CA2116676A1 - Method for delivering nucleic acids into cells - Google Patents

Method for delivering nucleic acids into cells

Info

Publication number
CA2116676A1
CA2116676A1 CA002116676A CA2116676A CA2116676A1 CA 2116676 A1 CA2116676 A1 CA 2116676A1 CA 002116676 A CA002116676 A CA 002116676A CA 2116676 A CA2116676 A CA 2116676A CA 2116676 A1 CA2116676 A1 CA 2116676A1
Authority
CA
Canada
Prior art keywords
group
cholesteryl
lipid
beta
cationic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002116676A
Other languages
French (fr)
Inventor
Richard M. Epand
Remo Bottega
Leaf Huang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
McMaster University
University of Tennessee Research Foundation
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2116676A1 publication Critical patent/CA2116676A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J41/00Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring
    • C07J41/0033Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005
    • C07J41/0055Normal steroids containing one or more nitrogen atoms not belonging to a hetero ring not covered by C07J41/0005 the 17-beta position being substituted by an uninterrupted chain of at least three carbon atoms which may or may not be branched, e.g. cholane or cholestane derivatives, optionally cyclised, e.g. 17-beta-phenyl or 17-beta-furyl derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle

Abstract

A method for facilitating the transfer of nucleic acids into cells comprising preparing a mixed lipid dispersion of a cationic lipid with a co-lipid in a suitable carrier solvent. The lipid has a structure which includes a lipophilic group derived from cholesterol, a linker bond, a spacer arm including from about 1 to about 20 carbon atoms in a branched or unbranched linear alkyl chain, and a cationic amino group selected from the group consisting of primary, secondary, tertiary and quaternary amino groups. The method further comprises adding the nucleic acids to the dispersion to form a complex. The cells are then treated with the complex.
There is also disclosed a novel cationic amphiphile useful for this purpose.

Description

2 PCT/USg2/07290 hilG'~7(i METHOD FOR DELIVERING NUCLEIC ACIDS INTO CELLS

The present invention relates to methods for facilitating the transfer of nucleic acids into cells and to a novel cationic amphiphile useful for this purpose.
Some but not all cationic amphiphiles are known to facilitate the transfer of DNA into cells, i.e., transfection. Although the mechanism of this activity is not yet clear, it probably involves the binding of the DNA/lipid complex with the cell surface via the excess positive charges on the complex. Cell surface bound complex is probably internalized and the DNA is released into the cytoplasm of the cell from an endocytic compartment. How the released DNA moves into the nucleus is ~ot known.
A cationic amphiphile contains the following four important structural elements:

lipophilic - Linker - Spacer - Amino group bond arm group The amino group is positively charged at neutral pH. It may be a primary, secondary, tertiary or quaternary ammonium group. The spacer arm is usually a hydrophilic, 2 to 15-atom moiety which connects the amino group to the lipophilic group via the linker bond. The linker bond is either an ether, ester, amide or other hydrolyzable bond.
The lipophilic group is a hydrophobic moiety which allows the insertion of the cationic amphiphile into the membranes of the cell or liposome. It serves as an 21i~ 76 2 anchor for the cationic ammonium group to attach to the surface of a cell or liposome.
N-[l-(2,3-dioleoxyloxy) propyl]-N,N,N-trimethyl ammoni~m chloride (DOTMA) is the first cationic amphiphile exhibiting the activity of transfection. Its lipophilic group is a double-chain, Cl8:l aliphatic group. It contains a quaternary ammonium group connected to the lipophilic group via a 3-carbon spacer arm with two ether linker bonds. Although the molecule is effective in transfection, it is not biodegradable and is rather toxic to cells.
Another series of cationic amphiphilès used in transfection is the quaternary ammonium detergents. Either single chain (such as cetyltrimethylammonium bromide) or double chain (such as dimethyldioctadecylammonium bromide) detergents exhibit activity to transfect animal cells. The amino group in these amphiphiles is quaternary and is connected to the lipophilic group without the spacer ar~ or linker bonds. Another single-chain detergent, stearylamine, contains a primary amino group connected to a single Cl8:0 chain without a spacer arm or linker bond.
This group ~f amphiphiles is also toxic to the cells.
Two other groups of cationic amphiphiles for transfection have been reported. The first group contains two Cl8:l chains as the lipophilic group. Both groups contain a quaternary ammonium group, but the spacer arm structure varies. In one case, the trimethylammonium group is directly connected to the two Cl8:l chains via a 3-carbon spacer arm and ester bond. The amphiphile, l,2-WO93/05162 211 ~ S 7 G PCT/US92/07290 dioleoxy-3-(trimethylammonio)propane, (DOTAP) is a close analog of DOTMA. In other cases, such as 1,2-dioleoyl-3-(4'-trimethylammonio) butanoyl-sn-glycerol, DOBT, or cholesteryl (4'-trimethylammonio) butanoate, ChOTB, the trimethylammonium group is connected via a butanoyl spacer arm to either the double-chain (for DOTB) or cholesteryl (for ChOTB) group. Other amphiphiles, i.e., l,2-dioleoyl-3-succinyl-sn-glycerol choline ester (DOSC) and cholesteryl hemisuccinate choline ester, ChOSC, contain a choline moiety as the quaternary ammonium group which is connected to the double-chain (for DOSC) or cholesteryl (for ChOSC) group via a succinyl spacer arm. The transfection activities of these amphiphiles are generally weak.
Yet another class of amphiphiles, called "lipopolyamine" has alæo been reported. The ammonium group is L-5-carboxyspermine which contains 2 primary and 2 secondary ammonium groups. Two examples of this lipopolyamine are dioctadecylamidologlycylspermine, DbGS, and dipalmitoyl phosphatidylethanolamidospermine, DPPES.
The cationic group is connected to two different double-chain, Cl6:0 lipophilic group via an amidoglycyl (for DOGS) or phosphorylethanolamine (for DPPES) spacer arm. These compounds are especially efficient in transfecting the primary endocrine cells without cellular toxicity.
A lipopolylysine reagent for transfection has also been reported. The reagent contains a polylysine moiety as the ammonium group which is connected to a phospholipid (N-glutarylphosphatidylethanolamine).
Therefore, the spacer arm is the side chain of lysine and W093fO5162 PCT/US92/07290 7..11~iGr~i the head group of the phospholipid. The lipophilic group is a double-chain, C18:1 group connecting to the spacer arm via two ester bonds. Although the reagents is efficient in transfection and non-toxic to cells, the activity requires scraping the treated cells. This is clearly not a convenient step and cannot be done for in vivo experiments.
An ideal transfection reagent should exhibit a high level of transfection activity without scraping or any other mechanical or physical manipulations of the cells or tissues. The reagent should be non-toxic or minimally toxic at the effective doses. It should also be biodegradable to avoid any long-term adverse side-effects on the treated cells.
Many reagents which fulfill these criteria contain a linker bond that is hydrolyzable in the cell.
For example, DOBT and DOSC, both contain ester linker bonds, can be metabolized and catabolized into other lipid species in the treated cells. However, cationic amphiphiles containing ester linker bonds are not stable when stored in an aqueous solution. This is probably due to a base-catalyzed hydrolysis reaction madiated by the amino group of the amphiphile.
Another key factor on the cellular toxicity of the cationic amphiphiles is their inhibitory effects on the activity of protein kinase C (PKC). PKC is a key enzyme which plays a crucial role in cellular signal transduction.
Cationic amphiphiles inhibit PKC activity by mimicking the endogenous inhibitor, sphingosine. PKC activity is also important for the cellular endocytosis pathway which is WO93/05162 PCT/U~92/072~
2 11 ~ G 7 G

likely to be involved in the action of the cationic amphiphiles to facilitate the entry of ~NA into cells.
Recently it has been reported that a PKC a~tivator, phorbolmyristateacetate, can stimulate the transfection efficiency of DNA mediated by the calcium phosphate precipitates.
The present inventors have therefore synthesized a series of novel cationic amphiphiles and screened their activities to inhibit PKC. Several amphiphiles which exhibit weak inhibitory activities towards PKC are particularly suitable for transfections. In addition, there has been prepared cationic reagents with a carbamoyl linker bond in order to overcome the problem of instability in solution. The stability of the bond in aqueous solution is much greater than that of the ester bond, yet it is hydrolyzable in the cell.
In brief, the present invention provides a method for facilitating the transfer of nucleic acids into cells.
The method comprises preparing a mixed lipid dispersion of a cationic lipid with a co-lipid in a suitable carrier solvent, such as distilled water or normal saline solution.
The cationic lipid has a structure which includes a lipophilic group derived from cholesterol, a linker bond, a spacer arm including from about l to about 20 carbon atoms in a branched or unbranched linear alkyl chain, and a cationic amino group. The amino group is selected from the group consisting of primary, secondary, tertiary and quaternary amino groups. The method further comprises W093/05162 PCT/US92/072~
~11(;~76 adding the nucleic acids to the dispersion to form a complex. The cells are then treated with the complex.
In a preferred embodiment of the invention, the dispersion has particles with an average diameter of about 150nm. The cationic lipid is preferentially selected from the group consisting of cholesteryl-3~-carboxyl-amidoethylenetrimethylammonium iodide, l-dimethylamino-3-trimethylammonio-DL-2-propyl-cholesteryl carboxylate iodide, cholesteryl-3~-carboxyamidoethyleneamine, cholesteryl-3~-oxysuccinamidoethylenetrimethylammonium iodide, l-dimethylamino-3-trimethylammonio-DL-2-propyl-cholesteryl-3~-oxysuccinate iodide, 2-[(2-trimethlyammonio)-ethylmethylamino] ethyl-cholesteryl-3~-oxysuccinate iodide, 3~[N-(N', N'-dimethylaminoethane)-carbamoyl~cholesterol, and 3~-~N-(polyethyleneimine~-carbamoyl]cholesterol. In a preferred embodiment, the co-lipid is a neutral or acidic phospholipid which may be preferentially selected from the group consisting 'of phosphatidyl choline and phosphaticlyl ethanolamine.
In addition, the present invention also provides a substantially non-toxic,substantially non-hydrolyzable cationic lipid for facilitating the transfer of nucleic acids into cells. The lipid comprises a lipophilic group derived from cholesterol, a linker bond, a spacer arm including from about 1 to about 20 carbon atoms in a branched or unbranched linear alkyl chain, and a cationic amino group. The amino group is selected from the group comprising primary, secondary, tertiary or quaternary amino groups.

WO 93/05162 2 1 1 ~ ~ 7 G Pcr/US92/07290 The cationic lipid is preferably selected from the group consisting of cholesteryl-3~-carboxyamidoethylenetrimethylammonium iodide, l-dimethylamino-3 -trimethylammonio-DL-2-propyl -cholesteryl carboxylate iodide, cholesteryl-3~-carboxyamidoethyleneamine, cholesteryl-3,~-oxysuccinamidoethylenetrimethylammonium iodide, l-dimethylamino-3 -trimethylammonio-DL-2 -propyl-cholesteryl-3~-oxysuccinate iodide, 2 - t ( 2-trimethyl-ammonio) ethylmethylamino] ethyl-~holesteryl-3~-oxysuccinate iodide, 3,B ~ N- ( N ', N ' dimethyl aminoethane ) -carbamoyl ] -cholesterol, and 3,~ [N- (polyethyleneimine) -carbamoyl ] cholesterol .
The present invention may be better understood by reference to the following Examples when considering in conjunction with the drawings in which: `
FIGUR~ 1 is the synthetic scheme for cholesteryl carboxylate analogues;
FIGURE 2 is the synthetic scheme f or cholesteryl hemisuccinate analogues;
FIGURE 3 is the synthetic scheme for cholesteryl formate analogues;
FIGURE 4 is a graph of the effect of different co-lipids on the transfection activity of a cationic lipid dispersion in L929 cells;
FIGURE 5 is a graph of the effect of the ratio of co-lipid to a cationic lipid of the present invention on the transfection activity in L929 cells;

WO93/0~162 PCT/US92/07290 2 ~ 1 G ~7 (;

FIGURE 6 is a graph of the effect of lipid dose on the transfection activity in L929 cells;
FIGURE 7 is a graph of the effect of DNA dose on the tr~nsfection activity of the lipid dispersion in L929 cells;
FIGURE 8 is a representation of a gel showing complex formation of DNA with the cationic lipid dispersion: and FIGURE 9 is a graph of the transfection efficiency and toxicity of a cationic lipid of the. present invention.
In order to facilitate a further understanding of the present invention, the fo~lowing Examples are given primarily for the purposes of illustrating certain more specific details thereof.
MatexiLals:
Cholesterol (99 ~ ~ grade), cholesterol hemisuccinate, 1,1'-carbonyldiimidazole, were purchased from Sigma Chemical Co., St. Louis, MO. Magnesium powder-50 mesh (99 + ~), thionyl bromide ~97%), 1,3--propane sulfone (99%), iodomethane (99%), trans-1,2-dichloroethylene (98%), M,M-dimethylaniline (99~), N,N-dimethylethylenediamine (95%), 1,3-bis-dimethylamino-2-p r o p a n o l ( 9 7 % ) , 2 - ( ~ 2 -(dimethylamino)ethyl~methylamino)ethanol (98%), were obtained from Aldrich Chemical Co., Milwaukee, WI.
Cholesteryl chloroformate (95%), and polyethyleneimine were obtained from Fluka. Methanol, dichloromethane, and WO93/05162 2 1 1 6 ~ 7 ~ PCT/USg2/072gO

acetonitrile were HPLC grade solvents. All other chemicals and solvents, unless specified were reagent grade.
A synthetic scheme for cholesteryl carboxylate analogues is shown in FIGURE 1.
EXAMPLE I
Cholestervl Bromide (I):
Cholesterol, (25 g, 64.6 mmol) was dissolved in mL of dimethylaniline (78.9 mmol) and 5 mL of chloroform. While stirring on ice; small quantities of thionyl bromide (~ mL, 77.6 mmol) dissolved in 20 mL of cold chloroform was added slowly over a period of 15 minutes. After the addition of thionly bromide was comple~e, the mixture was stirred for an additional 2 hours at room temperature. The resulting solution was poured into 200 mL of ice cold 95% ethanol and left on ice for 2 hour until crystallization was complete. The product was filtered and washed with 25 mL of ice cold 95% ethanol.
A small amount of product was recovered from the filt~ate with the addition of 75 mL distilled water followed by refrigeration. Finally, the product was recrystallized from 120 mL of acetone gi~ing 21.8 g of cholesteryl bromide (yield, 75%) with a melting point of 93-9S%C (lit 97-98-C).
The identity of the product was confirmed with mass spectrometry (EI) which showed an intense peak with an m/z of 448, corresponding to the molecular ion (~ ) of cholesteryl bromide. Also, the bromide molecular weig~t pattern characteristic of the two different isotopes of bromine (79Br:81Br,1:1) was observed.

r u ~ ~

EXAMPLE II
Cholest-5-ene-3~-Carboxylic Acid ~
The synthesis of cholesteryl-3~-carboxylate was performed using a Grignard reaction. All glassware was oven dried at 110C overnight. In a 500 mL three-neck flask set up for reflux, a solution of methyl magnesium iodide was freshly prepared by treating 9 g of oven dried (llOC) magnesium powder in 100 mL anhydrous diethyl ether with 10 mL of methyl iodide. After the vigorous reaction subsided, cholesteryl bromide (25 9, 56 mmol) dissolved in 100 mL of anhydrous diethyl ether was slowly added to the methyl magnesium iodide solution over a three hour period.
The solution was refluxed for 36 hours with enough heat required to bring the diethyl ether to a boil. Subsequent to cooling, the Grignard reagent was added to finely ground solid carbon dioxide, and after 1 hour, the complex was hydrolyzed by treatment with ice cold 1 M sulfuric acid.
After the steroid was extracted with diethyl ether (3 x'250 mL), the ethereal layer was washed with 10 mM sodium thiosulfate (3 x 50 mL) to remove a persistent orange color. After removing the water layer, the ether layer was washed with distilled water and filtered to remove an insoluble residue. The ether layer was subsequently dried over anhydrous sodium sulfate and rotary e~aporated to give a white-yellow oily suspension. Tituration with pentane yielded 8~6 g of cholesteryl-3~-carboxylate (yield, 37%) as a fine powder with a melting point of 212-215-C (lit 218-220 C). Mass spectrometry (EI) showed an m/z of 414 of the molecular ion (M~). The product was characterized by W093/05162 2 1 ~ G 6 7 6 PCT/USg2/07290 2proton NMR. The product was lyophilized overnight to give an anhydrous starting material for acylation reactions.
EXAMPT~ III
Cholestervl-3~-CarboxYamidoe~h~lenedimethvlamine (III~:
The acylation of cholesteryl carboxylate was carried out under a dry argon or nitrogen atmosphere in oven dried glassware. Cholesterol carboxylate (2 g, 4.8 mmol) was suspended in S mL of dichloromethane (HPLC grade under 4 A molecule sieves). A 1.5 molar excess of 1,1'-carbonyldiimidazole (CDI, 1~2 g) dissolved in 15 mL
dichloromethane was added to the cholesteryl carboxylate suspension is small volumes with intermittent shaking.
When the reaction subsided, the solution was stirred overnight. N,N-dimethylethylenediamine (5 mL, 43.2 mmol) was subsequently added and the resulting solution was stirred for 36 hours at room temperature. Dichloromethane was removed by rotary evaporation, after which the rea~tion was quenched with a small volume of distilled water. 'The acylated steroid was extracted with diethylether (4 x 50 mL). Subsequently, the pooled ether fractions were back extractPd with distilled water (3 x 50 mL), dried over anhydrous sodium sulfate, and rotary evaporated under reduced pressure. The residue was then triturated with pentane and the product collected on a sintered glass funnel. A voluminous powder ~1.7 g, 73% yield) was obtained and found to be pure by TLC (Rf=0.72) using chloroform:methanol:water (65:25:4,v/v/v) as the developing solvent. The product gave a melting point of 167-169-C.
Mass spectrometry (FAB~) showed an intense peak at an m/z 2~i6(;7~

of 485 which corresponds to the protonated molecular ion (M+H)~. The product was characterized by proton NMR.
EXAMPLE IV
Cholestervl--3~-Carboxvamidoethvlenetrimethvlammonium Iodide (IV):
The quaternization of Compound III was performed using methyl iodide and potassium bicarbonate. Briefly, 1 g (2.1 mmol) of compound III was dissolved in 40 mL of methanol in the presence of 2 g (20 mmol) of potassium bicarbonate and 2 mL (32.1 mmol) of methyl iodide. The reaction was stirred for 24 hours at room temperature. The solvent was subsequently removed under vacuum and the remaining bicarbona~e was neutralized with 1 M HCl until the solution gave a pH reading of 7. Water was removed by lyophilization and the product was extracted from inorganic salt impurities using a small volume of ice cold methanol.
After evaporating the solvent, the product was recrystallized from absolute ethanol and was fur~her purified on a reverse phase column using an acetonitrile/0.1% trifluoroa~etic acid gradient (100~ to 85% acetonitrile in 60 minutes). The powder was shown to be pure with TLC (RfzO.10) using chloroform:methanol:water (65:25:4 v/v/v~ as the developing solvent. It was shown to melt with decomposition at about 190-C, and had a molecular ion with an m/z of 500 (~ ) according to mass spectrometry (FAB+). The product was characterized by proton NMR.

WO93/05162 2 1 1 6 S 7 ~ PCT/US92/07290 EXAMPLE v 1.3-Bis-DimethylaminQ-2-Pro~Yl-choleste~yl-3~-çarboxylate (V):
Acylation was performed using CDI activated cholesteryl-3~-carboxylate analogous to the method described for compound III, except that 2,3-bis-dimethylamino-2-propanol (8 mL, 47.6 mmol) was the nucleophile. After the addition of the nucleophile, the reaction was stirred at room temperature for 72 hours. The dichloromethane was removed and the remainin~ oily residue was dissolved in chloroform. Impurities precipitated with a large volume of petroleum ether (bp, 35-60 C). The filtrate was rotary evaporated to dryness, re-dissolved in pentane, and filtered once again. After drying, the pentane soluble material was dried and re-dissolved in a small volume of diethyl ether and added to a large volume of hot diethyl ether:acetonitrile (30:70, v/v). The product crystallized at -20~C after allowing some of'the ether to evaporate. Mass spectroscopy (FAB+) gave an m/z of 543 for the protonated molecular ion (M+H)+'. The product was characterized by proton NMR.
EXAMPLE VI
l-Dimethylamlno-3-Trimethvlamm.onio-DL,-2-Propyl Cholesteryl Carboxvlate Iodide Salt fVI):
The methoidide of compound y was prepared by gently refluxing coumpound V (0.5 g, 0.9 mmol) and methyl iodide (2 mL, 32.l mmol) in 20 mL of ethanol for one hour.
After cooling, the precipitate (0.5 g, yield 79%) was recrystallized twice from absolute methanol. The product 211~S7G

melted with decomposition at about ~32 C and ran as a single spot on a TLC plate (Rf=0.22) using chloroform:methanol:water (65:25:4, v/v/v) as the developing solvent. The product had a molecular ion with an m/z of 557 (~ ) with FAB~ mass spectroscopy, consistent with the alkylation of one of the possible two tertiary amine sites. The product was characterized by proton NMR.
EXAMPLE VII
Cholester~1-3~-Carboxyamidoethyleneamine (VII~:
To a solution of ethylenediamine ~S.ll g, 85 mmol) in 20 mL dichloromethane, a solution of CDI activated cholesteryl carboxylate (O.7 g, 1.7 mmol) in 5 mL of dich~oromethane was added dropwise over a 1.5 hour period.
When the addition of the activated sterol was complete, the reaction was stirred for 48 hours under nitrogen. After removing the solvent under reduced pressure, the residue was dissolved in chloroform:methanol (2:1, v/v) and extracted against water (3 x 50 mL). The chloroform p~ase was subsequently dried with anhydrous sodium sulfate, the solvent removed and the residue purified by preparative TLC
using chloroform:methanol:water (65:25:4l v/v/v) as the developing solvent. The band at about Rf = 0.3 was collected, extracted with chloroform:methanol (1:1, v/v) and dried under reduced pressure. The product (0.65 g, yield, 81%) ran as a single spot (Rf = 0.33) and melted with decomposition at about 194'C. Mass spectrometry (FAB~) gave an m/z of 457 for the protonated molecular ion (M+H)~. The product was characterized by proton NMR.

WO93/0~162 2 1 1 6 6 7 6 PCT/US92/07290 A scheme describing the various steps for producing cholesterol hemisuccinate analogues is depicted in FIGURE 2.
EXAMPLE VIII
Cholestervl-3~-Oxysuccinamidoethyle~edimethYlamine VIII:
The synthesis of compound VIII first required the acyl imidazolide of cholesteryl hemisuccinate which was prepared by reacting cholesterol hemisuccinate with N,N-ca~bonyldiimidazole (CDI) as described for the synthesis of compound III. Briefly, to cholesterol hemisuccinate (2 g, 4.1 mmol) suspended in 5 mL of dichloromethane was added 1.5 equivalents of CDI (1 g~ dissolved in 15 mL of dichloromethane. The solution was stirred overnight after which N,N-dimethylethylenediamine (5 mL, 43.2 mmol) was added. Dichloromethane was subsequently removed by rotary evaporation, distilled water was added and the acylated sterol was extracted with diethyl ether (4 x 50 mL).
Subsequently, the ether fractions were washed ~ith distilled water (3 x 50 mL) and dried over anhydrous sodium culfate. The ether was removed by rotary evaporation. The product was washed with 200 mL of pentane, and minor impurities were removed using preparative silica gel TLC.
After developing with chloroform:methanol:water (65:25:4), v/v/v) the band present at about an Rf = 0.80 was collected and extracted with chloroform/methanol ~2:1 v~v). The residue was purified further using chloroform:ethyl acetate (1:1, v/v) as the second developing solvent. The band at about Rf = 0.2 was extracted with chloroform/methanol (2:1 v/v). The lyophilized product ran as a single spot on TLC

o ~ ~ r r with an Rf of o.75 using chloroform:methanol:water (65:25:4, v/v/v) as the developing solvent and had a melting point of 11g-111 C. Mass spectrometry (FAB+) showed an m/z of 557 which would correspond to the protonated molecular ion (M+H)~. The product was characterized by proton NMR.
EXAMPLE IX
Cholesteryl-3*-Oxvsuccinamidoethylenetrimethvlammonium ~odide (IX):
The quaternization of compound VIII was carried out with methyl iodide in absolute ethanol as described earlier for the synthesie of compound VI. Allowing the solution to cool to room temperature afforded 0.5 g (80%
yield) of the quaternary ammonium salt. Subsequently, the product was recrystallized from absolute ethanol giving a fine white powder which melted ~ith decomposition at about 196-C. The product ran as a single spot on a TLC plate (R
= 0.43) using chloroform:methanol:water (65:25:4) as 'the developing solvent. Mass spectrometry (FA+) indicated a molecular ion with an m/z of 572 (M~ ). The product was characterized by proton NN.
EXAMPLE X
1.3~B,i-Dimethylamino-2-Propyl-CholesterYl-3p~-oxysuccinate (X):
Acylation was performed using CDI activated cholesteryl hemisuccinate according to the procedure described earlier for compound V. After the addition of 1,3-bi-dimethylamino-2-propanol (7 mL, 41.6 mmol), the mixture was stirred for 72 hours, after which the solvent WO93~05162 2 1 1 6 ~ 7 6 PCT/US92/07290 was removed under vacuum. The product, extracted from the residue with diethyl ether (3 x 75 mL), gave an oil following removal of the ether. The addition of pentane precipitated additional impurities; after rotary evaporation, the resulting oil could not be successfully crystallized using a variety of solvents or by lyophilization. Mass spectrometry (FA+) indicated a protonated molecular ion with an m/z of 616 (M+H)~. the product was characterized by proton NM.
EXAMPLE XI
l-Dimethylamino-3-Trimethylammonio-DL-2-PropYl-CholesterYl-3B-Oxvsuccinate Iodide Sal~ (XI:
Methylation of compound X was performed using the method described pre~iously (Example VI). After l hour, the solution was cooled and the methiodide recrystallized twice from absolute methanol to give needle shaped crystals which melted with decomposition at about 222 C. The product ran as a single spot on a TLC plate (R = 0~17) using chloroform:methanol:water (65:25:4, v/v~v) as the developing solvent. Mass spectrometry tFA+) indicated a molecular ion with an m/z of S29 (M~) consistent with the methylation of l of a possible 2 tertiary amine sites. The product was characterized by proton NM.
EXAMPLE XII
2-~ r 2-~dimeth~lam m o)ethyl1methylamino~ethyl-CholesterYl-3B-OxYsuccinate (XII):
The synthesis of compound XII was analogous to the method described for the acylation of compound VIII
except that 2-~2-dimethylamino)ethyl]-methylamino)ethanol 211~r)7G 1~

(7 mL, 42.o mmol) was the amino alcohol used as the nucleophile. After extraction with diethylether, the product was lyophilized dry and further purified by preparative TLC using chloroform:methanol:water (65:2S:4, v/v/v). After the band present at R = 0.80 was collected and extracted with chloroform:methanol, (2:1, v/v), the residue was purified further using chloroform:ethyl acetate (1:1; v/v) as the second TLC developing solvent. The band which was present at about R = 0.2 was collected and the silica was extracted with chloroform:methanol (2:1, v/v).
The product, which ran as a single spot on a TLC plate (R
s 0.72) using chloroform:methanol:water (65:2s:4 v/v/v) as the developing solvent gave a melting point of 50-52'C.
Mass spectrometry (FA+) showed a protonated molecular ion with an m/z of 615 (M~H)~. The product was characterized by proton NMR.
~XANPLE~
2- ~ r 2-TrimethYlammoniolet~vlmçthylamino)ethyl-Cholester~l-3~-Oxvsuccinate Iodine Salt lXIII~:
The acylation of compound ~II (O.5 g, 0.8 mmol) was carried out under reflux conditions with methyl iodide in absolute ethanol as described in Example VI. The precipitate was recrystallized twice from absolute methanol and stained as a single spot on a TLC plate (~ = 0.22) using chloroform:methanol:water (65:25:4; v/v/v) as the developing solvent. The crystals melted with decomposition at about 172'C. Mass spectrometry ~FA~) gave an m/z of 629 for the molecular ion (~') consistent with the methylation WO93/0516~ PCT/US92/07290 2~1~S76 of only one the possible two tertiary amine sites. The product was characterized by proton NM.
The scheme for the synthesis of cholesteryl formate analogues is shown in FIGURE 3.
EXAMPLE XIV
3~rN-(N'.N'-dimethylaminQethane)-carbamoyllcholesterol (XIV):
Compound XIV was synthesized by mixing a solution of cholesteryl chloroformate (0.5 mmol) in chloroform with a solution of N,N-dimethylethylenediamine (9.1 mmol) in chloroform in a dry ice-ethanol bath. The solvent and the unreacted amine were removed in vacuo. Compound XIV was purified by two successive recrystallizations in ethanol.
(Yield, 65%) TLC (chloroform;methanol=65:35) showed a single spot (R = 0.37) when developed with iodine. The product was characterized by proton NM.
EXAMPLE XV
3~ r~- (~olyethYlenei~ine)-~arbamovllcholesterol XV): ' Synthesis of compound XV was similar to that of compound XIV. Cholesterol chloroformate (O.1 mmol) and polyet~yleneimine 600 (6 g) were mixed in chloroform in a dry ice-ethanol bath. After the volatile material of the reaction mixture was removed in vacuo, the solid crude product was dialyzQd against 4L distilled water for 3 days (during which the water was changed several times).
Finally, the product was lyophilized to dryness, giving an estimated yield of 81%. Compound XIV ran as a single spot on TLC (chloroform:methanol=65:35).

WO93/05162 PCT/US9~/07290 211~76 EXAMPLE XVI
PreDaration of cationic li~id dis~ersions:
cationic cholesterol derivatives were mixed with a phospholipid in chloroform solution at different molar ratios. The solvent was removed by evaporation under a stream of N2 gas and desiccated in vacuo for at least 30 minutes. The dry lipid film was hydrated in 20 mM Hepes buffer, Ph 7.8, overnight. The suspension was sonicated in a bath-type sonicator (Laboratory Supplies, Hicksville, NY) to generate small particle dispersions (average diameter =
150 nm).
EXAMPLE XVII
Transf~ction of cells:
Plasmid pUCSV2CAT (approximately 5kb in size) containing the structural gene of E. coli chloramphenicol acetyl transferase (CAT) driven by the SV40 virus early promoter was used as a model for the polyanions to be delivered by the cationic lipid dispersions. DNA was miXed with cationic lipid dispersions in 1 ml serum-free M199 medium or McCoy's medium to form DNA~lipid complex.
Cultured mammalian cells of about 80-100% confluency in a 6-well plate were washed once with serum-free medium. The DNA/lipid complex was added to the washed cells which were incubated at 37'C for 5 hours. The cells were washed again and the serum-containing medium was added. Cells were harvested 30-72 hours later and extracted for cellular proteins. The CAT activity in the extracted protein was measured by using either rl4C] chloramphenicol or [3H~ acetyl CoA as a radiolabeled substrate. One activity unit of CAT

WO93~05162 ~ 6 7 6 PCT/US92/07290 is defined as nmole of radiolabeled substrate converted to the radiolabeled product in one minute. Protein content in the cell extracts was measured by the Bradford (BIORAD) assay.
EXAMPLE XVIII
Isolation of ~rotein kinase C:
As rapidly as possible, brains for 25 Sprague-Dawley rats (150-200 g) were removed, washed with 100 mL of 20 mM TRIS, 1 mM EDTA, lmM EGTA, Ph 7.5, and homo~enized in 150 mL of ice cold 20 mM TRIS, 10 mM EGT~, 2 mM EDTA, 10 mM
DTT, 0.2S M sucrose, 2 mM PMSF and 100 ~g/mL leupeptin, pH
7.5. The homogenate was immediately centrifuged at 100,000 g for 40 minutes at 4' C in a Beckman Ti 50 . 2 rotor. The supernatant was applied to a 2~ 5 X 20 cm column of DEAE
Sepharose (fast flow) containing 60 mL of resin equilibrated with 20 mM TRIS, 1 mM EDTA, ? mM DTT, pH 7.5 (buffer A). The column was washed with 300 mL of buffer A
and an additional 200 mL of buffer A containing 0.03 M kCl.
Protein kinase C was eluted with a 500 mL continuous KCl gradient (0.03 - 0.3 M KCl). Fractions of 5 mL volumes were collected. Fractions showing calcium and phospholipid dependence were pooled; the salt concentration was adjusted to 1.5 KCl with the appropriate quantity of solid KCl. The crude sample containing 1.5 M KCl was stirred for 15 minutes and subsequently loaded onto a 1 X 10 cm column c.ontaining 9 mL Phenyl separose equilibrated with 1.5 M XCl in 20 mM TRIS, 0.5 mM EGTA, 1 mM DTT, pH 7.5 (buffer B).
The column was washed with 90 mL of buffer B containing 1.5 M KCl. PKC was eluted with a 100 mL continuous KCl 211~o76 gradient (1.5 - 0 M KCl). Fractions of 3 mL volumes were collected. The column was washed with an additional 50 mL
of buffer B. Most of the enzyme activity eluted during this stage. Fractions showing calcium and phospholipid dependence were pooled and concentrated to 4 mL using an Amicon ultrafiltration cell fitted with a YM-10 filter.
The concentrated sample was loaded onto a 2.5 X 100 cm column containing 400 ml of Sephacryl S-200 HR beads equilibrated with buffer B containing 10% glycerod (buffer C). Fractions of 3 mL volumes were collected. About 150 mL
of buffer was run through; PKC eluted very close to the column void volume. The fractions showing calcium and phospholipid dependence were pooled and loaded onto a 0.5 x 5 cm column containing 2.5 mL polylysine agarose equilibrated with buffer C. PKC was eluted with a 40 mL
continuous KCl gradient (0-0.8 M KCl). Fractions of 1 mL
volumes were collected. The first few active fractions were contaminated. The uncontaminated fractions were pooled, concentrated, and diluted with buffer C to remove the high salt content. After reconcentrating, the sample was divided into working portions, frozen in liquid nitrogen and stored at -80-C. Full activity was regained after rapid thawing. Trace impurities (116 k, 66 k, and 50 k Mr) could still be detected when the gel was silver stained heavily. The enzyme gave a specific activity of 200 nmoles phosphate incorporated per minute per milligram of protein when assayed for histone phosphorylation using the Triton mixed micelle assay with 6.5 mole %
phosp~atidylserine, 2.5 mole % nAG and 100 ~M calcium WO93/05162 ~ 1 1 6 S 7 ~ PCT/US92/07290 present. Specific activities ranging from 30 nmoles/min/mg to 600 nmoles/min/mg have been observed for PKC using the Triton mixed micelle assay under the same conditions.
EXAMPLE XIX
Mixed micelle assav of ~rotein kinase C:
Phosphatidylserine and 1,2-diolein with and without additive were dissolved in a solution of chloroform/methanol (2:1, v/v). Solvent was evaporated with a stream of nitrogen and last traces removed using a vacuum desiccator at 40-C. The lipid films were then solubilized by the addition of 3% Triton X-100, vortexed vigorously for 30 seconds and then incubated at 30 C for 10 minutes to allcw for equilibration. At 25 ~L, an aliquot of this solution was used in a final assay volume of 250 ~L, containing 20 mM TR~S-HCl, pH 7.5, 10 mN MgCL2, 200 ~g/mL histone III-S, 100 ~M CaCl2, 10 ~N~y-32P~ adenosine 5' triphosphate, 2.75 mM Triton X-100, with 300 ~M (6.5 mole percent) phosphatidylserine and 107 ~M (2.5 mole per~ent 1,2-diolein. For controls, 25 ~L of 20 mM EGTA replaced the CaCl2. To initiate the reaction, 150 ng of protein was added. After briefly mixing, the tubes were incubated for 10 minutes at 30C. The reaction was terminated by adding 1 mL of cold 0.5 mg/mL BSA and 1 ~L of cold 25%
trichloroacetic acid. This mixture was passed through a GF/C Whatman filter and washed five times with 2 mL of 25%
trichloroacetic acid. After drying, the filters were counted with 6 mL ACS scintillation fluid.

WO93/05162 PCT/US92/072gO
2 1 1 ~3 ~ 7 6 24 EXAMPLE XX
Formation of homoqenous _ disPersion with cationic cholesterol derivatives None of the cationic cholesterol derivatives by themselves form stable homogenous dispersion by sonication in a low io~ic strength buffer. It was necessary to add a phospholipid, acidic or neutral, to form mixed lipid dispersion~ For example, compound VIII requires a minimal of 1 part of PC or PC and 9 parts of compound VIII to form a uniform dispersion. In the case of compound XIV, a minimal ration of phosphatidyl choline (PC) or phosphatidyl ethanolamine (PE) to XIV = 4:6 is required. Such non-cationic lipid used in the dispersion is called co-lipid.
EXAMPLE XXI
Delivery of DNA into mammalian ~ells bv cationic lipi~
dis~ersions Plasmid DNA, pUCSV2CAT, was used as a model compound for polyanions because it contains a structu~al gene for CAT. The efficiency of intracellular delivery can be readily assayed by the expression of C~T activity in the extracted proteins of the treated cells. Table l lists the CAT activity of mouse L929 cells which have been transfected with this plasmid DNA as mediated by various c~tionic lipid dispersions. In addition, we have also measured the inhibitory activity of the pure cationic cholesterol derivatives on diolein, phosphatidyl serine (PS), and Ca2~ stimulated protein kinase C. This activity was expressed as an ICSo, which is the concentration at which 50% of PXC activity was inhibited. As can be seen W O 93/05162 2 1 1 ~ S 7 o PCT/US92/07290 from Table I, derivatives giving low ICso values, i.e., those strong PKC inhibitors, were not a good delivery vehicle for DNA. For example, compounds IV, XI, VI and XIII, all having a IC50 value less than 20 ~M, produced minimal CAT activities in the treated cells. Among the ones which gave rise to high CAT activities, derivatives with a single tertiary amino group (compounds VIII, VI and III) were more effective in delivering DNA than similar analogs containing a single quaternary ami~o group (compounds IX and IV). Furthermore, among the derivatives with the same amino head group, those containing a longer spacer arm (compounds VIII and IX) delivered a greater quantity of DNA than those containing a shorter spacer arm (compounds X, XI, V, VI and XV) were generally less effective delivery vehicles.
Compound VII deserves some special attention. It contains only a single primary amino group with a short spacer arm, yet the transfection activity was relati~ely high.
~ABLE I

PKC Inhibition Relative CAT
Com~ound IC.~uM) Activit~

IV 12 0.7 X 408 0.5 XIV __ 2 1 ~ 5 G76 EXAMPLE XXII
The im~ortance of the co-lipid The experiments described in Example XXI were done with a lipid dispersion containing a cationic cholesterol derivative and a co-lipid dioleoyl phosphatidylethanolamine (DOPE). We have studied the role of co-lipid in the delivery efficiency. FIGURE 4 shows the data of an experiment in which compound VIII was mixed with a variety of different co-lipid, neutral and acidic, at a molar ratio of l:l. The DNA delivery activity of these mixed dispersions were then studied. As can be seen, only DOPE supported the delivery activity of compound VIII.
Other neutral lipids such as dioleoyl phosphatidylcholine (NOPC), N-methyl-DOPE, N,N-dimethyl DOPE had little or no activity. None of the acidic lipids, such as PS and phosphatidylglycerol (PG) showed any activity.
The molar ratio of DOPE and compound VIII in the dispersion also played an important role. FIGURE 5 shows that maximal DNA delivery activity of the dispersion occurred when the dispersion contained 20-50% compound VIII. Too much or too little of compound VIII in the mixed dispersion did not yield ~ood delivery activity.
EX~MPLE XXIII
oDtimization Qf dis~ersion-to-DNA ratio for deliveEy A l:l mixture of compound VIII and DOPE were used to study the optimal ratio of dispersion-to-DNA for delivery. FIGURE 6 shows the data of an experiment in which various amounts of dispersion were added to a fixed amount of DNA (5 ~g) for transfection. Maximal activities WO93~05162 2 1 1 6 ~ 7 6 PCT/US92/07290 occurred at 69-80 nmoles of dispersion. We then used 70 nmoles dispersion and varied the amount of DNA for transfection (Fig. 7). The bell-shaped curve in the figure indicates that a 5 ~g DNA gave the maximal activity. Thus the optimal ratio of dispersion-to-DNA was 70 nmole lipid for 5 ~g DNA.

ComDlex formation of DNA with cationic lipid dispersions It was expected that polyanions complex with the cationic lipid dispersion via electrostatic interactions.
Again, a l:l mixture of compound VIII and DOPE was used for the study. We have characterized the dispersion/DNA
complexes by agarose gel electrophoresis. As shown in FIGURE 8, 1 ~g plasmid DNA electrophoresed as two closely located bands in the gel (lane l), which could be completely digested if DNAse was included in the incubation bu~fer (lane 7). Incubation mixtures containing increasing amounts of dispersion showed decreasing intensities of~DNA
bands (lanes 2, 3, 4, 5 and 6). Furthermore, all of the uncomplexed, free DNA could be digested by DNA se, but only a portion of the complexed DNA was digested (lanes 8, 9, lO, ll and 12). These results clearly showed that the lipid dispersion form complexes with DNA which are either larger in size and/or less negatively charged such that the complex does not enter the gel during electrophoresis.
Furthermore, the complex is partially resistant to DNAse, whereas the free, uncomplexed DNA is not. It should be noted that at the optimal dispersion/DNA ration nearly all DNA ware complexed with liposomes (not shown in FIGURE 8).

211~7~ ~

EXAMPLE ~XV
Relationshi~ between deliverY activitv and cytotoxicity of the cationic li~id complex This was studied by using a dispersion composed of compound XIV and DOPE ~3:2, molar ratio). A431 human epidermoid carcinoma cells were used for the transfection experiments. A fixed amount of DNA (4 ~g) was mixed with an increasing amount of cationic lipid dispersion or a commercially available transfection reagent, Lipofectin, and added to the A431 cells for tran fection (FIGURE 9).
The toxicity of the treatment to the cells was measured as the total amount of cellular protein extractable at the time of CAT activity assay. As can be seen from the Figure, Lipofectin treated cells showed a greatly reduced protein content with 50% inhi~ition occurring at about 7 ~g lipid/ml. Cells treated with the diæpersion `containing compound XIV and DOPE showed less toxicity; the I~o occurred at about 2S ~g lipid/ml. The novel cationic cholesterol dispersion had also produced higher CAT
activities than Lipofectin. It is important to note that maximal C~T activity of cells treated with Lipofectin occurred at the Lipofectin concentration of 15 ~g/ml. At this concentration only about 12% of the total cellular proteins could be recovered from the culture. On the other hand, maximal CAT activity of cells treated with the cationic cholesterol dispersion occurred at 20 ~g/ml; about 80~ of the total cellular protein still remained in the culture at this concentration. Thus, the novel cationic WO93/05162 ~ 1 1 6 6 7 6 PCT/US92/07290 cholesterol dispersion is more potent int he delivery activity and is also less toxic to the treated cells.
EXAMPLE XXVI
Stability of the cationic cholesterol derivatives Lipid dispersions were prepared with various cationic cholesterol derivatives and DOPE (about l:l molar ratio). The transfection activitie~ of the dispersions were tested at different times after the dispersions were stored at 4-C in PBS, pH 7.5. Of the derivatives listed in Table I, only the dispersions containing compounds XIV and XV were stable after storage: their transfection activities did not change for at least 2 months. On the other hand, the dispersions composed of other derivati~es lose activity after 2-3 days in storage. Compounds XIV and XV contain a carbamoyl linker bond whereas other compounds contain either an ester bond or an amide bond. It is known that ester and amide bonds are more sensitive than the carbamoyl bond to hydrolysis particularly in the presence of ba~es.
The cationic derivatives may catalyze the hydrolysis of each other's ester bonds, leading to the inactivat~on of the delivery actîvity. Compounds containing carbamoyl linker bonds are less sensitive to the base-catalyzed hydrolysis, yet they can still be hydrolysed by cellular enzymes, i.e., they are biodegradable. This is in contrast to the non-degradable ether bond in DOTMA which is the active ingredient of Lipofectin. Thus, a carbamoyl bond seems to be the best choice for the linker bond of the cationic lipids as a delivery vehicle for polyanions.

W O 93/05162 P ~ /US92/07290 21' 6076 Various of the features of the invention which are believed to be new are set forth in the appended claims.

Claims (6)

THE CLAIMS:
1. A method for facilitating the transfer of nucleic acids into mammalian cells, the method comprising:
preparing a mixed lipid dispersion of a cationic lipid with a co-lipid in a carrier solvent, the cationic lipid having a structure including a lipophilic group derived from cholesterol, a linker bond, selected from the group consisting of carboxy amides and carbamoyls, a spacer arm including from 1 to 20 carbon atoms in a linear branched or unbranched alkyl chain, and a cationic amino group selected from the group consisting of primary, secondary, tertiary and quaternary amino groups, wherein the cationic lipid is a weak protein kinase C (PKC) inhibitor and wherein the colipid is selected from the group consisting of phosphatidyl choline (PC), phosphatidyl ethanolamine (PE), and dioleoyl phosphatidyl ethanolamine (DOPE);
adding the nucleic acids to said dispersion to form a complex: and treating the cell with the complex.
2. The method of Claim 1 wherein said dispersion of the cationic lipid has particles with an average diameter of about 150 nm.
3. The method of Claim 1 wherein the carrier solvent is selected from the group consisting of distilled water, normal saline, and buffered saline.
4. The method of Claim 1 wherein the cationic lipid is selected from the group consisting of cholesteryl-3.beta.-carboxyamidoethylenetrimethylammonium iodide, cholesteryl-3.beta.-carboxyamidoethyleneamine, cholesteryl-3.beta.-oxysuccinamidoethylenetrimethylammonium iodide, 3.beta.[N-(N',N'-dimethylaminoethane)carbamoyl]cholesterol, and 3.beta.-N-(polyethyleneimine)-carbamoyl)cholesterol.
5. A substantially non-toxic cationic lipid for facilitating the transfer of nucleic acids into cells, the cationic lipid comprising;
a lipophilic group derivred from cholesterol;
a linker bond;
a spacer arm including from about 1 to about 20 carbon atoms in a linear branched or unbranched alkyl chain; and a cationic amino group selected from the group comprising primary, secondary, tertiary and quaternary amino groups.
6. The cationic lipid of Claim 5 selected from the group consisting of cholesteryl-3.beta.-carboxyamido-ethylenetrimethylammonium iodide, cholesteryl-3.beta.-carboxyamidoethyleneamine, cholesteryl-3.beta.-oxysuccinamidoethylenetrimethylammonium iodide, 3.beta.[N-(N',N'-dimethylaminoethane)carbamoyl]-cholesterol, and 3.beta.-{N- (polyethyleneimine)-carbamoyl}cholesterol.
CA002116676A 1991-08-28 1992-08-28 Method for delivering nucleic acids into cells Abandoned CA2116676A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/751,873 US5283185A (en) 1991-08-28 1991-08-28 Method for delivering nucleic acids into cells
US751,873 1991-08-28

Publications (1)

Publication Number Publication Date
CA2116676A1 true CA2116676A1 (en) 1993-03-18

Family

ID=25023883

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002116676A Abandoned CA2116676A1 (en) 1991-08-28 1992-08-28 Method for delivering nucleic acids into cells

Country Status (12)

Country Link
US (1) US5283185A (en)
EP (1) EP0663013B1 (en)
JP (2) JP3258661B2 (en)
AT (1) ATE292690T1 (en)
AU (1) AU665029B2 (en)
CA (1) CA2116676A1 (en)
DE (1) DE69233497T2 (en)
DK (1) DK0663013T3 (en)
ES (1) ES2240958T3 (en)
HK (1) HK1014736A1 (en)
SG (1) SG50630A1 (en)
WO (1) WO1993005162A1 (en)

Families Citing this family (440)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6509032B1 (en) * 1991-08-28 2003-01-21 Mcmaster University Cationic amphiphiles
US5795870A (en) * 1991-12-13 1998-08-18 Trustees Of Princeton University Compositions and methods for cell transformation
US5693769A (en) * 1991-12-13 1997-12-02 Transcell Technologies, Inc. Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
IL101241A (en) * 1992-03-16 1997-11-20 Yissum Res Dev Co Pharmaceutical or cosmetic composition comprising stabilized oil-in-water type emulsion as carrier
DE69333434T2 (en) * 1992-04-03 2005-03-03 The Regents Of The University Of California, Oakland SELF-ORGANIZING SYSTEM FOR THE ADMINISTRATION OF POLYNUCLEOTIDES CONTAINING AN AMPHIPHATIC PEPTIDE
US6806084B1 (en) * 1992-06-04 2004-10-19 The Regents Of The University Of California Methods for compositions for in vivo gene delivery
US5981175A (en) * 1993-01-07 1999-11-09 Genpharm Internation, Inc. Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
EP1624068A1 (en) * 1993-06-01 2006-02-08 Life Technologies Inc. Genetic immunization with cationic lipids
US5674908A (en) 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US5928944A (en) * 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US6075012A (en) * 1994-02-11 2000-06-13 Life Technologies, Inc. Reagents for intracellular delivery of macromolecules
US6989434B1 (en) * 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
NZ268146A (en) * 1994-04-12 1997-10-24 Liposome Co Inc Liposome compositions and medicinal uses
US5866135A (en) * 1994-04-21 1999-02-02 North American Vaccine, Inc. Group A streptococcal polysaccharide immunogenic compositions and methods
US5777153A (en) * 1994-07-08 1998-07-07 Gilead Sciences, Inc. Cationic lipids
FR2722506B1 (en) * 1994-07-13 1996-08-14 Rhone Poulenc Rorer Sa COMPOSITION CONTAINING NUCLEIC ACIDS, PREPARATION AND USES
US6468981B1 (en) * 1994-07-29 2002-10-22 Emory University Compositions and methods for targeting pharmaceutically active materials to cells containing androgen receptors
US5908635A (en) * 1994-08-05 1999-06-01 The United States Of America As Represented By The Department Of Health And Human Services Method for the liposomal delivery of nucleic acids
US5837533A (en) * 1994-09-28 1998-11-17 American Home Products Corporation Complexes comprising a nucleic acid bound to a cationic polyamine having an endosome disruption agent
US5785992A (en) * 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5753613A (en) * 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
FR2726764B1 (en) * 1994-11-14 1997-01-31 Pasteur Merieux Serums Vacc ADJUVANT FOR VACCINE COMPOSITION
US5939401A (en) * 1994-12-09 1999-08-17 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US5910487A (en) * 1994-12-09 1999-06-08 Genzyme Corporation Cationic amphiphiles and plasmids for intracellular delivery of therapeutic molecules
US6071890A (en) * 1994-12-09 2000-06-06 Genzyme Corporation Organ-specific targeting of cationic amphiphile/DNA complexes for gene therapy
US5767099A (en) * 1994-12-09 1998-06-16 Genzyme Corporation Cationic amphiphiles containing amino acid or dervatized amino acid groups for intracellular delivery of therapeutic molecules
US5747471A (en) * 1994-12-09 1998-05-05 Genzyme Corporation Cationic amphiphiles containing steroid lipophilic groups for intracellular delivery of therapeutic molecules
US6331524B1 (en) 1994-12-09 2001-12-18 Genzyme Corporation Organ-specific targeting of cationic amphiphile / DNA complexes for gene therapy
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5840710A (en) * 1994-12-09 1998-11-24 Genzyme Corporation Cationic amphiphiles containing ester or ether-linked lipophilic groups for intracellular delivery of therapeutic molecules
US5719131A (en) * 1994-12-09 1998-02-17 Genzyme Corporation Cationic amphiphiles containing dialkylamine lipophilic groups for intracellular delivery of therapeutic molecules
US6383814B1 (en) 1994-12-09 2002-05-07 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5650096A (en) * 1994-12-09 1997-07-22 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5786214A (en) * 1994-12-15 1998-07-28 Spinal Cord Society pH-sensitive immunoliposomes and method of gene delivery to the mammalian central nervous system
WO1996020208A2 (en) * 1994-12-28 1996-07-04 Max-Delbrück-Centrum für Molekulare Medizin New cholesterol derivative for liposomal gene transfer
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5830430A (en) 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
CN1136920C (en) 1995-02-28 2004-02-04 加利福尼亚大学董事会 Gene transfer-mediated angiogenesis therapy
US6752987B1 (en) 1995-02-28 2004-06-22 The Regents Of The University Of California Adenovirus encoding human adenylylcyclase (AC) VI
US20030148968A1 (en) * 1995-02-28 2003-08-07 Hammond H. Kirk Techniques and compositions for treating cardiovascular disease by in vivo gene delivery
US5658588A (en) * 1995-03-31 1997-08-19 University Of Cincinnati Fibrinogen-coated liposomes
FR2732895B1 (en) * 1995-04-11 1997-05-16 Pasteur Merieux Serums Vacc USE OF A CATIONIC AMPHIPATHIC COMPOUND AS A TRANSFECTING AGENT, AS A VACCINE ADDITIVE, OR AS A MEDICINAL PRODUCT
US5859228A (en) * 1995-05-04 1999-01-12 Nexstar Pharmaceuticals, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
JPH11508231A (en) * 1995-05-26 1999-07-21 ソマティックス セラピー コーポレイション Delivery vehicles containing stable lipid / nucleic acid complexes
AU701106B2 (en) * 1995-06-07 1999-01-21 Promega Biosciences, Inc. Novel carbamate-based cationic lipids
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5753262A (en) * 1995-06-07 1998-05-19 Aronex Pharmaceuticals, Inc. Cationic lipid acid salt of 3beta N- (N', N'-dimethylaminoethane) - carbamoyl!cholestrol and halogenated solvent-free preliposomal lyophilate thereof
AU723163B2 (en) * 1995-06-07 2000-08-17 Tekmira Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
CA2223179A1 (en) * 1995-06-07 1996-12-19 Bob Dale Brown Phosphonic acid-based cationic lipids
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US5981501A (en) * 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5705385A (en) * 1995-06-07 1998-01-06 Inex Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US6120794A (en) * 1995-09-26 2000-09-19 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
AUPN741696A0 (en) * 1996-01-05 1996-01-25 Commonwealth Scientific And Industrial Research Organisation Delivery of nucleic acids ii
US5789244A (en) * 1996-01-08 1998-08-04 Canji, Inc. Compositions and methods for the treatment of cancer using recombinant viral vector delivery systems
US20040014709A1 (en) * 1996-01-08 2004-01-22 Canji, Inc. Methods and compositions for interferon therapy
US6392069B2 (en) 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
US7002027B1 (en) 1996-01-08 2006-02-21 Canji, Inc. Compositions and methods for therapeutic use
FR2744453B1 (en) * 1996-02-05 1998-07-10 Transgene Sa LIPOPOLYAMINE COMPOUNDS, PHARMACEUTICAL COMPOSITIONS COMPRISING SAME, NUCLEIC ACID TRANSFER VECTORS AND PREPARATION METHOD
FR2745569B1 (en) * 1996-03-01 1998-05-07 Centre Nat Rech Scient COMPOUNDS RELATED TO THE AMIDINIUM FAMILY, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND THEIR APPLICATIONS
FR2751972B1 (en) * 1996-07-30 1998-09-04 Centre Nat Rech Scient COMPOUNDS RELATED TO THE AMIDINIUM FAMILY, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND THEIR APPLICATIONS
IL125926A (en) * 1996-03-01 2004-09-27 Centre Nat Rech Scient Compounds related to the amidinium family, pharmaceutical compositions containing same and uses thereof
US6432635B1 (en) 1996-03-21 2002-08-13 Helsinki University Licensing Ltd. Oy Cystatin B mutants
US6258792B1 (en) 1996-04-12 2001-07-10 University Of Pittsburgh Cationic cholesteryl derivatives containing cyclic polar groups
EP0904282B1 (en) * 1996-04-12 2001-12-05 University Of Pittsburgh Novel cationic cholesteryl derivatives containing cyclic polar groups
DE69727384T2 (en) * 1996-05-24 2004-11-04 IC-VEC Ltd. POLYCATONIC STERIN DERIVATIVES FOR TRANSFECTION
US5935936A (en) * 1996-06-03 1999-08-10 Genzyme Corporation Compositions comprising cationic amphiphiles and co-lipids for intracellular delivery of therapeutic molecules
AU736143B2 (en) * 1996-06-10 2001-07-26 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US6093816A (en) 1996-06-27 2000-07-25 Isis Pharmaceuticals, Inc. Cationic lipids
JP2001503735A (en) * 1996-07-03 2001-03-21 ユニバーシティ オブ ピッツバーグ Emulsion formulation for hydrophilic active reagent
US6750010B1 (en) 1996-08-23 2004-06-15 The Regents Of The University Of California Methods for treating bipolar mood disorder associated with markers on chromosome 18p
CA2264140A1 (en) 1996-08-26 1998-03-05 Transgene S.A. Cationic lipid-nucleic acid complexes
CA2268365A1 (en) * 1996-10-18 1998-04-30 Jeff Nordstrom Il-12 gene expression and delivery systems and uses
CA2268276A1 (en) * 1996-10-18 1998-04-30 Jeff Nordstrom Gene expression and delivery systems and uses
CA2270396C (en) * 1996-11-04 2008-03-11 Qiagen Gmbh Cationic reagents for transfection
US5877220A (en) * 1997-03-06 1999-03-02 Genta, Incorporated Amide-based oligomeric cationic lipids
US7262173B2 (en) * 1997-03-21 2007-08-28 Georgetown University Chemosensitizing with liposomes containing oligonucleotides
US6126965A (en) * 1997-03-21 2000-10-03 Georgetown University School Of Medicine Liposomes containing oligonucleotides
US20030229040A1 (en) * 1997-03-21 2003-12-11 Georgetown University Cationic liposomal delivery system and therapeutic use thereof
US5925628A (en) * 1997-03-31 1999-07-20 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5912239A (en) * 1997-04-04 1999-06-15 Genzyme Corporation Imidazole-containing cationic amphiphiles for intracellular delivery of therapeutic molecules
US5981275A (en) * 1997-04-14 1999-11-09 Genzyme Corporation Transgene expression system for increased persistence
US5948878A (en) * 1997-04-15 1999-09-07 Burgess; Stephen W. Cationic polymers for nucleic acid transfection and bioactive agent delivery
FR2762787B1 (en) * 1997-04-30 2000-10-06 Pasteur Merieux Serums Vacc ANTI-HELICOBACTER VACCINE COMPOSITION COMPRISING A TH1 ADJUVANT
FR2762788B1 (en) * 1997-04-30 2000-10-06 Pasteur Merieux Serums Vacc ANTI-HELICOBACTER VACCINE COMPOSITION FOR SUB-DIAPHRAGMATIC SYSTEMIC USE
BR9809381A (en) * 1997-04-30 2000-07-04 Merieux Oravax Anti-helicobacter vaccine composition comprising a th1-type adjuvant
US5948925A (en) * 1997-05-06 1999-09-07 Genzyme Corporation Cationic amphiphiles containing linkers derived from neutral or positively charged amino acids
US5952516A (en) * 1997-05-08 1999-09-14 Genzyme Corporation Cationic amphiphiles containing multiplesteroid lipophilic groups
US5942634A (en) * 1997-05-09 1999-08-24 Genzyme Corporation Cationic amphiphiles for cell transfections
US6287591B1 (en) * 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
JP2001500897A (en) * 1997-06-10 2001-01-23 ジエンザイム コーポレイション Cationic amphiphilic compositions for intracellular delivery of therapeutic molecules
US6635623B1 (en) 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
US20050096288A1 (en) * 1997-06-13 2005-05-05 Aragene, Inc. Lipoproteins as nucleic acid vectors
FR2765877B1 (en) * 1997-07-10 1999-09-10 Oreal COMPOSITION COMPRISING AN AQUEOUS DISPERSION OF LIPID VESICLES BASED ON CHOLESTERYL CHAIN CARBAMATES, USE IN PARTICULAR IN COSMETICS AND NEW COMPOUNDS
US6395713B1 (en) 1997-07-23 2002-05-28 Ribozyme Pharmaceuticals, Inc. Compositions for the delivery of negatively charged molecules
US20030073640A1 (en) * 1997-07-23 2003-04-17 Ribozyme Pharmaceuticals, Inc. Novel compositions for the delivery of negatively charged molecules
AU8428998A (en) * 1997-07-24 1999-02-16 Inex Pharmaceuticals Corporation Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
US20020058795A1 (en) * 1997-09-08 2002-05-16 Russ J. Mumper Hydrophobic glycosylamine derivatives, compositions, and methods for use
US6066626A (en) 1997-10-29 2000-05-23 Genzyme Corporation Compositions and method for treating lysosomal storage disease
US5998482A (en) * 1997-11-10 1999-12-07 David; Sunil A. Use of synthetic polycationic amphiphilic substances with fatty acid or hydrocarbon substituents as anti-sepsis agents
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US20010003580A1 (en) * 1998-01-14 2001-06-14 Poh K. Hui Preparation of a lipid blend and a phospholipid suspension containing the lipid blend
FR2774394B1 (en) * 1998-01-30 2002-04-26 Rhone Poulenc Rorer Sa TRANSFECTANT COMPOUNDS SENSITIVE TO REDUCING CONDITIONS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM, AND THEIR APPLICATIONS
CA2318512C (en) 1998-01-30 2009-10-13 Aventis Pharma S.A. Transfecting compositions sensitive to reducing conditions, pharmaceutical compositions containing them, and their applications
BR9909350A (en) * 1998-04-02 2000-12-12 Aventis Pharma Sa Compounds, composition, use of a compound, and processes for preparing the compounds, for transferring nucleic acids to cells and for treating diseases by administering a nucleic acid
FR2777017B1 (en) * 1998-04-02 2002-08-23 Rhone Poulenc Rorer Sa NOVEL NUCLEIC ACID TRANSFER AGENTS, COMPOSITIONS CONTAINING THEM AND USES THEREOF
FR2781160B1 (en) 1998-07-03 2000-08-18 Pasteur Merieux Serums Vacc USE OF AN AMPHIPATHIC COMPOUND TO ADJUST A SUBUNIT VACCINE
AU5366099A (en) 1998-08-20 2000-03-14 Connaught Laboratories Limited Nucleic acid molecules encoding inclusion membrane protein of (chlamydia)
US6686339B1 (en) * 1998-08-20 2004-02-03 Aventis Pasteur Limited Nucleic acid molecules encoding inclusion membrane protein C of Chlamydia
US6693087B1 (en) 1998-08-20 2004-02-17 Aventis Pasteur Limited Nucleic acid molecules encoding POMP91A protein of Chlamydia
AU5911899A (en) * 1998-09-09 2000-03-27 Genzyme Corporation Methylation of plasmid vectors
US6135976A (en) 1998-09-25 2000-10-24 Ekos Corporation Method, device and kit for performing gene therapy
EP1129064B1 (en) * 1998-11-12 2008-01-09 Invitrogen Corporation Transfection reagents
AU776150B2 (en) * 1999-01-28 2004-08-26 Medical College Of Georgia Research Institute, Inc. Composition and method for (in vivo) and (in vitro) attenuation of gene expression using double stranded RNA
ATE375391T1 (en) * 1999-05-03 2007-10-15 Sanofi Pasteur Ltd CHLAMYDIA ANTIGENS AND CORRESPONDING DNA FRAGMENTS AND THEIR USES
KR20010069179A (en) * 1999-05-28 2001-07-23 박종상 Cationic lipids for gene transfer and Preparation Method thereof
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
US20010048940A1 (en) * 1999-06-18 2001-12-06 Jennifer D. Tousignant Cationic amphiphile micellar complexes
FR2797188A1 (en) * 1999-08-04 2001-02-09 Univ Paris Nord Laboratoire De CATIONIC LIPID COMPOUNDS AND THEIR USE FOR THE TRANSFER OF NEGATIVELY CHARGED SUBSTANCES OF THERAPEUTIC INTEREST
EP1808180A3 (en) 1999-10-22 2010-12-22 Sanofi Pasteur Limited Modified GP 100 and uses thereof
EP1233671A4 (en) * 1999-11-29 2005-11-02 Mirus Corp Compositions and methods for drug delivery using amphiphile binding molecules
GB9930533D0 (en) * 1999-12-23 2000-02-16 Mitsubishi Tokyo Pharm Inc Nucleic acid delivery
EP1255822A2 (en) * 1999-12-27 2002-11-13 The Regents Of The University Of California Modified adenylylcyclase type vi useful in gene therapy for congestive heart failure
EP1261620A2 (en) * 2000-02-07 2002-12-04 Roche Diagnostics Corporation Cationic amphiphiles for use in nucleic acid transfection
ATE529435T1 (en) 2000-03-29 2011-11-15 Dgi Biotechnologies L L C AGONISTS AND ANTAGONISTS OF INSULIN AND IGF-1 RECEPTORS
AU2001242190A1 (en) * 2000-03-31 2001-10-15 Aventis Pasteur Limited Immunogenic peptides derived from prostate-specific membrane antigen (psma) and uses thereof
US7189705B2 (en) 2000-04-20 2007-03-13 The University Of British Columbia Methods of enhancing SPLP-mediated transfection using endosomal membrane destabilizers
EP1792995A3 (en) 2000-05-08 2007-06-13 Sanofi Pasteur Limited Chlamydia secretory locus orf and uses thereof
AU2001258102B2 (en) * 2000-05-10 2007-03-01 Aventis Pasteur Limited Immunogenic polypeptides encoded by mage minigenes and uses thereof
US20040204379A1 (en) * 2000-06-19 2004-10-14 Cheng Seng H. Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
EP1313771B1 (en) * 2000-08-25 2007-03-21 Aventis Pasteur Limited Haemophilus influenzae lipopolysaccharide inner-core oligosaccharide epitopes as vaccines for the prevention of haemophilus influenzae infections
US6696038B1 (en) 2000-09-14 2004-02-24 Expression Genetics, Inc. Cationic lipopolymer as biocompatible gene delivery agent
US20040142474A1 (en) * 2000-09-14 2004-07-22 Expression Genetics, Inc. Novel cationic lipopolymer as a biocompatible gene delivery agent
KR100373844B1 (en) * 2000-09-21 2003-02-26 굿젠 주식회사 Cationic lipids and method for preparing the same
KR100373845B1 (en) * 2000-09-21 2003-02-26 굿젠 주식회사 Cationic lipids and method for preparing the same
KR100381512B1 (en) * 2000-09-21 2003-04-26 굿젠 주식회사 Cationic lipids and use thereof
FR2814958B1 (en) * 2000-10-06 2003-03-07 Aventis Pasteur VACCINE COMPOSITION
CA2325088A1 (en) * 2000-12-01 2002-06-01 Fusogenix Inc. Novel membrane fusion proteins derived from poikilothermic reovirus
TWI255272B (en) 2000-12-06 2006-05-21 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
CA2364392A1 (en) * 2000-12-12 2002-06-12 Ic Vec Limited Compound
EP1669366A1 (en) * 2000-12-12 2006-06-14 Mitsubishi Chemical Corporation Lipids comprising an aminoxy group
US20020188023A1 (en) * 2000-12-12 2002-12-12 Michael Jorgensen Compound
CA2369944A1 (en) * 2001-01-31 2002-07-31 Nucleonics Inc. Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
DE10109898A1 (en) 2001-02-21 2002-09-05 Novosom Gmbh Variable charge lipids
US6613534B2 (en) 2001-03-20 2003-09-02 Wake Forest University Health Sciences MAP-2 as a determinant of metastatic potential
US20030096414A1 (en) * 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
AU2002256398A2 (en) * 2001-04-30 2002-11-11 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
US20030077829A1 (en) * 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
US20040143104A1 (en) * 2001-08-08 2004-07-22 Wadsworth Samuel C. Methods of treating diabetes and other blood sugar disorders
JP2005509409A (en) * 2001-08-08 2005-04-14 ジェンザイム・コーポレーション Methods for treating diabetes and other glycemic disorders
MXPA04003761A (en) * 2001-10-22 2005-04-08 Sterrenbeld Biotechnologie North America Inc Amphiphilic cationic lipids derived from cholesterol.
US7255874B1 (en) 2001-12-21 2007-08-14 Closure Medical Corporation Biocompatible polymers and adhesives: compositions, methods of making and uses related thereto
DE60315628T2 (en) 2002-04-09 2008-06-05 Sanofi Pasteur Ltd., Toronto MODIFIED CEA NUCLEIC ACID AND EXPRESSION VECTORS
EP1356820A1 (en) 2002-04-26 2003-10-29 Institut National De La Sante Et De La Recherche Medicale (Inserm) DNA vaccine combined with an inducer of tumor cell apoptosis
US20040180438A1 (en) * 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
EP1549352A4 (en) * 2002-05-06 2005-07-27 Nucleonics Inc Methods for delivery of nucleic acids
EP1509203B1 (en) 2002-05-15 2016-04-13 California Pacific Medical Center Delivery of nucleic acid-like compounds
ES2354607T3 (en) 2002-06-28 2011-03-16 Protiva Biotherapeutics Inc. PROCEDURE AND APPLIANCE TO PRODUCE LIPOSOMES.
WO2004011624A2 (en) * 2002-07-31 2004-02-05 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
NZ539061A (en) * 2002-08-29 2008-03-28 Univ Singapore Targeting an allergen to the MHC class II processing and presentation pathway in a vaccination group induces a strong Th1 immune response
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US8506959B2 (en) 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20050026859A1 (en) * 2002-11-12 2005-02-03 John Hilfinger Methods and compositions of gene delivery agents for systemic and local therapy
EP1587908A4 (en) * 2003-01-09 2008-02-20 Invitrogen Corp Cellular delivery and activation polypeptide-nucleic acid complexes
US20070269891A9 (en) * 2003-01-13 2007-11-22 Yasunobu Tanaka Solid surface with immobilized degradable cationic polymer for transfecting eukaryotic cells
US20040138154A1 (en) * 2003-01-13 2004-07-15 Lei Yu Solid surface for biomolecule delivery and high-throughput assay
EP1594512A4 (en) * 2003-02-11 2007-07-11 Kemia Inc Compounds for the treatment of viral infection
ATE479752T1 (en) 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
AU2004227847A1 (en) 2003-03-31 2004-10-21 Alza Corporation Lipid particles having asymmetric lipid coating and method of preparing same
WO2004094595A2 (en) 2003-04-17 2004-11-04 Alnylam Pharmaceuticals Inc. MODIFIED iRNA AGENTS
US20070178066A1 (en) * 2003-04-21 2007-08-02 Hall Frederick L Pathotropic targeted gene delivery system for cancer and other disorders
US8052966B2 (en) 2003-04-21 2011-11-08 University Of Southern California Methods and compositions for treating metastatic cancer
AU2004245074A1 (en) * 2003-06-04 2004-12-16 Canji, Inc Methods and compositions for interferon therapy
JP4980716B2 (en) 2003-06-12 2012-07-18 アルナイラム ファーマシューティカルズ, インコーポレイテッド Conserved HBV and HCV sequences useful for gene silencing
CN101291653B (en) * 2003-07-16 2012-06-27 普洛体维生物治疗公司 Lipid encapsulated interfering rna
WO2005040388A2 (en) * 2003-08-22 2005-05-06 Nucleonics Inc. Eukariotic expression systems for expression of inhibitory rna in multiple intracellular compartments
CA2551022C (en) * 2003-09-15 2013-06-04 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
US8562970B2 (en) * 2003-10-08 2013-10-22 Sanofi Pasteur Limited Modified CEA/B7 vector
WO2005039558A1 (en) * 2003-10-24 2005-05-06 Transgene S.A. Targeted delivery of therapeutically active compounds
US20050107318A1 (en) * 2003-11-17 2005-05-19 Samuel Wadsworth Methods of treating diabetes and other blood sugar disorders
AU2005214091B2 (en) * 2004-02-24 2010-08-12 Abbvie B.V. Method for determining the risk of developing a neurological disease
US7303881B2 (en) * 2004-04-30 2007-12-04 Pds Biotechnology Corporation Antigen delivery compositions and methods of use
WO2005108981A1 (en) 2004-05-12 2005-11-17 The Walter And Eliza Hall Institute Of Medical Research A method of cell isolation
CA2569645C (en) 2004-06-07 2014-10-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
WO2005121348A1 (en) 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
WO2006002262A2 (en) 2004-06-21 2006-01-05 The Board Of Trustees Of The Leland Stanford Junior University Genes and pathways differentially expressed in bipolar disorder and/or major depressive disorder
CA2572439A1 (en) * 2004-07-02 2006-01-12 Protiva Biotherapeutics, Inc. Immunostimulatory sirna molecules and uses therefor
WO2006007712A1 (en) * 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
MX2007001679A (en) 2004-08-09 2007-05-23 Elan Pharm Inc Prevention and treatment of synucleinopathic and amyloidogenic disease.
US7985581B2 (en) 2004-08-23 2011-07-26 Alnylam Pharmaceuticals, Inc. Multiple RNA polymerase III promoter expression constructs
ATE505541T1 (en) 2004-09-24 2011-04-15 Alnylam Pharmaceuticals Inc TARGETING INTERMEDIATE PRODUCTS FOR COUNTER STRAND REPLICATION OF SINGLE STRAND VIRUSES BY RNAI
WO2006055635A2 (en) * 2004-11-15 2006-05-26 Mount Sinai School Of Medicine Of New York University Compositions and methods for altering wnt autocrine signaling
AU2005306533B2 (en) * 2004-11-17 2012-05-31 Arbutus Biopharma Corporation siRNA silencing of apolipoprotein B
US7964571B2 (en) 2004-12-09 2011-06-21 Egen, Inc. Combination of immuno gene therapy and chemotherapy for treatment of cancer and hyperproliferative diseases
EP2316942B1 (en) 2004-12-22 2021-04-21 Alnylam Pharmaceuticals, Inc. Conserved hbv and hcv sequences useful for gene silencing
GB0507997D0 (en) * 2005-02-01 2005-05-25 Powderject Vaccines Inc Nucleic acid constructs
EP2932982B1 (en) 2005-05-17 2018-10-03 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
US20060286082A1 (en) * 2005-06-20 2006-12-21 Kurzweil Raymond C Systems and methods for generating biological material
DK2407484T3 (en) 2005-06-24 2016-09-05 The Walter And Eliza Hall Inst Of Medical Res Therapeutic pro-apoptotic BH3-like molecules, and methods of generating and / or selecting those
US20100119525A1 (en) * 2005-08-01 2010-05-13 Mount Sinai Schoool Of Medicine Of New York University Method for extending longevity using npc1l1 antagonists
US20080184618A1 (en) * 2005-08-03 2008-08-07 Amcol International Virus-Interacting Layered Phyllosilicates and Methods of Use
US20070031512A1 (en) * 2005-08-03 2007-02-08 Amcol International Corporation Virus-interacting layered phyllosilicates and methods of inactivating viruses
US20100272769A1 (en) * 2005-08-03 2010-10-28 Amcol International Virus-, Bacteria-, and Fungi-Interacting Layered Phyllosilicates and Methods of Use
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
CN101346393B (en) 2005-11-02 2015-07-22 普洛体维生物治疗公司 Modified siRNA molecules and uses thereof
JP5366554B2 (en) 2005-11-12 2013-12-11 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー FGF2-related methods for diagnosing and treating depression
JP2009523739A (en) 2006-01-20 2009-06-25 ウィメンズ アンド チルドレンズ ヘルス リサーチ インスティテュート インコーポレーティッド Methods for treatment, prevention, and diagnosis of bone lesions
WO2008001166A2 (en) * 2006-02-07 2008-01-03 Council Of Scientific & Industrial Research Process for synthesis of glycomimicking cationic amphiphiles
EP2495327B1 (en) 2006-03-03 2016-09-28 ProMIS Neurosciences Inc. Methods and compositions to treat and detect misfolded-SOD1 mediated diseases
GB0606190D0 (en) 2006-03-28 2006-05-10 Isis Innovation Construct
US8357781B2 (en) 2006-06-01 2013-01-22 Janssen Alzheimer Immunotherapy Neuroactive fragments of APP
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
PT2118300E (en) 2007-02-23 2015-09-22 Univ California Prevention and treatment of synucleinopathic and amyloidogenic disease
PL2583978T3 (en) 2007-02-23 2016-07-29 Prothena Biosciences Ltd Co Prevention and treatment of synucleinopathic and amyloidogenic disease
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
JP5475643B2 (en) 2007-05-04 2014-04-16 マリーナ バイオテック,インコーポレイテッド Amino acid lipids and uses thereof
NZ597601A (en) 2007-05-16 2013-04-26 Mat Malta Advanced Technologies Ltd Treatment and prevention of influenza
US20080312174A1 (en) * 2007-06-05 2008-12-18 Nitto Denko Corporation Water soluble crosslinked polymers
EP2537529B1 (en) 2007-08-02 2018-10-17 Gilead Biologics, Inc. Loxl2 inhibitory antibodies and uses thereof
US9144546B2 (en) 2007-08-06 2015-09-29 Clsn Laboratories, Inc. Nucleic acid-lipopolymer compositions
WO2009033268A1 (en) 2007-09-11 2009-03-19 University Of Guelph Novel polysaccharide immunogens from clostridium difficile
CA2697992C (en) 2007-10-04 2017-08-22 Zymogenetics, Inc. B7 family member zb7h6 and related compositions and methods
EP2217221B1 (en) * 2007-10-17 2018-06-27 Korea Advanced Institute of Science and Technology Ldl-like cationic nanoparticles for deliverying nucleic acid gene, method for preparing thereof and method for deliverying nucleic acid gene using the same
CA2703947C (en) 2007-10-26 2018-12-04 Governing Council Of The University Of Toronto Therapeutic and diagnostic methods using tim-3
EP2217721A4 (en) * 2007-10-29 2013-01-09 Univ California Osteoarthritis gene therapy
AU2008340355B2 (en) 2007-12-04 2015-01-22 Tekmira Pharmaceuticals Corporation Targeting lipids
US9078812B2 (en) * 2007-12-06 2015-07-14 The Bay Zoltan Foundation For Applied Research Particulate drug carriers as desensitizing agents
US8779088B2 (en) 2007-12-17 2014-07-15 Marfl Ab Vaccine for the treatment of Mycobacterium related disorders
ES2535419T3 (en) 2007-12-27 2015-05-11 Protiva Biotherapeutics Inc. Polo kinase expression silencing using interfering RNA
CN102016059B (en) 2007-12-28 2014-10-22 依兰制药公司 Treatment and prophylaxis of amyloidosis
AU2009234266B2 (en) 2008-04-11 2015-08-06 Tekmira Pharmaceuticals Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
CA2721380A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Silencing of csn5 gene expression using interfering rna
JP5475753B2 (en) * 2008-04-15 2014-04-16 プロチバ バイオセラピューティクス インコーポレイティッド Lipid formulations for nucleic acid delivery
CN110075113A (en) 2008-04-17 2019-08-02 Pds生物科技公司 Immune response is stimulated by the enantiomer of cation lipid
ATE550024T1 (en) 2008-05-30 2012-04-15 Univ Yale TARGETED OLIGONUCLEOTIDE COMPOSITIONS FOR MODIFYING GENE EXPRESSION
EP2303318A2 (en) 2008-06-20 2011-04-06 Wyeth LLC Compositions and methods of use of orf1358 from beta-hemolytic streptococcal strains
WO2010011895A1 (en) * 2008-07-25 2010-01-28 Alnylam Pharmaceuticals, Inc. Enhancement of sirna silencing activity using universal bases or mismatches in the sense strand
EP2743265B1 (en) 2008-10-09 2017-03-15 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
SG196818A1 (en) 2008-10-16 2014-02-13 Marina Biotech Inc Processes and compositions for liposomal and efficient delivery of gene silencing therapeutics
EP2411519B1 (en) 2009-03-27 2015-07-22 Institut National de la Santé et de la Recherche Médicale Kanamycin antisense nucleic acid for the treatment of cancer
EP2429658B1 (en) 2009-05-14 2016-04-20 Sanofi Pasteur Method for adjuvating the lipopolysaccharide (LPS) of gram-negative bacteria
CA2761924C (en) 2009-05-14 2017-10-31 Sanofi Pasteur Menigococcus vaccine containing lipooligosaccharide (los) from modified strains of l6 immunotype neisseria meningitidis
CN102985548B (en) 2009-05-16 2016-10-26 崔坤元 For deliver treatment molecule comprise cationic amphiphilic thing and the compositions of auxiliary lipid
EP2432499A2 (en) 2009-05-20 2012-03-28 Schering Corporation Modulation of pilr receptors to treat microbial infections
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
US9018187B2 (en) 2009-07-01 2015-04-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
ES2613498T3 (en) 2009-07-01 2017-05-24 Protiva Biotherapeutics Inc. New lipid formulations for the delivery of therapeutic agents to solid tumors
WO2011011447A1 (en) 2009-07-20 2011-01-27 Protiva Biotherapeutics, Inc. Compositions and methods for silencing ebola virus gene expression
US9937128B2 (en) 2009-08-03 2018-04-10 The University Of North Carolina At Chapel Hill Liposomes comprising a calcium phosphate-containing precipitate
US8637468B2 (en) 2009-08-12 2014-01-28 The University Of Kansas Synthetic cholesterylamine-linker derivatives for agent delivery into cells
US20110059111A1 (en) 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
CA2775092A1 (en) 2009-09-23 2011-03-31 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
GB0918679D0 (en) 2009-10-23 2009-12-09 Iqur Ltd Influenza vaccine
GB0918782D0 (en) 2009-10-26 2009-12-09 St Georges Hosp Medical School A protein as an adjuvant for a vaccine
WO2011056682A1 (en) 2009-10-27 2011-05-12 The University Of British Columbia Reverse head group lipids, lipid particle compositions comprising reverse headgroup lipids, and methods for the delivery of nucleic acids
WO2011067745A2 (en) 2009-12-06 2011-06-09 Rosetta Green Ltd. Compositions and methods for enhancing plants resistance to abiotic stress
WO2011084357A1 (en) 2009-12-17 2011-07-14 Schering Corporation Modulation of pilr to treat immune disorders
WO2011088462A2 (en) 2010-01-18 2011-07-21 Albany Medical College Alpha-fetoprotein 'ring and tail' peptides
WO2011109427A2 (en) 2010-03-01 2011-09-09 Alnylam Pharmaceuticals, Inc. Improving the biological activity of sirna through modulation of its thermodynamic profile
GB201004475D0 (en) 2010-03-17 2010-05-05 Isis Innovation Gene silencing
WO2011120023A1 (en) 2010-03-26 2011-09-29 Marina Biotech, Inc. Nucleic acid compounds for inhibiting survivin gene expression uses thereof
WO2011133584A2 (en) 2010-04-19 2011-10-27 Marina Biotech, Inc. Nucleic acid compounds for inhibiting hras gene expression and uses thereof
WO2011139843A2 (en) 2010-04-28 2011-11-10 Marina Biotech, Inc. Multi-sirna compositions for reducing gene expression
WO2011141704A1 (en) 2010-05-12 2011-11-17 Protiva Biotherapeutics, Inc Novel cyclic cationic lipids and methods of use
JP2013527856A (en) 2010-05-12 2013-07-04 プロチバ バイオセラピューティクス インコーポレイティッド Cationic lipids and methods of use
EP2576613A1 (en) 2010-06-07 2013-04-10 Pfizer Inc. Her-2 peptides and vaccines
EP2580238A1 (en) 2010-06-09 2013-04-17 Zymogenetics, Inc. Dimeric vstm3 fusion proteins and related compositions and methods
CA2802994A1 (en) 2010-06-17 2011-12-22 The United States Of America As Represented By The Secretary, National I Nstitutes Of Health Compositions and methods for treating inflammatory conditions
US9006417B2 (en) 2010-06-30 2015-04-14 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
GB201014026D0 (en) 2010-08-20 2010-10-06 Ucl Business Plc Treatment
WO2012035557A1 (en) 2010-09-14 2012-03-22 Council Of Scientific & Industrial Research Novel cationic amphiphiles with mannose-mimicking head-groups and a process for the preparation thereof
WO2012078667A2 (en) 2010-12-06 2012-06-14 The Penn State Research Foundation Compositions and methods relating to proliferative diseases
AU2012272970A1 (en) 2011-06-21 2014-02-06 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (ANGPTL3) iRNA compositions and methods of use thereof
CA2845884A1 (en) 2011-08-22 2013-02-28 Cangene Corporation Clostridium difficile antibodies
EP3275461A1 (en) 2011-09-19 2018-01-31 Axon Neuroscience SE Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease field
HUE048622T2 (en) 2011-11-18 2020-08-28 Alnylam Pharmaceuticals Inc Rnai agents, compositions and methods of use thereof for treating transthyretin (ttr) associated diseases
ES2923787T3 (en) 2011-11-18 2022-09-30 Alnylam Pharmaceuticals Inc Modified RNAi Agents
US9035039B2 (en) 2011-12-22 2015-05-19 Protiva Biotherapeutics, Inc. Compositions and methods for silencing SMAD4
WO2013118120A2 (en) 2012-02-06 2013-08-15 Rosetta Green Ltd. Isolated polynucleotides expressing or modulating micrornas or targets of same, transgenic plants comprising same and uses thereof in improving nitrogen use efficiency, abiotic stress tolerance, biomass, vigor or yield of a plant
US9352042B2 (en) 2012-02-24 2016-05-31 Protiva Biotherapeutics, Inc. Trialkyl cationic lipids and methods of use thereof
US9879012B2 (en) 2012-03-29 2018-01-30 Regents Of The University Of Colorado, A Body Corporate Click nucleic acids
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
PE20142406A1 (en) 2012-05-04 2015-01-23 Pfizer ANTIGENS ASSOCIATED WITH PROSTATE AND VACCINE-BASED IMMUNOTHERAPY REGIMES
US9708607B2 (en) 2012-08-03 2017-07-18 Alnylam Pharmaceuticals, Inc. Modified RNAi agents
EP2897639A4 (en) 2012-09-21 2016-05-04 Frank Bedu-Addo Improved vaccine compositions and methods of use
BR112015013105B1 (en) 2012-12-05 2022-02-08 Alnylam Pharmaceuticals, Inc DOUBLE-STRAND RNAI AGENT CAPABLE OF INHIBITING PCSK9 EXPRESSION, ITS USES, PHARMACEUTICAL COMPOSITION AND METHOD OF INHIBITING PCSK9 EXPRESSION IN A CELL IN VITRO
JP6674888B2 (en) 2013-03-13 2020-04-01 プロセナ バイオサイエンシーズ リミテッド Tau immunotherapy
CN105246337B (en) 2013-03-14 2019-02-15 吉恩维沃公司 Thymidine kinase diagnostic analysis for gene therapy application
PT2970974T (en) 2013-03-14 2017-11-29 Alnylam Pharmaceuticals Inc Complement component c5 irna compositions and methods of use thereof
JP6392315B2 (en) 2013-03-14 2018-09-19 イミュソフト コーポレーション In vitro memory B cell differentiation method and transduction method using VSV-G pseudotype virus vector
CA2905696A1 (en) 2013-03-15 2014-09-18 The Penn State Research Foundation Compositions and methods including leelamine and arachidonyl trifluoromethyl ketone relating to treatment of cancer
EP3003295A1 (en) 2013-03-15 2016-04-13 The Penn State Research Foundation Compositions and methods including celecoxib and plumbagin relating to treatment of cancer
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
SG10201804472YA (en) 2013-05-22 2018-07-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20160122420A1 (en) 2013-06-06 2016-05-05 Rowlands David J Antibody display
US20160151284A1 (en) 2013-07-23 2016-06-02 Protiva Biotherapeutics, Inc. Compositions and methods for delivering messenger rna
AR097738A1 (en) 2013-09-23 2016-04-13 Alnylam Pharmaceuticals Inc METHODS TO TREAT OR PREVENT DISEASES ASSOCIATED WITH TRANSTIRETINE (TTR)
EP3798306A1 (en) 2013-12-12 2021-03-31 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
US10119136B2 (en) 2014-01-09 2018-11-06 Alnylam Pharmaceuticals, Inc. RNAi agents modified at the 4′-C position
KR20230152154A (en) 2014-02-11 2023-11-02 알닐람 파마슈티칼스 인코포레이티드 KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3107567A4 (en) 2014-02-19 2017-10-25 Cangene Corporation Methods of modulating an immune response
EP3110401A4 (en) 2014-02-25 2017-10-25 Merck Sharp & Dohme Corp. Lipid nanoparticle vaccine adjuvants and antigen delivery systems
SG10202104570TA (en) 2014-05-22 2021-06-29 Alnylam Pharmaceuticals Inc Angiotensinogen (agt) irna compositions and methods of use thereof
CN106456772A (en) 2014-06-11 2017-02-22 吉利德科学公司 Methods for treating cardiovascular diseases
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
EP3808846A1 (en) 2014-08-20 2021-04-21 Alnylam Pharmaceuticals, Inc. Modified double-stranded rna agents
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
US10415037B2 (en) 2014-10-02 2019-09-17 Arbutus Biopharma Corporation Compositions and methods for silencing hepatitis B virus gene expression
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
US20170304459A1 (en) 2014-10-10 2017-10-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhalation delivery of conjugated oligonucleotide
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
EP3904519A1 (en) 2014-10-30 2021-11-03 Genzyme Corporation Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
CA2970138A1 (en) 2014-12-17 2016-06-23 Pioneer Hi-Bred International, Inc. Modulation of yep6 gene expression to increase yield and other related traits in plants
EP3234107B1 (en) 2014-12-19 2022-09-14 Immusoft Corporation B cells for in vivo delivery of therapeutic agents
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
CA2984154A1 (en) 2015-05-01 2016-11-10 Onl Therapeutics, Inc. Peptide compositions and methods of use
WO2016179342A2 (en) 2015-05-06 2016-11-10 Alnylam Pharmaceuticals, Inc. Factor xii (hageman factor) (f12), kallikrein b, plasma (fletcher factor) 1 (klkb1), and kininogen 1 (kng1) irna compositions and methods of use thereof
US20180245074A1 (en) 2015-06-04 2018-08-30 Protiva Biotherapeutics, Inc. Treating hepatitis b virus infection using crispr
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
CN108350455A (en) 2015-07-29 2018-07-31 阿布特斯生物制药公司 Composition for making hepatitis B virus silenced gene expression and method
CN108366966A (en) 2015-08-24 2018-08-03 光环生物干扰疗法公司 Polynucleotides nano particle and application thereof for adjusting gene expression
SG10202007937SA (en) 2015-09-02 2020-09-29 Alnylam Pharmaceuticals Inc PROGRAMMED CELL DEATH 1 LIGAND 1 (PD-L1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3906943A1 (en) 2015-09-04 2021-11-10 Primatope Therapeutics Inc. Humanized anti-cd40 antibodies and uses thereof
EP3350328A1 (en) 2015-09-14 2018-07-25 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
EP3352800B1 (en) 2015-09-24 2022-01-05 The University of North Carolina at Chapel Hill Methods and compositions for reducing metastases
US10149887B2 (en) 2015-10-23 2018-12-11 Canbas Co., Ltd. Peptides and peptidomimetics in combination with t cell activating and/or checkpoint inhibiting agents for cancer treatment
KR20180085736A (en) 2015-11-09 2018-07-27 더 유니버시티 오브 브리티쉬 콜롬비아 Amyloid beta mid-region epitopes and structurally selective antibodies thereto
EP3374379A4 (en) 2015-11-09 2019-05-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
CN108350053A (en) 2015-11-09 2018-07-31 英属哥伦比亚大学 Amyloid beta epitope and its antibody
AU2016354590B2 (en) 2015-11-13 2023-11-23 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
EP3387129A1 (en) 2015-12-10 2018-10-17 Alnylam Pharmaceuticals, Inc. STEROL REGULATORY ELEMENT BINDING PROTEIN (SREBP) CHAPERONE (SCAP) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
BR112018013967A2 (en) 2016-01-19 2019-02-05 Pfizer cancer vaccines
US11406721B2 (en) 2016-02-22 2022-08-09 The Regents Of The University Of California Compositions and methods for imaging cell populations
PT3452507T (en) 2016-05-02 2022-12-20 Prothena Biosciences Ltd Tau immunotherapy
JP7194985B2 (en) 2016-05-02 2022-12-23 プロセナ バイオサイエンシーズ リミテッド Tau-recognizing antibody
KR102471787B1 (en) 2016-05-02 2022-11-29 프로테나 바이오사이언시즈 리미티드 Tau recognition antibody
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
WO2018006052A1 (en) 2016-06-30 2018-01-04 Protiva Biotherapeutics, Inc. Compositions and methods for delivering messenger rna
EP3528827A4 (en) 2016-10-21 2020-11-04 Merck Sharp & Dohme Corp. Influenza hemagglutinin protein vaccines
US20180125920A1 (en) 2016-11-09 2018-05-10 The University Of British Columbia Methods for preventing and treating A-beta oligomer-associated and/or -induced diseases and conditions
CN109996809A (en) 2016-11-14 2019-07-09 诺华股份有限公司 Composition relevant to fusogenic protein MINION, method and therapeutical uses
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
AU2017376950B2 (en) 2016-12-16 2024-02-22 Alnylam Pharmaceuticals, Inc. Methods for treating or preventing TTR-associated diseases using transthyretin (TTR) iRNA compositions
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
EP3461487A1 (en) 2017-09-29 2019-04-03 Nlife Therapeutics S.L. Compositions and methods for the delivery of mrna to hepatic cells
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
EP3714054A1 (en) 2017-11-20 2020-09-30 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US11597932B2 (en) 2017-12-21 2023-03-07 Alnylam Pharmaceuticals, Inc. Chirally-enriched double-stranded RNA agents
MX2020007945A (en) 2018-01-29 2020-09-24 Merck Sharp & Dohme Stabilized rsv f proteins and uses thereof.
RU2020133851A (en) 2018-03-16 2022-04-19 Иммьюсофт Корпорейшн B-CELLS GENETICALLY MODIFIED FOR FOLLISTATIN SECRETION AND METHODS OF THEIR USE TO TREAT FOLLISTATIN-RELATED DISEASES, CONDITIONS, DISORDERS AND TO INCREASE MUSCLE GROWTH AND STRENGTH
CN112533629A (en) 2018-06-19 2021-03-19 阿尔莫生物科技股份有限公司 Compositions and methods for combined use of IL-10 agents with chimeric antigen receptor cell therapy
GB201811382D0 (en) 2018-07-11 2018-08-29 Taylor John Hermon Vaccine
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
WO2020060986A1 (en) 2018-09-18 2020-03-26 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
US20220056444A1 (en) 2018-12-05 2022-02-24 Empirico Inc. Process to inhibit or eliminate eosinophilic diseases of the airway and related conditions
CN113631709A (en) 2018-12-20 2021-11-09 普拉克西斯精密药物股份有限公司 Compositions and methods for treating KCNT 1-related disorders
WO2020150265A1 (en) 2019-01-15 2020-07-23 Empirico Inc. Prodrugs of alox-15 inhibitors and methods of using the same
EP3935083A4 (en) 2019-03-03 2022-11-30 Prothena Biosciences Limited Antibodies recognizing tau
US20220175723A1 (en) 2019-04-22 2022-06-09 The Penn State Research Foundation Methods and compositions relating to inhibition of aldehyde dehydrogenases for treatment of cancer
US20220211743A1 (en) 2019-05-17 2022-07-07 Alnylam Pharmaceuticals, Inc. Oral delivery of oligonucleotides
IT201900007060A1 (en) 2019-05-21 2020-11-21 St Superiore Di Sanita Engineered tumor cells and their uses
IT201900012540A1 (en) 2019-07-22 2021-01-22 Humanitas Mirasole Spa CHI3L1 inhibitors and their uses
EP4013870A1 (en) 2019-08-13 2022-06-22 Alnylam Pharmaceuticals, Inc. Small ribosomal protein subunit 25 (rps25) irna agent compositions and methods of use thereof
AR120341A1 (en) 2019-11-01 2022-02-09 Alnylam Pharmaceuticals Inc COMPOSITIONS OF RNAi AGENTS AGAINST HUNTINGTINE (HTT) AND THEIR METHODS OF USE
US20230022576A1 (en) 2019-11-19 2023-01-26 Protalix Ltd. Removal of constructs from transformed cells
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
EP4073251A1 (en) 2019-12-13 2022-10-19 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
CN115315273A (en) 2020-01-14 2022-11-08 辛德凯因股份有限公司 IL-2 orthologs and methods of use thereof
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
US20230123584A1 (en) 2020-02-14 2023-04-20 Merck Sharp & Dohme Llc Hpv vaccine
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
CN116209759A (en) 2020-03-26 2023-06-02 阿尔尼拉姆医药品有限公司 Coronavirus iRNA compositions and methods of use thereof
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
KR20230018377A (en) 2020-04-27 2023-02-07 알닐람 파마슈티칼스 인코포레이티드 Apolipoprotein E (APOE) IRNA preparation composition and method of use thereof
EP4150090A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
CA3162416C (en) 2020-05-15 2023-07-04 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
BR112022023366A2 (en) 2020-05-19 2023-05-02 Othair Prothena Ltd MULTIPLE EPITOPE VACCINE FOR THE TREATMENT OF ALZHEIMER DISEASE
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
BR112023000428A2 (en) 2020-07-10 2023-03-14 Inst Nat Sante Rech Med METHODS AND COMPOSITIONS TO TREAT EPILEPSY
CN116322754A (en) 2020-08-05 2023-06-23 辛德凯因股份有限公司 IL10RA binding molecules and methods of use
EP4192489A2 (en) 2020-08-05 2023-06-14 Synthekine, Inc. Il2rb binding molecules and methods of use
CA3190420A1 (en) 2020-08-05 2022-02-10 Synthekine, Inc. Compositions and methods related to il27 receptor binding
WO2022031884A2 (en) 2020-08-05 2022-02-10 Synthekine, Inc. Il2rg binding molecules and methods of use
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
EP4232581A1 (en) 2020-10-21 2023-08-30 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
EP4251170A1 (en) 2020-11-25 2023-10-04 Akagera Medicines, Inc. Lipid nanoparticles for delivery of nucleic acids, and related methods of use
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
TW202245835A (en) 2021-02-04 2022-12-01 美商默沙東有限責任公司 Nanoemulsion adjuvant composition for pneumococcal conjugate vaccines
WO2022174000A2 (en) 2021-02-12 2022-08-18 Alnylam Pharmaceuticals, Inc. Superoxide dismutase 1 (sod1) irna compositions and methods of use thereof for treating or preventing superoxide dismutase 1- (sod1-) associated neurodegenerative diseases
JP2024509783A (en) 2021-02-25 2024-03-05 アルナイラム ファーマシューティカルズ, インコーポレイテッド Prion protein (PRNP) IRNA compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
CA3214499A1 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
BR112023025224A2 (en) 2021-06-04 2024-02-27 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 (C9ORF72) OPEN READING BOARD 72 IRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
US20230044997A1 (en) 2021-07-15 2023-02-09 Turn Biotechnologies, Inc. Synthetic, persistent rna constructs and methods of use for cell rejuvenation and for treatment
WO2023288288A1 (en) 2021-07-15 2023-01-19 Turn Biotechnologies, Inc. Synthetic, persistent rna constructs with on/off mechanism for controlled expression and methods of use
WO2023288285A1 (en) 2021-07-15 2023-01-19 Turn Biotechnologies, Inc. Polycistronic expression vectors
IL309905A (en) 2021-07-23 2024-03-01 Alnylam Pharmaceuticals Inc Beta-catenin (ctnnb1) irna compositions and methods of use thereof
AU2022331279A1 (en) 2021-08-19 2024-02-15 Merck Sharp & Dohme Llc Thermostable lipid nanoparticle and methods of use thereof
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023122762A1 (en) 2021-12-22 2023-06-29 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023220603A1 (en) 2022-05-09 2023-11-16 Regeneron Pharmaceuticals, Inc. Vectors and methods for in vivo antibody production
US20230392188A1 (en) 2022-06-02 2023-12-07 Perkinelmer Health Sciences, Inc. Electrophoresis-mediated characterization of dna content of adeno-associated virus capsids
WO2023235818A2 (en) 2022-06-02 2023-12-07 Scribe Therapeutics Inc. Engineered class 2 type v crispr systems
WO2023240076A1 (en) 2022-06-07 2023-12-14 Scribe Therapeutics Inc. Compositions and methods for the targeting of pcsk9
WO2023240074A1 (en) 2022-06-07 2023-12-14 Scribe Therapeutics Inc. Compositions and methods for the targeting of pcsk9
WO2023240277A2 (en) 2022-06-10 2023-12-14 Camp4 Therapeutics Corporation Methods of modulating progranulin expression using antisense oligonucleotides targeting regulatory rnas
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4544545A (en) * 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
WO1988000824A1 (en) * 1986-07-28 1988-02-11 Liposome Technology, Inc. Liposomes with enhanced retention on mucosal tissue
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides

Also Published As

Publication number Publication date
US5283185A (en) 1994-02-01
JP3258661B2 (en) 2002-02-18
DK0663013T3 (en) 2005-08-01
JPH07500963A (en) 1995-02-02
ES2240958T3 (en) 2005-10-16
DE69233497T2 (en) 2006-01-26
SG50630A1 (en) 1998-07-20
DE69233497D1 (en) 2005-05-12
HK1014736A1 (en) 1999-09-30
WO1993005162A1 (en) 1993-03-18
JP2002153270A (en) 2002-05-28
EP0663013B1 (en) 2005-04-06
AU665029B2 (en) 1995-12-14
EP0663013A4 (en) 1994-11-28
ATE292690T1 (en) 2005-04-15
EP0663013A1 (en) 1995-07-19
AU2656592A (en) 1993-04-05

Similar Documents

Publication Publication Date Title
US5283185A (en) Method for delivering nucleic acids into cells
US6627218B2 (en) Cationic amphiphiles of cholesterol
US5955365A (en) Self-assembling polynucleotide delivery system
US6716882B2 (en) Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US5840710A (en) Cationic amphiphiles containing ester or ether-linked lipophilic groups for intracellular delivery of therapeutic molecules
US6075012A (en) Reagents for intracellular delivery of macromolecules
US5948767A (en) Cationic amphiphile/DNA complexes
US5925628A (en) Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5719131A (en) Cationic amphiphiles containing dialkylamine lipophilic groups for intracellular delivery of therapeutic molecules
US5939401A (en) Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
HUT77171A (en) Lipopolyamines as transfection agents and pharmaceuticals containing them
JP2014208687A (en) Lipids and lipid assemblies comprising transfection enhancer elements
US6649780B1 (en) Cationic lipids
US5942634A (en) Cationic amphiphiles for cell transfections
US5912239A (en) Imidazole-containing cationic amphiphiles for intracellular delivery of therapeutic molecules
US5795870A (en) Compositions and methods for cell transformation
AU2006202596B8 (en) Novel compounds, methods and derivatives
AU3018397A (en) Novel compounds, methods and derivatives
US5952516A (en) Cationic amphiphiles containing multiplesteroid lipophilic groups
AU734980B2 (en) Cationic amphiphiles and plasmids for intracellular delivery of therapeutic molecules

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued