction of the activated carbonate with a melittin amine connects the targeting compound, the asialoglycoprotein receptor ligand, to the melittin peptide via a peptidase cleavable dipeptide-amidobenzyl carbamate linkage. Enzyme cleavage of the dipeptide removes the targeting ligand from the peptide and triggers an elimination reaction which results in regeneration of the peptide amine.


Dipeptides Glu-Gly, Ala-Cit, Phe-Cit (“Cit” is the amino acid citrulline) are shown in Example 3. While charged amino acids also permissible, neutral amino acids are preferred.


A preferred masking agent provides targeting function through affinity for cell surface receptors, i.e. the masking agent contains a ligand for a cell surface receptor. Preferred masking agents contain saccharides having affinity for the ASGPr, including but not limited to: galactose, N-Acetyl-galactosamine and galactose derivatives. Galactose derivatives having affinity for the ASGPr are well known in the art. An essential feature of the reversibly modified melittin is that more than 80% of the melittin amines (in a population of peptide) are modified by attachment of ASGPr ligands via physiologically labile, reversible covalent linkages.


In another embodiment, the melittin peptides of the invention are further modified, at the amino or carboxyl termini, by covalent attachment of a steric stabilizer or an ASGPr ligand-steric stabilizer conjugate. The amino or carboxy terminal modifications may be linked to the peptide during synthesis using methods standard in the art. Alternatively, the amino or carboxy terminal modifications may be done through modification of cysteine residues on melittin peptide having amino or carboxy terminal cysteine residues. A preferred steric stabilizer is a polyethylene glycol. Preferred polyethylene glycols have 1-120 ethylene units. In another embodiment, preferred polyethylene glycols are less than 5 kDa in size. For ASGPr ligand-steric stabilizer conjugates, a preferred steric stabilizer is a polyethyleneglycol having 1-24 ethylene units.


The RNAi polynucleotide conjugate and delivery peptide are administered to a mammal in pharmaceutically acceptable carriers or diluents. In one embodiment, the delivery peptide and the RNAi polynucleotide conjugate may be combined in a solution prior to administration to the mammal. In another embodiment, the delivery peptide and the RNAi polynucleotide conjugate may be co-administered to the mammal in separate solutions. In yet another embodiment, the delivery peptide and the RNAi polynucleotide conjugate may be administered to the mammal sequentially. For sequential administration, the delivery peptide may be administered prior to administration of the RNAi polynucleotide conjugate. Alternatively, for sequential administration, the RNAi polynucleotide conjugate may be administered prior to administration of the delivery peptide.


Further objects, features, and advantages of the invention will be apparent from the following detailed description when taken in conjunction with the accompanying drawings.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 Table listing melittin peptides suitable for use in the invention.



FIG. 2. Drawing illustrating linkage of GalNAc cluster to RNA.



FIG. 3. Graph illustrating (A) blood urea nitrogen (BUN) levels and (B) creatinine levels in primates treated with reversibly modified melittin siRNA delivery peptides and siRNA-cholesterol conjugates.



FIG. 4. Graph illustrating (A) aspartate aminotransferase (AST) levels and (B) alanine transaminase (ALT) levels in primates treated with reversibly modified melittin siRNA delivery peptides and siRNA-cholesterol conjugates.



FIG. 5. Graph illustrating knockdown of endogenous Factor VII levels in primates treated with reversibly modified melittin siRNA delivery peptides and siRNA-cholesterol conjugates.





DETAILED DESCRIPTION OF THE INVENTION

Described herein is an improved method for delivering RNA interference (RNAi) polynucleotides to liver cells in a mammal in vivo. We describe an in vivo RNAi polynucleotide delivery system employing a small delivery peptide, melittin, derived from bee venom peptide and an independently targeting RNAi polynucleotide. By using liver targeted RNAi polynucleotide conjugate molecules and asialoglycoprotein receptor targeted reversibly inhibited melittin peptides, efficient RNAi polynucleotide delivery to liver is observed.


Because the melittin and RNAi polynucleotide are independently targeted to hepatocytes, the concentration of the melittin and polynucleotides and the ratio between them is limited only by the solubility of the components rather than the solubility of the associated complex or ability to manufacture the complex. Also, the polynucleotide and melittin may be mixed at anytime prior to administration, or even administered separately, thus allowing the components to be stored separately, either in solution or dry.


The invention includes conjugate delivery systems of the composition:





Y-Melittin-(L-M)xplus N-T,


wherein N is a RNAi polynucleotide, T is a polynucleotide targeting moiety (either a hydrophobic group having 20 or more carbon atoms or a galactose cluster), Melittin is a bee venom melittin peptide or a derivative as describe herein, and masking agent M contains an ASGPr ligand as described herein covalently linked to Melittin via a physiologically labile reversible linkage L. Cleavage of L restores an unmodified amine on Melittin. Y is optional and if present comprises a polyethyleneglycol (PEG) or a ASGPr ligand-PEG conjugate linked to the amino terminus, the carboxy terminus, or an amino or carboxy terminal cysteine of Melittin. Attachment of Y to the amino terminus or an amino terminal cysteine is preferred. x is an integer greater than 1. In its unmodified state, Melittin is membrane active. However, delivery peptide Melittin-(L-M)x is not membrane active. Reversible modification of Melittin primary amines, by attachment of M reversibly inhibits or inactivates membrane activity of Melittin. Sufficient percentage of Melittin primary amines are modified to inhibit membrane activity of the polymer and provide for hepatocyte targeting. Preferably x has a value greater than 80%, and more preferably greater than 90%, of the primary amines on Melittin, as determined by the quantity of amines on Melittin in the absence of any masking agents. More specifically, x has a value greater than 80% and up to 100% of the primary amines on Melittin. It is noted that melittin typically contains 3-5 primary amines (the amino terminus (if unmodified) and typically 2-4 Lysine residues). Therefore, modification of a percentage of amines is meant to reflect the modification of a percentage on amines in a population of melittin peptides. Upon cleavage of reversible linkages L, unmodified amines are restored thereby reverting Melittin to its unmodified, membrane active state. A preferred reversible linkage is a pH labile linkage. Another preferred reversible linkage is a protease cleavable linkage. Melittin-(L-M)x, an ASGPr-targeted reversibly masked membrane active polymer (delivery peptide), and T-N, a polynucleotide-conjugate, are synthesized or manufactured separately. Neither T nor N are covalently linked directly or indirectly to Melittin, L, or M. Electrostatic or hydrophobic association of the polynucleotide or the polynucleotide-conjugate with the masked or unmasked polymer is not required for in vivo liver delivery of the polynucleotide. The masked polymer and the polynucleotide conjugate can be supplied in the same container or in separate containers. They may be combined prior to administration, co-administered, or administered sequentially.


Hydrophilic groups indicate in qualitative terms that the chemical moiety is water-preferring. Typically, such chemical groups are water soluble, and are hydrogen bond donors or acceptors with water. A hydrophilic group can be charged or uncharged. Charged groups can be positively charged (anionic) or negatively charged (cationic) or both (zwitterionic). Examples of hydrophilic groups include the following chemical moieties: carbohydrates, polyoxyethylene, certain peptides, oligonucleotides, amines, amides, alkoxy amides, carboxylic acids, sulfurs, and hydroxyls.


Hydrophobic groups indicate in qualitative terms that the chemical moiety is water-avoiding. Typically, such chemical groups are not water soluble, and tend not to form hydrogen bonds. Lipophilic groups dissolve in fats, oils, lipids, and non-polar solvents and have little to no capacity to form hydrogen bonds. Hydrocarbons containing two (2) or more carbon atoms, certain substituted hydrocarbons, cholesterol, and cholesterol derivatives are examples of hydrophobic groups and compounds.


Hydrophobic groups are preferably hydrocarbons, containing only carbon and hydrogen atoms. However, non-polar substitutions or non-polar heteroatoms which maintain hydrophobicity, and include, for example fluorine, may be permitted. The term includes aliphatic groups, aromatic groups, acyl groups, alkyl groups, alkenyl groups, alkynyl groups, aryl groups, aralkyl groups, aralkenyl groups, and aralkynyl groups, each of which may be linear, branched, or cyclic. The term hydrophobic group also includes: sterols, steroids, cholesterol, and steroid and cholesterol derivatives.


As used herein, membrane active peptides are surface active, amphipathic peptides that are able to induce one or more of the following effects upon a biological membrane: an alteration or disruption of the membrane that allows non-membrane permeable molecules to enter a cell or cross the membrane, pore formation in the membrane, fission of membranes, or disruption or dissolving of the membrane. As used herein, a membrane, or cell membrane, comprises a lipid bilayer. The alteration or disruption of the membrane can be functionally defined by the peptide's activity in at least one the following assays: red blood cell lysis (hemolysis), liposome leakage, liposome fusion, cell fusion, cell lysis, and endosomal release. Membrane active peptides that can cause lysis of cell membranes are also termed membrane lytic peptides. Peptides that preferentially cause disruption of endosomes or lysosomes over plasma membranes are considered endosomolytic. The effect of membrane active peptides on a cell membrane may be transient. Membrane active peptides possess affinity for the membrane and cause a denaturation or deformation of bilayer structures.


Delivery of a polynucleotide to a cell is mediated by the melittin peptide disrupting or destabilizing the plasma membrane or an internal vesicle membrane (such as an endosome or lysosome), including forming a pore in the membrane, or disrupting endosomal or lysosomal vesicles thereby permitting release of the contents of the vesicle into the cell cytoplasm.


Endosomolytic peptides are peptides that, in response to an endosomal-specific environmental factors, such as reduced pH or the presence of lytic enzymes (proteases), are able to cause disruption or lysis of an endosome or provide for release of a normally cell membrane impermeable compound, such as a polynucleotide or protein, from a cellular internal membrane-enclosed vesicle, such as an endosome or lysosome. Endosomolytic polymers undergo a shift in their physico-chemical properties in the endosome. This shift can be a change in the polymer's solubility or ability to interact with other compounds or membranes as a result in a shift in charge, hydrophobicity, or hydrophilicity. Exemplary endosomolytic peptides have pH-labile or enzymatic-sensitive groups or bonds. A reversibly masked membrane active peptide, wherein the masking agents are attached to the polymer via pH labile bonds, can therefore be considered to be an endosomolytic polymer.


Melittin, as used herein, is a small amphipathic membrane active peptide, comprising about 23 to about 32 amino acids, derived from the naturally occurring in bee venom peptide melittin. The naturally occurring melittin contains 26 amino acids and is predominantly hydrophobic on the amino terminal end and predominantly hydrophilic (cationic) on the carboxy terminal end. Melittin of the invention can be isolated from a biological source or it can be synthetic. A synthetic polymer is formulated or manufactured by a chemical process “by man” and is not created by a naturally occurring biological process. As used herein, melittin encompasses the naturally occurring bee venom peptides of the melittin family that can be found in, for example, venom of the species: Apis florea, Apis mellifera, Apis cerana, Apis dorsata, Vespula maculifrons, Vespa magnifica, Vespa velutina, Polistes sp. HQL-2001, and Polistes hebraeus. As used herein, melittin also encompasses synthetic peptides having amino acid sequence identical to or similar to naturally occurring melittin peptides. Specifically, melittin amino acid sequence encompass those shown in FIG. 1. In addition to the amino acids which retain melittin's inherent high membrane activity, 1-8 amino acids can be added to the amino or carboxy terminal ends of the peptide. Specifically, cysteine residues can be added to the amino or carboxy termini. The list in FIG. 1 is not meant to be exhaustive, as other conservative amino acid substitutions are readily envisioned. Synthetic melittin peptides can contain naturally occurring L form amino acids or the enantiomeric D form amino acids (inverso). However, a melittin peptide should either contain essentially all L form or all D form amino acids but may have amino acids of the opposite stereocenter appended at either the amino or carboxy termini. The melittin amino acid sequence can also be reversed (retro). Retro melittin can have L form amino acids or D form amino acids (retroinverso). Two melittin peptides can also be covalently linked to form a melittin dimer. Melittin can have modifying groups, other that masking agents, that enhance tissue targeting or facilitate in vivo circulation attached to either the amino terminal or carboxy terminal ends of the peptide. However, as used herein, melittin does not include chains or polymers containing more than two melittin peptides covalently linked to one another other or to another polymer or scaffold.


Masking

The melittin peptides of the invention comprise reversibly modified melittin peptides wherein reversible modification inhibits membrane activity, neutralizes the melittin to reduce positive charge and form a near neutral charge polymer, and provides cell-type specific targeting. The melittin is reversibly modified through reversible modification of primary amines on the peptide.


The melittin peptides of the invention are capable of disrupting plasma membranes or lysosomal/endocytic membranes. Membrane activity, however, leads to toxicity when the peptide is administered in vivo. Therefore, reversible masking of membrane activity of melittin is necessary for in vivo use. This masking is accomplished through reversible attachment of masking agents to melittin to form a reversibly masked melittin, i.e. a delivery peptide. In addition to inhibiting membrane activity, the masking agents provide cell-specific interactions, i.e. targeting.


It is an essential feature of the masking agents that, in aggregate, they inhibit membrane activity of the polymer and provide in vivo hepatocyte targeting. Melittin is membrane active in the unmodified (unmasked) state and not membrane active (inactivated) in the modified (masked) state. A sufficient number of masking agents are linked to the peptide to achieve the desired level of inactivation. The desired level of modification of melittin by attachment of masking agent(s) is readily determined using appropriate peptide activity assays. For example, if melittin possesses membrane activity in a given assay, a sufficient level of masking agent is linked to the peptide to achieve the desired level of inhibition of membrane activity in that assay. Modification of ≧80% or ≧90% of the primary amine groups on a population of melittin peptides, as determined by the quantity of primary amines on the peptides in the absence of any masking agents, is preferred. It is also a preferred characteristic of masking agents that their attachment to the peptide reduces positive charge of the polymer, thus forming a more neutral delivery peptide. It is desirable that the masked peptide retain aqueous solubility.


As used herein, melittin is masked if the modified peptide does not exhibit membrane activity and exhibits cell-specific (i.e. hepatocyte) targeting in vivo. Melittin is reversibly masked if cleavage of bonds linking the masking agents to the peptide results in restoration of amines on the peptide thereby restoring membrane activity.


It is another essential feature that the masking agents are covalently bound to melittin through physiologically labile reversible bonds. By using physiologically labile reversible linkages or bonds, the masking agents can be cleaved from the peptide in vivo, thereby unmasking the peptide and restoring activity of the unmasked peptide. By choosing an appropriate reversible linkage, it is possible to form a conjugate that restores activity of melittin after it has been delivered or targeted to a desired cell type or cellular location. Reversibility of the linkages provides for selective activation of melittin. Reversible covalent linkages contain reversible or labile bonds which may be selected from the group comprising: physiologically labile bonds, cellular physiologically labile bonds, pH labile bonds, very pH labile bonds, extremely pH labile bonds, and proetease cleavable bonds.


As used herein, a masking agent comprises a preferrably neutral (uncharged) compound having an ASGPr ligand and an amine-reactive group wherein reaction of the amine-reactive group with an amine on a peptide results in linkage of the ASGPr ligand to the peptide via a reversible physiologically labile covalent bond. Amine reactive groups are chosen such the cleavage in response to an appropriate physiological condition (e.g., reduced pH such as in an endosome/lysosome, or enzymatic cleavage such as in an endosome/lysosome) results in regeneration of the melittin amine. An ASGPr ligand is a group, typically a saccharide, having affinity for the asialoglycoprotein receptor. Preferred masking agents of the invention are able to modify the polymer (form a reversible bond with the polymer) in aqueous solution.


A preferred amine-reactive group comprises a disubstituted maleic anhydride. A preferred masking agent is represented by the structure:




embedded image


wherein in which R1 comprises an asialoglycoprotein receptor (ASGPr) ligand and R2 is an alkyl group such as a methyl (—CH3) group, ethyl (—CH2CH3) group, or propyl (—CH2CH2CH3) group.


In some embodiments, the galactose ligand is linked to the amine-reactive group through a PEG linker as illustrated by the structure:




embedded image


wherein n is an integer between 1 and 19.


Another preferred amine-reactive group comprises a dipeptide-amidobenzyl amine reactive carbonate derivative represented by the structure:




embedded image


wherein:

    • R1 is the R group of amino acid 1,
    • R2 is the R group of amino acid 2,
    • R3 is —CH2—O—C(O)—O—Z, wherein Z is halide,




embedded image




    • and R4 comprises the ASGPr ligand.





Reaction of the activated carbonate with a melittin amine connects the ASGPr ligand to the melittin peptide via a peptidase cleavable dipeptide-amidobenzyl carbamate linkage.




embedded image


Enzymatic cleavage of the dipeptide removes the targeting ligand from the peptide and triggers an elimination reaction which results in regeneration of the peptide amine. While the structure above shows a single masking agent linked to a melittin peptide, in practice, several masking agents are linked to the melittin peptide; preferably such that more than 80% of the amines on a population of melittin peptides are modified.


Dipeptides Glu-Gly, Ala-Cit, Phe-Cit (“Cit” is the amino acid citrulline) are shown in Example 3. With respect to the above structure, Glu-Gly, Ala-Cit, Phe-Cit represent R2-R1. While charged amino acids are permissible, neutral amino acids are preferred. Other amino acid combinations are possible, provided they are cleaved by an endogenous protease. In addition, 3-5 amino acids may be used as the linker between the amido benzyl group and the targeting ligand.


As with maleic anhydride-based masking agents, the ASGPr ligand can be linked to the peptidase cleavable dipeptide-amidobenzyl carbonate via a PEG linker.


The membrane active polyamine can be conjugated to masking agents in the presence of an excess of masking agents. The excess masking agent may be removed from the conjugated delivery peptide prior to administration of the delivery peptide.


In another embodiment, the melittin peptides of the invention are further modified, at the amino or carboxyl termini, by covalent attachment of a steric stabilizer or an ASGPr ligand-steric stabilizer conjugate. Modification of the hydrophobic terminal end is preferred; the amino terminal end for melittin having “normal sequence” and the carboxyl terminal end for retro-melittin. A preferred steric stabilizer is a polyethylene glycol. The amino or carboxy terminal modifications may be linked to the peptide during synthesis using methods standard in the art. Alternatively, the amino or carboxy terminal modifications may be done through modification of cysteine residues on melittin peptides having amino or carboxy terminal cysteine residues. Preferred polyethylene glycols have 1-120 ethylene units. In another embodiment, preferred polyethylene glycols are less than 5 kDa in size. For ASGPr ligand-steric stabilizer conjugates (NAG-PEG modification), a preferred steric stabilizer is a polyethyleneglycol having 1-24 ethylene units. Terminal PEG modification, when combined with reversible masking, further reduces toxicity of the melittin delivery peptide. Terminal NAG-PEG modification enhances efficacy.


Steric Stabilizer

As used herein, a steric stabilizer is a non-ionic hydrophilic polymer (either natural, synthetic, or non-natural) that prevents or inhibits intramolecular or intermolecular interactions of a molecule to which it is attached relative to the molecule containing no steric stabilizer. A steric stabilizer hinders a molecule to which it is attached from engaging in electrostatic interactions. Electrostatic interaction is the non-covalent association of two or more substances due to attractive forces between positive and negative charges. Steric stabilizers can inhibit interaction with blood components and therefore opsonization, phagocytosis, and uptake by the reticuloendothelial system. Steric stabilizers can thus increase circulation time of molecules to which they are attached. Steric stabilizers can also inhibit aggregation of a molecule. A preferred steric stabilizer is a polyethylene glycol (PEG) or PEG derivative. PEG molecules suitable for the invention have about 1-120 ethylene glycol monomers.


ASGPr Ligand

Targeting moieties or groups enhance the pharmacokinetic or biodistribution properties of a conjugate to which they are attached to improve cell-specific distribution and cell-specific uptake of the conjugate. Galactose and galactose derivates have been used to target molecules to hepatocytes in vivo through their binding to the asialoglycoprotein receptor (ASGPr) expressed on the surface of hepatocytes. As used herein, a ASGPr ligand (or ASGPr ligand) comprises a galactose and galactose derivative having affinity for the ASGPr equal to or greater than that of galactose. Binding of galactose targeting moieties to the ASGPr(s) facilitates cell-specific targeting of the delivery peptide to hepatocytes and endocytosis of the delivery peptide into hepatocytes.


ASGPr ligands may be selected from the group comprising: lactose, galactose, N-acetylgalactosamine (GalNAc), galactosamine, N-formylgalactosamine, N-acetyl-galactosamine, N-propionylgalactosamine, N-n-butanoylgalactosamine, and N-iso-butanoyl-galactosamine (Iobst, S. T. and Drickamer, K. J.B.C. 1996, 271, 6686). ASGPr ligands can be monomeric (e.g., having a single galactosamine) or multimeric (e.g., having multiple galactosamines).


In one embodiment, the melittin peptide is reversibly masked by attachment of ASGPr ligand masking agents to ≧80% or ≧90% of primary amines on the peptide.


Labile Linkage

A linkage or linker is a connection between two atoms that links one chemical group or segment of interest to another chemical group or segment of interest via one or more covalent bonds. For example, a linkage can connect a masking agent to a peptide. Formation of a linkage may connect two separate molecules into a single molecule or it may connect two atoms in the same molecule. The linkage may be charge neutral or may bear a positive or negative charge. A reversible or labile linkage contains a reversible or labile bond. A linkage may optionally include a spacer that increases the distance between the two joined atoms. A spacer may further add flexibility and/or length to the linkage. Spacers may include, but are not be limited to, alkyl groups, alkenyl groups, alkynyl groups, aryl groups, aralkyl groups, aralkenyl groups, aralkynyl groups; each of which can contain one or more heteroatoms, heterocycles, amino acids, nucleotides, and saccharides. Spacer groups are well known in the art and the preceding list is not meant to limit the scope of the invention.


A labile bond is a covalent bond other than a covalent bond to a hydrogen atom that is capable of being selectively broken or cleaved under conditions that will not break or cleave other covalent bonds in the same molecule. More specifically, labile bond is a covalent bond that is less stable (thermodynamically) or more rapidly broken (kinetically) under appropriate conditions than other non-labile covalent bonds in the same molecule. Cleavage of a labile bond within a molecule may result in the formation of two molecules. For those skilled in the art, cleavage or lability of a bond is generally discussed in terms of half-life (t1/2) of bond cleavage (the time required for half of the bonds to cleave). Thus, labile bonds encompass bonds that can be selectively cleaved more rapidly than other bonds a molecule.


Appropriate conditions are determined by the type of labile bond and are well known in organic chemistry. A labile bond can be sensitive to pH, oxidative or reductive conditions or agents, temperature, salt concentration, the presence of an enzyme (such as esterases, including nucleases, and proteases), or the presence of an added agent. For example, increased or decreased pH is the appropriate conditions for a pH-labile bond.


The rate at which a labile group will undergo transformation can be controlled by altering the chemical constituents of the molecule containing the labile group. For example, addition of particular chemical moieties (e.g., electron acceptors or donors) near the labile group can affect the particular conditions (e.g., pH) under which chemical transformation will occur.


As used herein, a physiologically labile bond is a labile bond that is cleavable under conditions normally encountered or analogous to those encountered within a mammalian body. Physiologically labile linkage groups are selected such that they undergo a chemical transformation (e.g., cleavage) when present in certain physiological conditions.


As used herein, a cellular physiologically labile bond is a labile bond that is cleavable under mammalian intracellular conditions. Mammalian intracellular conditions include chemical conditions such as pH, temperature, oxidative or reductive conditions or agents, and salt concentration found in or analogous to those encountered in mammalian cells. Mammalian intracellular conditions also include the presence of enzymatic activity normally present in a mammalian cell such as from proteolytic or hydrolytic enzymes. A cellular physiologically labile bond may also be cleaved in response to administration of a pharmaceutically acceptable exogenous agent. Physiologically labile bonds that are cleaved under appropriate conditions with a half life of less than 45 min. are considered very labile. Physiologically labile bonds that are cleaved under appropriate conditions with a half life of less than 15 min are considered extremely labile.


Chemical transformation (cleavage of the labile bond) may be initiated by the addition of a pharmaceutically acceptable agent to the cell or may occur spontaneously when a molecule containing the labile bond reaches an appropriate intra- and/or extra-cellular environment. For example, a pH labile bond may be cleaved when the molecule enters an acidified endosome. Thus, a pH labile bond may be considered to be an endosomal cleavable bond. Enzyme cleavable bonds may be cleaved when exposed to enzymes such as those present in an endosome or lysosome or in the cytoplasm. A disulfide bond may be cleaved when the molecule enters the more reducing environment of the cell cytoplasm. Thus, a disulfide may be considered to be a cytoplasmic cleavable bond.


As used herein, a pH-labile bond is a labile bond that is selectively broken under acidic conditions (pH<7). Such bonds may also be termed endosomally labile bonds, since cell endosomes and lysosomes have a pH less than 7. The term pH-labile includes bonds that are pH-labile, very pH-labile, and extremely pH-labile.


Reaction of an anhydride with an amine forms an amide and an acid. For many anhydrides, the reverse reaction (formation of an anhydride and amine) is very slow and energetically unfavorable. However, if the anhydride is a cyclic anhydride, reaction with an amine yields an amide acid, a molecule in which the amide and the acid are in the same molecule. The presence of both reactive groups (the amide and the carboxylic acid) in the same molecule accelerates the reverse reaction. In particular, the product of primary amines with maleic anhydride and maleic anhydride derivatives, maleamic acids, revert back to amine and anhydride 1×109 to 1×1013 times faster than its noncyclic analogues (Kirby 1980).


Reaction of an Amine with an Anhydride to Form an Amide and an Acid



embedded image


Reaction of an Amine with a Cyclic Anhydride to Form an Amide Acid



embedded image


Cleavage of the amide acid to form an amine and an anhydride is pH-dependent and is greatly accelerated at acidic pH. This pH-dependent reactivity can be exploited to form reversible pH-labile bonds and linkers. Cis-aconitic acid has been used as such a pH-sensitive linker molecule. The γ-carboxylate is first coupled to a molecule. In a second step, either the α or β carboxylate is coupled to a second molecule to form a pH-sensitive coupling of the two molecules. The half life for cleavage of this linker at pH 5 is between 8 and 24 h.


Structures of Cis-Aconitic Anhydride and Maleic Anhydride



embedded image


The pH at which cleavage occurs is controlled by the addition of chemical constituents to the labile moiety. The rate of conversion of maleamic acids to amines and maleic anhydrides is strongly dependent on substitution (R2 and R3) of the maleic anhydride system. When R2 is methyl, the rate of conversion is 50-fold higher than when R2 and R3 are hydrogen. When there are alkyl substitutions at both R2 and R3 (e.g., 2,3-dimethylmaleicanhydride) the rate increase is dramatic: 10,000-fold faster than non-substituted maleic anhydride. The maleamate bond formed from the modification of an amine with 2,3-dimethylmaleic anhydride is cleaved to restore the anhydride and amine with a half-life between 4 and 10 min at pH 5. It is anticipated that if R2 and R3 are groups larger than hydrogen, the rate of amide-acid conversion to amine and anhydride will be faster than if R2 and/or R3 are hydrogen.


Very pH-labile bond: A very pH-labile bond has a half-life for cleavage at pH 5 of less than 45 min. The construction of very pH-labile bonds is well-known in the chemical art.


Extremely pH-labile bonds: An extremely pH-labile bond has a half-life for cleavage at pH 5 of less than 15 min. The construction of extremely pH-labile bonds is well-known in the chemical art.


Disubstituted cyclic anhydrides are particularly useful for attachment of masking agents to melittin peptides of the invention. They provide physiologically pH-labile linkages, readily modify amines, and restore those amines upon cleavage in the reduced pH found in cellular endosomes and lysosome. Second, the α or β carboxylic acid group created upon reaction with an amine, appears to contribute only about 1/20th of the expected negative charge to the polymer (Rozema et al. Bioconjugate Chemistry 2003). Thus, modification of the peptide with the disubstituted maleic anhydrides effectively neutralizes the positive charge of the peptide rather than creates a peptide with high negative charge. Near neutral delivery peptides are preferred for in vivo delivery.


RNAi Polynucleotide Conjugate

We have found that conjugation of an RNAi polynucleotide to a polynucleotide targeting moiety, either a hydrophobic group or to a galactose cluster, and co-administration of the RNAi polynucleotide conjugate with the delivery peptide described above provides for efficient, functional delivery of the RNAi polynucleotide to liver cells, particularly hepatocytes, in vivo. By functional delivery, it is meant that the RNAi polynucleotide is delivered to the cell and has the expected biological activity, sequence-specific inhibition of gene expression. Many molecules, including polynucleotides, administered to the vasculature of a mammal are normally cleared from the body by the liver. Clearance of a polynucleotide by the liver wherein the polynucleotide is degraded or otherwise processed for removal from the body and wherein the polynucleotide does not cause sequence-specific inhibition of gene expression is not considered functional delivery.


The RNAi polynucleotide conjugate is formed by covalently linking the RNAi polynucleotide to the polynucleotide targeting moiety. The polynucleotide is synthesized or modified such that it contains a reactive group A. The targeting moiety is also synthesized or modified such that it contains a reactive group B. Reactive groups A and B are chosen such that they can be linked via a covalent linkage using methods known in the art.


The targeting moiety may be linked to the 3′ or the 5′ end of the RNAi polynucleotide. For siRNA polynucleotides, the targeting moiety may be linked to either the sense strand or the antisense strand, though the sense strand is preferred.


In one embodiment, the polynucleotide targeting moiety consists of a hydrophobic group More specifically, the polynucleotide targeting moiety consists of a hydrophobic group having at least 20 carbon atoms. Hydrophobic groups used as polynucleotide targeting moieties are herein referred to as hydrophobic targeting moieties. Exemplary suitable hydrophobic groups may be selected from the group comprising: cholesterol, dicholesterol, tocopherol, ditocopherol, didecyl, didodecyl, dioctadecyl, didodecyl, dioctadecyl, isoprenoid, and choleamide. Hydrophobic groups having 6 or fewer carbon atoms are not effective as polynucleotide targeting moieties, while hydrophobic groups having 8 to 18 carbon atoms provide increasing polynucleotide delivery with increasing size of the hydrophobic group (i.e. increasing number of carbon atoms). Attachment of a hydrophobic targeting moiety to an RNAi polynucleotide does not provide efficient functional in vivo delivery of the RNAi polynucleotide in the absence of co-administration of the delivery peptide. While siRNA-cholesterol conjugates have been reported by others to deliver siRNA (siRNA-cholesterol) to liver cells in vivo, in the absence of any additional delivery vehicle, high concentrations of siRNA are required and delivery efficacy is poor. When combined with the delivery peptides described herein, delivery of the polynucleotide is greatly improved. By providing the siRNA-cholesterol together with a delivery peptide of the invention, efficacy of siRNA-cholesterol is increased about 100 fold.


Hydrophobic groups useful as polynucleotide targeting moieties may be selected from the group consisting of: alkyl group, alkenyl group, alkynyl group, aryl group, aralkyl group, aralkenyl group, and aralkynyl group, each of which may be linear, branched, or cyclic, cholesterol, cholesterol derivative, sterol, steroid, and steroid derivative. Hydrophobic targeting moieties are preferably hydrocarbons, containing only carbon and hydrogen atoms.


However, substitutions or heteroatoms which maintain hydrophobicity, for example fluorine, may be permitted. The hydrophobic targeting moiety may be attached to the 3′ or 5′ end of the RNAi polynucleotide using methods known in the art. For RNAi polynucleotides having 2 strands, such as siRNA, the hydrophobic group may be attached to either strand.


In another embodiment, the polynucleotide targeting moiety comprises a galactose cluster (galactose cluster targeting moiety). As used herein, a galactose cluster comprises a molecule having two to four terminal galactose derivatives. As used herein, the term galactose derivative includes both galactose and derivatives of galactose having affinity for the asialoglycoprotein receptor equal to or greater than that of galactose. A terminal galactose derivative is attached to a molecule through its C-1 carbon. The asialoglycoprotein receptor (ASGPr) is unique to hepatocytes and binds branched galactose-terminal glycoproteins. A preferred galactose cluster has three terminal galactosamines or galactosamine derivatives each having affinity for the asialoglycoprotein receptor. A more preferred galactose cluster has three terminal N-acetyl-galactosamines. Other terms common in the art include tri-antennary galactose, tri-valent galactose and galactose trimer. It is known that tri-antennary galactose derivative clusters are bound to the ASGPr with greater affinity than bi-antennary or mono-antennary galactose derivative structures (Baenziger and Fiete, 1980, Cell, 22, 611-620; Connolly et al., 1982, J. Biol. Chem., 257, 939-945). Mulivalency is required to achieve nM affinity. The attachment of a single galactose derivative having affinity for the asialoglycoprotein receptor does not enable functional delivery of the RNAi polynucleotide to hepatocytes in vivo when co-administered with the delivery peptide.




embedded image


A galactose cluster contains three galactose derivatives each linked to a central branch point. The galactose derivatives are attached to the central branch point through the C-1 carbons of the saccharides. The galactose derivative is preferably linked to the branch point via linkers or spacers. A preferred spacer is a flexible hydrophilic spacer (U.S. Pat. No. 5,885,968; Biessen et al. J. Med. Chem. 1995 Vol. 39 p. 1538-1546). A preferred flexible hydrophilic spacer is a PEG spacer. A preferred PEG spacer is a PEG3 spacer. The branch point can be any small molecule which permits attachment of the three galactose derivatives and further permits attachment of the branch point to the RNAi polynucleotide. An exemplary branch point group is a di-lysine. A di-lysine molecule contains three amine groups through which three galactose derivatives may be attached and a carboxyl reactive group through which the di-lysine may be attached to the RNAi polynucleotide. Attachment of the branch point to the RNAi polynucleotide may occur through a linker or spacer. A preferred spacer is a flexible hydrophilic spacer. A preferred flexible hydrophilic spacer is a PEG spacer. A preferred PEG spacer is a PEG3 spacer (three ethylene units). The galactose cluster may be attached to the 3′ or 5′ end of the RNAi polynucleotide using methods known in the art. For RNAi polynucleotides having 2 strands, such as siRNA, the galactose cluster may be attached to either strand.


A preferred galactose derivative is an N-acetyl-galactosamine (GalNAc). Other saccharides having affinity for the asialoglycoprotein receptor may be selected from the list comprising: galactose, galactosamine, N-formylgalactosamine, N-acetylgalactosamine, N-propionyl-galactosamine, N-n-butanoylgalactosamine, and N-iso-butanoylgalactos-amine. The affinities of numerous galactose derivatives for the asialoglycoprotein receptor have been studied (see for example: Iobst, S. T. and Drickamer, K. J.B.C. 1996, 271, 6686) or are readily determined using methods typical in the art.




embedded image


One Embodiment of a Galactose Cluster



embedded image


Galactose Cluster with PEG Spacer Between Branch Point and Nucleic Acid

The term polynucleotide, or nucleic acid or polynucleic acid, is a term of art that refers to a polymer containing at least two nucleotides. Nucleotides are the monomeric units of polynucleotide polymers. Polynucleotides with less than 120 monomeric units are often called oligonucleotides. Natural nucleic acids have a deoxyribose- or ribose-phosphate backbone. A non-natural or synthetic polynucleotide is a polynucleotide that is polymerized in vitro or in a cell free system and contains the same or similar bases but may contain a backbone of a type other than the natural ribose or deoxyribose-phosphate backbone. Polynucleotides can be synthesized using any known technique in the art. Polynucleotide backbones known in the art include: PNAs (peptide nucleic acids), phosphorothioates, phosphorodiamidates, morpholinos, and other variants of the phosphate backbone of native nucleic acids. Bases include purines and pyrimidines, which further include the natural compounds adenine, thymine, guanine, cytosine, uracil, inosine, and natural analogs. Synthetic derivatives of purines and pyrimidines include, but are not limited to, modifications which place new reactive groups on the nucleotide such as, but not limited to, amines, alcohols, thiols, carboxylates, and alkylhalides. The term base encompasses any of the known base analogs of DNA and RNA. A polynucleotide may contain ribonucleotides, deoxyribonucleotides, synthetic nucleotides, or any suitable combination. Polynucleotides may be polymerized in vitro, they may be recombinant, contain chimeric sequences, or derivatives of these groups. A polynucleotide may include a terminal cap moiety at the 5′-end, the 3′-end, or both the 5′ and 3′ ends. The cap moiety can be, but is not limited to, an inverted deoxy abasic moiety, an inverted deoxy thymidine moiety, a thymidine moiety, or 3′ glyceryl modification.


An RNA interference (RNAi) polynucleotide is a molecule capable of inducing RNA interference through interaction with the RNA interference pathway machinery of mammalian cells to degrade or inhibit translation of messenger RNA (mRNA) transcripts of a transgene in a sequence specific manner. Two primary RNAi polynucleotides are small (or short) interfering RNAs (siRNAs) and micro RNAs (miRNAs). RNAi polynucleotides may be selected from the group comprising: siRNA, microRNA, double-strand RNA (dsRNA), short hairpin RNA (shRNA), and expression cassettes encoding RNA capable of inducing RNA interference. siRNA comprises a double stranded structure typically containing 15-50 base pairs and preferably 21-25 base pairs and having a nucleotide sequence identical (perfectly complementary) or nearly identical (partially complementary) to a coding sequence in an expressed target gene or RNA within the cell. An siRNA may have dinucleotide 3′ overhangs. An siRNA may be composed of two annealed polynucleotides or a single polynucleotide that forms a hairpin structure. An siRNA molecule of the invention comprises a sense region and an antisense region. In one embodiment, the siRNA of the conjugate is assembled from two oligonucleotide fragments wherein one fragment comprises the nucleotide sequence of the antisense strand of the siRNA molecule and a second fragment comprises nucleotide sequence of the sense region of the siRNA molecule. In another embodiment, the sense strand is connected to the antisense strand via a linker molecule, such as a polynucleotide linker or a non-nucleotide linker. MicroRNAs (miRNAs) are small noncoding RNA gene products about 22 nucleotides long that direct destruction or translational repression of their mRNA targets. If the complementarity between the miRNA and the target mRNA is partial, translation of the target mRNA is repressed. If complementarity is extensive, the target mRNA is cleaved. For miRNAs, the complex binds to target sites usually located in the 3′ UTR of mRNAs that typically share only partial homology with the miRNA. A “seed region”—a stretch of about seven (7) consecutive nucleotides on the 5′ end of the miRNA that forms perfect base pairing with its target—plays a key role in miRNA specificity. Binding of the RISC/miRNA complex to the mRNA can lead to either the repression of protein translation or cleavage and degradation of the mRNA. Recent data indicate that mRNA cleavage happens preferentially if there is perfect homology along the whole length of the miRNA and its target instead of showing perfect base-pairing only in the seed region (Pillai et al. 2007).


RNAi polynucleotide expression cassettes can be transcribed in the cell to produce small hairpin RNAs that can function as siRNA, separate sense and anti-sense strand linear siRNAs, or miRNA. RNA polymerase III transcribed DNAs contain promoters selected from the list comprising: U6 promoters, H1 promoters, and tRNA promoters. RNA polymerase II promoters include U1, U2, U4, and U5 promoters, snRNA promoters, microRNA promoters, and mRNA promoters.


Lists of known miRNA sequences can be found in databases maintained by research organizations such as Wellcome Trust Sanger Institute, Penn Center for Bioinformatics, Memorial Sloan Kettering Cancer Center, and European Molecule Biology Laboratory, among others. Known effective siRNA sequences and cognate binding sites are also well represented in the relevant literature. RNAi molecules are readily designed and produced by technologies known in the art. In addition, there are computational tools that increase the chance of finding effective and specific sequence motifs (Pei et al. 2006, Reynolds et al. 2004, Khvorova et al. 2003, Schwarz et al. 2003, Ui-Tei et al. 2004, Heale et al. 2005, Chalk et al. 2004, Amarzguioui et al. 2004).


The polynucleotides of the invention can be chemically modified. Non-limiting examples of such chemical modifications include: phosphorothioate internucleotide linkages, 2′-O-methyl ribonucleotides, 2′-deoxy-2′-fluoro ribonucleotides, 2′-deoxy ribonucleotides, “universal base” nucleotides, 5-C-methyl nucleotides, and inverted deoxyabasic residue incorporation. These chemical modifications, when used in various polynucleotide constructs, are shown to preserve polynucleotide activity in cells while at the same time increasing the serum stability of these compounds. Chemically modified siRNA can also minimize the possibility of activating interferon activity in humans.


In one embodiment, a chemically-modified RNAi polynucleotide of the invention comprises a duplex having two strands, one or both of which can be chemically-modified, wherein each strand is about 19 to about 29 nucleotides. In one embodiment, an RNAi polynucleotide of the invention comprises one or more modified nucleotides while maintaining the ability to mediate RNAi inside a cell or reconstituted in vitro system. An RNAi polynucleotide can be modified wherein the chemical modification comprises one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) of the nucleotides. An RNAi polynucleotide of the invention can comprise modified nucleotides as a percentage of the total number of nucleotides present in the RNAi polynucleotide. As such, an RNAi polynucleotide of the invention can generally comprise modified nucleotides from about 5 to about 100% of the nucleotide positions (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the nucleotide positions). The actual percentage of modified nucleotides present in a given RNAi polynucleotide depends on the total number of nucleotides present in the RNAi polynucleotide. If the RNAi polynucleotide is single stranded, the percent modification can be based upon the total number of nucleotides present in the single stranded RNAi polynucleotide. Likewise, if the RNAi polynucleotide is double stranded, the percent modification can be based upon the total number of nucleotides present in the sense strand, antisense strand, or both the sense and antisense strands. In addition, the actual percentage of modified nucleotides present in a given RNAi polynucleotide can also depend on the total number of purine and pyrimidine nucleotides present in the RNAi polynucleotide. For example, wherein all pyrimidine nucleotides and/or all purine nucleotides present in the RNAi polynucleotide are modified.


An RNAi polynucleotide modulates expression of RNA encoded by a gene. Because multiple genes can share some degree of sequence homology with each other, an RNAi polynucleotide can be designed to target a class of genes with sufficient sequence homology. Thus, an RNAi polynucleotide can contain a sequence that has complementarity to sequences that are shared amongst different gene targets or are unique for a specific gene target. Therefore, the RNAi polynucleotide can be designed to target conserved regions of an RNA sequence having homology between several genes thereby targeting several genes in a gene family (e.g., different gene isoforms, splice variants, mutant genes, etc.). In another embodiment, the RNAi polynucleotide can be designed to target a sequence that is unique to a specific RNA sequence of a single gene.


The term complementarity refers to the ability of a polynucleotide to form hydrogen bond(s) with another polynucleotide sequence by either traditional Watson-Crick or other non-traditional types. In reference to the polynucleotide molecules of the present invention, the binding free energy for a polynucleotide molecule with its target (effector binding site) or complementary sequence is sufficient to allow the relevant function of the polynucleotide to proceed, e.g., enzymatic mRNA cleavage or translation inhibition. Determination of binding free energies for nucleic acid molecules is well known in the art (Frier et al. 1986, Turner et al. 1987). A percent complementarity indicates the percentage of bases, in a contiguous strand, in a first polynucleotide molecule which can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second polynucleotide sequence (e.g., 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary). Perfectly complementary means that all the bases in a contiguous strand of a polynucleotide sequence will hydrogen bond with the same number of contiguous bases in a second polynucleotide sequence.


By inhibit, down-regulate, or knockdown gene expression, it is meant that the expression of the gene, as measured by the level of RNA transcribed from the gene or the level of polypeptide, protein or protein subunit translated from the RNA, is reduced below that observed in the absence of the blocking polynucleotide-conjugates of the invention. Inhibition, down-regulation, or knockdown of gene expression, with a polynucleotide delivered by the compositions of the invention, is preferably below that level observed in the presence of a control inactive nucleic acid, a nucleic acid with scrambled sequence or with inactivating mismatches, or in absence of conjugation of the polynucleotide to the masked polymer.


In Vivo Administration

In pharmacology and toxicology, a route of administration is the path by which a drug, fluid, poison, or other substance is brought into contact with the body. In general, methods of administering drugs and nucleic acids for treatment of a mammal are well known in the art and can be applied to administration of the compositions of the invention. The compounds of the present invention can be administered via any suitable route, most preferably parenterally, in a preparation appropriately tailored to that route. Thus, the compounds of the present invention can be administered by injection, for example, intravenously, intramuscularly, intracutaneously, subcutaneously, or intraperitoneally. Accordingly, the present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier or excipient.


Parenteral routes of administration include intravascular (intravenous, intraarterial), intramuscular, intraparenchymal, intradermal, subdermal, subcutaneous, intratumor, intraperitoneal, intrathecal, subdural, epidural, and intralymphatic injections that use a syringe and a needle or catheter. Intravascular herein means within a tubular structure called a vessel that is connected to a tissue or organ within the body. Within the cavity of the tubular structure, a bodily fluid flows to or from the body part. Examples of bodily fluid include blood, cerebrospinal fluid (CSF), lymphatic fluid, or bile. Examples of vessels include arteries, arterioles, capillaries, venules, sinusoids, veins, lymphatics, bile ducts, and ducts of the salivary or other exocrine glands. The intravascular route includes delivery through the blood vessels such as an artery or a vein. The blood circulatory system provides systemic spread of the pharmaceutical.


The described compositions are injected in pharmaceutically acceptable carrier solutions. Pharmaceutically acceptable refers to those properties and/or substances which are acceptable to the mammal from a pharmacological/toxicological point of view. The phrase pharmaceutically acceptable refers to molecular entities, compositions, and properties that are physiologically tolerable and do not typically produce an allergic or other untoward or toxic reaction when administered to a mammal. Preferably, as used herein, the term pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals and more particularly in humans.


The RNAi polynucleotide-targeting moiety conjugate is co-administered with the delivery peptide. By co-administered it is meant that the RNAi polynucleotide and the delivery peptide are administered to the mammal such that both are present in the mammal at the same time. The RNAi polynucleotide-targeting moiety conjugate and the delivery peptide may be administered simultaneously or they may be delivered sequentially. For simultaneous administration, they may be mixed prior to administration. For sequential administration, either the RNAi polynucleotide-targeting moiety conjugate or the delivery peptide may be administered first.


For RNAi polynucleotide-hydrophobic targeting moiety conjugates, the RNAi conjugate may be administered up to 30 minutes prior to administration of the delivery peptide. Also for RNAi polynucleotide-hydrophobic targeting moiety conjugates, the delivery peptide may be administered up to two hours prior to administration of the RNAi conjugate.


For RNAi polynucleotide-galactose cluster targeting moiety conjugates, the RNAi conjugate may be administered up to 15 minutes prior to administration of the delivery peptide. Also for RNAi polynucleotide-galactose cluster targeting moiety conjugates, the delivery peptide may be administered up to 15 minutes prior to administration of the RNAi conjugate.


Therapeutic Effect

RNAi polynucleotides may be delivered for research purposes or to produce a change in a cell that is therapeutic. In vivo delivery of RNAi polynucleotides is useful for research reagents and for a variety of therapeutic, diagnostic, target validation, genomic discovery, genetic engineering, and pharmacogenomic applications. We have disclosed RNAi polynucleotide delivery resulting in inhibition of endogenous gene expression in hepatocytes. Levels of a reporter (marker) gene expression measured following delivery of a polynucleotide indicate a reasonable expectation of similar levels of gene expression following delivery of other polynucleotides. Levels of treatment considered beneficial by a person having ordinary skill in the art differ from disease to disease. For example, Hemophilia A and B are caused by deficiencies of the X-linked clotting factors VIII and IX, respectively. Their clinical course is greatly influenced by the percentage of normal serum levels of factor VIII or IX: <2%, severe; 2-5%, moderate; and 5-30% mild. Thus, an increase from 1% to 2% of the normal level of circulating factor in severe patients can be considered beneficial. Levels greater than 6% prevent spontaneous bleeds but not those secondary to surgery or injury. Similarly, inhibition of a gene need not be 100% to provide a therapeutic benefit. A person having ordinary skill in the art of gene therapy would reasonably anticipate beneficial levels of expression of a gene specific for a disease based upon sufficient levels of marker gene results. In the hemophilia example, if marker genes were expressed to yield a protein at a level comparable in volume to 2% of the normal level of factor VIII, it can be reasonably expected that the gene coding for factor VIII would also be expressed at similar levels. Thus, reporter or marker genes serve as useful paradigms for expression of intracellular proteins in general.


The liver is one of the most important target tissues for gene therapy given its central role in metabolism (e.g., lipoprotein metabolism in various hypercholesterolemias) and the secretion of circulating proteins (e.g., clotting factors in hemophilia). In addition, acquired disorders such as chronic hepatitis (e.g. hepatitis B virus infection) and cirrhosis are common and are also potentially treated by polynucleotide-based liver therapies. A number of diseases or conditions which affect or are affected by the liver are potentially treated through knockdown (inhibition) of gene expression in the liver. Such liver diseases and conditions may be selected from the list comprising: liver cancers (including hepatocellular carcinoma, HCC), viral infections (including hepatitis), metabolic disorders, (including hyperlipidemia and diabetes), fibrosis, and acute liver injury.


The amount (dose) of delivery peptide and RNAi-polynucleotide-conjugate that is to be administered can be determined empirically. We have shown effective knockdown of gene expression using 0.1-10 mg/kg animal weight of siRNA-conjugate and 5-60 mg/kg animal weight delivery peptide. A preferred amount in mice is 0.25-2.5 mg/kg siRNA-conjugate and 10-40 mg/kg delivery peptide. More preferably, about 12.5-20 mg/kg delivery peptide is administered. The amount of RNAi polynucleotide-conjugate is easily increased because it is typically not toxic in larger doses.


As used herein, in vivo means that which takes place inside an organism and more specifically to a process performed in or on the living tissue of a whole, living multicellular organism (animal), such as a mammal, as opposed to a partial or dead one.


EXAMPLES
Example 1
Melittin Synthesis

All melittin peptides were made using peptide synthesis techniques standard in the art. Suitable melittin peptides can be all L-form amino acids, all D-form amino acids (inverso). Independently of L or D form, the melittin peptide sequence can be reversed (retro).


Example 2
Melittin Modification

Amino Terminal Modification of Melittin Derivatives.


Solutions of CKLK-Melittin (20 mg/ml), TCEP-HCl (28.7 mg/ml, 100 mM), and MES-Na (21.7 mg/ml, 100 mM) were prepared in dH2O. In a 20 ml scintillation vial, CKLK-Melittin (0.030 mmol, 5 ml) was reacted with 1.7 molar equivalents TCEP-HCl (0.051 mmol, 0.51 ml) and left to stir at room temperature for 30 min. MES-Na (2 ml) and Water (1.88 ml) were then added in amounts to yield final concentrations of 10 mg/ml Melittin and 20 mM MES-Na. The pH was checked and adjusted to pH 6.5-7. A solution of NAG-PEG2-Br (100 mg/ml) was prepared in dH2O. NAG-PEG2-Br (4.75 eq, 0.142 mmol, 0.61 ml) was added, and the solution was left to stir at room temperature for 48 h.


Alternatively, in a 20 ml scintillation vial, Cys-Melittin (0.006 mmol, 1 ml) was reacted with 1.7 molar equivalents TCEP-HCl (0.010 mmol, 100 μl) and left to stir at room temperature for 30 min. MES-Na (400 μl) and water (390 μl) were added in amounts to yield final concentrations of 10 mg/ml Melittin and 20 mM MES-Na. The pH was checked and adjusted to pH 6.5-7. A solution of NAG-PEG8-Maleimide (100 mg/ml) was prepared in dH2O. NAG-PEG8-Maleimide (2 eq, 0.012 mmol, 110 μl) was added, and the solution was left to stir at room temperature for 48 h.


Samples were purified on a Luna 10μ C18 100 Å 21.2×250 mm column. Buffer A: H2O 0.1% TFA and Buffer B: MeCN, 10% Isopropyl Alcohol, 0.1% TFA. Flow rate of 15 ml/min, 35% A to 62.5% B in 20 min.


Other amino terminal modifications were made using similar means. Carboxyl terminal modifications were made substituting melittin peptides having carboxyl terminal cysteines for melittins having amino terminal cysteines.


Compounds used to modified Cys-Melittin or Melittin-Cys:




embedded image




    • n is an integer from 1 to 120 (PEG molecular weight up to about 5 kDa)







embedded image


Peptides having acetyl, dimethyl, stearoyl, myristoyl, and PEG amino or carboxyl terminal modifications, but not terminal cysteine residues, were generated on resin during peptide synthesis using methods typical in the art.


Example 3
Masking Agents Synthesis

A. pH Labile Masking Agents: Steric Stabilizer CDM-PEG and Targeting Group CDM-NAG (N-Acetyl Galactosamine) Syntheses.


To a solution of CDM (300 mg, 0.16 mmol) in 50 mL methylene chloride was added oxalyl chloride (2 g, 10 wt. eq.) and dimethylformamide (5 μl). The reaction was allowed to proceed overnight, after which the excess oxalyl chloride and methylene chloride were removed by rotary evaporation to yield the CDM acid chloride. The acid chloride was dissolved in 1 mL of methylene chloride. To this solution was added 1.1 molar equivalents polyethylene glycol monomethyl ether (MW average 550) for CDM-PEG or (aminoethoxy)ethoxy-2-(acetylamino)-2-deoxy-β-D-galactopyranoside (i.e. amino bisethoxyl-ethyl NAG) for CDM-NAG, and pyridine (200 μl, 1.5 eq) in 10 mL of methylene chloride. The solution was then stirred 1.5 h. The solvent was then removed and the resulting solid was dissolved into 5 mL of water and purified using reverse-phase HPLC using a 0.1% TFA water/acetonitrile gradient.




embedded image


Generic Disubstituted Maleic Anhydride Masking Agent

R1 comprises a neutral ASGPr ligand. Preferably the Masking Agent in uncharged.




embedded image


R is a methyl or ethyl, and n is an integer from 2 to 100. Preferably, the PEG contains from 5 to 20 ethylene units (n is an integer from 5 to 20). More preferably, PEG contains 10-14 ethylene units (n is an integer from 10 to 14). The PEG may be of variable length and have a mean length of 5-20 or 10-14 ethylene units. Alternatively, the PEG may be monodisperse, uniform or discrete; having, for example, exactly 11 or 13 ethylene units.




embedded image


n is an integer from 1 to 10. As shown above, a PEG spacer may be positioned between the anhydride group and the ASGPr ligand. A preferred PEG spacer contains 1-10 ethylene units.


Alternatively an alkyl spacer may be used between the anhydride and the N-Acetylgalactosamine.




embedded image


n is a integer from 0 to 6.


Other spacers or linkers may be used bet between the anhydride and the N-Acetyl-galactosamine. However, a hydrophilic, neutral (preferably uncharged) spacer or linker is preferred)


B. Protease (Peptidase) Cleavable Masking Agents.


Melittin peptide can also be reversibly modified using specialized enzyme cleavable linkers. These enzyme cleavable linkers employ a dipeptide connected to an amidobenzyl activated carbonate moiety. Reaction of the activated carbonate with a peptide amine connects a targeting compound, such as asialoglycoprotein receptor ligand, to the melittin peptide via a peptidase cleavable dipeptide-amidobenzyl carbamate linkage. Enzyme cleavage of the dipeptide removes the targeting ligand from the peptide and triggers an elimination reaction which results in regeneration of the peptide amine. The following enzymatically cleavable linkers were synthesized:




embedded image


Dipeptides Glu-Gly, Ala-Cit, Phe-Cit are shown (“Cit” is the amino acid citrulline). Other amino acid combinations are permissible. In addition, 3-5 amino acids may be used as the linker between the amido benzyl group and the targeting ligand. Further, other activated carbonates known in the art are readily substituted for the para-nitrophenol used in the above compounds.


Example 4
Reversible Modification/Masking of Melittin

A. Modification with Maleic Anhydride-Based Masking Agents.


Prior to modification, 5× mg of disubstituted maleic anhydride masking agent (e.g. CDM-NAG) was lyophilized from a 0.1% aqueous solution of glacial acetic acid. To the dried disubstituted maleic anhydride masking agent was added a solution of ×mg melittin in 0.2×mL of isotonic glucose and 10×mg of HEPES free base. Following complete dissolution of anhydride, the solution was incubated for at least 30 min at RT prior to animal administration. Reaction of disubstituted maleic anhydride masking agent with the peptide yielded:




embedded image


wherein R is melittin and R1 comprises a ASGPr ligand (e.g. NAG). The anhydride carboxyl produced in the reaction between the anhydride and the polymer amine exhibits ˜ 1/20th of the expected charge (Rozema et al. Bioconjugate Chemistry 2003). Therefore, the membrane active polymer is effectively neutralized rather than being converted to a highly negatively charged polyanion.


In some embodiments, the masked Melittin peptide (MLP-(CDM-NAG)) was in a solution containing 125 mg Melittin, 500 mg dextran 1K, 3.18 mg sodium carbonate, 588 mg sodium bicarbonate in 5 ml water. In some embodiments, the MLP-(CDM-NAG) was lyophilized.


B. Modification with Protease Cleavable Masking Agents.


1×mg of peptide and 10×mg HEPES base at 1-10 mg/mL peptide was masked by addition of 2-6×mg of amine-reactive p-nitrophenyl carbonate or N-hydroxysuccinimide carbonate derivatives of the NAG-containing protease cleavable substrate. The solution was then incubated at least 1 h at room temperature (RT) before injection into animals.


Example 5
siRNAs

The siRNAs had the following sequences:









Factor VII-rodent


sense: 


(SEQ ID 97)


(Chol)-5′ GfcAfaAfgGfcGfuGfcCfaAfcUfcAf(invdT) 3′





antisense: 


(SEQ ID 98)


5′ pdTsGfaGfuUfgGfcAfcGfcCfuUfuGfcdTsdT 3′


or





sense


(SEQ ID 99)


5′ GGAUfCfAUfCfUfCfAAGUfCfUfUfACfdTsdT 3′





antisense


(SEQ ID 100)


5′ GUfAAGACfUfUfGAGAUfGAUfCfCfdTsdT 3′





Factor VII = primate


Sense


(SEQ ID 101)


(chol)-5′ uuAGGfuUfgGfuGfaAfuGfgAfgCfuCfaGf


(invdT) 3′





Antisense


(SEQ ID 102)


5′ pCfsUfgAfgCfuCfcAfuUfcAfcCfaAfcdTsdT 3′





ApoB siRNA:


sense


(SEQ ID 103)


(cholC6SSC6)-5′ GGAAUCuuAuAuuuGAUCcAsA 3′





antisense


(SEQ ID 104)


5′ uuGGAUcAAAuAuAAGAuUCcscsU 3′





siLUC


sense


(SEQ ID 105)


(chol)5′-uAuCfuUfaCfgCfuGfaGfuAfcUfuCfgAf


(invdT)-3′





antisense


(SEQ ID 106)


5′-UfcGfaAfgUfaCfuCfaGfcGfuAfaGfdTsdT-3′


lower case = 2′-O-CH3 substitution


s = phosphorothioate linkage


f after nucleotide = 2′-F substitution


d before nucleotide = 2′-deoxy






RNA synthesis was performed on solid phase by conventional phosphoramidite chemistry on an ÄKTA Oligopilot 100 (GE Healthcare, Freiburg, Germany) and controlled pore glass (CPG) as solid support.


Example 6
siRNA-Targeting Molecule Conjugates

A. Synthesis of GalNAc Cluster.


A GalNAc cluster polynucleotide targeting ligand was synthesized as described in US Patent Publication 20010207799.




embedded image


B. GalNAc Cluster-siRNA Conjugates.


The GalNAc cluster of Example 6A above was conjugated to siRNA as shown in FIG. 2 and as described below.


(1) Compound 1


(150 mg, 0.082 mmol, FIG. 2) was dissolved in dry methanol (5.5 ml) and 42 μL sodium methylate were added (25% solution in MeOH). The mixture was stirred under an argon atmosphere for 2 h at RT. An equal amount of methanol was added as well as portions of an anionic exchange material Amberlite IR-120 to generate a pH ˜7.0. The Amberlite was removed by filtration. The solution was dried with Na2SO4, and the solvent was removed under reduced pressure. Compound 2 was obtained in quantitative yield as a white foam. TLC (SiO2, dichloromethane (DCM)/MeOH 5:1+0.1% CH3COOH): Rf 2=0.03; for detection a solution of sulfuric acid (5%) in MeOH was used followed by heating. ESI-MS, direct injection, negative mode; [M−H]−1calculated: 1452.7; [M−H]1−measured: 1452.5.


(2) Compound 2


(20 mg, 0.014 mmol, FIG. 2) was co-evaporated with pyridine and dichloromethane. The residue was dissolved in dry DMF (0.9 ml) and a solution of N-Hydroxysuccinimide (NHS) in DMF (1.6 mg, 0.014 mmol) was added while stirring under an argon atmosphere. At 0° C. a solution of N,N′-Dicyclohexylcarbodiimide (DCC) in DMF (3.2 mg, 0.016 mmol) was slowly added. The reaction was allowed to warm to RT and stirred overnight. Compound 3 was used without further purification for conjugation to RNA.


(3) Synthesis of Amino-Modified RNA.


RNA equipped with a C-6-amino linker at the 5′-end of the sense strand was produced by standard phosphoramidite chemistry on solid phase at a scale of 1215 μmol using an ÄKTA Oligopilot 100 (GE Healthcare, Freiburg, Germany) and controlled pore glass as solid support. RNA containing 2′-O-methyl nucleotides were generated employing the corresponding phosphoramidites, 2′-O-methyl phosphoramidites and TFA-hexylaminolinker amidite. Cleavage and deprotection as well as purification was achieved by methods known in the field (Wincott F., et al, NAR 1995, 23, 14, 2677-84).


The amino-modified RNA was characterized by anion exchange HPLC (purity: 96.1%) and identity was confirmed by ESI-MS ([M+H]1+calculated: 6937.4; [M+H]1+measured: 6939.0. Sequence: 5′-(NH2C6)GGAAUCuuAuAuuuGAUCcAsA-3′ (SEQ ID 149); u,c: 2′-O-methyl nucleotides of corresponding bases, s: phosphorothioate.


(4) Conjugation of GalNAc Cluster to RNA.


RNA (2.54 μmol) equipped with a C-6 amino linker at the 5′-end was lyophilized and dissolved in 250 μL sodium borate buffer (0.1 mol/L sodium borate, pH 8.5, 0.1 mol/L KCl) and 1.1 mL DMSO. After addition of 8 μL N,N-Diisopropylethylamine (DIPEA), a solution of compound 3 (theoretically 0.014 mmol, FIG. 2) in DMF was slowly added under continuous stirring to the RNA solution. The reaction mixture was agitated at 35° C. overnight. The reaction was monitored using RP-HPLC (Resource RPC 3 ml, buffer: A: 100 mM Triethylammonium acetate (TEAA, 2.0 M, pH 7.0) in water, B: 100 mM TEAA in 95% acetonitrile, gradient: 5% B to 22% B in 20 CV). After precipitation of RNA using sodium acetate (3 M) in EtOH at −20° C., the RNA conjugate was purified using the conditions described above. The pure fractions were pooled, and the desired conjugate compound 4 was precipitated using sodium acetate/EtOH to give the pure RNA conjugate. Conjugate 4 has been isolated in 59% yield (1.50 μmol). The purity of conjugate 4 was analyzed by anion exchange HPLC (purity: 85.5%) and identity was confirmed by ESI-MS ([M+H]1+calculated: 8374.4; [M+H]1+measured: 8376.0.


(5) Conjugate 4 (Sense Strand) was Annealed with an 2′-O-Methyl-Modified Antisense Strand.


The siRNA conjugate was generated by mixing an equimolar solution of complementary strands in annealing buffer (20 mM sodium phosphate, pH 6.8; 100 mM sodium chloride), heated in a water bath at 85-90° C. for 3 min, and cooled to RT over a period of 3-4 h. Duplex formation was confirmed by native gel electrophoresis.


C. Hydrophobic Group-siRNA Conjugates.


(1) siRNA Conjugation to Alkyl Groups.


A 5′-C10-NHS ester modified sense strand of siRNA (NHSC10-siRNA, or COC9-siRNA) was prepared employing 5′-Carboxy-Modifier C10 amidite from Glen Research (Virginia, USA). The activated RNA, still attached to the solid support was used for conjugation with lipophilic amines listed in Table 1 below. 100 mg of the sense strand CPG (loading 60 μmol/g, 0.6 μmol RNA) were mixed with 0.25 mmol of the corresponding amine obtained from, Sigma Aldrich Chemie GmbH (Taufkirchen, Germany) or Fluka (Sigma-Aldrich, Buchs, Switzerland).









TABLE 1







Lipophilic amines used in forming hydrophobic group-siRNA conjugates











Nr
Lipophilic Amine
mg
mmol
solvent





2
N-Hexylamine
25
0.25
1 mL CH2Cl2


3
Dodecylamine
50
0.25
0.55 mL CH3CN, 0.45 mL CH2Cl2


4
Octadecylamine
67
0.25
1 mL CH2Cl2


5
Didecylamine
74
0.25
1 mL CH2Cl2


6
Didodecylamine
88
0.25
1 mL CH2Cl2


7
Dioctadecylamine
67
0.12
0.45 mL CH2Cl2, 0.45 mL






Cyclohexan









The mixture was shaken for 18 h at 40° C. The RNA was cleaved from the solid support and deprotected with an aqueous ammonium hydroxide solution (NH3, 33%) at 45° C. overnight. The 2′-protecting group was removed with TEA×3HF at 65° C. for 3.5 h. The crude oligoribonucleotides were purified by RP-HPLC (Resource RPC 3 ml, buffer: A: 100 mM TEAA in water, B: 100 mM TEAA in 95% CH3CN, gradient: 3% B to 70% B in 15 CV, except for Nr 7: gradient from 3% B to 100% B in 15 CV).


To generate siRNA from RNA single strand, equimolar amounts of complementary sense and antisense strands were mixed in annealing buffer (20 mM sodium phosphate, pH 6.8; 100 mM sodium chloride), heated at 80° C. for 3 min, and cooled to RT over a period of 3-4 h. The siRNA, which are directed against factor VII mRNA were characterized by gel electrophoresis.


(2) siRNA Conjugation to Cholesterol—


siRNA-cholesterol conjugates were synthesized using methods standard in the art. Cholesterol can be attached to the 5′ or 3′ termini of the sense or antisense strand of the siRNA. A preferred attachment is to the 5′ end of the sense strand of the siRNA. siRNA-Cholesterol can also be made post siRNA synthesis using RNA strands containing a reactive group (e.g. thiol, amine, or carboxyl) using methods standard in the art.


In Vivo siRNA Delivery
Example 7
Administration of RNAi Polynucleotides In Vivo, and Delivery to Hepatocytes

RNAi polynucleotide conjugates and masked melittin peptides were synthesized as described above. Six to eight week old mice (strain C57BL/6 or ICR, ˜18-20 g each) were obtained from Harlan Sprague Dawley (Indianapolis Ind.). Mice were housed at least 2 days prior to injection. Feeding was performed ad libitum with Harlan Teklad Rodent Diet (Harlan, Madison Wis.). Mice were injected with 0.2 mL solution of delivery peptide and 0.2 mL siRNA conjugates into the tail vein. For simultaneous injection of delivery peptide and siRNA, the siRNA-conjugate was added to modified peptide prior to injection and the entire amount was injected. The composition was soluble and nonaggregating in physiological conditions. Solutions were injected by infusion into the tail vein. Injection into other vessels, e.g. retro-orbital injection, are predicted to be equally effective.


Wistar Han rats, 175-200 g were obtained from Charles River (Wilmington, Mass.). Rats were housed at least 1 week prior to injection. Injection volume for rats was typically 1 ml.


Serum ApoB Levels Determination.


Mice were fasted for 4 h (16 h for rats) before serum collection by submandibular bleeding. For rats blood was collected from the jugular vein. Serum ApoB protein levels were determined by standard sandwich ELISA methods. Briefly, a polyclonal goat anti-mouse ApoB antibody and a rabbit anti-mouse ApoB antibody (Biodesign International) were used as capture and detection antibodies respectively. An HRP-conjugated goat anti-rabbit IgG antibody (Sigma) was applied afterwards to bind the ApoB/antibody complex. Absorbance of tetramethyl-benzidine (TMB, Sigma) colorimetric development was then measured by a Tecan Safire2 (Austria, Europe) microplate reader at 450 nm.


Plasma Factor VII (F7) Activity Measurements.


Plasma samples from animals were prepared by collecting blood (9 volumes) (by submandibular bleeding for mice or from jugular vein for rats) into microcentrifuge tubes containing 0.109 mol/L sodium citrate anticoagulant (1 volume) following standard procedures. F7 activity in plasma is measured with a chromogenic method using a BIOPHEN VII kit (Hyphen BioMed/Aniara, Mason, Ohio) following manufacturer's recommendations. Absorbance of colorimetric development was measured using a Tecan Safire2 microplate reader at 405 nm.


Example 8
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide—does Response of Melittin Peptide

Melittin was reversibly modified with CDM-NAG as described above. The indicated amount of melittin was then co-injected with the 200 μg ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above.









TABLE 2







Inhibition of ApoB activity in normal liver cells in mice treated with


ApoB-siRNA-cholesterol conjugate and CDM-NAG vs. CDM-PEG


reversibly inhibited Melittin peptide.













μg
μg
%


Peptide Name
Modification
siRNA
peptide
knockdowna















Apis florea

5× CDM-PEG
200
800
0


(SEQ ID 1)
5× CDM-NAG
200
100
25


L form

200
200
51




200
400
78




200
800
87




200
1200
94






aKnockdown relative to isotonic glucose injected animals







Example 9
In Vivo Knockdown of Endogenous Factor VII Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Rats

The indicated melittin was reversibly modified with 5× CDM-NAG as described above. The indicated amount of melittin, in mg per kg animal weight, was then co-injected with the 3 mg/kg cholesterol-Factor VII siRNA. Effect on Factor VII levels were determined as described above.









TABLE 3







Inhibition of Factor VII activity in normal liver


cells in rats treated with Factor VII-siRNA-


cholesterol conjugate and CDM-NAG reversibly


inhibited melittin.










SEQ

ug
Factor VII


ID
Peptide
peptidea
knockdownb





 1
GIGAILKVLATGLPTLISWIKNKRKQ
 1
30




 3
83




10
90




20
95





11
YIGAILKVLATGLPTLISWIKNKRKQ
 1
93




 3
97






amg peptide per kilogram animal weight




bKnockdown relative to isotonic glucose injected animals







Example 10
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, L-Form Vs. D-Form Melittin

Melittin was reversibly modified with CDM-NAG as described above. The indicated amount of melittin was then co-injected with 50 μg ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above.









TABLE 4







Inhibition of ApoB activity in normal liver cells in mice treated with


ApoB-siRNA cholesterol conjugate and the indicated CDM-NAG


reversibly inhibited melittin peptide.













μg
μg
%


Peptide Name
Modification
siRNA
peptide
knockdown














Leu-Melittin L form
5× CDM-NAG
50
25
15


(SEQ ID 7)

50
50
70




50
100
90




50
200
90




50
400
90


Leu-Melittin D form
5× CDM-NAG
50
25
30


(SEQ ID 150)

50
50
80




50
100
90









Example 11
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, Normal Vs. Reversed (Retro) Sequence

Melittin was reversibly modified with CDM-NAG (5×) as described above. The indicated amount of melittin was then co-injected with the indicated amount of ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above.









TABLE 5







Inhibition of ApoB activity in normal liver cells in mice treated with


ApoB-siRNA cholesterol conjugate and the indicated CDM-NAG reversibly


inhibited Melittin peptide.











SEQ



percent


ID
modification
Peptide
siRNA
knockdown





 1

GIGAILKVLATGLPTLISWIKNKRKQ
200 μg
90





400 μg
80





95
Retroinversoa
QQRKRKIWSILAALGTTLVKLVAGIC-NH2
 30 mg/kg
39



Methoxy








92
retroinverso
QQRKRKIWSILAPLGTTLVKLVAGIC-NH2
400 μg
85





 20 mg/kg
94





95
retroinverso
QQRKRKIWSILAALGTTLVKLVAGIC-NH2
 20 mg/kg
91





93
retroinverso
QQKKKKIWSILAPLGTTLVKLVAGIC-NH2
 20 mg/kg
70





96
retroinverso
QKRKNKIWSILTPLGTALVKLIAGIG-NH2
 20 mg/kg
70






a-retroinverso = normal melittin amino acid sequence is reversed and all amino acids are D-form amino acids (Glycine (G) is achiral)







Example 12
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, Melittin Modification Level

Melittin was reversibly modified with the indicated amount of CDM-NAG as described above. 50 μg melittin was then co-injected with the 100 μg ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above.


Percent melittin amine modification was determined by TNBS Assay for free amines on the peptide. 20 μg peptide was pipetted into 96 well clear plate (NUNC 96) containing 190 μL 50 mM BORAX buffer (pH 9) and 16 μg TNBS. Sample were allowed to react with TNBS for ˜15 minutes at RT and then the A420 is measured on a Safire plate reader. Calculate the % amines modified as follows: (Acontrol−Asample)/(Acontrol−Ablank)×100. Modification of more than 80% of amines provided optimal melittin masking and activity.









TABLE 6







Inhibition of ApoB activity in normal liver cells in mice treated with ApoB-siRNA


cholesterol conjugate and Melittin reversibly modified at the


indicated levels with CDM-NAG.














μg
μg
% amines
%


Peptide Name
Modification
siRNA
peptide
modifieda
knockdown





Leu-Melittin (SEQ ID 7)
1× CDM-NAG
100
50
68
74


L form
2× CDM-NAG
100
50
88
88



5× CDM-NAG
100
50
98
82






adetermined by TNBS assay







Example 13
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, Melittin Peptide Derivatives

Melittin peptides having the indicated sequence were reversibly modified with CDM-NAG (5×) as described above. The indicated amount of melittin was then co-injected with the indicated amount of ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above.









TABLE 7







Inhibition of ApoB activity in normal liver cells in mice treated with


ApoB-siRNA cholesterol conjugate and the indicated CDM-NAG reversibly


inhibited Melittin peptide.











μg
μg
percent


Peptide Name
peptidea
siRNAb
knockdown





CBZ-Mel (SEQ ID 108)
100
 80
96





Mel-NH2 (SEQ ID 116)
 50
100
86





Acetyl-dMel-NH2 (SEQ ID 107)
100
100
89





G1A (SEQ ID 2)
100
100
88





G1C (SEQ ID 3)
100
100
37





G1F dMel (SEQ ID 4)
100
 50
94





G1H (SEQ ID 5)
400
100
78





G1dI (D form Ile at 1st position, SEQ ID 6)
 50
100
34





GIL d-Mel (D-form, SEQ ID 150)
 50
100
91





GIL d(1-11)-1(12-26) (SEQ ID 109)
100
100
70





G1Nle (SEQ ID 8)
100
100
96





G1V (SEQ ID 9)
100
100
91





G1W (SEQ ID 10)
200
200
96





G1Y dMel (SEQ ID 11)
100
 50
95





G1Y-Mel-NH2 (SEQ ID 110)
200
200
94





G12L (SEQ ID 13)
 80
100
58





G12W (SEQ ID 14)
 80
100
51





N22T Mel-NH2 (SEQ ID 15)
 50
100
34





G1Y, K7N (SEQ ID 16)
 80
100
32





G1Y, K7A (SEQ ID 17)
400
100
83





G1L, K7S (SEQ ID 18)
100
100
89





G1L, K7R (SEQ ID 19)
100
100
92





G1L, K7H (SEQ ID 20)
100
100
97





G1L, T11C dMel (SEQ ID 21)
100
 50
81





G1L, G12L (SEQ ID 22)
400
100
93





G1L, T15C dMel (SEQ ID 24)
100
100
95





G1L, S18C (SEQ ID 25)
100
100
93





G1L, K21A (SEQ ID 28)
100
100
95





G1Y, K23A (SEQ ID 29)
100
100
42





G1L, R24A (SEQ ID 30)
100
100
87





GlY, K25A (SEQ ID 31)
100
100
77





G1Y, Q26C (SEQ ID 32)
100
100
93





G1Y, K7A, K21A (SEQ ID 35)
100
100
14





G1L, T11C, S18C dMel (SEQ ID 38)
100
100
88





T11G, T15G, S18G (SEQ ID 39)
 50
100
32





T11A, T15A, S18A (SEQ ID 40)
 50
100
38





G1L, I2L, 15L, I17L, I20L (SEQ ID 43)
400
100
96





G1L, I2Nle, I5Nle, I17Nle, I20Nle 
100
100
99


(SEQ ID 44)








G1L, I2V, I5V, I17V, I20V (SEQ ID 45)
100
100
24





dimethyl-dMel I2L, I5L, T11C, I17L, 
100
100
87


I20L dMel (SEQ ID 46)








dimethyl-dMel I2Nle, I5Nle, T11C, Il7Nle, 
100
100
78


I20Nle dMel (SEQ ID 47)








Apis Mellifera (Big Honey Bee; SEQ ID 50)
400
100
72





C-Mel G1L (SEQ ID 51)
100
100
89





C-dMel G1Nle (SEQ ID 52)
100
100
84





Dimethyl G-Mel G1L (SEQ ID 53)
100
100
91





PEG(5k)-KLK-dMel G1Y (SEQ ID 56)
300
100
72





CKLK-Mel G1L (SEQ ID 57)
150
100
91





myristoyl-CKLK-Mel G1L (SEQ ID 111)
 80
100
96





CKLK-dMel G1Nle (SEQ ID 112)
200
100
84





Acetyl-CKLK-dMel G1Nle (SEQ ID 113)
100
200
97





PEG24-GKLK-Mel G1L (SEQ ID 59)
 50
100
85





Mel-Cys (SEQ ID 62)
400
100
83





G1L Mel-Cys (SEQ ID 63)
400
100
82





G1L dMel-C (SEQ I)
 50
100
93





G1Nle Mel-C (SEQ ID 64)
400
 50
89





G1L Mel-KLKC (SEQ ID 65)
100
100
97





G1Y Mel-PLGIAGQC (SEQ ID 66)
100
100
79





G1L, Mel-KKKKK (SEQ ID 67)
400
100
96





G1Y dMel-GFKGC (SEQ ID 68)
400
100
96





CFK-G1L dMel-C (SEQ ID 69)
100
100
79





G1L Mel (1-23) (SEQ ID 71)
400
100
69





G1L, L5V, A10T, T15A Mel (1-23) (SEQ ID 72)
400
100
69





G1L, L5V, A10T, T15A, N22G, K23E dMel (1-23)
400
100
92


(SEQ ID 73)








G1L retroMel-KLK-Stearoyl (SEQ ID 75)
400
100
50





G1L retroMel-Stearoyl (SEQ ID 74)
400
100
56





G1L retro-dMel-KLK-PEG(5k) (SEQ ID 115)
100
100
32


QQRKRKIWSILAPLGTTLVKLVAGIC-








(N-PDP-PE)-NH2dMel (SEQ ID 92)
400
200
55


(PE = dioleolyl-phosphatidyl-ethanolamine)








Ac-CIGAVLKVLTTGLPALISWIKRKRQQ-NH2
400
200
85


(SEQ ID 90)








(Ac-CIGAVLKVLTTGLPALISWIKRKRQQ-NH2)2
400
200
45


(SEQ ID 90)






g peptide per mouse




g siRNA per mouse



dMel = Melittin peptide having D-form amino acids






Example 14
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, Enzymatically Cleavable Masking Agents

Melittin was reversibly modified with the indicated amount of enzymatically cleavable masking agents as described above. 200-300 μg masked melittin was then co-injected with the 50-100 μg ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above. Peptidase cleavable dipeptide-amidobenzyl carbamate modified melittin was an effective siRNA delivery peptide. The use of D-from melittin peptide is preferred in combination with the enzymatically cleavable masking agents. While more peptide was required for the same level of target gene knockdown, because the peptide masking was more stable, the therapeutic index was either not altered or improved (compared to masking of the same peptide with CDM-NAG).









TABLE 8







Inhibition of Factor VII activity in normal liver cells in mice treated with


Factor VII-siRNA cholesterol conjugate and G1L-Melittin (D form)


(SEQ ID 150) reversibly inhibited with the indicated enzymatically


cleavable masking agent.












NAG-linkage
μg
μg
percent












Peptide
amounta
type
peptide
siRNA
knockdown















G1L

CDM-NAG
200
100
97


d-Mel

NAG-AlaCit
200
50
96


(SEQ

NAG-GluGly
200
50
96


ID 150)

NAG-PEG4-PheCit
200
50
94




NAG-PEG7-PheCit
200
50
86




CDM-NAG
300
50
98




NAG-GluGly
300
50
95




NAG-GluGly
300
50
95




NAG-GluGly
300
50
82






aAmount of masking agent per Melittin amine used in the masking reaction.







Example 15
In Vivo Knockdown of Endogenous ApoB Levels Following Delivery of ApoB siRNA with Melittin Delivery Peptide in Mice, Amine Modified Melittin Peptides

Melittin peptides containing the indicated PEG amino terminal modifications were synthesized as described above. The PEG amino terminal modified melittin peptides were then reversibly modified with 5× CDM-NAG as described above. The indicated amount of Melittin was then co-injected with the 100-200 μg ApoB siRNA-cholesterol conjugate. Effect on ApoB levels were determined as described above. Addition of PEG less than 5 kDa in size decreased toxicity of the melittin peptides. Amino terminal modification with PEG greater than 5 kDa resulted in decreased efficacy (data not shown).









TABLE 9







Inhibition of ApoB activity in normal liver cells in mice treated with


ApoB-siRNA cholesterol conjugate and the indicated CDM-NAG


reversibly inhibited Melittin peptide.













NAG

μg
μg
percent


Peptide
amount
PEG
peptide
siRNA
knockdown















G1L


25
100
0


(SEQ ID 7)


50
100
72





100
100
94


CKLK-Mel


150
100
91


G1L


400
100
97


(SEQ ID 57)

NAG-(PEG)2
25
100
15





50
100
83




NAG-(PEG)4
25
100
58





50
100
81





100
100
94




NAG-(PEG)8
25
100
58





50
100
89





100
100
96


Acetyl-


100
200
90


CKLK-


200
100
90


dMel G1Nle

PEG (1k)
150
100
93


(SEQ ID 58)


CRLR-Mel

PEG (1k)
100
100
93


CKFR-Mel

PEG (1k)
100
100
81


CKLK-Mel

PEG (5k)
100
100
90


G1L


(SEQ ID 57)









Example 16
Other Melittin Derivative Sequences Known to have Membrane Activity









TABLE 10







Melittin peptides having membrane activity.









SEQ




ID
Sequence
Peptide Name





76
GIGAVLKVLTTGLPALISWISRKKRQQ
I5V, A10T, T15A, N22R,




R24K, K25R Mel-Q





77
GIGARLKVLTTGLPR ISWIKRKRQQ
I5R, A10T, T15R, L164,




N22R, K25Q





78
GIGAILKVLSTGLPALISWIKRKRQE
A10S, T15A, N22R, K25Q,




Q26E





79
GIGAVLKVLTTGLPALIGWIKRKRQQ
I5V, A10T, T15A, 518G,




N22R, K25Q





80
GIGAVLKVLATGLPALISWIKRKRQQ
I5V, T15A, N22R, K25Q





81
GIGAVLKVLSTGLPALISWIKRKRQQ
I5V, A10S T15A, N22R,




K25Q





82
GIGAILRVLATGLPTLISWIKNKRKQ
K7R





83
GIGAILKVLATGLPTLISWIKRKRKQ
N22R





84
GIGAILKVLATGLPTLISWIKKKKQQ
N22K, R24K, K25Q





85
GIGAILKVLATGLPTLISWIKNKRKQGSKKKK
Mel-GSKKKK





86
KKGIGAILKVLATGLPTLISWIKNKRKQ
KK-Mel





87
GIGAILEVLATGLPTLISWIKNKRKQ
K7E Mel





88
GIGAVLKVLTTGLPALISWIKRKR
I5V, T15A, N22R, 25-264





89
GIGAVLKVLTTGLPALISWIKR
I5V, T15A, N22R, 23-264





94
QKRKNKIWSILTPLGTALVKLIAGIG-NH2
Q25K reverse Mel









Example 17
Factor VII Knockdown in Primate Following Factor VII siRNA Delivery by Melittin Delivery Peptide

NAG-PEG2-G1L melittin was masked by reaction with 10× CDM-NAG as described above. G1L melittin was masked by reaction with 5× CDM-NAG as described above. On day 1, 1 mg/kg masked NAG-PEG2-G1L melittin, 1 mg/kg masked G1L melittin, or 3 mg/kg masked G1L melittin were co-injected with 2 mg/kg chol-Factor VII siRNA into Cynomolgus macaque (Macaca fascicularis) primates (male, 3.0 to 8.0 kg). 2 ml/kg was injected into the saphenous vein using a 22 to 25 gauge intravenous catheter. As a control, another set of primates were co-injected with 10 mg/kg G1L melittin and 2 mg/kg of a control siRNA, chol-Luciferasr siRNA. At the indicated time points (indicated in FIG. 3-5), blood samples were drawn and analyzed for Factor VII and toxicity markers. Blood was collected from the femoral vein and primates are fasted overnight before all blood collections. Blood tests for blood urea nitrogen (BUN), alanine transaminase (ALT), aspartate aminotransferase (AST), and creatinine were performed on a Cobas Integra 400 (Roche Diagnostics) according to the manufacturer's recommendations. Factor VII levels were determined as described above. Significant knockdown of Factor VII was observed at less than 1 mg/kg peptide dose. No significant toxicity was observed at a dose of 10 mg/kg peptide. Thus, the masked melittin peptides have a therapeutic index of 5-10.


Example 18
ApoB Knockdown in Primate Following ApoB siRNA Delivery by Melittin Delivery Peptide

G1L melittin was masked by reaction with 5× CDM-NAG as described above. On day 1, 2 mg/kg masked G1L melittin was co-injected with 2 mg/kg chol-ApoB siRNA into Cynomolgus macaque (Macaca fascicularis) primates. At the indicated time points (Table 11), blood samples were drawn and analyzed for ApoB protein levels and toxicity markers. Blood tests for blood urea nitrogen (BUN), alanine transaminase (ALT), aspartate aminotransferase (AST), and creatinine were performed on a Cobas Integra 400 (Roche Diagnostics) according to the manufacturer's recommendations. ApoB levels were determined as described above. No increases in BUN, Creatinine, or AST were observed. Only a transient, minor elevation in AST was observed on day 2 (1 day after injection). Knockdown of ApoB reached nearly 100% at day 11 and remained low for 31 days.









TABLE 11







Inhibition of ApoB activity in normal liver cells in primate treated with


ApoB-siRNA cholesterol conjugate and CDM-NAG masked G1L melittin.









day


















4
1
2
4
8
11
15
18
25
31





















BUN (mg/dl)
21
26
22
23
27
27
28
22
22
22


Creatinine
0.8
0.9
0.9
0.7
0.8
0.8
0.9
0.9
0.9
0.9


(mg/dl)


AST (U/L)
25
27
71
30
37
27
32
29
39
50


ALT (U/L)
34
33
58
49
50
46
46
41
39
44


apoB (mg/dl)
1072
1234
198
23
4
0
34
43
76
184









Example 19
Reduction in Hepatitis B Virus (HBV) In Vivo Following Delivery of HBV siRNAs with Melittin Delivery Peptide

A) pHBV Model Mice:


At day −42, 6 to 8 week old female NOD.CB17-Prkdscid/NcrCrl (NOD-SCID) mice were transiently transfected in vivo with MC-HBV1.3 by hydrodynamic tail vein injection (Yang P L et al. “Hydrodynamic injection of viral DNA: a mouse model of acute hepatitis B virus infection.” Proc Natl Acad Sci USA 2002 Vol. 99: p. 13825-13830). MC-HBV1.3 is a plasmid-derived minicircle that contains the same terminally redundant human hepatitis B virus sequence HBV1.3 as in the HBV1.3.32 transgenic mice (GenBank accession #V01460) (Guidotti L G et al. “High-level hepatitis B virus replication in transgenic mice. J Virol 1995 Vol. 69, p 6158-6169.). 10 μg MC-HBV1.3 in Ringer's Solution in a total volume of 10% of the animal's body weight was injected into mice via tail vein to create pHBV model of chronic HBV infection. The solution was injected through a 27-gauge needle in 5-7 seconds as previously described (Zhang G et al. “High levels of foreign gene expression in hepatocytes after tail vein injection of naked plasmid DNA.” Human Gene Therapy 1999 Vol. 10, p 1735-1737.). At day −21, three weeks transfection, Hepatitis B surface antigen (HBsAg) HBsAg expression levels in serum were measured by ELISA and the mice were grouped according to average HBsAg expression levels.


B) HBV siRNAs:


HBV siRNA mediate RNA interference to inhibit the expression of one or more genes necessary for replication and/or pathogenesis of Hepatitis B Virus. In particular, HBV siRNAs inhibition viral polymerase, core protein, surface antigen, e-antigen and/or the X protein, in a cell, tissue or mammal. HBV siRNAs can be used to treat hepatitis B virus infection. HBV siRNAs can also be used to treat or prevent chronic liver diseases/disorders, inflammations, fibrotic conditions and proliferative disorders, like cancers, associated with hepatitis B virus infection. Preferably, the sequence is at least 13 contiguous nucleotides in length, more preferably at least 17 contiguous nucleotides, and most preferably at least 18 contiguous nucleotides.









HBV siRNA 9


sense strand 


SEQ ID 117


Chol-C6-uAuCfuGfuAfgGfcAfuAfaAfuUfgGfuAf(invdT)





anti-sense


SEQ ID 118


dTAfcCfaAfutiuAfuGfcCfuAfcAfgdTsdT





HBV siRNA 10


sense strand 


SEQ ID 119


Chol-C6-uAuAfcCfuCfuGfcCfuAfaUfcAfuCfuAf(invdT)





anti-sense


SEQ ID 120


dTAfgAfuGfaUfuAfgGfcAfgAfgGfudTsdT







n=2′-O-methyl substitution, Nf=2′-Fluoro substitution, N=Ribose, dN=deoxyribose, inv=inverted, s=phosphorothioate bond, Chol=cholesterol. C6: —(CH2)6











HBV siRNA 9 unmodified sequence



unmodified sense



SEQ ID 121



CUGUAGGCAUAAAUUGGUA







unmodified antisense 



SEQ ID 122



UACCAAUUUAUGCCUACAG







HBV siRNA 10 unmodified sequence



unmodified sense



SEQ ID 123



ACCUCUGCCUAAUCAUCUA







unmodified antisense 



SEQ ID 124



UAGAUGAUUAGGCAGAGGU






Structure of the Cholesterol-C6-siRNA:



embedded image


HBV siRNAs 9 and 10 were synthesized, purified, hydridized (sense and anti-sense strands), and combined at a 1:1 molar ratio. The combined siRNAs were used for all subsequent procedures.


Suitable hepatitis B virus siRNAs are described in US Patent Publication US 2013-0005793 (U.S. Pat. No. 8,809,293), which is incorporated herein by reference.


C) Melittin Delivery Peptide:


CDM-NAG was added to Melittin, SEQ ID 7 (G1L melittin, L-form), in a 250 mM HEPES-buffered aqueous solution at a 5:1 (w/w) ratio at room temperature and incubated for 30 min to yield Melittin delivery peptide. The reaction mixture was adjusted to pH 9.0 with 4 M NaOH. The extent of the reaction was assayed using 2,4,6-trinitrobenzene-sulfonic acid and determined to be >95%. Melittin delivery peptide was purified by tangential flow in 10 mM bicarbonate buffer, pH 9.0, to which 10% dextran (w/w) was added. The final purified material was lyophilized.


D) Formation of HBV siRNA Delivery Composition:


5 mg lyophilized Melittin delivery peptide was resuspended with 1 mL water. Melittin delivery peptide was then combined with HBV siRNAs at a 1:1 ratio (w/w) (˜5.49:1 molar ratio). Isotonic glucose was added as necessary to bring the volume of each injection to 200 μl.


In some embodiments, the HBV siRNAs were in at a concentration of 26 g/L in a solution that also contained 0.069 g/L sodium phosphate monobasic monohydrate and 0.071 g/L sodium phosphate dibasic heptahydrate.


In some embodiments, a 4.8 ml injected solution contained 25.0 g/L HBV siRNAs, 25.0 g/LMLP-(CDM-NAG), 0.066 g/L sodium phosphate monobasic monohydrate, 0.068 g/L sodium phosphate dibasic heptahydrate, 0.1 g/L dextran 1K, 0.318 g/L sodium carbonate and 0.588 g/L sodium bicarbonate.


E) siRNA Delivery:


At day 1, each mouse was then given a single IV administration via tail vein of 200 μl containing 2, 4, or 8 mg/kg Melittin delivery peptide+HBV siRNAs, isotonic glucose, or 8 mg/kg Melittin delivery peptide.


F) Analyses:


At various times, before and after administration of melittin delivery peptide+HBV siRNAs, isotonic glucose, or melittin delivery peptide alone, serum HBsAg, serum HBV DNA, or liver HBV RNA were measured. HBV expression levels were normalized to control mice injected with isotonic glucose.


i) Serum Collection:


Mice were anesthetized with 2-3% isoflurane and blood samples were collected from the submandibular area into serum separation tubes (Sarstedt AG & Co., Nümbrecht, Germany). Blood was allowed to coagulate at ambient temperature for 20 min. The tubes were centrifuged at 8,000×g for 3 min to separate the serum and stored at 4° C.


ii) Serum Hepatitis B Surface Antigen (HBsAg) Levels:


Serum was collected and diluted 10 to 2000-fold in PBS containing 5% nonfat dry milk. Secondary HBsAg standards diluted in the nonfat milk solution were prepared from serum of ICR mice (Harlan Sprague Dawley) that had been transfected with 10 μg HBsAg-expressing plasmid pRc/CMV-HBs (Aldevron, Fargo, N. Dak.). HBsAg levels were determined with a GS HBsAg EIA 3.0 kit (Bio-Rad Laboratories, Inc., Redmond, Wash.) as described by the manufacturer. Recombinant HBsAg protein, ayw subtype, also diluted in nonfat milk in PBS, was used as a primary standard (Aldevron).


HBsAg expression for each animal was normalized to the control group of mice injected with isotonic glucose in order to account for the non-treatment related decline in expression of MC-HBV1.3. First, the HBsAg level for each animal at a time point was divided by the pre-treatment level of expression in that animal (Day −1) in order to determine the ratio of expression “normalized to pre-treatment”. Expression at a specific time point was then normalized to the control group by dividing the “normalized to pre-treatment” ratio for an individual animal by the average “normalized to pre-treatment” ratio of all mice in the isotonic glucose control group.


iii) Serum HBV DNA Levels:


Equal volumes of serum from mice in a group were pooled to a final volume of 100 μL. DNA was isolated from serum samples using the QIAamp MinElute Virus Spin Kit (Qiagen, Valencia, Calif.) following the manufacturer's instructions. Sterile 0.9% saline was added to each sample to a final volume of 200 μL. Serum samples were added to tubes containing buffer and protease. Carrier RNA was added to aid in the isolation of small amounts of DNA. 1 ng of pHCR/UbC-SEAP plasmid DNA (Wooddell C I, et al. “Long-term RNA interference from optimized siRNA expression constructs in adult mice.” Biochem Biophys Res Commun (2005) 334, 117-127) was added as a recovery control. After incubating 15 min at 56° C., nucleic acids were precipitated from the lysates with ethanol and the entire solution applied to a column. After washing, the samples were eluted into a volume of 50 μL Buffer AVE.


The number of copies of HBV genomes in DNA isolated from the pHBV mouse model serum was determined by qPCR. Plasmid pSEAP-HBV353-777, encoding a short segment of the HBV genome within the S gene (bases 353-777 of GenBank accession #V01460), was used to create a six log standard curve. Samples with recovery of DNA below 2 standard deviations from the average, based on detection of pHCR/UbC-SEAP were omitted. TaqMan chemistry-based primers and probes with fluor/ZEN/IBFQ were utilized:









HBV primers:


(SEQ ID 125)


5′-GCCGGACCTGCATGACTA-3′


and





(SEQ ID 126)


5′-GGTACAGCAACAGGAGGGATACATA-3′





HBV probe: 6-carboxyfluorescein (FAM)-labeled 


reporter:


(SEQ ID 127)


5′-FAM/CTGCTCAAGGAACCTC-3′





hHCR (HCR/UbC-SEAP) primers:


(SEQ ID 128)


5′-CATGCCACCTCCAACATCCACTC-3′





(SEQ ID 129)


5-GGCATAGCCACTTACTGACGACTC-3′,





hHCR probe


(SEQ ID 130)


5′-FAM/TTGTCCTGGC/ZEN/GTGGTTTAGGTAGTGTGA/IBFQ-3′






qPCR assays were performed on a 7500 Fast or StepOne Plus Real-Time PCR system (Life Technologies). For evaluation of HBV DNA in serum, DNA was isolated from duplicate purification steps from pooled group serum samples. Quantitations of HBV DNA and recovery control plasmid were determined by qPCR reactions performed in triplicate. The probes to quantitate HBV and pHCR/UbC-SEAP were included in each reaction.


iv) HBV RNA Analysis:


At various times, mice were euthanized and the liver was excised and placed into a 50-mL conical tube containing 12 ml of TRI Reagent RT (Molecular Research Center, Inc., Cincinnati, Ohio). Total RNA was isolated following the manufacturer's recommendation. Briefly, livers in TRI Reagent were homogenized using a Bio-Gen PRO200 tissue homogenizer (Pro Scientific, Inc., Oxford, Conn.) for approximately 30 seconds. 1 ml homogenate was added to 0.2 ml chloroform, mixed, and phases were separated by centrifugation. 0.1 ml of aqueous phase was removed, precipitated with isopropyl alcohol, and centrifuged. The resultant pellet was washed with 75% ethanol and resuspended in 0.4-0.6 ml nuclease-free water. Total RNA (50-500 ng) was reverse transcribed using the High Capacity cDNA Reverse Transcription Kit (Life Technologies, Grand Island, N.Y.). The cDNA was then diluted 1:50 and multiplex RT-qPCR was performed using 5′ exonuclease chemistry with forward primer 5′-GCCGGACCTGCATGACTA-3′ (SEQ ID 125), reverse primer 5′-GGTACAGCAACAGGAGGGATACATA-3′ (SEQ ID 126), and 6-carboxyfluorescein (FAM)-labeled reporter 5′-CTGCTCAAGGAACCTC-3′ (SEQ ID 127) for detection of HBV.


The RT-qPCR probe binds to all HBV RNA except the gene X transcript, which is expressed at nearly undetectable levels. Thus, the probe measured total HBV RNA. Gene expression assays for HBV, mouse β-actin, and Gene Expression Master Mix (Life Technologies, Grand Island, N.Y.) were utilized. Gene expression data were analyzed using the comparative CT method of relative quantification (Livak K J et al. “Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T))” Method. Methods 2001 Vol. 25, p 402-408).


Total RNA from each animal was reverse transcribed to generate cDNA. The cDNA was assayed by duplicate qPCR reactions that measured the HBV total RNA and the endogenous control, mouse β-actin mRNA, in the same reaction.





ΔΔCT=(CTtarget−CTcontrol)sample−(CTtarget−CTcontrol)reference


Relative Expression=2−ΔΔCT

Relative Expression of an individual=GEOMEAN of replicates


Low Range and High Range refer to 2−Avg.ΔΔCT+S.D.ΔCT and 2−Avg.ΔΔCT−S.D.ΔCT.


v) Quantitation of siRNA in Tissues:


The levels of total guide strand, total full-length guide strand, and 5′-phosphorylated full length guide strand for HBV siRNAs 9 and 10 in the liver were measured at various times by fluorescent PNA probe hybridization and HPLC anion exchange chromatography. The guide strand becomes 5′-phosphorylated by endogenous cytoplasmic CLP1 kinase (Weitzer S et al “The human RNA kinase hCLp1 is active on 3′ transfer RNA exons and short interfering RNAs.” Nature 2007 Vol. 447, p 222-227.). A fluorescently-labeled, sequence-specific peptide-nucleic acid (PNA) probe that hybridized to the guide strand was added to homogenized liver tissue. The probe-guide strand hybrid was analyzed by HPLC anion exchange chromatography that separated the guide strand based on charge.


Tissues were collected and immediately frozen in liquid nitrogen. Tissue samples were pulverized while frozen. Up to 25 mg frozen powder was solubilized in 1 mL of diluted Affymetrix Lysis Solution (one part Affymetrix Lysis Solution, two parts nuclease-free water) containing 50 μg/ml proteinase K. Samples were sonicated with a micro stick sonicator and incubated at 65° C. for 30 min. If samples needed further dilution, this was performed before the hybridization step, using the Affymetrix Lysis Solution diluted as described above. Serial dilutions of siRNA standards were also prepared in diluted Lysis Solution.











siRNA standard: RD74 (HBV siRNA 9)



sense



SEQ ID 131



(NH2C6)CfuGfuAfgGfcAfuAfaAfuUfgGfuAf(invdT)







anti-sense 



SEQ ID 132



pdTAfcCfaAfutNuAfuGfcCfuAfcAfgdTsdT







siRNA standard: RD77 (HBV siRNA 10)



sense



SEQ ID 133



(NH2C6)AfcCfuCfuGfcCfuAfaUfcAfuCfuAf(invdT)







anti-sense 



SEQ ID 134



pdTAfgAfuGfaUfuAfgGfcAfgAfgGfudTsdT







n=2′-O-methyl, Nf=2′-Fluoro, dN=deoxyribose, inv=inverted, s=phosphorothioate bond.


SDS was precipitated from the standards and samples by adding 10 μl of 3M KCl to 100 μl of the tissue sample solution. After incubating 10 min on ice, samples were centrifuged for 15 min at 2,700×g. Quantitation of siRNA was performed with the supernatant.


Sequence-specific peptide-nucleic acid (PNA) probes containing a fluorescent Atto 425 label at the N-terminus attached to the PNA chain via two ethylene glycol linkers (OO=PEG2; PNA Bio, Thousand Oaks, Calif.) were designed to bind to the antisense strand of each HBV siRNA.











Peptide-nucleic acid (PNA) probes



AD9 (HBV siRNA 9)



SEQ ID 135



Atto425-OO-CTGTAGGCATAAATT







AD10 (HBV siRNA 10) 



SEQ ID 136



Atto425-OO-ACCTCTGCCTAATCA






To 55 μl diluted serum sample was added 143 μL nuclease-free water, 11 μl 200 mM Tris-HCl (pH 8), and 11 μl 1 μM AD9 or AD10 PNA-probe solution in 96-well conical-bottom plates. The plate was sealed and incubated at 95° C. for 15 min in a thermal cycler. The temperature of the thermal cycler was reduced to 54° C. and samples were incubated for another 15 min. After incubation, samples were stored at 4° C. until they were loaded onto an autosampler for HPLC analysis.


HPLC analysis was carried out using a Shimadzu HPLC system equipped with an LC-20AT pump, SIL-20AC autosampler, RF-10Axl fluorescence detector, and a CTO-20Ac column oven (Shimadzu Scientific Instruments, Columbia, Md.). The 96-well plate from the hybridization step was loaded onto the autosampler. Injection volumes of 100 μl were made onto a DNAPac PA-100 4×250 mm analytical column (#DX043010; Fisher Scientific, Pittsburgh, Pa.) with an attached 4×50 mm guard column (#DXSP4016; Fisher Scientific, Pittsburgh, Pa.). Analysis was carried out at a flow rate of 1 ml/min with a column oven temperature of 50° C. A gradient elution using mobile phase A (10 mM Tris-HCl (pH 7), 100 mM NaCl, 30% (v/v) Acetonitrile) and mobile phase B (10 mM Tris-HCl (pH 7), 900 mM NaCl, 30% (v/v) Acetonitrile) was used following the program in Table 12 Error! Reference source not found.


Fluorescence detection was set to an excitation of 436 nm and an emission of 484 nm with a medium gain setting of 4. Concentrations of analytes eluted in the 7-10 min range were calculated using a 12-point external standard calibration curve. Calibration curves were generated with PNA-hybridized full length phosphorylated siRNA RD74 and RD77.









TABLE 12







Gradient and elution times for PNA probe hybridization and


HPLC anion exchange chromatography analysis of siRNA in liver.












Time (min)
% Eluent A
% Eluent B
Curve
















0
80
20




1.00
80
20
Linear



11.00
40
60
Linear



11.50
0
100
Linear



13.00
0
100
Linear



14.50
80
20
Linear



23.00
80
20
Linear










iv) Clinical Chemistry:


Clinical chemistry markers in mouse serum were measured using a COBAS Integra 400 (Roche Diagnostics, Indianapolis, Ind.) chemical analyzer according to the manufacturer's instructions.


G) Hepatitis B Virus (HBV) Knockdown In Vivo:


HBV DNA: Maximum HBV DNA knockdown occurred at days 8 and 15 in mice treated with 8 mg/kg Melittin delivery peptide+HBV siRNAs. Total HBV DNA in serum was reduced by 294-fold and 345-fold, respectively. On day 29, HBV DNA in serum of mice remained 13.5-fold lower than untreated control mice. Total HBV DNA was reduced 91.8-fold and 6.5-fold on day 8 in mice treated with 4 mg/kg and 2 mg/kg Melittin delivery peptide+HBV siRNAs, respectively.


HBsAg in Serum:


Maximum knockdown occurred at days 8 and 15 in mice treated with 8 mg/kg Melittin delivery peptide+HBV siRNAs. HBsAg in serum was reduced by 270-fold and 139-fold, respectively. On day 29, HBsAg in serum was 7.3-fold lower than untreated control mice. HBsAg in serum was reduced 71.4-fold and 5.4-fold and on day 8 in mice treated with 4 mg/kg and 2 mg/kg Melittin delivery peptide+HBV siRNAs, respectively.


The duration of effect from a single 8 mg/kg dose was at least 28 days. HBsAg and HBV DNA were reduced by more than 95% through Day 22. HBV DNA and HBsAg levels in serum from mice that were injected with Melittin delivery peptide (without HBV siRNAs) remained comparable to levels in mice that received a single injection of isotonic glucose (Table 13).


HBV RNA in Liver:


Maximum knockdown occurred at day 8 in mice treated with 8 mg/kg Melittin delivery peptide+HBV siRNAs. Total HBV RNA in liver was reduced by an average of 12.5-fold. On day 29, total HBV RNA in the liver was 3.4-fold lower than the average of the untreated control group. Total HBV RNA was reduced 5.8-fold and 1.6-fold on day 8 in mice treated with 4 mg/kg and 2 mg/kg Melittin delivery peptide+HBV siRNAs, respectively (Table 13).


Quantitation of siRNA in Tissues:


Injection of 8 mg/kg Melittin delivery peptide+HBV siRNAs into pHBV model mice resulted in approximately 80 ng/g HBV siRNAs in the cytoplasm of hepatocytes on day 8, as evidenced by 5′ phosphorylation of about 40 ng/g each full-length HBV siRNA 9 and HBV siRNA10 guide strands. The resulting pharmacodynamic effects on day 8 were 93% knockdown of total HBV RNA and greater than 99% reduction in HBsAg and HBV DNA in the serum. On day 22, almost all of the guide strand in the liver was 5′ phosphorylated and full-length (Table 13).


Clinical Chemistry:


Liver and renal functions were evaluated on day −1 (pre-injection) and day 2 (24 hours post-injection). There were no Melittin delivery peptide+HBV siRNAs-related changes in clinical chemistry nor was there any evidence of toxicity from either Melittin delivery peptide+HBV siRNAs or Melittin delivery peptide alone administration.









TABLE 13







Knockdown of HBsAg and HBV RNA and presence of 5′ phosphorylated


siRNA in liver following intravascular administration of melittin delivery


peptide + HBV siRNAs in HBV mouse model.












melittin






delivery



peptide +
HBsAg
HBV RNA
5′ phosphorylated



HBV siRNAs
relative
relative
siRNA guide strand


day
(mg/kg)
knockdown
knockdown
(ng/g liver tissue)














8
8
99.6 ± 0.4%
93%
76


15
8
99.3 ± 1.4%
80%
27


22
8
97 ± 5%
76%
12


29
8
 86 ± 15%
71%
2-15


8
4
99%
83%
28


8
2
82%
36%
 7









Example 20
Antiviral Efficacy of RNAi in Chronic HBV Infection in Chimpanzee

A single chimpanzee chronically infected with HBV genotype B (chimpanzee 4x0139; genotype B; viral load ˜7×109 GE/ml, 51.3-51.5 kg) was given the melittin delivery peptide+HBV siRNAs (HBV siRNA 9 and HBV siRNA 10) by IV infusion. The viral HBV DNA titer of this animal for 2 years preceding this trial ranged from 4×109 to 1.3×1010 Genome Equivalents/ml (baseline value for this study). Blood samples was taken at health check (day −7) and again immediately before dosing to serve as the baseline samples (day 1). The health check included physical exam, CBC, and whole blood chemistries. 2 mg/kg melittin delivery peptide+HBV siRNAs (20.6 ml of 5 mg/ml melittin delivery peptide) was administered at day 1 by IV push over 3 minutes. 3 mg/kg melittin delivery peptide+HBV siRNAs (30.9 ml of 5 mg/ml melittin delivery peptide) was administered at day 15 by IV push over 3 minutes. Blood samples were obtained on days 4, 8, 11, 15, 22, 29, 36, 43, 57, 64, 71, 78, and 85. Liver biopsies were obtained three times, at health check, day 29 and day 57. Animals were sedated for all procedures. Sedations for bleeds and dosing were accomplished with Telazol™ (2 mg/kg) and xylazine (100 mg) administered intramuscularly as immobilizing agents. Yohimbine is used as a reversal agent for Xylazine at the end of the procedure.


Assays for Serum and Liver HBV DNA.


HBV DNA levels were determined for serum and liver biopsy samples (baseline and days 29 and 57) using a TaqMan assay targeting the core and X regions. Both assays should detect all genomes. DNA was purified from 100 μl of serum or homogenized liver tissue using the Qiagen QiaAmp DNA Mini Kit (cat#51304), according to the manufacturer's protocol. DNA samples were analyzed by real time PCR using TaqMan technology with primers and probe designed against the HBV core gene.











forward primer, HBV core F 



(SEQ ID 137)



5′ CGAGGCAGGTCCCCTAGAAG 3′;







reverse primer, HBV core R 



(SEQ ID 138)



5′ TGCGACGCGGYGATTG 3′;







probe, HBV core probe 



(SEQ ID 139)



5′ 6-FAM/AGAACTCCCTCGCCTCGCAGACG-6-TAM 3′.








    • Liver DNA and RNA was also analyzed with primers and probe designed against the HBV X gene forward primer, HBV X F-CCGTCTGTGCCTTCTCATCTG (SEQ ID 140) reverse primer, HBV X R-AGTCCAAGAGTYCTCTTATGYAAGACCTT (SEQ ID 141) probe, HBV X 5′ 6-FAM/CCGTGTGCACTTCGCTTCACCTCTGC-6-TAM 3′ (SEQ ID 142)





A plasmid containing an HBV DNA insert was used to generate a standard curve for each TaqMan assay ranging from 10 GE to 1 million GE. Samples were analyzed in TaqMan assays using an ABI 7500 sequence detector using the following cycle parameters: 2 min at 50° C./10 min at 95° C./45 cycles of 15 sec at 95° C./1 min at 60° C.


Liver HBV DNA levels were decreased 2.4-fold (core region PCR assay) and 2.7-fold (X region PCR assay) below baseline levels on day 29.


Serum HBV DNA levels dropped rapidly after the first dose with a 17-fold decline by day 4. The levels increased between days 8-15 from 18.8 to 6.7-fold below baseline. Following the second dose on day 15, a drop in viral DNA was observed, reaching 35.9-fold decline from baseline on day 22.


Serum HBsAg and HBeAg Analyses.


HBsAg levels were determined using an ELISA kit from BioRad (GS HBsAg EIA 3.0). Quantification of surface antigen was determined by comparing OD to known surface antigen standards. HBeAg quantification was determined for all bleeds using an ELISA kit from DiaSorin (ETI-EBK Plus).


HBsAg levels were markedly reduced, declining from a baseline level of 824 μg/ml to 151 μg/ml on day 29. Values had declined significantly by day 4 following the first dose of ARC 520 (18% decrease compared to baseline values). The values continued to drop through day 15 to 53% of baseline (2.1-fold), and reached the maximum decline of 81% (5.2-fold) on day 29.


Serum levels of HBeAg were 136 ng/ml at baseline and dropped to 12.5 ng/ml (10.9-fold) by day 4 following the first injection of ARC 520. Levels increased to 46 ng/ml (2.9-fold below baseline) on day 15. Following the second injection, the levels declined again to 28 ng/ml on day 22.


RT-PCR Analysis of Cytokine and Chemokines.


The transcript levels for ISG15, CXCL11 (I-TAC), CXCL10 (IP-10), CXCL9 (Mig), Interferon gamma (IFNγ) and GAPDH were determined by quantitative RT-PCR. Briefly, 200 ng of total cell RNA from liver was analyzed by qRT-PCR assay using primers and probe from ABI Assays-on-Demand™ and an ABI 7500 TaqMan sequence analyzer (Applied Biosystems/Ambion, Austin, Tex.). The qRT-PCR was performed using reagents from the RNA UltraSense™ One-Step Quantitative RT-PCR System (Invitrogen Corporation, Carlsbad, Calif.), and the following cycle settings: 48° C., 30 min; 95° C., 10 min; and 95° C., 15 sec; and 60° C., 1 min, the latter two for 45 cycles. Liver biopsies were immediately placed in RNAlater® Stabilization Reagent and processed as described by the manufacturer and RNA was extracted using RNA-Bee (Tel-Test, Inc Friendswood, Tex.) for total cell RNA. No substantial induction of these genes was noted.


Luminex Analysis of Cytokines and Chemokines.


Monitoring of cytokines and chemokines was performed using a Luminex 100 with the xMAP (multi-analyte platform) system using a 39-plex human cytokine/chemokine kit (Millipore; Billerica, Mass.). Dilutions of standards for each cytokine were evaluated in each assay. Dilutions of standards for each cytokine were evaluated in each run to provide quantification. The following cytokines/chemokines were evaluated in serum samples using a luminex method: EGF, Eotaxin, FGF-2, Flt-3 Ligand, Fractalkine (CX3CL1), G-CSF, GM-CSF, GRO, IFNα2, IFNγ, IL-10, IL-12p40, IL-12p70, IL-13, IL-15, IL-17, IL-1α, IL-1β, IL-1 Receptor antagonist, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, MCP-1 (CCL2), MCP-3 (CCL7), MDC (CCL22), MIP-1α (CCL3), MIP-1β (CCL4), sCD40L, sIL-2 Receptor antagonist, TGFα, TNFα, TNFβ, VEGF. Similar to the hepatic transcripts, no substantial changes in chemokines and cytokines were observed during the therapy.


Clinical Pathology.


Blood chemistries were determined with a Unicel DxC 600 Analyzer (Beckman Coulter, Inc., and Diagnostic Chemicals Ltd, Oxford, Conn., USA). Whole blood chemistries had the following measurements: Na, K, Cl, Ca, CO2, Phos., ALT, AST, GGT, LDH, Direct Bilirubin, Total Bilirubin, Alk Phos, BUN, Creatine, Creatine Kinase, Glucose, Total protein, Albumin, Cholesterol, Triglycerides. Values from uninfected animals from the same colony were used to establish normal ranges. Liver biopsies were taken from the anesthetized animal by a standard procedure. Biopsy material was divided immediately into a fraction for histopathology, and DNA and RNA analysis. Sections for histopathology were processed for fixation in 10% formalin in PBS, paraffin embedded and stained with hematoxylin and eosin. Fractions for DNA analysis were snap frozen. Fractions for RNA analysis were placed in RNAlater® Stabilization Reagent.


Immunohistochemical Staining of Liver.


Liver biopsies were fixed in buffered-formalin, paraffin embedded, and sectioned at 4 microns. Slides were de-paraffinized in EZ-DeWax (BioGenex; HK 585-5K) 2× for 5 min and rinsed with water. Antigen retrieval was performed in a microwave pressure cooker for 15 min at 1000 Watts and 15 min at 300 Watts in citrate buffer (antigen retrieval solution; BioGenex; HK 086-9K). Cooled slides were rinsed with water and PBS and treated sequentially with peroxidase suppressor, universal block, and avidin (all reagents from Pierce 36000 Immunohisto Peroxidase Detection Kit). Slides were incubated sequentially for 1 h at room temperature with primary antibody diluted in universal block containing a biotin block, for 0.5 h with biotinylated goat anti-mouse IgG, and for 0.5 h with avidin-biotin complex (ABC). Slides were developed with Immpact Nova Red peroxidase substrate (Vector, SK-4805; Burlingame Calif.), counter stained Mayers (Lillie's) hematoxylin (DAKO, S3309), dehydrated and mounted in non-aqueous mounting media (Vector, VectaMount; H-5000). Rabbit anti-HBV core was prepared from purified core particles expressed in baculovirus.


Most hepatocytes were positive for HBV core antigen with intense staining of the cytoplasm and some staining of the nucleus. A decline in staining occurred at day 29 that was considered significant.


Example 21
Reduction in Hepatitis B Virus (HBV) In Vivo Transgenic Mouse Model Following Delivery of HBV siRNAs Using Melittin Delivery Peptide

A) Transgenic HBV Model Mice:


Transgenic HBV1.3.32 mice contain a single copy of the terminally redundant, 1.3-genome length human HBV genome of the ayw strain (GenBank accession number V01460) integrated into the mouse chromosomal DNA. High levels of HBV replication occur in the livers of these mice (Guidotti L G et al. “High-level hepatitis B virus replication in transgenic mice.” J Virol 1995 Vol. 69, p 6158-6169).


Mice were selected for the study on the basis of the HBeAg level in their serum upon weaning. Mice were grouped such that the average HBeAg levels was similar in each group. Student's T-test was used to assure there were no significant differences between any of the groups relative to the control siLuc group.


Melittin delivery peptide HBV siRNA delivery composition (melittin delivery peptide+HBV siRNAs were prepared as described in example 19. HBV siRNA 9, HBV siRNA 10, RD74 (HBV siRNA 9), and siRNA standard: RD77 (HBV siRNA 10) were prepared as in example 19.











siLuc (firefly Luciferase siRNA)



sense strand 



SEQ ID 143



Chol-uAuCfuUfaCfgCfuGfaGfuAfcUfuCfgAf(invdT)







anti-sense



SEQ ID 144



UfsCfgAfaGfuAfcUfcAfgCfgUfaAfgdTsdT






B) HBV siRNA Delivery:


Female HBV1.3.32 mice, 1.8-7.7 months old, were given a single IV injection into the retro-orbital sinus of 200 μl per 20 g body weight of 3 mg/kg or 6 mg/kg melittin delivery peptide+HBV siRNAs on day 1. Control mice injected with isotonic glucose or 6 mg/kg melittin delivery peptide+siLuc.


Serum Collection:


Mice were briefly anesthetized with 50% CO2 and blood samples were collected from the retro-orbital sinus using heparinized Natelson micro blood collecting tubes (#02-668-10, Fisher Scientific, Pittsburgh, Pa.). Blood was transferred to microcentrifuge tubes, remaining at ambient temperature for 60-120 min during collection. Samples were then centrifuged at 14,000 rpm for 10 min to separate the serum, which was then stored at −20° C.


C) HBcAg Knockdown:


A qualitative assessment of HBV core antigen (HBcAg) distribution in the cytoplasm of hepatocytes following melittin delivery peptide mediated delivery of HBV siRNAs was performed by immunohistochemical staining of liver sections. The presence of cytoplasmic HBcAg indicates that the protein is being actively expressed. Tissue samples were fixed in 10% zinc-buffered formalin, embedded in paraffin, sectioned (3 μm), and stained with hematoxylin (Chisari F V et al. “Expression of hepatitis B virus large envelope polypeptide inhibits hepatitis B surface antigen secretion in transgenic mice.” J Virol 1986 Vol. 60, p 880-887). The intracellular distribution of HBcAg was assessed by the labeled-avidin-biotin detection procedure (Guidotti L G et al. “Hepatitis B virus nucleocapsid particles do not cross the hepatocyte nuclear membrane in transgenic mice.” J Virol 1994 Vol. 68, 5469-5475). Paraffin-embedded sections in PBS, pH 7.4, were treated for 10 min at 37° C. with 3% hydrogen peroxide and washed with PBS. After the sections were blocked with normal goat serum for 30 min at room temperature, rabbit anti-HBcAg (Dako North America, Inc., Carpinteria, Calif.) primary antiserum was applied at a 1:100 dilution for 60 min at 37° C. After a wash with PBS, a secondary antiserum consisting of biotin-conjugated goat anti-rabbit immunoglobulin G F(ab9)2 (Sigma-Aldrich Co. LLC., St. Louis, Mo.) was applied at a 1:100 dilution for 30 min at 37° C. The antibody coated slides were washed with PBS, treated with the streptavidin-horseradish peroxidase conjugate (ExtrAvidin; Sigma-Aldrich Co. LLC., St. Louis, Mo.) at a 1:600 dilution for 30 min at 37° C., stained with 3-amino-9-ethyl carbazole (AEC; Shandon-Lipshaw, Pittsburgh, Pa.), and counterstained with Mayer's hematoxylin before being mounted. HBcAg levels and distribution within the hepatocytes were visually assessed. Cytoplasmic HBcAg was greatly reduced relative to nuclear HBcAg at days 15 and 29 following injection of 6 mg/kg melittin delivery peptide+HBV siRNAs, indicating knockdown of HBcAg expression.









TABLE 14







Qualitative assessment of HBcAg staining in the nucleus (n) compared


to HBcAg staining in the cytoplasm (c).











nuclear (n) vs.




cytoplasmic (c)


Treatment
day
distribution












Isotonic glucose
8
n = c



8
n = c


6 mg/kg melittin delivery peptide + siLuc
8
n = c


6 mg/kg melittin delivery peptide +
8
n = c


HBV siRNAs
8
n = c



15
n >> c



15
n >> c



29
n >> c



29
n >> c









D) HBeAg Knockdown:


The effect of melittin delivery peptide mediated delivery of HBV siRNA delivery on HBV e antigen (HBeAg) was determined by ELISA. Serum was collected from the mice at pre-injection day −1, 6 hours post-injection, and on days 3, 8, 15, 22, and 29. HBeAg analysis was performed with the HBe enzyme linked immunosorbent assay (ELISA) as described by the manufacturer (Epitope Diagnostics, San Diego, Calif.) using 2 μl of mouse serum. The level of antigen was determined in the linear range of the assay. The HBeAg levels for each animal and at each time point were normalized to the day −1 pre-dose level. The melittin delivery peptide+HBV siRNAs treatment groups were separately compared to the isotonic glucose group or the siLuc group. Paired T-tests were used to evaluate changes in HBeAg expression from day 3 to day 8.


The levels of HBeAg was reduced by 85-88% (7-8 fold) and day 3 and approximately 71-73% at day 8 for both dose levels. HBeAg remained reduced ˜66% at day 29 in animals treated with 6 mg/kg melittin delivery peptide+HBV siRNAs. These transgenic mice are known to produce HBeAg in their kidneys. The level of circulating HBeAg originating from the kidneys is not known.









TABLE 15







Relative HBeAg expression normalized to day −1 and mean of combined


control groups on day 3 or day 8










day










treatment
3
8





Isotonic glucose
1.09 ± 0.35
0.86 ± 0.09


6 mg/kg melittin delivery peptide +
0.91 ± 0.04
1.14 ± 0.21


siLuc


3 mg/kg melittin delivery peptide +
0.15 ± 0.05
0.29 ± 0.12


HBV siRNAs


6 mg/kg melittin delivery peptide +
0.12 ± 0.07
0.27 ± 0.17


HBV siRNAs
















TABLE 16







Relative HBeAg expression normalized to day −1 of each group









day














treatment
−1
0.25
3
8
15
22
29





Isotonic glucose
1.00
1.37 ± 0.26
1.75 ± 0.65
1.08 ± 0.14





6 mg/kg melittin delivery
1.00
1.43 ± 0.09
1.46 ± 0.07
1.43 ± 0.30





peptide + siLuc


3 mg/kg melittin delivery
1.00
1.01 ± 0.26
0.24 ± 0.08
0.37 ± 0.16
0.51 ± 0.15




peptide + HBV siRNAs


6 mg/kg melittin delivery
1.00
0.96 ± 0.25
0.20 ± 0.11
0.34 ± 0.22
0.32 ± 0.14
0.25 ± 0.13
0.34 ± 0.18


peptide + HBV siRNAs









E) HBV RNA Knockdown:


HBV produces at least 6 mRNA species that are in length: 3.5 kilobases (kb) (2 types), 2.4 kb, 2.1 kb (2 types) and 0.7 kb. One 3.5 kb mRNA that encodes HBeAg. HBeAg is a secreted protein. The other 3.5 kb mRNA is the pre-genomic RNA (pgRNA), which is translated to produce the core protein (HBcAg) and the polymerase. The pgRNA is reverse transcribed to generate the virion DNA. HBcAg protein monomers assemble to form the capsid that encloses the virion DNA. The 2.4 kb and 2.1 kb mRNAs encode the envelope (S) protein that are also called S antigen (HBsAg). The HBsAg proteins form the envelope around the viral capsid (Because transgenic HBV1.3.32 mice produce antibodies to the this protein, HBsAg was not measured.). The 0.7 kb mRNA encodes X protein and is usually undetectable in transgenic mice.


After mice were sacrificed, liver tissue was frozen in liquid nitrogen and stored at −70° C. prior to total RNA extraction. RNA was isolated and levels of the HBV transcripts were evaluated and quantitated relative to the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH) by Northern blotting and by quantitative real-time PCR (RT-qPCR).


Northern Analysis.


RNA (Northern) filter hybridization analyses were performed using 10 μg of total cellular RNA. Filters were probed with 32P-labeled HBV (strain ayw) genomic DNA to detect HBV sequences and mouse glyceraldehyde-3-phosphate dehydrogenase (GAPDH) cDNA to detect the GAPDH transcript used as an internal control. The radioactive hybridization signals corresponding to the 3.5 kb HBV RNA and the 2.1 kb RNA bands in the Northern blot were normalized to the signal corresponding to the GAPDH mRNA band from the same animal. The 2.1 kb HBV RNA:GAPDH ratio from each animal was divided by the average of this ratio in the combined controls groups, consisting of 4 mice injected with isotonic glucose and 4 mice treated with melittin delivery peptide+siLuc, to determine treatment-specific changes in the 2.1 kb HBV RNA. The 3.5 kb HBV RNA was analyzed by the same method. In both cases error is shown as the standard deviation of the ratio. Statistical significance was determined by a Student's two-tailed t-test. Results from RNA filter hybridization (Northern blot) analyses of total cellular RNA from liver tissue are shown in Table. Melittin delivery peptide+HBV siRNAs treatment reduced viral RNA content in liver. No effects on viral RNA levels in liver were observed in animals receiving isotonic glucose or melittin delivery peptide+siLuc treatments.









TABLE 17







Northern blot analysis of knockdown of 2.1 kb HBV RNA encoding HBsAg


following single does melittin delivery peptide + HBV siRNAs


treatment in transgenic mice.














HBV RNA/
fold

% RNA


treatment
day
GAPDH
reduction
P-valuea
knockdownb















Isotonic glucose
8
2.79 ± 0.70





6 mg/kg melittin delivery
8
2.91 ± 0.20


peptide + siLuc


3 mg/kg melittin delivery
8
0.527 ± 0.111
5.4
<0.0001
81.5 ± 3.4


peptide + HBV siRNAs
15
1.23 ± 0.84
2.3
0.002
56.7 ± 2.6


6 mg/kg melittin delivery
8
0.0487 ± 0.0419
58.4
<0.0001
98.3 ± 1.3


peptide + HBV siRNAs
15
0.0301 ± 0.0159
94.3
<0.0001
 98.9 ± 0.05



29
0.324 ± 0.220
8.8
<0.0001
88.6 ± 6.7






aComparison of the mean of the treatment group against the combined mean of the control groups using a two-tailed unpaired t test.




bHBV RNA levels normalized to combined average of control groups.














TABLE 18







Northern blot analysis of knockdown of 3.5 kb HBV RNA following


single does melittin delivery peptide + HBV siRNAs treatment


in transgenic mice.













HBV RNA/

fold


treatment
day
GAPDH
P-valuea
reductionb














Isotonic glucose
8
1.72 ± 0.47




6 mg/kg melittin delivery
8
1.72 ± 0.11


peptide + siLuc


3 mg/kg melittin delivery
8
0.949 ± 0.458
0.006
1.8


peptide + HBV siRNAs
15
1.11 ± 0.64
0.045
1.6


6 mg/kg melittin delivery
8
0.335 ± 0.226
<0.0001
5.1


peptide + HBV siRNAs
15
0.795 ± 0.340
0.0009
2.2



29
0.969 ± 0.483
0.008
1.8






aComparison of the mean of the treatment group against the combined mean of the control groups using a two-tailed unpaired t test.




bHBV RNA levels normalized to combined average of control groups.







RT-qPCR Analysis.


Quantitative PCR following a reverse transcription step (RT-qPCR) was used to measure the level of GAPDH and HBV 3.5 kb transcripts in HBV1.3.32 mouse liver RNA. After DNase I treatment, 1 μg of RNA was used for cDNA synthesis using the TaqMan reverse transcription reagents (Life Technologies, Grand Island, N.Y.) followed by qPCR quantification using SYBR Green and an Applied Biosystems 7300 Real-Time PCR System. Thermal cycling consisted of an initial denaturation step for 10 min at 95° C. followed by 40 cycles of denaturation (15 sec at 95° C.) and annealing/extension (1 min at 60° C.). The relative HBV 3.5 kb RNA expression levels were estimated using the comparative CT (ΔCT) method with normalization to mouse GAPDH RNA. The PCR primers used were 5′-GCCCCTATCCTATCAACACTTCCGG-3′ SEQ ID 145 (HBV 3.5 kb RNA sense primer, coordinates 2,311 to 2,335), 5′-TTCGTCTGCGAGGCGAGGGA-3′ SEQ ID 146 (HBV 3.5 kb RNA antisense primer, coordinates 2401 to 2382), 5′-TCTGGAAAGCTGTGGCGTG-3′ SEQ ID 147 (mouse GAPDH sense primer), and 5′-CCAGTGAGCTTCCCGTTCAG-3′ SEQ ID 148 (mouse GAPDH antisense primer), respectively.









TABLE 19







RT-qPCT analysis of knockdown of 3.5 kb HBV RNA following


single does melittin delivery peptide + HBV siRNAs


treatment in transgenic mice.













HBV RNA/

fold


treatment
day
GAPDH
P-valuea
reductionb














Isotonic glucose
8
2.88 ± 2.60




6 mg/kg melittin delivery
8
2.36 ± 0.69


peptide + siLuc


6 mg/kg melittin delivery
8
0.292 ± 0.280
0.45
8.8


peptide + HBV siRNAs
15
0.452 ± 0.285
0.03
5.7



29
1.98 ± 1.45
0.55
1.3






aComparison of the mean of the treatment group against the combined mean of the control groups using a two-tailed unpaired t test.




bHBV RNA levels normalized to combined average of control groups.







F) HBV DNA Replication Intermediate Knockdown:


After mice were sacrificed, liver tissue was frozen in liquid nitrogen and stored at −70° C. prior to DNA extraction. DNA was isolated from the liver and the HBV replicative intermediates were evaluated and quantitated relative to the transgene by Southern blotting. Southern blot analysis of 20 μg HindIII-digested total cellular DNA was performed using a 32P-labelled HBV (strain ayw) genomic DNA. Relative levels of HBV replicative intermediates, the relaxed circular DNA (HBV RC DNA) and single-stranded DNA (HBV SS DNA), were normalized to levels of the HBV transgene (HBV transgene DNA) in the same animal following phosphorimager quantitation. The signal from the combined HBV RC and SS DNA: HBV Tg DNA from each animal was divided by the average of this ratio in the combined controls groups, consisting of 4 mice injected with isotonic glucose and 4 mice co-injected with ARC-EX1 and siLuc, to determine treatment-specific changes in the replicative intermediates. Southern blot analysis indicated that all groups treated with melittin delivery peptide+HBV siRNAs had reduced levels of HBV replicative intermediates (Tables). HBV DNA replication intermediates remained greatly suppressed for four weeks after a single injection of 6 mg/kg melittin delivery peptide+HBV siRNAs. Replicative intermediates were reduced 98-99% (64-74 fold) at one and two weeks and 97% (29-fold) at four weeks.









TABLE 20







HBV replication intermediate levels normalized to a


combined average of control groups













fold


treatment
day

reduction













Isotonic glucose
8
0.959 ± 0.495



6 mg/kg melittin delivery peptide +
8
1.042 ± 0.236


siLuc


3 mg/kg melittin delivery peptide +
8
0.145 ± 0.029
6.9


HBV siRNAs
15
0.240 ± 0.079
4.2


6 mg/kg melittin delivery peptide +
8
0.016 ± 0.027
63.5


HBV siRNAs
15
0.013 ± 0.004
74.1



29
0.034 ± 0.033
29.1
















TABLE 21







Ratio of HBV Replication Intermediates/HBV Tg DNA as evaluated by


Southern blot analysis.












Ratio





HBV Replication




Intermediates/HBV


treatment
day
Transgene DNA
P-value













Isotonic glucose
8
37.3 ± 22.3



6 mg/kg melittin deliverypeptide +
8
40.5 ± 10.6


siLuc


combined average

38.9


3 mg/kg melittin delivery peptide +
8
5.63 ± 1.29
0.0006


HBV siRNAs
15
9.33 ± 3.54
0.001


6 mg/kg melittin delivery peptide +
8
0.61 ± 1.23
0.0003


HBV siRNAs
15
0.52 ± 0.17
0.0003



29
1.34 ± 1.47
0.0003









G) Quantitation of HBV siRNA in Liver:


The amounts of HBV siRNA guide strands in the livers of melittin delivery peptide+HBV siRNAs treated mice were quantitated by hybridization with a fluorescent peptide nucleic acid (PNA) probe as described in example 19. The PNA-hybridization method allowed quantitation of the total amount of guide strand, including metabolites of HBV siRNAs 9 and 10 (total, total full-length, 5′ phosphorylated full-length, and non-phosphorylated full-length) per weight of tissue. The presence of full length 5′ phosphorylated guide strand indicated efficient delivery of the siRNA to the target cell cytoplasm.









TABLE 22







HBV siRNA guide strand measured in liver homogenates.











melittin
HBV siRNA 9 guide strand
HBVsiRNA 10 guide strand



delivery peptide +
(ng/g tissue)
(ng/g tissue)















HBV siRNAs
5′ phosph.
total full

5′ phosph
total full



day
(mg/kg)
full length
length
total
full length
length
total

















8
3
3.8 ± 1.4
3.8 ± 1.4
14.8 ± 3.8
0.8 ± 1.3
0.8 ± 1.3
0.8 ± 1.3


8
6
17.9 ± 8.2 
21.3 ± 10.2
76.8 ± 34.1
11.5 ± 6.7 
12.6 ± 7.6 
18.8 ± 11.4


15
3
0.0 ± 0.0
0.0 ± 0.0
4.6 ± 1.6
0.0 ± 0.0
0.0 ± 0.0
3.4 ± 2.0


15
6
9.5 ± 2.2
9.5 ± 2.2
35.0 ± 15.7
4.8 ± .09
4.8 ± .09
5.9 ± 1.5


29
6
0.5 ± 0.8
0.5 ± 0.8
2.3 ± 2.4
0.0 ± 0.0
0.0 ± 0.0
2.1 ± 2.2









H) Clinical Chemistry:


Serum for clinical chemistry and cytokine evaluation was collected from each mouse at day −1 prior to injection and at 6 hr and 48 hr post-injection. Clinical chemistry analysis of alanine aminotransferase (ALT), Aspartate aminotransferase (AST), blood urea nitrogen (BUN), and creatinine was measured using a COBAS Integra 400 (Roche Diagnostics, Indianapolis, Ind.) chemical analyzer according to the manufacturer's instructions. Each assay required 2-23 μL serum, depending on the test. Clinical chemistries from all groups of animals were compared before and after injection by one-way ANOVA. Bonferroni's Multiple Comparison Test was used to compare individual group values before and after injection. There were no increases in ALT, AST, BUN, or creatinine 48 hr post-injection. A panel of 25 mouse cytokines were evaluated using a MILLIPLEX MAP Mouse Cytokine/Chemokine Magnetic Bead Panel-Premixed 25 Plex-Immunology Multiplex Assay (Catalog #MCYTOMAG-70K-PMX, EMD Millipore Corporation, Billerica, Mass.): granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), interferon gamma (IFN-γ), interleukin-1 alpha (IL-1α), interleukin-1 beta (IL-1β), interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin-5 (IL-5), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-9 (IL-9), interleukin-10 (IL-10), interleukin-12 subunit p40 (IL-12p40), interleukin-12 subunit p70 (IL-12p70), interleukin-13 (IL-13), interleukin-15 (IL-15), interleukin-17 (IL-17), interferon gamma-induced protein-10 (IP-10), keratinocyte-derived cytokine (KC), monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein-1 alpha (MIP-1α), macrophage inflammatory protein-1 beta (MIP-1β), macrophage inflammatory protein-2 (MIP2), regulated on activation, normal T cell expressed and secreted (RANTES) and tumor necrosis factor alpha (TNF-α). A few cytokines were elevated by the handling procedures, but appeared unrelated to melittin delivery peptide+HBV siRNAs treatment.


IL-6 levels were elevated in all groups at 6 h post-injection. Elevation was higher in mice receiving 3 mg/kg melittin delivery peptide+HBV siRNAs and highest—8-fold above the upper limit of normal (up to 170 pg/ml)—in mice receiving 6 mg/kg melittin delivery peptide+HBV siRNAs. IL-6 levels returned to normal by day 3, 48 hr after injection.


KC levels were elevated at 6 h, up to 40-fold above the upper limit of normal (103 pg/ml), but this elevation was similar in all treatment groups.


IP-10 levels were elevated less than 2-fold at 6 h and in some samples at 48 h. However, elevations were also in the isotonic glucose control group.


MIP2 is normally undetectable in mouse serum, but levels were elevated after injection in all groups, primarily at 6 hr.


G-CSF levels, while slightly elevated, 3-4 fold average at 6 hr post-injection, the group averages remained within normal range.


TNF-α and MCP-1 were elevated in all groups at 6 h, but remained well below the upper limit of normal.


One out of 12 mice injected with 6 mg/kg melittin delivery peptide+HBV siRNAs had an IL-7 level approximately 3-fold higher than the upper limit of normal at 6 h: 80 pg/ml.


Evaluation of liver or kidney toxicity showed minimal adverse effects. There were no increases relative to pre-injection in clinical chemistry markers for liver or kidney. Elevation of some cytokines was observed pre-dosing and a few cytokines were elevated by handling procedures that appeared to be unrelated to melittin delivery peptide+HBV siRNAs treatment.

Claims
  • 1. A composition comprising: a first component and a second component wherein comprises Melittin-(L-T)x and the second component comprises an siRNA, and wherein Melittin is a melittin peptide,-L-T has the structure represented by —CO—C(CH3)═C(T)—COOH or —CO—C(T)═C(CH3)—COOH, wherein T comprises a targeting ligand having affinity for the an asialoglycoprotein receptorx is greater than 80% of the number of primary amines of a population of melittin peptides, andthe siRNA comprises a first siRNA wherein said first siRNA inhibits expression of a hepatitis B virus gene.
  • 2. The composition of claim 1 wherein the melittin peptide comprises the amino acid sequence of SEQ ID 1, SEQ ID 7, SEQ ID 11, SEQ ID 51, SEQ ID 57, SEQ ID 58, SEQ ID 92, or SEQ ID 96.
  • 3. The composition of claim 2 wherein the melittin peptide comprises the amino acid sequence of SEQ ID 7.
  • 4. The composition of claim 3 wherein T comprises N-acetylgalactosamine (GalNAc).
  • 5. The composition of claim 4 wherein -(L-T) has the structure represented by:
  • 6. The composition of claim 5 wherein a cholesterol moiety is covalently linked to the siRNA.
  • 7. The composition of claim 6 wherein the first siRNA comprises the nucleotide sequence of SEQ ID 122 or SEQ ID 124.
  • 8. The composition of claim 7 wherein at least one nucleotide of the first siRNA is modified.
  • 9. The composition of claim 7 wherein the first siRNA comprises SEQ ID 118 or SEQ ID 120
  • 10. The composition of claim 9 wherein the first siRNA comprises SEQ ID 118 and SEQ ID 117 or SEQ ID 120 and SEQ ID 119.
  • 11. The composition of claim 7 wherein the second component comprises a second siRNA wherein said second siRNA inhibits expression of a hepatitis B virus gene.
  • 12. The composition of claim 11 wherein the first siRNA comprises SEQ ID 122 and the second siRNA comprises SEQ ID 124.
  • 13. The composition of claim 12 wherein at least one of the nucleotides is modified.
  • 14. The composition of claim 13 wherein the first siRNA comprises SEQ ID 118 and SEQ ID 117 and the second siRNA comprises SEQ ID 120 and SEQ ID 119.
  • 15. The composition of claim 14 wherein the first component and the second component are provided in separate vials.
  • 16. The composition of claim 15 wherein the first component, the second component, or the first and second components contains a pharmaceutically acceptable carrier
  • 17. The composition of claim 16 wherein the pharmaceutically acceptable carrier comprises dextran.
  • 18. The composition of claim 17 wherein the first component, the second component, or the first and second components are lyophilized.
  • 19. A method of inhibiting expression of a hepatitis B virus gene in a patient comprising administering to said patient the components of claim 1.
  • 20. The method of claim 19 wherein the melittin peptide comprises the amino acid sequence of SEQ ID 7.
  • 21. The method of claim 20 wherein -(L-T) has the structure represented by:
  • 22. The method of claim 21 wherein the first siRNA comprises the nucleotide sequence of SEQ ID 122 or SEQ ID 124.
  • 23. The method of claim 22 wherein at least one of the nucleotides is modified.
  • 24. The method of claim 23 wherein the first siRNA comprises SEQ ID 118 or SEQ ID 120
  • 25. The method of claim 24 wherein the second component comprises a second siRNA wherein said second siRNA inhibits expression of a hepatitis B virus gene.
  • 26. The method of claim 25 wherein the first siRNA comprises SEQ ID 118 and the second siRNA comprises SEQ ID 120.
  • 27. The method of claim 13 wherein the first siRNA comprises SEQ ID 118 and SEQ ID 117 and the second siRNA comprises SEQ ID 120 and SEQ ID 119.
  • 28. A method of treating a patient having a hepatitis B virus infection comprising: resuspending the components of claim 28 in water, combining the resuspended components, and administering to said patient a therapeutic amount of the resuspended components.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation-in-part of U.S. application Ser. No. 14/789,142, filed 7 Jul. 2015, pending, which is a continuation of Ser. No. 13/926,380, filed 25 Jun. 2013, issued as Pat. No. 9,107,957, which is a continuation of U.S. application Ser. No. 13/326,433, 15 Dec. 2011, issued as U.S. Pat. No. 8,501,930, which claims the benefit of U.S. Provisional Application No. 61/424,191, filed 17 Dec. 2010. Each of Ser. No. 14/789,142, 13/926,380 and 13/326,433 is incorporated herein by reference.

Provisional Applications (1)
Number Date Country
61424191 Dec 2010 US
Continuations (2)
Number Date Country
Parent 13926380 Jun 2013 US
Child 14789142 US
Parent 13326433 Dec 2011 US
Child 13926380 US
Continuation in Parts (1)
Number Date Country
Parent 14789142 Jul 2015 US
Child 15200441 US