CA2213417A1 - Novel cationic lipids and the use thereof - Google Patents

Novel cationic lipids and the use thereof

Info

Publication number
CA2213417A1
CA2213417A1 CA002213417A CA2213417A CA2213417A1 CA 2213417 A1 CA2213417 A1 CA 2213417A1 CA 002213417 A CA002213417 A CA 002213417A CA 2213417 A CA2213417 A CA 2213417A CA 2213417 A1 CA2213417 A1 CA 2213417A1
Authority
CA
Canada
Prior art keywords
independently
carbons
compound according
cationic lipid
integer
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
CA002213417A
Other languages
French (fr)
Inventor
Evan C. Unger
Dekang Shen
Guanli Wu
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.)
ImaRx Pharmaceutical Corp
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 CA2213417A1 publication Critical patent/CA2213417A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C237/10Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by nitrogen atoms not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/10Phosphatides, e.g. lecithin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • C07K14/501Fibroblast growth factors [FGF] acidic FGF [aFGF]
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/829Liposomes, e.g. encapsulation
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/797Lipid particle
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/797Lipid particle
    • Y10S977/798Lipid particle having internalized material
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/797Lipid particle
    • Y10S977/798Lipid particle having internalized material
    • Y10S977/799Containing biological material
    • Y10S977/80Nucleic acid, e.g. DNA or RNA
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/797Lipid particle
    • Y10S977/798Lipid particle having internalized material
    • Y10S977/799Containing biological material
    • Y10S977/801Drug
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/906Drug delivery
    • Y10S977/907Liposome

Abstract

Cationic lipid compound which comprise at least two cationic groups. The cationic lipid compound are particularly suitable for use as carriers in the intracellular delivery of bioactive agents, including pharmaceuticals and genetic material. Compositions of the present cationic lipid compounds include suspensions, micelles and liposomes.

Description

W O96/26179 PCTrUS96/01474 NOVEL CATIONIC LIPIDS AND THE USE THEREOF

Field of the Invention The present invention relates to novel cationic lipids and the use thereof. More particularly, the present invention relates to novel cationic lipids and their use in the delivery of biologically active agents.

Back~round of the Invention The intracellular delivery of biologically active agents, for example, pharmacologically active materials and diagnostic agents, is generally desirable in connection with the treatment and/or diagnosis of various diseases. For example, cell function can be influenced at the subcellular or molecular level by delivering the biologically active agent intracellularly.
Various methods have been developed for the delivery of biologically active agents directly into living cells. Included among such methods is the "carrier method"
which involves the use of a carrier to promote intracellular delivery of a bioactive agent to specifically targeted cells, for example, diseased cells. The intracellular delivery of therapeutic agents is referred to herein also as "transfection".
Various carriers have been developed for use in the transfection of biologically active agents. For example, liposomes and polymers have been developed for the transfection of genetic materials, including deoxyribo-nucleic acid (DNA) and ribonucleic acid (RNA). However, the currently available carriers, including liposomes and W O96/26179 PCT~US96/01471 -- 2 polymers, are generally ineffective for the intracellular delivery of biologically active materials in vivo.
Moreover, the currently available carriers have limited use in connection with the transfection of cells in vitro.
In addition to the carrier method, alternative methods have been developed for the transfection of biologically active agents, including genetic material, directly into cells. These methods include, for example, calcium phosphate precipitation and electroporation.
However, these methods are also generally ineffective for the intracellular delivery of biologically active agents in vi vo .
Great strides have been made in connection with the characterization and understanding of various diseases, for example, genetic diseases, and their associated protein transcription, in humans and other ~nim~l s. This has led to the development or postulation of improved methods for the treatment of such diseases with biologically active agents.
Various of these methods involve or re~uire that the biologically active agent be delivered intracellularly. As noted above, however, current methods for the transfection of cells with biologically active agents in vivo are generally ineffective. This is thwarting the study and implementation of improved methods for the treatment of various diseases.
The cellular membrane is a selective barrier which prevents random introduction of substances into the cell.
Accordingly, a major difficulty in the intracellular delivery of biologically active agents is believed to involve the transfer of the agent ~rom the extracellular space to the intracellular space. Localization of the biologically active agent at the surface of selected cell membranes has been difficult to achieve also.
Carriers have been engineered also from viral vectors. Specifically, vectors for the transfection of genetic material have been developed from whole viruses, including adenoviruses and retroviruses. However, only a CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/0117 limited amount of biologically active materials can be placed inside of a viral capsule. Moreover, in the case of ~ biologically active materials which comprise genetic material, undesired interaction of the viral carrier may occur with the encapsulated genetic material and the patient.
To m;nim; ze the potential interactions associated with viruses, attempts have been made to use only certain components of a virus. This is difficult to achieve in vivo inasmuch as the virus components must be able to recognize and reach the targeted cells. Despite extensive work, a successfully targeted, viral-mediated vector for the delivery of biologically active materials into cells in vivo has not been adequately achieved.
As noted above, liposomes have been used as a carrier for the intracellular delivery of biologically active agents, including genetic material. One of the original methods for the use of liposomes as carriers for biologically active agents is disclosed in Szoka and Papahadjopoulos, Ann. Rev. Biophysic. Bioeng., Vol. 9, pp.
467-508 (1980). The disclosed method involves the preparation of liposomes by the addition of an aqueous solution of genetic material to phospholipids which are dissolved in ether. Evaporation of the ether phase provides genetic material encapsulated in lipid vesicles.
Another method for encapsulating biologically active agents in liposomes involves the extrusion of dehydration-rehydration vesicles. Other methods, in addition to those described above, are known for the encapsulation by liposomes of biologically active agents.
More recently, liposomes have been developed from cationic lipids, such as N-[1-~2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride ("DOTMA") or lipids which comprise cationic polymers, for example, polysine. See, e.g., Xiaohuai and Huang, Biochimica et Biophysica Acta, Vol. 1189, pp. 195-203 (1994). Liposomes which are prepared from cationic materials (referred to hereinafter as W O96/26179 PCTrUS96/01474 -- 4 -- ~
"cationic liposomes") have been developed, inter alia, to transfect cells with genetic material, including DNA. It is believed that the cationic liposomes bind with the negatively charged phosphate group(s) of the nucleotides in DNA. Studies have shown that cationic liposomes mediate transfection of cells with genetic material in vitro more efficiently than other carriers, for example, cationic polymers. In addition, in vitro studies have shown also that cationic liposomes provide improved transfection of cells relative to other delivery methods, including electroporation and calcium phosphate precipitation.
However, the currently available cationic lipids and cationic liposomes are generally ineffective for the intracellular delivery of biologically active agents in vivo. Moreover, they are generally ineffective for the intracellular delivery of biologically active agents in serum. This is a serious drawback inasmuch as cells require serum for viability. In fact, it is generally necessary to remove serum from tissue culture baths during gene transfection studies involving cationic lipids and cationic liposomes. After transfection, the serum is replaced. This involves additional processing steps which render transfection of cells with cationic lipids and cationic liposomes complex and cumbersome.
2~ New and/or better cationic lipids useful, inter alia, for the intracellular delivery of bioactive agents are needed. The present invention is directed to this as well as other important ends.

s. -~y o~ the In~ention The present invention is directed to cationic lipids which comprise at least one, and preferably at least t two, cationic groups and which may be useful for the intracellular delivery of bioactive agents.
Specifically, in one embodiment, the present invention relates to a cationic lipid compound of the formula -W O 96/26179 PCTrUS96/01474 ~R4 - Y3)z Y1- (Rl- xl)x- R2 tY2- R3]y-(Xl- Rl)x- Y

wherein:
each of x, y and z is independently an integer from 0 to about 100;
each X1 is independently -O-, -S-, -NRs-, 5 -C(=X2) -, -C(=X2) -N(R5)-, -N(R5) -C(=X2)-, -C (=X2) -O-, -O-C (=X2) - or -X2-(R5X2) P(=X2) -X2-;
each X2 is independently 0 or S;
each Yl is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer ~0 from 1 to 3;
each Y2 is independently -N(R6)b-, -S(R6)b- or -P (R6) b- ~ wherein b is an integer from 0 to 2;
each Y3 is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer ~5 from 1 to 3;
each of R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 10 carbons; and each R6 is independently -[R7-X3]c-R8 or -Rg- [X4-Rlo] d-Q ~ wherein:
each of c and d is independently an integer ~rom 0 to about 100;

W O 96t26179 PCTrUS96/01474 each Q is independently a phosphate residue, ~N(Rll)q, ~S(Rll)q, ~P(Rll)q or -C02R6, wherein q is an integer ~rom 1 to 3;
each o~ X3 and X4 is independently -O-, -S-, 5 -NR5-, -C (=X2) -, -C (=X2) -N (R5) -, -N (R5) -C (=X2) -, -C (=X2) -O-, -O-C (=X2) - or -X2- (R5X2) P(=X2) -X2-;
each R7 is independently alkylene o~ 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of ~0 1 to about 60 carbons;
each o~ Rg and Rlo is independently alkylene o~
1 to about 20 carbons; and each Rll is independently -[R7-X3] C-R8 or -R9- [X4-Rlo] d-W, wherein:
each W is independently a phosphate residue, -N(Rl2)w, -S(Rl2)w~ -P(Rl2)w or -C02R6, wherein w is an integer ~rom 1 to 3; and Rl2 iS - [R7-X3]C-R8; with the proviso that the compound o~ ~ormula (I) comprises at least one, and pre~erably at least two, quaternary salts.
In another embodiment, the invention relates to a cationic lipid compound of the ~ormula Yr (~

wherein:

each Yl is independently a phosphate residue, N(R2)a-, S(Rz)a-, P(R2)a- or -C02R2, wherein a is an integer from 1 to 3;
R1 is alkylene of 1 to about 60 carbons cont~;n;ng o to about 30 -O-, -S-, -NR3- or -X2- (R3X2) P (=X2) -X2- heteroatoms or heteroatom groups;
R2 is a residue of the formula -R4- [ (Xl-Rs) X-Y2] y~R6~ wherein:
each of x and y is independently an integer from 0 to about 100;
each Xl is independently a direct bond, -O-, -S-, -NR3-, -C (=X2) -, -C (=X2) -N (R3) -, -N (R3) -C (=X2) -, -C (=X2) -O-, -O-C (=X2) - or -X2- (R3X2) P (=X2) -X2-;
each X2 is independently O or S;
lS each Y2 is independently -S(R2) b- ~ -N(R2) b- or -P (R2) b- ~ wherein b is an integer from 0 to 2;
each R3 is independently hydrogen or alkyl of 1 to about lO carbons;
each of R4 and R5 is independently a direct bond or alkylene of 1 to about 30 carbons containing 0 to about 15 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups; and each R6 is independently hydrogen or alkyl of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- or -X2- (R3X2) P (=X2) -X2- heteroatoms or heteroatom groups;
with the proviso that the compound of formula (II) comprises at least one, and preferably at least two, quaternary salts.

CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 In yet another embodiment, the present invention relates to a cationic lipid compound o~ the ~ormula ~ - Y3)z IYi--~Rr- x~ R2--[Y2~R3]y--(Xl--Rl)~-IY
( I rXl)~ rXl)~
lR2 lR2 Y i(Rl- Xl)x- ~3-1Y2]y- R2-(Xl-Rl)~- Y
~ - Y3)z wherein:
each of x, y and z is independently an integer ~rom O to about 100;
each X1 is independently -O-, -S-, -NR5-, -C (=X2) - ~ -C (=X2) -N(R5)-, -N(R5)-C (=X2) - ~ -C (=X2) -O--O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-i each X2 iS independently O or S;
each Yl is independently -O-, -N(R6) a~ ~ -S (R6) a~
or -P(R6) a~ ~ wherein a is an integer ~rom O to 2;
each Y2 is independently -N(R6) a~ ~ -S (R6) a~ or -P(R6)~-, wherein a is an integer ~rom O to 2;
each Y3 iS independently a phosphate residue, N(R6)b-, S(R6)b-, P(R6)b- or -C02R6, wherein b is an integer ~rom 1 to 3;
each o~ R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;

each R5 is independently hydrogen or alkyl o~
1 to about 10 carbons; and W O 96/26179 PCTrUS96/01474 g each R6 is independently -[R,-X3] C-R8 or -Rg-[X~-Rlo] d- Q, wherein:
each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, -N(Rll) q~ -S (Rll) q~ -P (Rll) q or -CO2Rll, wherein q is an integer from 1 to 3;
each of X3 and X4 iS independently -O-, -S-, -NR5-, -C(=X2)-~ -c(=x2)-N(R5)-~ -N(R5)-C(=X2)-, -C(=X2)-O-, lO -O-C(=X2)- or -X2-(R5x2)P(=x2)-x2-;
each R7 is independently alkylene of 1 to about 20 carbonsi each R8 is independently hydrogen or alkyl of 1 to about 60 carbonsi each of Rg and R1o is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R,-X3] C-R8 or -Rg- [X4- Rlo]d~W~ wherein:
each W is independently a phosphate residue, 20 -N(Rl2)w~ -S(Rl2)w/ -P (Rl2)W or -CO2Rl2, wherein w is an integer from 1 to 3; and Rl2 is -tR,-X3] c~R8i with the proviso that the compound of formula (III) comprises at least one, and pre~erably at least two, quaternary salts.
Cationic lipid compounds which comprise at least one, and preferably at least two, cationic groups are also the subject of the present invention.

CA 022l34l7 l997-08-20 W O96/26179 PCT~US96/01474 Another aspect o~ the present invention are cationic lipid compositions which are composed o~ cationic lipid compounds that comprise at least one, and pre~erably at least two, cationic groups.
Yet another aspect of the present invention is a cationic lipid formulation for the intracellular delivery of a bioactive agent. The ~ormulation comprises, in combination with a bioactive agent, a cationic lipid compound that comprises at least one, and preferably at least two cationic groups.
Still another aspect of the present invention relates to a process for the preparation of a cationic lipid formulation ~or the intracellular delivery of a bioactive agent. The process comprises combining together a bioactive agent and a cationic lipid composition which comprises a cationic lipid compound having at least one, and preferably at least two, cationic groups.
Also encompassed by the present invention is a method ~or delivering intracellularly a bioactive agent.
The method comprises contacting cells with a cationic lipid compound having at least one, and pre~erably at least two, cationic groups and a bioactive agent.
These and other aspects of the invention will become more apparent from the present specification and claims.

W O96/26179 PCT~US96/01474 Brief Description of the Drawin~s FIGS. 1 and 2 are graphical representations of the amount of protein expressed in transfection experiments involving cationic lipid compounds of the present invention and compounds disclosed in the prior art.

Detailed Description of the Invention As employed above and throughout the disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings.
"Alkyl" refers to an aliphatic hydrocarbon group which may be straight or branched having 1 to about 60 carbon atoms in the chain. "Lower alkyl" refers to an alkyl group having 1 to about 8 carbon atoms. "Higher alkyl"
refers to an alkyl group having about 10 to about 20 carbon atoms. The alkyl group may be optionally substituted with one or more alkyl group substituents which may be the same or different, where "alkyl group substituent" includes halo, aryl, hydroxy, alkoxy, aryloxy, alkyloxy, alkylthio, arylthio, aralkyloxy, aralkylthio, carboxy alkoxycarbonyl, oxo and cycloalkyl. There may be optionally inserted along the alkyl group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is lower alkyl. "Branched" refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain. Exemplary alkyl groups include methyl, ethyl, i-propyl, n-butyl, t-butyl, n-pentyl, heptyl, octyl, decyl, dodecyl, tridecyl, CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/0147 - 12 ~
tetradecyl, pentadecyl and hexadecyl. Pre~erred alkyl groups include the lower alkyl groups of 1 to about 4 carbons and the higher alkyl groups of about 12 to about 16 carbons. Preferred alkyl groups include also alkyl groups which are substituted with one or more halo atoms.
Fluoroalkyl groups are preferred among the halo-substituted alkyl groups, including, for example, fluoroalkyl groups of the formula CF3(CF2) n(CH2)m~, wherein each of m and n is independently an integer from 0 to about 22. Exemplary fluoroalkyl groups include perfluoromethyl, perfluoroethyl, perfluoropropyl, perfluorobutyl, perfluorocyclobutyl, perfluoropentyl, perfluorohexyl, perfluoroheptyl, perfluorooctyl, perfluorononyl, perfluorodecyl, perfluoroundecyl and perfluorododecyl.
"Alkenyl" refers to an alkyl group containing at least one carbon-carbon double bond. The alkenyl group may be optionally substituted with one or more "alkyl group substituents". Exemplary alkenyl groups include vinyl, allyl, n-pentenyl, decenyl, dodecenyl, tetradecadienyl, heptadec-8-en-1-yl and heptadec-8,11-dien-1-yl.
~ Alkynyl~ refers to an alkyl group containing a carbon-carbon triple bond. The alkynyl group may be optionally substituted with one or more "alkyl group substituents". Exemplary alkynyl groups include ethynyl, propargyl, n-pentynyl, decynyl and dodecynyl. Pre~erred alkynyl groups include the lower alkynyl groups.
"Cycloalkyl" re~ers to a non-aromatic mono- or multicyclic ring system of about 4 to about 10 carbon atoms.

W O96/26179 PCTrUS96/01474 The cycloalkyl group may be optionally partially unsaturated. The cycloalkyl group may be also optionally substituted with an aryl group substituent, oxo and/or alkylene. Preferred monocyclic cycloalkyl rings include cyclopentyl, cyclohexyl and cycloheptyl. Preferred multicyclic cycloalkyl rings include adamantyl, octahydronaphthyl, decalin, camphor, camphane, noradamantyl, bicyclo[2.2.2.]oct-5-ene, cis- 5-norbornene, 5-norbornene, (lR) - (- ) -myrtentane, norbornane and anti-3-oxo-tricyclo[2.2.1. o2,6] heptane.
"Aryl" refers to an aromatic carbocyclic radicalcont~;n;ng about 6 to about 10 carbon atoms. The aryl group may be optionally substituted with one or more aryl group substituents which may be the same or different, where "aryl group substituent" includes alkyl, alkenyl, alkynyl, aryl, aralkyl, hydroxy, alkoxy, aryloxy, aralkoxy, carboxy, aroyl, halo, nitro, trihalomethyl, cyano, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, acyloxy, acylamino, aroylamino, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylthio, alkylthio, alkylene and -NRR', where R and R~ are each independently hydrogen, alkyl, aryl and aralkyl.
Exemplary aryl groups include substituted or unsubstituted phenyl and substituted or unsubstituted naphthyl.
"Acyl" re~ers to an alkyl-CO- group wherein alkyl is as previously described. Preferred acyl groups comprise - alkyl of~ 1 to about 30 carbon atoms. Exemplary acyl groups include acetyl, propanoyl, 2-methylpropanoyl, butanoyl and palmitoyl.

W O96/26179 PCTrUS96/01474 ~ Aroyl" means an aryl-CO- group wherein aryl is as previously described. Exemplary aroyl groups include benzoyl and 1- and 2-naphthoyl.
"Alkoxy" refers to an alkyl-O- group wherein alkyl is as previously described. Exemplary alkoxy groups include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy and heptoxy.
"Aryloxy" refers to an aryl-O- group wherein the aryl group is as previously described. Exemplary aryloxy groups include phenoxy and naphthoxy.
~Alkylthio~ refers to an alkyl-S- group wherein alkyl is as previously described. Exemplary alkylthio groups include methylthio, ethylthio, i-propylthio and heptylthio.
"Arylthio" refers to an aryl-S- group wherein the aryl group is as previously described. Exemplary arylthio groups include phenylthio and naphthylthio.
"Aralkyl" refers to an aryl-alkyl- group wherein aryl and alkyl are as previously described. Exemplary aralkyl groups include benzyl, phenylethyl and naphthylmethyl.
"Aralkyloxy" refers to an aralkyl-O- group wherein the aralkyl group is as previously described. An exemplary aralkyloxy group is benzyloxy.
"Aralkylthio~ refers to an aralkyl-S- group wherein the aralkyl group is as previously described. An exemplary aralkylthio group is benzylthio.
"Dialkylamino" refers to an -NRR' group wherein each o~ R and R~ is independently an alkyl group as W O96/26179 PCTrUS96/01474 previously described. Exemplary alkylamino groups include ethylmethylamino, dimethylamino and diethylamino.
"Alkoxycarbonyl" refers to an alkyl-O-CO- group.
Exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, butyloxycarbonyl and t-butyloxycarbonyl.
"Aryloxycarbonyl" refers to an aryl-O-CO- group.
Exemplary aryloxycarbonyl groups include phenoxy- and naphthoxy-carbonyl.
"Aralkoxycarbonyl" refers to an aralkyl-O-CO-group. An exemplary aralkoxycarbonyl group isbenzyloxycarbonyl.
"Carbamoyl" refers to an H2N-CO- group.
"Alkylcarbamoyl" re~ers to a R'RN-CO- group wherein one of R and R' is hydrogen and the other of R and R' is alkyl as previously described.
~ Dialkylcarbamoyl" refers to R'RN-CO- group wherein each of R and R' is independently alkyl as previously described.
~ 'Acyloxy" refers to an acyl-O- group wherein acyl is as previously described.
"Acylamino" refers to an acyl-NH- group wherein acyl is as previously described.
"Aroylamino" refers to an aroyl-NH- group wherein aroyl is as previously described.
"Alkylene" refers to a straight or branched bivalent aliphatic hydrocarbon group having ~rom 1 to about 30 carbon atoms. The alkylene group may be straight, branched or cyclic. The alkylene group may be also W 096/26179 PCTrUS96/01474 optionally unsaturated and/or substituted with one or more "alkyl group substituents." There may be optionally inserted along the alkylene group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is alkyl as previously described. Exemplary alkylene groups include methylene (-CH2-), ethylene (-CH2CH2-), propylene (-(CH2) 3- ), cyclohexylene (-C6Hlo~)~ -CH=CH-CH=CH-, -CH=CH-CH2-, -tCF2)n(CH2)m~, wherein n is an integer from about 1 to about 22 and m is an integer from 0 to about 22, -(CH2)n-N(R)-tCH2)m~, wherein each of m and n is independently an integer ~rom 0 to about 30 and R iS hydrogen or alkyl, methylenedioxy (-O-CH2-O-) and ethylenedioxy (-O-(CHz)2-O-).
It is preferred that the alkylene group has about 2 to about 1~ 3 carbon atoms.
"Halo" or "halide" refers to fluoride, chloride, bromide or iodide.
"Heteroatom group" refers to a radical which contains at least one heteroatom.
"Amino Acid" refers to a naturally occurring or synthetic amino acid.
"Polypeptide" refers to a biologically active series of two or more amino acid residues bonded by peptide linkages. Polypeptides having about 3 to about 40 amino 2~ acid residues are preferred.
"Phosphate residue" refers to a substituent group which is derived from phosphoric acid (O=P(OH) 3) .
Preferably, the phosphate residue is an ester of phosphoric W O96/26179 PCTrUS96/01474 acid which is substituted with one or more alkyl and/or alkenyl groups. Preferred phosphate esters include phospholipids. Pre~erred among the phospholipids are - phosphoglycerides, with diacylglycerol phosphates being especially pre~erred. An exemplary diacylglycerol phosphate is 1,2-dioleoylglycero-3-phosphoethyl.
"Quaternary salt" refers to a type of ammonium, sul~onium or phosphonium compound in which the hydrogen atoms o~ the ammonium, sul~onium or phosphonium ion are replaced by alkyl groups. With respect to quaternary ammonium and phosphonium compounds, the molecular structure includes a nitrogen or phosphorous atom joined to ~our organic groups, ~or example, alkyl groups. The molecular structure o~ a quaternary sul~onium compound includes a 1~ sul~ur atom joined to three organic groups. These molecular structures are positively charged and are generally re~erred to as cations or cationic groups. The cations are typically, although not necessarily, associated with a negatively charged acid radical. The negatively charged radical is generally referred to as an anion or an anionic group. Exemplary anions include, for example, halides.
Quaternary salts are generally the product o~ the ~inal stage o~ alkylation o~ nitrogen, sul~ur or phosphorous.
~Lipid" re~ers to a synthetic or naturally-2~ occurring amphipathic compound which comprises a hydrophilic _ component and a hydrophobic component. Lipids include, ~or example, ~atty acids, neutral ~ats, phosphatides, W O 96/26179 PCTrUS96101474 glycolipids, aliphatic alcohols and waxes, terpenes and steroids.
~ Cationic lipid compound~ refers to a lipid which comprises a cationic group and which functions generally as a positively charged ion, for example, in solution.
Preferred cationic lipid compounds are lipids which comprise at least one cationic group, with lipids which comprise at least two or more cationic groups being more preferred.
"Cationic group" refers to a group which is positively charged. Preferred cationic groups include the positively charged ions of quaternary salts. Exemplary quaternary salts are ammonium, phosphonium and sulfonium salts.
"Counter ion" refers to an anion. An anion which 1~ is "pharmaceutically-acceptable" is substantially non-toxic and does not render the associated cation pharmaceutically unacceptable.
"Cationic lipid composition" refers to a composition which comprises a cationic lipid compound.
Exemplary cationic lipid compositions include suspensions, emulsions, vesicular compositions and hexagonal H II phase structures. "Cationic lipid formulation" refers to a composition which comprises a cationic lipid compound and a bioactive agent.
~Charge density~ refers to charge per unit mass or volume.
~Vesicle~ or ~vesicular species" refers to a spherical entity which is characterized by the presence of CA 022l34l7 l997-08-20 W O96/26179 PCTAU$96/01474 an internal void. Preferred vesicles or vesicular species are formulated from lipids, including the cationic lipid compounds of the present invention. In any given vesicle or - vesicular species, the lipids may be in the form of a monolayer or bilayer, and the mono- or bilayer lipids may be used to form one or more mono- or bilayers. In the case of more than one mono- or bilayer, the mono- or bilayers are generally concentric. The lipid vesicles or vesicular species include such entities commonly referred to as liposomes, micelles and the like. Thus, the lipids may be used to form a unilamellar vesicle (comprised of one monolayer or bilayer), an oligolamellar vesicle (comprised of about two or about three monolayers or bilayers) or a multilamellar vesicle (comprised of more than about three monolayers or bilayers). The internal void of the vesicles are generally filled with a liquid, including, for example, an aqueous liquid, a gas, a gaseous precursor, and/or a solid material, including, for example, a bioactive agent.
"Cationic vesicle" or "cationic vesicular composition" refers to a vesicle or vesicular species which is ~ormulated from a cationic lipid compound.
"Cationic vesicle formulation" refers to a composition of a vesicle or vesicular species and a bioactive agent.
~Liposome~ re~ers to a generally spherical cluster or aggregate of amphipathic compounds, including lipid compounds, typically in the form of one or more concentric layers, ~or example, bilayers.

W O96/26179 PCTrUS96/0147 "Emulsion" refers to a lipoidal mixture of two or more liquids and is generally in the form of a colloid. The lipids may be heterogeneously dispersed throughout the emulsion. Alternatively, the lipids may be aggregated in the form of, for example, clusters or layers, including mono- or bilayers.
"Suspension" refers to a mixture of finely divided colloidal particles floating in a liquid.
"Hexagonal H II phase structure" refers to a generally tubular aggregation of lipids in liquid media, for example, aqueous media, in which the hydrophilic portion(s) of the lipids generally face inwardly in association with a liquid environment inside the tube. The hydrophobic portion(s) of the lipids generally radiate outwardly and the complex assumes the shape of a hexagonal tube. A plurality of tubes is generally packed together in the hexagonal phase structure.
"Patient", as used herein, refers to animals, including m~mm~1 S, preferably humans.
~Bioactive agent" refers to a substance which is capable of exerting a biological effect in vitro and/or in vivo. The biological effect is preferably therapeutic in nature. As used herein, "bioactive agent" re~ers also to substances which are used in connection with an application which is diagnostic in nature, such as in methods for diagnosing the presence or absence of a disease in a patient. The bioactive agents may be neutral or positively or negatively charged. Preferably, the bioactive agents are W O96/26179 PCTrUS96/01474 negatively charged. Examples of suitable bioactive agents include pharmaceuticals and drugs, synthetic organic molecules, proteins, vitamins, steroids, polyanions, nucleosides, nucleotides, polynucleotides and diagnostic agents, such as contrast agents for use in connection with magnetic resonance imaging, ultrasound or computed tomography of a patient.
"Anionic group" refers to a group which is negatively charged. Preferred anionic groups include phosphate (P04-) groups.
"Anionic bioactive agent" refers to a bioactive agent that comprises at least one anionic group. Certain genetic materials, for example, polynucleotides, are exemplary anionic bioactive agents.
"Genetic material" refers generally to nucleotides and polynucleotides, including deoxyribonucleic acid (DNA) and ribonucleic acid (RNA). The genetic material may be made by synthetic chemical methodology known to one of ordinary skill in the art, or by the use of recombinant technology, or by a combination of the two. The DNA and RNA
may optionally comprise unnatural nucleotides and may be single or double stranded. "Genetic material" refers also to sense and anti-sense DNA and RNA, that is, a nucleotide sequence which is complementary to a specific sequence of nucleotides in DNA and/or RNA.
_ ~Pharmaceutical~ or ~'drug~ refers to any therapeutic or prophylactic agent which is used in the prevention, diagnosis, alleviation, treatment or cure of a W O96/26179 PCTrUS96/01471 malady, a~liction, disease or injury in a patient.
Therapeutically use~ul polynucleotides and polypeptides are included within the definition of drug.
"In combination with~ re~ers to the incorporation oi~ a bioactive agent with a cationic lipid compound of the present invention. The cationic lipid compound can be combined with the bioactive agent in any of a variety of dif~erent ways. For example, when the cationic lipid compound is in the form of a cationic vesicle or a cationic vesicular composition, the bioactive agent may be entrapped within the internal void of the vesicle. It is also contemplated that the bioactive agent may be integrated within the layer(s) or wall(s) of the vesicle, for example, by being interspersed among lipids which are contained within the vesicular layer(s) or wall(s). In addition, it is contemplated that the bioactive agent may be located on the surface o~ a vesicle. In this case, the bioactive agent may interact chemically with the surface of the vesicle and remain substantially adhered thereto. Such interaction may take the ~orm o~, ~or example, electrostatic interactions, hydrogen bonding, van der Waal's ~orces or covalent bonding.
Alternatively, or in addition to, the bioactive agent may interact with the surface o~ the vesicle in a limited ~nn~, Such limited interaction would permit migration o~
the bioactive agent, for example, from the sur~ace of a ~irst vesicle to the sur~ace of a second vesicle.
"Intracellular" or "intracellularly" refers to the area within the plasma membrane of a cell, including the W O96/26179 PCTrUS96/0147 protoplasm, cytoplasm and/or nucleoplasm. "Intracellular delivery" refers to the delivery of a bioactive agent into the area within the plasma membrane of a cell.
_"Cell" refers to any one of the minute protoplasmic masses which make up organized tissue, comprising a mass of protoplasm surrounded by a membrane, including nucleated and unnucleated cells and organelles.
"Immune competence" refers to the ability of the immune system to protect against pathogens or infectious agents.
The present invention is directed, in part, to a new class of cationic lipid compounds which are highly useful in connection with the intracellular delivery of one or more bioactive agents. The new class of lipids are described in more detail below.
Specifically, in one embodiment, the present invention relates to a cationic lipid compound of the formula ~- Y3)z Yl (Rl- Xl~- R2- [Y2- R3]y-(Xl- Rl)x- Y

wherein:
20each of x, y and z is independently an integer from 0 to about 100;
each X1 is independently -O-, -S-, -NRs-, -C(=X2)-, -C(=X2)-N(R5)-, -N(Rs)-C(=X2)-, -C(=X2)-O-, W O 96/26179 PCTrUS96/01474 -O-C(=X2) - or -X2-(R5X2) P(=X2) -X2-;
each X2 is independently O or S;
each Y1 is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer ~rom 1 to 3;
each Y2 is independently -N(R6) b- ~ -S (R6) b- or -P (R6) b- ~ wherein b is an integer ~rom 0 to 2;
each Y3 iS independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer ~0 ~rom 1 to 3;
each o~ Rl, R2, R3 and R4 is independently alkylene o~ 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl o~
1 to about 10 carbons; and each R6 is independently -[R7-X3] C-R8 or -Rg- [X4-Rlo] d- Q, wherein:
each of c and d is independently an integer ~rom 0 to about 100;
each Q is independently a phosphate residue, ~N(R11)q, ~S(Rl1)q, ~P(Rll)q or -CO2Rll, wherein q is an integer ~rom 1 to 3;
each of X3 and X4 is independently -O-, -S-, -NR5-, -C (=X2) -, -C (=X2) -N(R5) -, -N(R5) -C(=X2) -, -C (=X2) -O-, -O-C(=X2) - or -X2- (R5x2) P(=X2) -X2-;
each R7 is independently alkylene of 1 to about 20 carbons each R8 is independently hydrogen or alkyl o~
1 to about 60 carbons;

W O 96126179 PCTrUS96/01474 each of Rg and Rlo is independently alkylene of 1 to about 20 carbons; and each R1l is independently -[R7-X3]C-R8 or ~ -Rg-[X4-Rlo] d-W, wherein:
each W is independently a phosphate residue, ~N(Rl2)w, -S(Rl2)W~ -P(Rl2)w or -COzRl2, wherein w is an integer from 1 to 3; and Rl2 is -[R7-X3]C-R8; with the proviso that the compound of formula (I) comprises at least one, and preferably at least two, quaternary salts.
In the above formula (I), each of x, y and z is independently an integer from 0 to about 100. Preferably, each of x, y and z is independently an integer of from 0 to about 50, with integers from 0 to about 20 being more preferred. Even more preferably, each of x, y and z is independently an integer from 0 to about 10, with integers from 0 to about 5 being still more preferred. In certain particularly preferred embodiments, x is 1, y is 2 or 3 and z is 0 or 1.
In the above formula (I), each Xl is independently -O-, -S-, -NRs-, -C(=X2)-, -C(=X2)-N(Rs)-, -N(Rs)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(RsX2)P(=X2)-X2-. Pre~erably, each Xl is independently -C(=O)-NRs-, -NRs-C(=O)-, -C(=O)-O-or -O-C(=O)-.
Each X2 in the definitions of Xl, X3 and X4 above is independently O or S. Preferably, X2 is O.
In the above ~ormula (I), each Yl is independently a phosphate residue, N(R6)~-, s(R6)a-~ P(R6)a- or -CO2R6, W O 96/26179 PCT~US96/01474 wherein a is an integer from 1 to 3. Pre~erably, each Yl is independently a phosphate residue, N(R6)a- or -CO2R6, wherein a is 2 or 3. Pre~erably, a is 3.
Each Y2 in ~ormula (I) above is independently -N(R6) b- ~ -S (R6) b- or -P(R6) b- ~ wherein b is an integer from 0 to 2. Preferably, Y2 is -N(R6) b- ~ wherein b is 1 or 2.
In the above formula (I), each Y3 is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer from 1 to 3. Pre~erably, each Y3 is independently a phosphate residue, N(R6) a~ or -CO2R6, wherein a is 2 or 3. Preferably, a is 3.
In the above formula (I), each of Rl, R2, R3 and R4 is independently alkylene of~ 1 to about 20 carbons.
Preferably, each of Rl, R2, R3 and R4 is independently straight chain alkylene of 1 to about 10 carbons or cycloalkylene o~ about 4 to about 10 carbons. More preferably, each of~ Rl, R2, R3 and R4 is independently straight chain alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons. Even more preferably, each of Rl, R2, R3 and R4 is independently methylene, ethylene or cyclohexylene.
In the above definitions o~ Xl, X3 and X4, each R5 is independently hydrogen or alkyl of 1 to about 10 carbons.
Preferably, each R5 is independently hydrogen or alkyl of 1 to about 4 carbons. More preferably, R5 is hydrogen.
In the above definitions of Yl, Y2 and Y3, each R6 is independently -[R,-X3] C-R8 or -Rg- [X4-Rlo] d-Q, wherein each of c and d is independently an integer from 0 to about 100.

W O96/26179 PCTrUS96/0147 Preferably, each of c and d is independently an integer from 0 to about 50, with integers from 0 to about 20 being more preferred. Even more preferably, each of c and d is _ independently an integer from 0 to about 10, with integers from 0 to about 5 being still more preferred. In certain particularly preferred embodiments, c is 0 or 1 and d is 1.
Each Q in R6 above is independently a phosphate residue, ~N(Rll)ql ~S(Rll)q, ~P(Rll)q or -CO2Rll, wherein q is an integer from 1 to 3. Preferably, each Q is independently a phosphate residue, -N(Rll)q or -CO2Rll, wherein q is 2 or 3.
Preferably, q is 3.
Also in the above definition of R6, each of X3 and X4 is independently -O-, -S-, -NR5-, -C (=X2) -, -C (=X2)-N(Rs)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C (=X2) - or -X2-(R5X2)P(=X2)-X2-, wherein each of X2 and R5 is independently as previously described. Preferably, each of X3 and X4 iS independently -C(=O)-NR5-, -NR5-C(=O)-, -C(=O)-O- or -O-C(=O)-.
In the definitions o~ R6, Rll and Rl2 above, each R7 is independently alkylene of 1 to about 20 carbons.
Preferably, each R7 is independently alkylene of 1 to about 10 carbons, with alkylene of 1 to about 4 carbons being preferred. More preferably, each R7 is independently methylene or ethylene.
Also in the definitions of R6, Rll and Rl2 above, each R8 is independently hydrogen or alkyl of 1 to about 60 carbons. Preferably, each R8 is independently hydrogen or alkyl of 1 to about 40 carbons, with hydrogen or alkyl o~ 1 to about 20 carbons being more preferred. Even more W O96126179 PCTrUS96/01474 - 28 -pre~erred, each R8 is independently hydrogen or alkyl of 1 to about 16 carbons. In certain particularly pre~erred embodiments, each R8 is independently hydrogen, methyl, dodecyl or hexadecyl.
Each o~ Rg and R1o in the definitions of R6 and R
above is independently alkylene o~ 1 to about 20 carbons.
Preferably, each o~ Rg and R~o is independently alkylene of 1 to about 10 carbons. More preferably, each of Rg and R1o is independently alkylene of l to about 4 carbons. Even more pre~erably, each o~ Rg and R1o is independently methylene or ethylene.
Each R11 in Q above is independently -[R7-X3]C-R8 or -Rg-[X4-Rlo]d~W, wherein each of c, d, X3, X4, R7, R8, Rg and R1o is independently as previously described.
Each W in R11 above is independently a phosphate residue, -N(R12)W, -S(R12)W, -P(R12)W or -CO2R12, wherein w is an integer ~rom 1 to 3. Pre~erably, W is a phosphate residue, -N(R12)W or -CO2R12, wherein w is 2 or 3. Pre~erably, w is 3.
In the above de~inition of W, R12 is -[R7-X3]c~R8 wherein each of c, X3, R7 and R8 is independently as previously described.
In another embodiment of the present invention, there is provided a cationic lipid compound of the formula YiRiY

wherein:

WO 96/26179 PCT/US96/0147~1 each Yl is independently a phosphate residue, N(R2)a-, S(R2)a-, P(R2)a- or -CO2R2, wherein a is an integer from 1 to 3;
- Rl is alkylene of 1 to about 60 carbons cont~'n'ng 0 to about 30 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
R2 is a residue of the formula -R4 - [ (Xl-R5) X-Y2] y~R6~ wherein:
each of x and y is independently an integer from 0 to about 100;
each Xl is independently a direct bond, -O-, -S-, -NR3-, -C (=X2) -, -C (=X2) -N (R3) -, -N (R3) -C (=X2) -, -C (=X2) -O-, -O-C (=X2) - or -X2- (R3X2) P (=X2) -X2-;
each X2 is independently O or S;
each Y2 is independentlY -S(R2) b- ~ -N (R2) b- or -P (R2) b- ~ wherein b is an integer ~rom 0 to 2;
each R3 is independently hydrogen or alkyl of 1 to about 10 carbons;
each of R4 and R5 is independently a direct bond or alkylene of 1 to about 30 carbons containing 0 to about 15 -O-, -S-, -NR3- or -X2- (R3X2) P (=X2) -X2- heteroatoms or heteroatom groups; and each R6 is independently hydrogen or alkyl of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -25 NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
with the proviso that the compound o~ formula (II) comprises at least one, and preferably at least two, ~uaternary salts.

W O 96/26179 PCTrUS96/01474 In the above formula (II), each Yl is independently a phosphate residue, N(R2)a-, S(R2)a-, P(R2)a- or -CO2R2, wherein a is an integer from 1 to 3. Preferably, each Yl is independently a phosphate residue, -N(R2) a~ or -CO2R2, wherein a is 2 or 3. Preferably, a is 3.
Also in the above formula (II), Rl is alkylene of 1 to about 60 carbons containing o to about 30 -o-, -S-, -NR3-or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups.
Preferably, R1 is alkylene of 1 to about 40 carbons, with lD alkylene of 1 to about 20 carbons being preferred. More preferably, Rl is straight chain alkylene of 1 to about 10 carbons or cycloalkylene of about 4 to about 10 carbons.
Even more preferably, Rl is straight chain alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons.
In the above definition of Yl, R2 is a residue of the formula -R4-[(X1-Rs)x-Y2]y~R6~ wherein each of x and y is independently an integer from 0 to about 100. Preferably, each of x and y is independently an integer from 0 to about 50, with integers from 0 to about 20 being more preferred.
Even more preferably, each of x and y is independently an integer from 0 to about 10.
In the above definition o~ R2, each Xl is independently a direct bond, -O-, -S-, -NR3-, -C(=X2)-, -C (=X2) -N(R3)-, -N(R3)-C(=X2)-, -C(=X2)-O-, -O-C (=X2) - or -X2-(R3X2)P(=X2)-X2-. Preferably, X1 is a direct bond, -C(=X2)-N(R3)-, -N(R3)-C(=X2)-, -C(=X2)-O or -o-C(=X2)-.

W O96/26179 PCTrUS96/01474 Each X2 in the above definitions of Xl, R1, R4, R5 and R6 is independently O or S. Preferably, X2 is O.
Each Y2 in the above definition of R2 is independently -S(R2) b- ~ -N~R2) b- or -P(R2) b- ~ wherein b is an integer of from 0 to 2. Preferably, Y2 is -N(R2) b- and b is or 2.
In the above definitions of Xl, Rl, R~, R5 and R6, each R3 is independently hydrogen or alkyl of 1 to about 10 carbons. Pre~erably, each R3 iS independently hydrogen or 10 alkyl of 1 to about 4 carbons. More preferably, R3 iS
hydrogen.
In the above definition of R2, each of R4 and R5 iS
independently a direct bond or alkylene of 1 to about 30 carbons containing 0 to about 15 -O-, -S-, -NR3- or 15 -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups.
Preferably, each of R4 and R5 iS independently a direct bond or alkylene o~ l to about 20 carbons. More preferably, each of R4 and R5 iS independently a direct bond, straight chain alkylene of 1 to about 10 carbons or cycloalkylene of 4 to 20 about 10 carbons. Even more preferably, each o~ R4 and R5 iS
independently a direct bond, straight chain alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons.
Each R6 in R2 above is independently hydrogen or 25 alkyl of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups. Preferably, each R6 is independently hydrogen or W O96/26179 PCTrUS96/01474 ~ 32 -alkyl of 1 to about 40 carbons. More preferably, each R6 is independently hydrogen or alkyl of 1 to about 20 carbons.
In yet another embodiment of the present invention, there is provided a cationic lipid compound of the formula ~ - Y3)z Y1 (Ri- Xl)X - R2 - tY2- R3]y-(Xl- Rl)x~lY1 (~l-Xl)~ X~),c lR2 IR2 Y i(Ri-Xl)X-tR3-lY2]y- R2-(xiRl)x- Y
~ - Y3)z wherein:
each of x, y and z is independently an integer from O to about 100;
each X1 is independently -O-, -S-, -NRs-, -C(=X2)-l -C(=X2)-N(R5)-~ -N(Rs)-C(=X2)-, -C(=X2)-O-~-O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-i each X2 iS independently O or S;
each Yl is independently -O-, -N(R6)a~, -S(R6)a~
or ~P(R6)a~, wherein a is an integer from O to 2;
each Y2 is independently -N(R6)a~, -S(R6)a~ or ~P(R6)a~, wherein a is an integer from O to 2;
each Y3 iS independently a phosphate residue, N(R6) b- ~ S (R6) b- ~ P (R6) b- or -C02R6, wherein b is an integer from 1 to 3;

CA 022l34l7 l997-08-20 W O 96/26179 PCTrUS96/01474 each of R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of ~ 1 to about 10 carbons; and each R6 is independently -[R,-X3] C-R8 or -Rg- [X4-Rlo] d-Q ~ wherein:
each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, ~N(R11)q, ~S~R11)q, ~P(R11)q or -CO2R11, wherein q is an integer from 1 to 3;
each of X3 and X4 iS independently -O-, -S-, -NR5-, -C(=X2) -, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-i each R, is independently alkylene of 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 60 carbons;
each of Rg and R1o is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R,-X3] C-R8 or -Rg- [X4-Rlo] d-W, wherein:
each W is independently a phosphate residue, ~N(R12)w, -S(R12)w~ -P(Rl2)w or -CO2R12, wherein w is an integer from 1 to 3; and - Rl2 iS - [R,-X3] C-R8i with the proviso that the compound of formula (III) comprises at least one, and preferably at least two, quaternary salts.

W O96/26179 PCT~US96/0147 - 34 -In the above formula (III), each of x, y and z is independently an integer ~rom 0 to about 100. Preferably, each of x, y and z is independently an integer from 0 to about 50, with integers from 0 to about 20 being more preferred. Even more preferably, each of x, y and z is independently an integer from 0 to about 10. Still more preferably, each of x, y and z is independently an integer from 0 to about 5. In certain particularly preferred embodiments, x is 1, y is 2 or 3 and z is 0 or 1.
In the above formula (III), each Xl is independently -O-, -S-, -NR5-, -C (=X2) ~ ~ ~C (=X2) -N(Rs)-, -N(Rs)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(RsX2)P(=X2)-X2-. Preferably, each Xl is independently -C(=O)-NRs-, -NRs-C(=O)-, -C(=O)-O-or -O-C(=O)-.
In the above definitions of Xl, X3 and X4, each X2 is independently O or S. Preferably, X2 is O.
Each Yl in formula (III) above is independently -O-, -N(R6) a~ ~ ~S (R6) a~ or -P(R6) a~ ~ wherein a is an integer from 0 to 2. Preferably, Yl is -N(R6) a~ ~ wherein a is 1 or 2.
Each Y2 in formula (III) above is independently -N(R6)a-, -S(R6)a- or -P(R6)a-, wherein a is an integer from o to 2. Preferably, Y2 is -N(R6) a~ -In the above formula (III), each Y3 iS
independently a phosphate residue, N(R6)b-, S(R6)b-, P(R6)b- or -CO2R6, wherein b is an integer ~rom 1 to 3. Preferably, each Y3 iS independently a phosphate residue or N(R6) b-wherein b is 2 or 3. Preferably, b is 3.

CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 - 35 -In the above formula (III), each of Rl, R2, R3 and R4 iS independently alkylene of 1 to about 20 carbons.
Preferably, each of Rl, R2, R3 and R4 is independently straight chain alkylene of 1 to about 10 carbons or cycloalkylene of about 4 to about 10 carbons. More preferably, each of Rl, R2, R3 and R4 iS independently straight chain alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons. Even more preferably, each of Rl, R2, R3 and R4 iS independently methylene, ethylene or cyclohexylene.
In the above definitions of Xl, X3 and X4, each R5 is independently hydrogen or alkyl of 1 to about 10 carbons.
Preferably, each R5 iS independently hydrogen or alkyl of 1 to about 4 carbons. More preferably, R5 is hydrogen.
In the above definitions of Yl, Y2 and Y3, each R6 is independently -[R7-X3] C-R8 or -Rg-[X4-Rlo] d-Q, wherein each of c and d is independently an integer from 0 to about 100.
Preferably, each of c and d is independently an integer ~rom 0 to about 50, with integers from 0 to about 20 being more preferred. Even more preferably, each of c and d is independently an integer from 0 to about 10, with integers from 0 to about 5 being still more preferred. In certain particularly preferred embodiments, c is 0 or 1 and d is 1.
Each Q in R6 above is independently a phosphate residue, ~N(Rll)q, ~S(Rll)q, ~P(Rll)q or -CO2Rll, wherein q is an integer from 1 to 3. Preferably, each Q is independently a phosphate residue, ~N(Rll)q or -CO2Rll, wherein q is 2 or 3.
Preferably, q is 3.

Also in the above definition of R6, each of X3 and X4 iS independently -O-, -S-, -NR5-, -C (=X2) - ~ -C (=X2) -N (R5) --N (R5) - C ( =X2) -, - C ( =X2) -O -, -O -C ( =X2 ) - or -X2-(R5X2)P(=X2)-X2-, wherein X2 and R5 are as previously described. Preferably, each of X3 and X4 iS independently -C(=O) -NR5-, -NR5-C (=O) -, -C(=O)-O- or -O-C(=O)-.
In the definitions of R6, Rll and Rl2 above, each R, is independently alkylene of 1 to about 20 carbons.
Preferably, each R7 is independently alkylene of l to about 10 carbons, with alkylene of 1 to about 4 carbons being preferred. More preferably, each R, is independently methylene or ethylene.
Also in the de~initions of R6, Rl1 and R12 above, each R8 is independently hydrogen or alkyl of 1 to about 60 carbons. Preferably, each R8 is independently hydrogen or alkyl of 1 to about 40 carbons, with hydrogen or alkyl o~ 1 to about 20 carbons being more preferred. In certain particularly preferred embodiments, each R8 is independently hydrogen, methyl, dodecyl or hexadecyl.
Each of Rg and R1o in the definitions o~ R6 and R
above is independently alkylene of 1 to about 20 carbons.
Preferably, each of Rg and Rlo is independently alkylene of 1 to about lo carbons. More preferably, each o~ Rg and Rlo is independently alkylene of 1 to about 4 carbons. Even more preferably, each of Rg and Rlo is independently methylene or ethylene.
In Q above, each Rll is independently -[R,-X3] C-R8 or W O96/26179 PCTAU$96/01474 -Rg-[X4-Rlo]d~W~ wherein each of c, d, X3, X4, R" R8, Rg and Rlo is independently as previously described.
Each W in Rll above is independently a phosphate residue, -N(Rl2)W, -S(Rl2)W, -P(Rl2)W or -CO2Rl2, wherein w is an integer from 1 to 3. Preferably, each W is independently a phosphate residue, -N(Rl2)W or -CO2Rl2, wherein w is 2 or 3.
Pre~erably, w is 3.
In W above, Rl2 is -[R7-X3]C-R8, wherein each of c, X3, R7 and R8 is independently as previously described.
In the above formulas, it is intended that when any symbol appears more than once in a particular formula or substituent, its m~nlng in each instance is independent of the other.
Also in the above formulas, it is intended that when each of two or more adjacent symbols is defined as being "a direct bond" to provide multiple, adjacent direct bonds, the multiple and adjacent direct bonds devolve into a single direct bond.
The compounds of formulas (I), (II) and (III) above are exemplary of the cationic lipid compounds which are the subject of the present invention. The cationic or positively charged properties of the cationic lipid compounds is due to the presence of at least one cationic group. In pre~erred embodiments, at least two cationic groups are present in the cationic lipid compounds o~ the _ present invention. The existence o~ the cationic groups imparts desirable and beneficial properties to the cationic lipid compounds, such properties being absent ~rom lipid W O 96/26179 PCTrUS96/01474 compounds known heretofore. In particular, the cationic lipid compounds of the present invention possess improved ability to bind and/or chelate with bioactive agents relative to lipid compounds of the prior art. This binding and/or chelation o~ the present cationic lipid compounds with bioactive agents is referred to generally hereinafter as "interaction". Accordingly, the cationic lipid compounds of the present invention are particularly suitable for use as carriers ~or bioactive agents and for the intracellular delivery of bioactive agents.
While the inventors do not wish to be bound by any theory or theories of operation, it is believed that the improved ability of the cationic lipid compounds of the present invention to interact with bioactive agents is due, at least in part, to the enhanced charge densities of the present lipid compounds. In this connection, the present cationic lipid compounds possess an increased, positive charge density due to the existence of at least one, and preferably at least two, cationic groups. As discussed in detail below, this enhanced charge density results in unexpectedly desirable interaction with bioactive agents.
Bioactive agents, whether neutral (uncharged) or positively or negatively charged, typically contain a dipole moment and/or one or more heteroatoms, ~or example, nitrogen, oxygen and sulfur atoms. These heteroatoms generally possess one or more unshared pairs of electrons.
It is believed that the positively charged lipid compounds o~ the present invention electrostatically interact with the W O 96126179 PCTrUS96/01474 negatively charged region of the dipole moment and/or with the unshared pair(s) of electrons on the heteroatoms.
The cationic lipid compounds of the present invention possess particularly improved abilities to interact with bioactive agents which are anionic and which contain one or more anionic groups. Such anionic bioactive agents possess a greater negative charge density relative to neutral or positively charged bioactive agents.
Due to the improved ability of the cationic lipid compounds of the present invention to interact with bioactive agents, the present lipid compounds are particularly suitable for use as carriers for the intracellular delivery of bioactive agents. Thus, the cationic lipid compounds of the present invention are particularly applicable for use in vitro and/or in vivo in methods for the treatment of diseases, including genetic diseases, which involve or require the intracellular delivery of bioactive agents.
As discussed in detail below, the cationic lipid compounds are also particularly suitable for use in the formulation of cationic vesicles, including micelles and liposomes. The inventors have found that cationic liposomes are also particularly suitable ~or use as carriers ~or the intracellular delivery of bioactive agents.
As noted above, the cationic lipid compounds of the present invention comprise at least one, and pre~erably at least two, cationic groups. In an alternate embodiment, the cationic lipid compounds comprise more than at least two W O96/26179 PCTrUS96/01474 cationic groups, for example, at least three cationic groups. In another alternate embodiment, the cationic lipid compounds comprise at least four cationic groups. In yet another alternate embodiment, the cationic lipid compounds comprise at least five cationic groups. In certain embodiments, the cationic lipid compounds comprise more than five cationic groups.
For purposes of illustration only, and not for purposes of limitation, cationic groups may be provided, for example, in the compounds of formula (I) by the group Yl.
Thus, for example, when Yl in formula (I) is N(R6)a- and a is 3, a quaternary salt is formed in that the nitrogen atom of Yl is bonded to four other carbon atoms. The nitrogen atom is therefore positively charged.
Other cationic groups, in addition to the cationic groups discussed above, would be apparent to one of ordinary skill in the art based on the present disclosure.
In embodiments in which the cationic group comprises a quaternary salt, the cationic lipid compound is generally, although not necessarily, associated with a counter ion. Preferably, the counter ion is a pharmaceutically-acceptable counter ion.
In certain preferred embodiments of the present invention, the counter ion is selected from the group consisting of halide, Rl3SO3-, Rl3CO2-, phosphate, sulfite, nitrate, gluconate, guluronate, galacturonate, estolate and mesylate, wherein Rl3 is hydrogen, alkyl of 1 to about 20 carbons or aryl of about 6 to about 10 carbons. Preferably, W O 96/26179 PCT~US96/01474 Rl3 is hydrogen or alkyl of 1 to about 10 carbons or phenyl.
In other preferred embodiments, the counter ion is halide (fluoride, chloride, bromide or iodide), with iodide being - preferred. Various other counter ions, including pharmaceutically acceptable counter ions, would be apparent to one skilled in the art based on the present disclosure.
As those skilled in the art will recognize, once placed in possession of the present invention, cationic lipid compositions may be readily formulated from the cationic lipid compounds. Depending on the desired physical properties, cationic lipid compositions may be prepared from the cationic lipid compounds, alone or in combination with other materials, for example, materials which act to stabilize the composition.
It is generally desirable to combine the cationic lipid compounds with other materials, including stabilizing materials, for example, additional amphipathic compounds, to stabilize and/or otherwise improve the properties of the compositions. Compositions which are prepared from the present cationic lipid compounds and additional amphipathic compounds include, for example, suspensions, emulsions, vesicles and hexagonal H II phase structures.
A wide variety of materials which act to stabilize the compositions of the present invention are readily available and would be apparent to a person skilled in the art based on the present disclosure. Included among such materials are additional amphipathic compounds, such as lipids, and ~atty materials. The particular stabilizing W O96/26179 PCTrUS96/01474 material which is ultimately combined with the present cationic lipid compounds may be selected as desired to optimize the properties of the resulting composition. It is believed that suitable stabilizing materials are readily identifiable and that compositions of the present cationic lipid compounds can be prepared by one skilled in the art without undue experimentation.
It is also desirable, in certain instances, to combine the cationic lipid compounds with a material which is capable of promoting fusion of the lipid with the cell membrane. Such materials enhance the ability of the cationic lipid compositions to deliver intracellularly the bioactive agent. Certain of such materials are capable also of promoting gene expression. These latter materials are particularly suitable for use in the transfection of genetic material. Examples of materials which are capable of promoting fusion of the cationic lipid composition with cell membranes include, for example, ammonium sulfate, cytochalasin B, chloroquine, glycerol, propylene glycol and poly(ethylene glycol).
In one embodiment of the invention, a cationic lipid composition is provided which comprises a cationic lipid suspension and/or emulsion. Lipid suspensions and emulsions are well known and may be prepared using conventional techniques. As those skilled in the art will recognize, a suspension is a mixture of finely divided particles floating in a liquid, and an emulsion is a colloidal mixture of two or more liquids. The components of W O96/26179 PCT~US96101474 the suspension/emulsion are generally mixed together by mechanical agitation, optionally but preferably in the presence of small amounts of additional substances known as - emulsifiers.
Typically, in preparing the suspension/emulsion, the cationic lipid compounds may be added to ethanol or chloroform or any other suitable organic solvent and agitated by hand or by using mechanical techniques. The solvent is then evaporated from the mixture leaving a dried glaze of cationic lipid. The lipids are resuspended in aqueous media, such as phosphate buffered saline, resulting in a suspension/emulsion. To achieve a more homogeneous size distribution of the involved lipids, the mixture may be sonicated using conventional sonication techniques as well as microfluidization (using, for example, a Microfluidizer, Newton, MA), and/or high pressure extrusion (such as, for example, 600 psi) using an Extruder Device (Lipex Biomembranes, Vancouver, Canada). The lipid may be also subjected to one or more alternating cycles o~ ~reezing and thawing to promote the formation of a substantially uniform suspension/emulsion. In addition, a salt, for example, sodium chloride, is optionally added to the suspension/emulsion in a concentration o~ about 0.05 molar (M) to about 1.0 M to promote the formation of substantially uniform dispersions. Bioactive agents may be added to the cationic lipid compounds during the preparation of the suspension/emulsion, such as at the stage where the lipids are added to the organic solvent or at other stages of W O96/26179 PCTrUS96/0147 preparation, or may be added a~ter the cationic lipid suspension/emulsion has been ~ormed, as desired. In preparing the suspensions/emulsions, particularly useful additives are, for example, soybean lecithin, glucose, Pluronic F-68, and D,L-~-tocopherol (Vitamin E), generally in an amount of about 0.03 to about 5 percent by weight.
These additives are particularly useful where intravenous applications are desired. Techniques and ingredients for formulating lipid suspensions/emulsions are well known in the art and are applicable to the present cationic suspensions/emulsions. Suitable procedures and suspension/emulsion ingredients are reported, for example, in Modern Pharmaceutics, pp. 505-507, Gilbert Baker and Christopher Rhodes, eds., Marcel Dekker Inc., New York, NY
(1990), the disclosures of which are hereby incorporated herein by reference in its entirety.
In another embodiment of the invention, a cationic lipid composition is provided which comprises a cationic ~esicular composition. The cationic vesicular composition may comprise micelles and/or liposomes. With particular reference to cationic micelle compositions, the following discussion is provided.
Micelles may be prepared using any one of a variety of conventional micellar preparatory methods which will be apparent to those skilled in the art. These methods typically involve suspension of the cationic lipid compound in an organic solvent, evaporation of the solvent, resuspension in an aqueous medium, sonication and CA 022l34l7 l997-08-20 W O 96/26179 PCTrUS96/01474 centrifugation. The foregoing methods, as well as others, are discussed, for example, in Canfield et al., Methods in Enzymology, Vol. 189, pp. 418-422 ~1990); El-Gorab et al, - Biochem. Biophys. Acta, Vol. 306, pp. 58-66 (1973);
Colloidal Surfactant, Shinoda, K., Nakagana, T~m~mll~hi and Isejura, Academic Press, NY (1963) (especially "The Formation of Micelles", Shinoda, Chapter 1, pp. 1-88);
Catalysis in Micellar and Macromolecular Systems, Fendler and Fendler, Academic Press, NY (1975). The disclosures of each of the foregoing publications are incorporated by reference herein, in their entirety. The micelles may be prepared in the presence of a bioactive agent or the bioactive agent may be added to pre-formed micelles.
It is generally desirable to include one or more stabilizing materials in the micellar compositions.
Exemplary materials which may be combined with the cationic lipid compounds to stabilize the micellar compositions produced therefrom include lauryltrimethylammonium bromide, cetyltrimethylammonium bromide, myristyltrimethylammonium bromide, alkyldimethylbenzylammonium chloride, wherein the alkyl group is about 12 to about 16 carbons, benzyldimethyldodecyl~m~on;um bromide or chloride, benzyldimethylhexadecylammonium bromide or chloride, benzyldimethyltetradecylammonium bromide or chloride, cetyldimethylethylammonium bromide or chloride, _ cetylpyridinium bromide and chloride and lauryl sulfate.
Other materials for stabilizing the micellar compositions, in addition to those exempli~ied above, would W O96/26179 PCTrUS96/01474 be apparent to one skilled in the art based on the present disclosure.
As noted above, the cationic vesicular composition may comprise cationic liposomes. Cationic liposomes are particularly effective as carriers for the intracellular delivery of bioactive agents and are therefore preferred cationic lipid compositions. The present cationic liposomes are highly stable and permit substantially complete entrapment of a bioactive agent within the vesicle. Thus, compositions which comprise cationic liposomes are highly e~fective carriers ~or the transfection of bioactive agents in that the liposomes are capable of (A) effectively interacting with the bioactive agent by virtue of electrostatic forces (as discussed above in connection with the cationic lipid compounds, generally); and (B) entrapping the bioactive agent within the liposome vesicle. The cationic liposomes are also highly biocompatible.
The cationic liposome compositions may comprise one or more cationic lipid compounds. In any given liposome, the cationic lipid compound~s) may be in the form of a monolayer or bilayer, and the mono- or bilayer lipids may be used to form one or more mono- or bilayers. In the case of more than one mono- or bilayer, the mono- or bilayers are generally concentric. Thus, the lipids may be used to form a unilamellar liposome (comprised of one monolayer or bilayer), an oligolamellar liposome (comprised of two or three monolayers or bilayers) or a multilamellar -CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 liposome (comprised o~ more than three monolayers or bilayers).
As with the suspensions/emulsions and micelles above, cationic liposome compositions are preferably formulated from both the present cationic lipid compounds and additional stabilizing materials, including additional amphipathic compounds. In the case of liposomes, the additional amphipathic compounds preferably comprise lipids.
A wide variety of additional lipids are available which may be incorporated into the liposome compositions. Preferably, the lipids are selected to optimize certain desirable properties o~ the liposomes, including serum stability and plasma half-life. The selection of suitable lipids in the preparation of cationic liposome compositions would be apparent to a person skilled in the art and can be achieved without undue experimentation, based on the present disclosure.
Lipids which may be used in combination with the present cationic lipid compounds and in the formulation of cationic liposome compositions include ZONYh~
fluorosurfactants (DuPont Chemicals, Wilmington, DE) and the fluorine-cont~;n;ng compounds which are described in the following publications: S. Gaentzler et al., New Journal of Chemistry, Vol. 17(5), pp. 337-344 (1993); C. Santaella et 25 al , New Journal of Chemistry, Vol. 16 (3), pp. 399-404 (1992); and h. sole-Violan, New Journal of Chemistry, Vol.
17(8, 9), pp. 581-583 (1993); the disclosures of each of which are hereby incorporated by re~erence, in their W O 96/26179 PCTrUS96/01474 entireties. Other exemplary lipids which may be used in the preparation of cationic liposome compositions include phosphatidylcholine with both saturated and unsaturated lipids, including dioleoylphosphatidylcholine, dimyristoyl-phosphatidylcholine, dipalmitoylphosphatidylcholine (DPPC)and distearoylphosphatidylcholinei phosphatidylethanol-amines, such as dioleoylphosphatidylethanolamine and dipalmitoylphosphatidylethanolamine (DPPE); phosphatidyl-serine; phosphatidylglycerol; sphingolipids; sphingomyelin;
lysolipids; glycolipids, such as ganglioside GM1;
glucolipids; sulfatides; glycosphingolipids; phosphatidic acids, such as dipalmitoylphosphatidic acid (DPPA); palmitic acid; stearic acid; arachidonic acid; oleic acid; fatty acids; lipids with ether and ester-linked fatty acids;
polymerizable lipids; cholesterol, cholesterol sulfate and cholesterol hemisuccinate; 12-{[(7'-diethylaminocoumarin-3-yl)carbonyl]methylamino}octadecanoic acid; N-[12-{t(7'-diethylaminocoumarin-3-yl)carbonyl]methylamino}-octadecanoyl]-2-aminopalmitic acid; cholesteryl-4'-trimethylaminobutanoate; N-succinyldioleoylphosphatidyl-ethanolamine; 1,2-dioleoyl-sn-glycerol; 1,2-dipalmitoyl-sn-3-succinylglycerol; 1,3-dipalmitoyl-2-succinyl-glycerol; 1-hexadecyl-2-palmitoylglycerophosphatidylethanolamine; and palmitoylhomocysteine.
Lipids bearing polymers, including the hydrophilic polymers poly(ethylene glycol) (PEG), polyvinylpyrrolidone, and poly(vinyl alcohol), may also be included in the liposome compositions o~ the present invention. Examples of W O96/26179 PCTrUS96/01474 suitable hydrophilic polymers include, ~or example, PEG
2,000, PEG 5,000 and PEG 8,000, which have molecular weights of 2,000, 5,000 and 8,000, respectively. Other suitable polymers, hydrophilic and otherwise, will be readily apparent to those skilled in the art based on the present disclosure. Polymers which may be incorporated via alkylation or acylation reactions onto the surface of the liposome are particularly useful for improving the stability and size distribution of the liposomes. Exemplary lipids which bear hydrophilic polymers include, for example, dipalmitoylphosphatidylethanolamine-PEG, dioleoyl-phosphatidylethanolamine-PEG and distearylphosphatidyl-ethanolamine-PEG.
Other materials ~or use in the preparation of cationic liposome compositions, in addition to those exempli~ied above, would be apparent to one skilled in the art based on the present disclosure.
The amount of stabilizing material, such as, ~or example, additional amphipathic compound, which is combined with the present cationic lipid compounds may vary depending upon a variety of ~actors, including the speci~ic cationic lipid compound(s) o~ the invention selected, the speci~ic stabilizing material(s) selected, the particular use ~or which it is being employed, the mode o~ delivery, and the like. The amount o~ stabilizing material to be combined _ with the present cationic lipid compounds in a particular situation, and the ratio o~ stabilizing material to cationic lipid compound, will vary and is readily determinable by one W O96/26179 PCTAUS96/01~74 skilled in the art based on the present disclosure. In general, for example, it has been ~ound that higher ratios, that is, ratios higher than about 4:1, 3:1 or 2:1, of cationic lipid compound to stabilizing lipid, are preferred.
A wide variety of methods are available in connection with the preparation of cationic liposome compositions. Accordingly, the cationic liposomes may be prepared using any one of a variety of conventional liposome preparatory techniques which will be apparent to those skilled in the art. These techniques include solvent dialysis, French press, extrusion (with or without freeze thaw), reverse phase evaporation, microemulsification and simple freeze-thawing. The liposomes may also be prepared by various processes which involve shaking or vortexing.
This may be achieved, for example, by the use of a mechanical shaking device, such as a Wig-L-Bug~ (Crescent Dental, Lyons, IL). Conventional microemulsification equipment, such as a Microfluidizer~ (Microfluidics, Woburn, MA) may be used also.
Additional methods for the preparation of liposome compositions from the cationic lipid compounds of the present invention include, for example, sonication, chelate dialysis, homogenization, solvent infusion, spontaneous formation, solvent vaporization, controlled detergent dialysis, and others, each involving the preparation of liposomes in various ~ashions. Methods which involve freeze-thaw techniques are pre~erred in connection with the preparation of liposomes ~rom the cationic lipid compounds W O 96/26179 PCTrUS96/0147 of the present invention. Suitable freeze-thaw techniques are described, ~or example, in copending U.S. application Serial No. 07/838,504, ~iled February 19, 1992, the disclosures of which are incorporated herein by reference in their entirety. Preparation of the liposomes may be carried out in a solution, such as an aqueous saline solution, aqueous phosphate buffer solution, or sterile water, containing one or more bioactive agents, so that the bioactive agent ls encapsulated in the liposome or incorporated into the liposome membrane. Alternatively, the bioactive agents may be added to previously formed liposomes.
The size of the liposomes can be adjusted, if desired, by a variety of techniques, including extrusion, filtration, sonication and homogenization. In addition, the size of the liposomes can be adjusted by the introduction of a l~m, n~ stream of a core of liquid into an immiscible sheath of liquid. Other methods for adjusting the size of the cationic liposomes and for modulating the resultant liposomal biodistribution and clearance of the liposomes would be apparent to one skilled in the art based on the present disclosure. Preferably, the size of the cationic liposomes is adjusted by extrusion under pressure through pores of a defined size. Although liposomes employed in the subject invention may be of any one of a variety of sizes, preferably the liposomes are small, that is, less than about 100 nanometer (nm) in outside diameter.

CA 022l34l7 l997-08-20 W O96126179 PCTrUS96/01474 Many of the ~oregoing liposomal preparatory techniques, as well as others, are discussed, ~or example, in U.S. Patent No. 4,728,578; U.K. Patent Application GB
2193095 A; U.S. Patent No. 4,728,575; U.S. Patent No.
4,737,323; International Application Serial No.
PCT/US85/01161; Mayer et al., Biochimica et Biophysica Acta, Vol. 858, pp. 161-168 (1986); Hope et al., Biochimica et Biophysica Acta, Vol. 812, pp. 55-65 (1985); U.S. Patent No.
4,533,254; Mayhew et al., Methods in Enzymology, Vol. 149, 10 pp. 64-77 (1987); Mayhew et al., Biochimica et Biophysica Acta, Vol 755, pp. 169-74 (1984); Cheng et al, Investigative Radiology, Vol. 22, pp. 47-55 (1987); International Application Serial No. PCT/US89/05040; U.S. Patent No.
4,162,282; U.S. Patent No. 4,310,505; U.S. Patent No.
15 4,921,706; and Liposome Technology, Gregoriadis, G., ed., Vol. I, pp. 29-31, 51-67 and 79-108 (CRC Press Inc., Boca Raton, FL 1984~, the disclosures o~ each oE which are hereby incorporated by re~erence herein, in their entirety.
Although any o~ a number of varying techniques can 20 be used, the liposomes o~ the present invention are preferably prepared using a shaking technique. Pre~erably, the shaking techniques involve agitation with a mechanical shaking apparatus, such as a Wig-L-Bug~ (Crescent Dental, Lyons, IL), such as those disclosed in copending U.S.
25 application Serial No. 160,232, ~iled November 30, 1993, the disclosures o~ which are hereby incorporated herein by re~erence in their entirety.

W O96/26179 PCTrUS96/0147J

As those skilled in the art will recognize, any of the cationic lipid compounds and compositions containing the cationic lipid compounds, with or without bioactive agents, may be lyophilized for storage, and reconstituted in, for example, an aqueous medium (such as sterile water or phosphate buffered solution, or aqueous saline solution), with the aid of vigorous agitation. To prevent agglutination or fusion of the lipids as a result of lyophilization, it may be useful to include additives which prevent such fusion or agglutination from occurring.
Additives which may be useful include sorbitol, mannitol, sodium chloride, glucose, trehalose, polyvinylpyrrolidone and poly(ethylene glycol), for example, PEG 400. These and other additives are described in the literature, such as in the U.S. Pharmacopeia, USP XXII, NF XVII, The United States Pharmacopeia, The National Formulary, United States Pharmacopeial Convention Inc., 12601 Twinbrook Parkway, Rockville, MD 20852, the disclosures of which are hereby incorporated herein by reference in their entirety.
Lyophilized preparations generally have the advantage of greater shelf life.
The inventors have found that intracellular delivery of bioactive agents through the use of cationic lipid compositions, including suspensions/emulsions and vesicular compositions, may be enhanced by the presence of a _ gaseous substance. It is contemplated that the gaseous substance promotes uptake by cells of the bioactive agent.
Thus, in certain preferred embodiments, a gas, such as an -W O96126179 PCTrUS96/01474 - 54 -inert gas, is incorporated in the cationic lipid compositions. Alternatively, a precursor to a gaseous substance may be incorporated in the cationic lipid compositions. Such precursors include, for example, materials which are capable ol~ converting in vivo to a gas, and pre~erably, to an inert gas.
Preferred gases are gases which are inert and which are biocompatible, that is, gases which are not injurious to biological ~unction. Preferable gases include those selected ~rom the group consisting o~ air, noble gases, such as helium, neon, argon and xenon, carbon dioxide, nitrogen, ~luorine, oxygen, sulfur hexa~luoride, ~luorocarbons, perfluorocarbons, and mixtures thereo~.
Other gases, including the gases exemplified above, would be readily apparent to one skilled in the art based on the present disclosure.
In pre~erred embodiments, the gas comprises a perfluorocarbon. Preferably, the per~luorocarbon is selected from the group consisting o~ perfluoromethane, per~luoroethane, per~luoropropane, per~luorobutane, perfluorocyclobutane, and mixtures thereo~. More preferably, the perfluorocarbon gas is per~luoropropane or perfluorobutane, with perfluoropropane being particularly pre~erred.
As noted above, it may also be desirable to incorporate in the cationic lipid compositions a precursor to a gaseous substance. Such precursors include materials that are capable o~ being converted in vivo to a gas.

W O96/26179 PCTrUS96/01474 Preferably, the gaseous precursor is biocompatible, and the gas produced in vivo is biocompatible also.
Among the gaseous precursors which are suitable for use in the present compositions are pH sensitive agents.
These agents include materials that are capable of evolving gas, for example, upon being exposed to a pH that is neutral or acidic. Examples of such pH sensitive agents include salts of an acid which is selected from the group consisting of inorganic acids, organic acids and mixtures thereof.
Carbonic acid (H2CO3) is an example of a suitable inorganic acid, and aminomalonic acid is an example of a suitable organic acid. Other acids, including inorganic and organic acids, would be readily apparent to one skilled in the art based on the present disclosure.
Preferably, the gaseous precursor is a salt which is selected from the group consisting of an alkali metal salt, an ammonium salt and mixtures thereof. More preferably, the salt is selected from the group consisting of carbonate, bicarbonate, sesquecarbonate, aminomalonate and mixtures thereo~.
Examples of gaseous precursor materials for use in the cationic lipid compositions of the present invention include lithium carbonate, sodium carbonate, potassium carbonate, lithium bicarbonate, sodium bicarbonate, potassium bicarbonate, magnesium carbonate, calcium carbonate, magnesium bicarbonate, ammonium carbonate, ~mmnn; um bicarbonate, ~mmnn; um sesquecarbonate, sodium sesquecarbonate, sodium aminomalonate and ammonium W O96126179 PCTrUS96/01474 - 56 -aminomalonate. Aminomalonate is well known in the art, and its preparation is described, for example, in Thanassi, Biochemistry, Vol. 9, no. 3, pp. 525-532 (1970); Fitzpatrick et al., Inorganic Chemistry, Vol. 13, no. 3 pp. 568-574 (1974); and Stelmashok et al., Koordinatsionnaya Khimiya, Vol. 3, no. 4, pp. 524-527 (1977). The disclosures o~ these publications are hereby incorporated herein by reference.
In addition to, or instead of, being sensitive to changes in pH, the gaseous precursor materials may also comprise compounds which are sensitive to changes in temperature. Such temperature sensitive agents include materials which have a boiling point of greater than about 37~C. Exemplary temperature sensitive agents are methyl lactate, perfluoropentane and perfluoroh~n~. The gaseous precursor materials may be also photoactivated materials, such as diazonium ion and aminomalonate. As discussed more ~ully hereinafter, certain lipid compositions, and particularly vesicular compositions, may be designed so that gas is formed at the target tissue or by the action of sound on the particle. Examples of gaseous precursors are described, for example, in U.S. Patent Nos. 5,088,499 and 5,149,319. These patents are hereby incorporated herein by reference in their entirety. Other gaseous precursors, in addition to those exemplified above, will be apparent to one skilled in the art based on the present disclosure.
In certain preferred embodiments, a gaseous agent, for example, air or a perfluorocarbon gas, is combined with a liquid perfluorocarbon, such as perfluorohexane, W O96/26179 PCTrUS96/0147 perfluoroheptane, perfluorooctylbromide (PFOB), perfluorodecalin, perfluorododecalin, perfluorooctyliodide, perfluorotripropylamine and perfluorotributylamine.
A preferred composition for use in the S intracellular delivery of a bioactive agent, for example, genetic material, comprises a bioactive agent, a perfluorocarbon gas and a gaseous precursor which has a boiling point of greater than about 37~C, such as perfluoropentane. As discussed in detail below, energy, for example, heat or ultrasound, is preferably applied to the patient after the administration of the composition and to assist in the intracellular delivery of the bioactive agent.
The gaseous substances and/or gaseous precursors are preferably incorporated in the cationic lipid compositions of the present invention irrespective of the physical nature of the composition. Thus, it is contemplated that the gaseous substances and/or precursors thereto are incorporated in compositions which are suspensions/emulsions or vesicular compositions, including micelles and liposomes. Incorporation of the gaseous substances and/or precursors thereto in the cationic lipid compositions may be achieved by using any o~ a number of methods. For example, the ~ormation of gas-filled vesicles can be achieved by shaking or otherwise agitating an aqueous mixture which comprises a gas or gas precursor and the cationic lipids of the present invention. This promotes the formation of stabilized vesicles within which the gas or gas precursor is encapsulated. Gas or gaseous precursor W O96/26179 PCT~US96/01474 cationic lipid compositions may be prepared in other manners similar to those discussed in connection with the incorporation of bioactive agents in vesicular compositions as earlier discussed.
The gaseous substances and/or precursors thereto may also be incorporated in the cationic lipid compositions using any conventional and well-known techniques. For example, a gas may be bubbled directly into an aqueous mixture of the present cationic lipid compounds, optionally in the presence of a bioactive agent. Alternatively, a gas instillation method can be used as disclosed, ~or example, in U.S. Patent Nos. 5,352,435 and 5,228,446, the disclosures of each of which are hereby incorporated herein by reference in their entireties. Suitable methods for incorporating the gas or gas precursor in cationic lipid compositions are disclosed also in U.S. Patent No. 4,865,836, the disclosure o~ which is hereby incorporated herein by reference. Other methods would be apparent to one skilled in the art based on the present disclosure.
In preferred embodiments, the gaseous substances and/or gaseous precursor materials are incorporated in vesicular compositions, with micelles and liposomes being preferred. Liposomes are particularly pre~erred because of their high stability and biocompatability. As discussed in detail below, vesicles in which a gas or gas precursor or both are encapsulated are advantageous in that they can be more easily monitored in vivo, for example, by monitoring techniques which involve ultrasound. Thus, the circulation CA 022l34l7 l997-08-20 W O 96/26179 PCTrUS96/01474 and delivery o~ the vesicles to the targeted tissue and/or cells can be observed via a non-invasive procedure. Gas precursor- or gas-filled vesicles are preferred also because the application of high energy ultrasound, radio frequency, optical energy, for example, laser light, and/or heat, to produce areas of hyperthermia, can be used to rupture in vivo the vesicles and thereby promote release of the entrapped gas (or precursor thereto) and bioactive agent.
Thus, vesicular compositions permit the controlled release of a bioactive agent in vivo.
In addition to being entrapped within the vesicle, it is contemplated that the bioactive agent may be located also, or instead o~, outside of the vesicles or in the lipid membranes. Thus, in certain embodiments, the bioactive agent may be coated on the surface of the liposomes or micelles and/or in the lipid membranes, in addition to, or instead of, being entrapped within the vesicles.
The bioactive agent which is incorporated in the present cationic lipid compositions is preferably a substance which is capable o~ exerting a therapeutic biological effect in vitro and/or in vivo. Particularly suitable bioactive agents for use in the methods and compositions of the present invention is genetic material.
Examples of genetic materials include, for example, genes carried on expression vectors, such as plasmids, phagemids, cosmids, yeast arti~icial chromosomes (YACs) and defective-or ~helper~ viruses; anti-sense and sense oligonucleotides;
phosphorothioate oligodeoxynucleotides; antigene nucleic CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 acids; and single and double stranded RNA and DNA, including DNA which encodes at least a portion of a gene, for example, DNA which encodes for human leukocyte antigen (HLA), dystrophin, cystic fibrosis transmembrane receptor (CFTR), interleukin-2 (IL-2), tumor necrosis factor (TNF) and granulocyte-macrophage colony stimulating factor (GMCSF).
The DNA can also encode certain proteins which may be used in the treatment of various types of pathologies or conditions, including those which are associated with the loss or deterioration of immune competence. Such pathologies or conditions involving immune competence include, for example, acquired immune deficiency syndrome (AIDS), cancer, chronic viral infections, and autoimmune disease.
Specifically, DNA may be selected which expresses adenosine de~min~e (ADA) for the treatment of ADA
deficiency; growth hormone for the treatment of growth deficiency or to aid in the healing of tissues; insulin for the treatment of diabetes; luteinizing hormone releasing hormone (LHRH) antagonist as a birth control agent; LHRH for the treatment of prostate or breast cancer; tumor necrosis factor and/or interleukin-2 for the treatment of advanced cancers; high-density lipoprotein (HDL) receptor for the treatment of liver disease; thymidine kinase for the treatment of ovarian cancer, brain tumors, or human immunode~iciency virus (HIV) in~ection; HLA-B7 for the treatment of malignant melanoma; IL-2 for the treatment of neuroblastoma, malignant melanoma or kidney cancer;

-W O 96/26179 PCTrUS96/01474 - 61 -interleukin-4 (IL-4) for the treatment of cancer; HIV env for the treatment of HIV infection; antisense ras/pS3 for the treatment of lung cancer; and Factor VIII for the treatment of Hemophilia B. Such therapies are described, for example, in Science, Vol. 258, pp. 744-746 (1992), the disclosure of which is incorporated herein by reference in its entirety.
As noted above, the present invention provides cationic lipid formulations which comprise cationic lipid compositions in combination with one or more bioactive agents. The cationic lipid compositions may comprise cationic suspensions/emulsions and/or cationic vesicular compositions, including cationic liposome compositions and/or cationic micelle compositions. In addition, the cationic lipid compositions can comprise one or more cationic lipid compounds optionally in combination with a stabilizing material, such as an amphipathic compound, and a gas or precursor thereto. These cationic lipid formulations may be prepared according to any of a variety o~ techniques.
For example, the cationic lipid formulations may be prepared ~rom a mixture of cationic lipid compounds, bioactive agent and gas or gaseous precursor. In the case of vesicular compositions, it is contemplated that the bioactive agent is entrapped within the vesicle of the liposome or micelles.
In certain cases, the bioactive agent can be incorporated _ also into the membrane walls o~ the vesicle. In the case of a suspension/emulsion, it is contemplated that the bioactive agent is generally dispersed homogeneously throughout the W O96/26179 PCTrUS96/01474 suspension/emulsion. Alternatively, the cationic lipid compositions may be preformed ~rom cationic lipid compounds and gas or gaseous precursor. In the latter case, the bioactive agent is then added to the lipid composition prior to use. For example, an aqueous mixture of liposomes and gas may be prepared to which the bioactive agent is added and which is agitated to provide the cationic liposome ~ormulation. The cationic liposome formulation is readily isolated also in that the gas- and/or bioactive agent-filled liposome vesicle generally float to the top of the aqueous solution. Excess bioactive agent can be recovered from the remaining aqueous solution.
The formulations of the present invention can be used in either in vi tro or in vivo applications. In the case of in vitro applications, including cell culture applications, the cationic lipid formulations can be added to the cells in cultures and then incubated. If desired, where liposomes are employed, energy, such as sonic energy, may be applied to the culture media to burst the liposomes and release any therapeutic agents.
With respect to in vivo applications, the ~ormulations o~ the present invention can be administered to a patient in a variety o~ forms adapted to the chosen route o~ administration, namely, parenterally, orally, or 2~ intraperitoneally. Parenteral administration, which is preferred, includes ~m; n; stration by the ~ollowing routes: -intravenous; intramuscular; interstitially; intra-arterially; subcutaneous; intraocular; intrasynovial;

CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 transepithelial, including transdermal; pulmonary via inhalation; ophthalmic; sublingual and buccal; topically, including ophthalmic; dermal; ocular; rectal; and nasal inhalation via insufflation. Intravenous administration is preferred among the routes o~ parenteral administration.
It is contemplated that the present cationic lipid formulations can be administered also by coating a medical device, for example, a catheter, such as an angioplasty balloon catheter, with a cationic lipid formulation.
Coating may be achieved, ~or example, by dipping the medical device into a cationic lipid formulation or a mixture of a cationic lipid formulation and a suitable solvent, for example, an aqueous-based buffer, an aqueous solvent, ethanol, methylene chloride, chloroform and the like. An 1~ amount of the formulation will naturally adhere to the sur~ace of the device which is subsequently administered to a patient, as appropriate. Alternatively, a lyophilized mixture o~ a cationic lipid ~ormulation may be speci~ically bound to the surface of the device. Such binding techniques are described, for example, in K. Ishihara et al., Journal of Biomedical Materials ~esearch, Vol. 27, pp. 1309-1314 (1993), the disclosures of which are incorporated herein by reference in their entirety.
The useful dosage to be administered and the particular mode of administration will vary depending upon such ~actors as the age, weight and the particular animal and region thereo~ to be treated, the particular bioactive agent and cationic lipid compound used, the therapeutic or W 096t26179 PCTAUS96/01474 diagnostic use contemplated, and the form of the formulation, for example, suspension, emulsion, micelle or liposome, as will be readily apparent to those skilled in the art. Typically, dosage is administered at lower levels and increased until the desirable therapeutic effect is achieved. The amount of cationic lipid compound that is administered can vary and generally depends upon the amount of bioactive agent being administered. For example, the weight ratio of cationic lipid compound to bioactive agent is pre~erably from about 1:1 to about 15:1, with a weight ratio of about 5:1 to about 10:1 being more pre~erred.
Generally, the amount of cationic lipid compound which is administered will vary from between about 0.1 milligram (mg) to about 1 gram (g) By way of general guidance, typically between about 0.1 mg and about 10 mg of the particular bioactive agent, and about 1 mg to about loO mg of the cationic lipid compositions, each per kilogram of patient body weight, is administered, although higher and lower amounts can be used.
After vesicular lipid formulations which comprise a gas or gaseous precursor and bioactive agent have been administered to a patient, energy, preferably in the ~orm o~
ultrasonic energy, can be applied to the target tissue to identify the location of the vesicles containing gas or gaseous precursor and bioactive agent. The applied energy may also be employed to effect release of the bioactive agent and facilitates cellular uptake of the bioactive agent. As one skilled in the art would recognize, this .

W O 96/26179 PCTrUS96/01474 method of mediating cellular transfection with ultrasonic energy is preferably effected with tissues whose acoustic window permits the transmission of ultrasonic energy. This is the case for most tissues in the body, including muscle and organ tissues, such as the heart and liver, as well as most other vital structures. With respect to brain tissue, it may be necessary to create a "surgical window" by removing part of the skull, inasmuch as ultrasonic energy generally does not transmit through bone. Intravascular and/or endoluminal ultrasound transducers may be used to apply the ultrasound energy to selected tissues and/or sites in the body, for example, the aorta and the esophagus.
Cationic lipid formulations can be formulated to be sufficiently stable in the vasculature such that they circulate throughout the body and provide blood pool equilibration. As one skilled in the art would recognize, the lipid formulations, including those which comprise suspensions/emulsions and vesicles, such as liposomes and micelles, may be coated with certain materials to minimize uptake by the reticuloendothelial system. Suitable coatings include, for example, gangliosides and glycolipids which bind saccharide moieties, such as glucuronate, galacturonate, guluronate, poly(ethylene glycol), poly(propylene glycol), polyvinylpyrrolidone, poly(vinyl alcohol), dextran, starch, phosphorylated and sulfonated mono-, di-, tri-, oligo- and polysaccharides and albumin.
Provided that the circulation half-life of the cationic lipid formulations is of a sufficient period of time, they W O96/26179 PCTrUS96/0147 will generally pass through the target tissue while passing through the body. In the case of lipid formulations which comprise gas or gaseous precursors, energy, for example, sonic energy, may be focused on the tissue to be treated, for example, diseased tissue. The bioactive agent will then be released locally in the target tissue. The inventors have found also that antibodies, carbohydrates, peptides, glycopeptides, glycolipids and lectins also assist in the targeting of tissue with the lipid formulations and the bioactive agents. Accordingly, these materials may be incorporated into the lipid formulations also.
Ultrasound can be used for both diagnostic and therapeutic purposes. In general, the levels of energy from diagnostic ultrasound are insufficient to cause rupture of vesicular species and to facilitate release and cellular uptake of the bioactive agents. Moreover, diagnostic ultrasound involves the application of one or more pulses of sound. Pauses between pulses permits the reflected sonic signals to be received and analyzed. The limited number of pulses used in diagnostic ultrasound limits the effective energy which is delivered to the tissue that is being studied.
On the other hand, higher energy ultrasound, for example, ultrasound which is generated by therapeutic ultrasound equipment, is generally capable of causing rupture of the vesicular species. In general, therapeutic ultrasound machines use from about 10 to about 100~ duty cycles, depending on the area of tissue to be treated with W O96/26179 PCTrUS96/01474 the ultrasound. Areas of the body which are generally characterized by larger amounts of muscle mass, for example, backs and thighs, as well as highly vascularized tissues, such as heart tissue, may require a larger duty cycle, for S example, up to about 100~.
In therapeutic ultrasound, continuous wave ultrasound is used to deliver higher energy levels. For the rupture of vesicular species, continuous wave ultrasound is pre~erred, although the sound energy may be pulsed also. If pulsed sound energy is used, the sound will generally be pulsed in echo train lengths of about 8 to about 20 or more pulses at a time. Preferably, the echo train lengths are about 20 pulses at a time. In addition, the frequency of the sound used may vary from about 0. 25 to about 100 megahertz (MHz). In general, frequency ~or therapeutic ultrasound ranges between about 0. 75 and about 3 MHz are pre~erred with about 1 and about 2 MHz being more preferred.
In addition, energy levels may vary from about 0. 05 Watt (W) to about 5.0 W, with energy levels of about 0.1 to about 0.5 W being preferred. For very small vesicular species, for example, species in which the vesicles have a diameter o~ less than about O .5 micron, higher ~requencies of sound are generally preferred. This is because smaller vesicular species are capable of absorbing sonic energy more e~ectively at higher ~requencies o~ sound. When very high frequencies are used, ~or example, greater than about 10 MHz, the sonic energy will generally penetrate fluids and tissues to a limited depth only. Thus, external application W O96/26179 PCTÇUS96/01471 - 68 -of the sonic energy may be suitable for skin and other superficial tissues. However, for deep structures it is generally necessary to focus the ultrasonic energy so that it is preferentially directed within a focal zone.
Alternatively, the ultrasonic energy may be applied via interstitial probes, intravascular ultrasound catheters or endoluminal catheters. Such probes or catheters may be used, for example, in the esophagus for the diagnosis and/or treatment of esophageal carcinoma.
The present invention is further described in the following examples. In these examples, examples 1 to 9 are actual examples. Examples 10 to 12 are prophetic examples.
These examples are for illustrative purposes only, and are not to be construed as limiting the appended claims.
Various of the starting materials used in the following examples are commercially available. N,N-dimethylethylenediamine and iodomethane were purchased from Aldrich Chemical Co. (Milwaukee, WI).

W O96/26179 PCT~US96/01474 Exa~nple 1 Synthesis of N,N'-Bis(dodecylaminocarbonylmethylene)-N,N'-bis(~-N,N,N-trimethylammoniumethyl-aminocarbonylmethylene)-N,N'-dimethylethylenediamine tetraiodide (EDTA-LA-TMA tetraiodide) H25C12~ NH

~3 ~ ~
ICH3 NH ~ O CH3 CH3 CH3 Cl2H2s 4I-Synthetic Route (i) Synthesis of N,N'-Bis(dodecylaminocarbonylmethylene)-ethylenediamine-N,N'-diacetic acid (EDTA-LA) H25Cl2~ NH
0//~ 0 ~~ N~--N~Jl'oH
OH ~ O
~ N H
H25Cl2 Dodecylamine (3.71 g, 0.02 mole) in dry methanol (60 mL) was added to a suspension of ethylenediaminetetra-acetic acid dianhydride (2.56 g, 0.01 mole) in dry methanol (30 mL). The mixture was stirred at 50~C ~or 6 hours. The resulting white solid precipitate was isolated by ~iltration CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/0l474 and dried under vacuum at room temperature to yield 3.43 g (64~) of the title compound. m.p. 156-158~ C.

IR: 3320 cm~l ~or OH; 1670cm~l f~or -C(=O)-.

(ii) Synthesis o~
5 N, N' -Bis(dodecylaminocarbonylmethylene)-N, N' -bis(~-N, N-dimethylaminoethylaminocarbonylmethylene)ethylenediamine (EDTA-LA-DMA) H25CI2~ NH
0~ 0 ~ N~ ,N ~ NH CIH3 H3C~ , ~_, ~ ~ o ~ CH3 CH3 ~ ~
C12H2s EDTA-LA (3.14 g, 0.005 mole) ~rom step (i), N,N-dimethylethylenediamine (0. 88 g, 0.01 mole) and CHCl3 (100 mL) were combined. A~ter dissolution of the solid materials, the solution was cooled to 5~C and a solution o~
1,3 -dicyclohexylcarbodiimide (DCC) (2.227 g, 0.011 mole) in CHCl3 (20 mL) was added dropwise. A precipitate was observed. The reaction mixture was stirred at room temperature ~or about 24 hours. The reaction mixture was ~iltered and the ~iltrate was washed with 0. 5~ acetic acid (100 mL) to decompose any excess DCC. A white milky solution was ~ormed which separated into two layers. The bottom organic layer was dried (Na2SO4) and concentrated in vacuo to yield 3.81 g of the title compound as a so~t solid.
IR: 3280 cm-1; 2900 cm-l; 1640 cm-l; 1530 cm-l.

W O 96/26179PCTrUS96/01474 (iii) Synthesis o~ EDTA-LA-TMA Tetraiodide A solution o~ EDTA-LA-DMA (3.66 g, 4.77 mmole) from step (ii), iodomethane (3.41 g, 24 mmole) and ethanol (30 mL) was re~luxed ~or 2 hours. The ethanolic solution was concentrated in vacuo and the resulting residue was lyophilized overnight. 3.98 g o~ EDTA-LA-TMA tetraiodide, a compound within the scope of the invention, was obtained as a yellow solid.
IR: 3260 cm~l; 1650 cm~1.

Example 2 Synthesis o~
N,N' '-Bis(hexadecylaminocarbonylmethylene)-N,N',N' '-tris(~-N,N,N-trimethylammonium-ethylaminocarbonylmethylene)-N,N',N''-trimethyl-diethylenetriamine hexaiodide (DTPA-HA-TME Hexaiodide) Cl H3 H3C-- I ~NH
CH3 f,~ 6I-o ICH3 ~ N ICH3 ÇH3 ~ CH3 I CH3 H3C--I ~-- ~ 0~ ~ I--CH3 CH3 HN~C ~H NH-Cl6H33 CH3 W O96/26179 PCTfUS96/01474 Synnthetic Route (i) Synthesis of N,N''-Bis(hexadecylaminocarbonylmethylene)-diethylenetriamine-N,N',N''-triacetic acid (DTPA-HA) OH
~bo ,~ ,N~ "-~N ~ O

OH ~ O O ~ OH

NH HN~

Hexadecylamine (4.82 g, 0.02 mole) in dry methanol(60 mL) was added to a suspension of diethylenetriamine pentaacetic acid dianhydride (3. 57 g, 0.01 mole) in dry methanol (30 mL). The resulting mixture was stirred at 50~C for 6 hours. The reaction mixture was cooled and the resulting white solid precipitate was collected by filtration. The white solid was dried under ~acuum to yield 5.9 g of the title compound.

W O96/26179 PCTrUS96/01471 (ii) Synthesis of N,N''-Bis(hexadecylaminocarbonylmethylene)-N,N',N''-tris(~-N,N-dimethylaminoethylamino-carbonylmethylene)diethylenetriamine (DTPA-HA-DMA) Cl H3 H3C ~/--NH
~bo q'~ N~ N~ N~O
H3C~N~\~NH l~o Oq~J HN~ IN,CH3 CH3 HN~ HN~ CH3 C~ 33 Cl6H33 A solution o~ DTPA-HA (4.2 g, 0.005 mole) ~rom step (i), N,N-dimethylethylenediamine (0.88 g, 0.01 mole) and CHCl3 (100 mL) was cooled to 0-5~ C. To this solution was added dropwise a solution o~ DCC (2.23 g, 0.011 mole) in CHCl3 (20 mL). The reaction mixture was stirred ~or 24 hours at room temperature. The resulting precipitate was removed by filtration and was washed with 0.5~ acetic acid (100 mL).
A white, milky solution was obtained which was ~iltered again, dried (Na2SO4), and concentrated in vacuo. The title compound was obtained as a soft solid (3. 5 g).

(iii) Synthesis o~ DTPA-HA-TMA Hexaiodide A solution o~ DTPA-HA-DMA (4. 7 g, 4.8 mmole) ~rom step (ii), iodomethane (3.41 g) and methanol (50 mL) was re~luxed ~or 2 hours. The methanolic solution was concentrated in vacuo and the resulting residue was lyophilized overnight. 6 g o~ DTPA-HA-TME hexaiodide, a W O96/26179 PCTrUS96/01474 compound within the scope o~ the invention, was obtained as a yellow solid.

Example 3 Synthesis of 5 N, N' -Bis (dodecylaminocarbonylmethylene) -N, N'-bis (~-N, N, N-trimethylammoniumethylaminocarbonylmethylene)-N,N'-dimethyl-cyclohexylene-1,4-diamine tetraiodide (CDTA-LA-TMA Tetraiodide) ICH3 NH ~ ~ ~ Nl-CH3 CH3~ ~ ~ CH3 H3C-<N ~ N> CH3 ~0 ~
H2sClf N N-C12H25 H H

S~nthetic Route (i) Synthesis of N,N'-Bis(dodecylaminocarbonylmethylene)-cyclohexylene-1,4-diamine-N,N'-diacetic acid (CDTA-LA) O O
~\ / \ /~
HO N ~ ~ N OH
~/ >
)co 0~
H2sClf N N-CI2H25 H H

A solution of dodecylamine (3.71 g, 0.02 mole) in dry methanol (60 ml) was added to a suspension of cyclohexane-1,4-diamine-N,N,N',N'-tetraacetic acid dianhydride (3.1 g, 0.01 mole) in dry methanol (30 mL). The resulting mixture was stirred at 50~C for 6 hours.
Filtration yielded the title compound (4.8 g) as a white solid.

5(ii) Synthesis o~
N, N' - Bi s ( dodecylaminocarbonylmethylene)-N, N' -bis(~-N,N-dimethylaminoethylaminocarbonyl methylene)cyclohexylene-1,4-diamine (CDTA-LA-DMA) O O
~\ / \ ~ </
\N ~ H ~ ~ > H ~ N\
H3C O O ~ CH3 H2sCl2- N N-Cl2H2s H H

A solution o~ CDTA-LA (3.4 g, 0.005 mole) ~rom 10step (i), N,N-dimethylethylenediamine (0.88 g, 0.01 mole) and CHCl3 (100 mL) was cooled to 0-5~C. To this solution was added dropwise a solution of DCC (2.23 g, 0.011 mole) in CHCl3 (20 mL). The reaction mixture was stirred for 24 hours at room temperature and ~iltered. The ~iltrate was washed 15with 0.5~ acetic acid (100 mL) to decompose any excess DCC
and was ~iltered again. The filtrate was dried (Na2SO4) and concentrated in vacuo to yield the title compound (4.2 g) as a so~t solid.

W O96126179 PCTrUS96101474 (iii) Synthesis of CDTA-LA-TMA Tetraiodide A solution o~ CDTA-LA-DMA (3 g) ~rom step (ii), iodomethane (3.5 g) and methanol (30 mL) was re~luxed ~or 2 hours. The methanolic mixture was concentrated in vacuo and the resulting residue was lyophilized overnight. 3.1 g o~
CDTA-LA-TMA tetraiodide, a compound within the scope o~ the invention, was obtained as a solid.

Example 4 Synthesis o~
1,1,7,7-tetra(~-N,N,N,N-tetramethylammonium-ethylaminocarbonylmethylene)-4-hexadecylaminocarbonyl-methylene-N,N',N''-trimethyl-1,4,7-triazaheptane heptaiodide (DTPA-MHA-TTMA Heptaiodide) H33Cl6~ NH
~ 7I-H3C~ H3CH3 CH3 --\ fH3 N--CH3 ~ --CH3 W O96/26179 PCTrUS96101471 Sy~lthetic Route (i) Synthesis o~
4- hexadecylaminocarbonylmethylene-1, 4,7-triazaheptane-1,1, 7,7 -tetraacetic acid (DTPA-MHA) H33C16~NH
~0 ,~ ,N~ "-~N ~ O

OH ~ O ~ OH

OH OH

A solution of diethylenetriaminepentaacetic acid (3.93 g, 0.01 mole), hexadecylamine (2. 4 g, 0.01 mole) and CHCl3 (200 mL) was cooled to 0-5~C. To this solution was added dropwise a solution o~ DCC (2.23 g, 0.011 mole) in CHCl3 (15 mL). The reaction mixture was stirred for 24 hours at room temperature and ~iltered. The ~iltrate was washed with 0.5~ acetic acid (100 mL) and ~iltered again. The ~iltrate was dried (Na2SO4) and concentrated in vacuo.
Recrystallization o~ the resulting residue ~rom water yielded the title compound as a white solid (3. 7 g).

W O96/26179PCTrUS96/0147 (ii) Synthesis of 1,1,7,7-tetra(~-N,N-dimethylamino-ethylaminocarbonylmethylene)-4-hexadecyl-aminocarbonylmethylene)-1,4, 7- triazaheptane (DTPA-MHA-TDMA) H33Cl6~NH
f~o ~ N,~ ,N~ "-~N ~ O
H3C~N~ NH ~~ ~~ HN~ N,CH3 CH3 HN ~ NH CH3 -~3 ~ N' 3 A solution o~ DTPA-MHA (3 g) ~rom step (i), N,N-dimethylethylenediamine (1.76 g) and CHCl3 (200 mL) was cooled to 0-5~ C. To this solution was added dropwise a solution o~ DCC (4.5 g, 0.02 mole) in CHCl3 (20 mL). The reaction mixture was stirred overnight at room temperature.
The resulting precipitate was collected by ~iltration and the ~iltrate was washed with 0.5~ acetic acid (100 mL) and ~iltered again. The ~iltrate was dried (Na2SO4) and concentrated in vacuo. The resulting residue was puri~ied on a silica gel column. The title compound was obtained as a soft solid (2.8 g).

(iii) Synthesis o~ DTPA-MHA-TTMA Heptaiodide A solution o~ DTPA-MHA-TDMA (2.24 g), iodomethane (3.5 g, 0.03 mole) and methanol (30 mL) was re~luxed ~or 2 hours. The methanolic solution was concentrated in vacuo and the resulting residue was lyophilized overnight to yield W O 96/26179 PCTrUS96/0147 2.4 g of DTPA-MHA-TTMA heptaiodide, a compound within the scope of the present invention.

Example 5 Using procedures similar to those in Examples 1 to 4, the following compounds within the scope of the invention were prepared.

Exam~le 5A

N, N' -Bis(dodecyloxycarbonylmethylene) -N, N' -bis(~- N, N, N- trimethylammoniumethylaminocarbonyl-methylene)ethylenediamine diiodide H25Cl2~
( ~

CH3 ~ ~NI CH3 W O96/26179 PCTrUS96/01474 Exaunple 5B
N,N,N'',N''-Tetra(~-N,N,N-trimethylammonium-ethylaminocarbonylmethylene)-N'-(1,2 -dioleoylglycero-3-phosphoethanolaminocarbonylmethylene)diethylenetriamine tetraiodide O
0--~0--IOH - OCO(CH2)7CH= CH(CH2)7CH3 O~ ~--OCO(CH2)7CH=CH(CH2)7CH3 '~ ~ N ~ / ~ N ~ NIH3CH3 CH3 ~ ICH3~ 1 3 CH3 Ex ~ ple 5C
N,N' -Bis(hexadecylaminocarbonylmethylene-N,N'-bis (trimethylammoniumethylaminocarbonyl methylene)ethylenediamine diiodide H33C16~ ~
0~ 0 CIH3 ~ ~ O CH3 E~ 2I-C1~33 W O96/26179 PCTrUS96/01471 Example 5D
N, N' -bis(hexadecyloxycarbonylmethylene) -N- (~-N,N,N-trimethylammoniumethylaminocarbonylmethylene)-N-methyl -N' -(carboxymethylene)ethylenediamine diiodide r OH

Oq~f~ ~NJ~o,Cl6H33 CH3 ~~ 2I-C1~33 Ex ~ ple 5E

N, N' - bis(hexadecylaminocarbonylmethylene)- N, N ' -bis(~-N, N, N-trimethylammoniumethylaminocarbonyl methylene)-N,N'-dimethylethylenediamine tetraiodide H33C16~ ~

o~ ~r~ - ~ NH C~3 Formulations o~ this invention are subjected to various biological tests, the results of which correlate to use~ul therapeutic activity. These tests are use~ul in determ;n;ng the ability o~ the present ~ormulations to deliver intracellularly bioactive agents, including genetic material. These tests are use~ul also in determ;n;ng the W O96/26179 PCTrUS96/01474 ability o~ the present ~ormulations to treat genetic diseases, including diseases which involve a pathology or condition which is associated with loss or deterioration o~
immune competence.

Exam~le 6 The following examples are directed to the intracellular delivery of genetic material with cationic lipid compounds of the present invention and compounds disclosed in the prior art. The genetic material involved in these trans~ection studies is DNA that codes ~or Chloramphenicol Acetyl Transferase ("CAT"). The amount of expressed CAT (nanograms per mL (ng/mL)) was assayed using the Boehringer Mannheim CAT ELISATM kit, the results of which are tabulated in FIGS. 1 and 2.
LIPOFECTAMINETM and LIPOFECTIN~ were purchased ~rom Gibco BRL, a division o~ Life Technologies, Inc.
(Gaithersburg, MD). LIPOFECTAMINETM is a 3:1 liposome formulation of N-[2-({2,5-bis(3-aminopropyl)amino]-1-oxypentyl}amino)ethyl-N,N-dimethyl-2,3-bis(9-octadecenyloxy)-1-prop~n~m;n'um trifluoroacetate and dioleoylphosphatidylethanolamine ("DOPE"). LIPOFECTIN~ is a liposome formulation of N- [1- (2,3-dioleoyloxy)propyl] -N,N,N-trimethylammonium chloride ("DOTMA") and DOPE. (See Proc.
Natl. Acad. Sci. USA, Vol. 84, p. 7413 (1987).) TRANSFECTAMTM was purchased ~rom Promega Corp (Madison, WI).
TRANSFECTAMTM is a cationic lipopolyamine compound which comprises a spermine headgroup. (See Proc . Na tl . Acad .

CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 Sci., Vol. 86, p. 6982 (1989); ~. Neurochem., Vol. 54, p.
1812 (1990); and DNA and Cell Biology, Vol. 12, p. 553 (1993).) "DOTAP" re~ers to 1,2-dioleoyl-3-propyl-N,N,N-trimethylammonium halide.

Example 6A
This example describes the biological testing of the compounds prepared in Examples 5C and 5D, Lipofectin, Lipofectamine and DOTAP and Transfectam in the absence of serum.
HeLa cells (American Type Culture Collection, Rockville, MD) were cultured in EMEM media (Mediatech, Washington, DC). The cells were grown in 6-well plates (Becton Dickinson, Lincoln Park, NJ) and at a density of 4X10s cells/well until they were 60-80~ confluent in a VWR
15 model 2500 CO2 incubator (VWR, Philadelphia, PA). DNA (1.7 ~g) was diluted to 50 ~L in HEPES bu~fered saline (HBS) (HEPES 20 mM, 150 mM NaCl, pH 7.4) for each well to be in~ected. 10 ~L solutions of the cationic lipid compounds prepared in Examples 5C and 5D, Lipofectin, Lipofectamine, DOTAP and Transfectam were each diluted to 50 ~L in HBS.
The DNA and lipid solutions were mixed by inverting and incubated at room temperature ~or 15 minutes. After incubating, each of the mixtures o~ DNA and cationic lipid (100 ~L) was added to 1.9 mL of media without serum and mixed by inverting. The media was removed ~rom the 6-well plates and replaced with the media containing lipid and DNA.
The cells were then incubated in a CO2 atmosphere at 37~C ~or W O96/26179 PCTrUS96/01471 5-6 hours After incubating, the lipid/DNA media was removed and replaced with complete media. The cells were then incubated ~or 48-72 hours and the level o~ expressed protein was assayed. The results o~ the assay were measured using an SLT Labinstruments SPECTRA Shell plate reader (ShT, Salzburg, Austria) which was linked to a Centris 650 computer (Apple Computer, Inc., Cupertino, CA) and controlled using DeltaSo~t II version 4.13s (Biometallics, Inc., Princeton, NJ). The results o~ the assay are depicted in FIG. l which show increased expression of CAT when the cationic lipid compounds o~ the present invention are used to trans~ect cells, relative to compounds o~ the prior art.
Accordingly, the experiments per~ormed in this example demonstrate that the cationic lipid compounds o~ the present invention provide use~ul and improved trans~ection o~ cells with bioactive agents as compared to compounds o~ the prior art.

Example 6B
This example describes the biological testing o~
the compound prepared in Example 5D, Lipo~ectin and Lipo~ectamine in the presence o~ serum.
HeLa cells were cultured in 4 mL o~ culture media as described above, except that the media was supplemented with enriched cal~ serum (Gibco BRL Life Technologies, Gaithersburg, MD) and Penicillin/Streptomycin (Boehringer Mannheim Biochemicals (BMB), Indianapolis, IN). The cells were grown in 6-well plates (Becton Dickinson, Lincoln Park, CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96/01474 NJ) and at a density of 4Xl05 cells/well until they were 60-80~ confluent in a VWR model 2500 CO2 incubator (VWR, Philadelphia, PA). DNA (3.3 ~Lg) was diluted to 100 ~L in HEPES buffered saline (HBS) (HEPES 20 mM, 150 mM NaCl, pH
7.4) for each well to be infected. 20 ~LL solutions were prepared of (1) the cationic lipid compound of Example 5D in combination with varying amounts of DOPE; (2) Lipofectin;
and (3) Lipofectamine. These were diluted to 100 ,LL in HBS
for each well. The compound of Example 5D and DOPE were combined in weight ratios of 5:1, 6:1, 7:1, 9:1, 11:1 and 14:1. The DNA and lipid solutions were mixed by inverting and incubated at room temperature for 15 minutes. After incubating, the mixtures of DNA and cationic lipid (200 ~L) were added to each well (except for a HeLa (CELLS) standard to which no DNA with cationic lipid was added) and the mixtures were agitated by pipetting several times upwards and downwards. The cells were then incubated in a CO2 atmosphere at 37~C for 48-72 hours. After incubating, the protein level was assayed as described above.
The results of the assay are depicted in FIG. 2 which show increased expression of CAT when the cationic lipid compounds of the present invention are used to transfect cells, relative to compounds of the prior art.
Particularly desirable transfection is observed in compositions which comprise a 6:1 ratio of the compound of Example 5D to DOPE. The experiments performed in this example demonstrate that the cationic lipid compounds of the present invention provide improved transfection of cells W O96/26179 PCTrUS96/01474 with bioactive agents as compared to compounds of the prior art.

Example 7 In vivo experiments in rats were performed which demonstrate the high effectiveness of the present cationic lipid compounds to deliver intracellularly genetic material.
The experiments demonstrate also the effectiveness of using ultrasound energy for targeting specific tissue in vivo with vesicular compositions containing genetic material.
Plasmid pSV ~-gal (Promega, Madison, WI) which contains the ~-galactosidase gene was combined with the cationic lipid compound prepared in Example 5D by mixing.
The resulting mixture was injected into each of three Sprague Dawley rats (rats (A), (B) and (C)) via the tail vein. Rat (A) was not subjected to ultrasound. Ultrasonic energy was applied to the inside of the hind leg during injection for each o~ rats (B) and (C). After 48 hours, the rats were euthanized and the tissues were removed. The tissues were fixed for 72 hours in 2~ formalin, sliced thin and placed in an X-gal solution. After 16 hours at 37~C, the tissues were inspected. The tissue ~rom rat (A) exhibited a blue color which is indicative of general transfection. The tissue from rats (B) and (C) exhibited blue color only at the site where ultrasound energy was applied. This indicates that localization of gene expression can be achieved with the compounds and methods of the present invention.

W O96/26179 PCTrUS96/01474 Exaunp~e 8 A cationic lipid composition according to the present invention was prepared ~rom six parts o~ the compound prepared in Example 5D and 1 part dipalmitoyl-phosphatidylethanolamine (DPPE) labeled with rhodamine (Avanti Polarlipids, Alabaster, AL). The cationic lipid composition was dissolved in ethanol and a Mansfield angioplasty catheter tip (Boston Scienti~ic Corp., Watertown, MA) was dipped into the ethanolic formulation, removed and allowed to dry. This procedure was repeated three times. The coated catheter tips were then placed onto a Nikon light microscope equipped with a ~ilter for rhodamine ~luorescence. A control catheter, which was not coated with the cationic lipid composition, was also placed onto the light microscope. Fluorescence o~ the coated catheter tips was observed, whereas the control catheter tips did not ~luoresce. This con~irmed the presence o~ a coating o~ the lipid composition on each o~ the coated catheters. The coated catheter tips were then dipped into 2~ normal saline, water and human serum ~or varying periods o~
time and viewed under the light microscope. Fluorescence o~
the catheter tips was observed again. This demonstrated that the coating of the rhodamine-labeled lipid composition adhered to the sur~ace o~ the catheters. Accordingly, lipid compositions o~ the present invention can be delivered to speci~ic locations within the body by coating the compositions onto catheters which are then administered to a patient, as appropriate.

W O96/26179PCTrUS96101474 Ex ~ ple 9 A cationic lipid formulation according to the present invention was prepared from DNA (5 ~g) fluorescently labelled with fluorescein-12 DUTP (deoxyuracil triphosphate, S commercially available from Boehringer Mannheim Biochemicals (BMB), Indianapolis, IN) using PCR, six parts of the compound prepared in Example 5D and 1 part DPPE. Catheters were subse~uently dipped into ethanolic solutions of the cationic lipid formulation as described (for the compositions) in Example 8. A control catheter was not coated with the subject cationic lipid formulation.
Fluorescence was induced and observed for~the coated catheters as described in Example 8. No fluorescence was observed with the control catheter. This demonstrated that lS the cationic lipid formulations of the present invention adhere to the surface of catheters. Accordingly, lipid formulations of the present invention can be delivered to specific locations within the body by coating the formulations onto catheters which are then administered to a patient, as appropriate.

Ex ~ ple 10 A cationic lipid formulation according to the present invention will be prepared from 6 parts of the compound of Example 5D, 1 part DPPE (dipalmitoyl-phosphatidylethanolamine) and 10 ~g of plasmid DNAcontaining the gene for endothelial cell growth factor and a Respiratory Syncytial Virus RSV) growth factor. The CA 022l34l7 l997-08-20 W O96/26179 PCTrUS96101474 formulation will be lyophilized, and 1 to 10 ~g of the lyophilized formulation will be coated on a balloon o~ an angioplasty catheter. Coating will be accomplished by r simply dipping the balloon into the formulation. The 5 angioplasty catheter will be introduced into the left anterior descending coronary artery of a patient to cross the region of a hemodynamically significant stenosis. The catheter will be inflated to 6 atmospheres of pressure with the coated balloon. The stenosis will be alleviated and the 10 lyophilized coating on the balloon will be deposited on the arterial wall. Transfection of endothelial cells results in localized production of endothelial cell growth factor.
Healing of the arterial wall will be improved and fibroblast proliferation will be reduced, resulting also in lessened 15 restenosis.

Example 11 The procedure described in Example 10 will be repeated except that the surface of the catheter balloon will be modified to improve the binding of the lyophilized 20 cationic lipid ~ormulation according to the procedure described in K. Ishihara et al., Journal of Biomedical Materials Research, Vol. 27, pp. 1309-1314 (1993).

Example 12 The procedure described in Example 10 will be 25 repeated except that a vascular stent comprising, ~or example, Dacron~ and/or wire mesh, is substituted ~or the W O96/26179 PCTrUS96/01474 angioplasty catheter Improved endothelialization is obtained along the sur~ace o~ the stent.

Claims (155)

1. A cationic lipid compound of the formula:

(I) wherein:
each of x, y and z is independently an integer from 0 to about 100;
each X1 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-;
each X2 is independently O or S;
each Y1 is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer from 1 to 3;
each Y2 is independently -N(R6)b- , -S(R6)b- or -P(R6)b- , wherein b is an integer from 0 to 2;
each Y3 is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -CO2R6, wherein a is an integer from 1 to 3;
each of R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 10 carbons; and each R6 is independently -[R7-X3]c-R8 or -R9-[X4-R10]d-Q, wherein:
each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, -N(R11)q, -S(R11)q, -P(R11)q or -CO2R11, wherein q is an integer from 1 to 3;
each of X3 and X4 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-;
each R7 is independently alkylene of 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 60 carbons;
each of R9 and R10 is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R7-X3]c-R8 or -R9-[X4-R10]d-W, wherein:
each W is independently a phosphate residue, -N(R12)w, -S(R12)w, -P(R12)w or -CO2R12, wherein w is an integer from 1 to 3; and R12 is -[R7-X3]c-R8; with the proviso that the compound of formula (I) comprises at least two quaternary salts.
2. A compound according to claim 1 wherein said quaternary salt comprises a pharmaceutically-acceptable counter ion.
3. A compound according to claim 2 wherein said counter ion is selected from the group consisting of halide, Rl3SO3-, Rl3CO2-, phosphate, sulfite, nitrate, gluconate, guluronate, galacturonate, estolate and mesylate, wherein Rl3 is hydrogen, alkyl of 1 to about 20 carbons or aryl of about 6 to about 10 carbons.
4. A compound according to claim 1 wherein:
each of c, d, x, y and z is independently an integer from O to about 50;
each Q is independently a phosphate residue, -CO2Rll or -N(Rll)q, wherein q is 2 or 3; and each W is independently a phosphate residue, -C02Rl2 or -N(Rl2)W~ wherein w is 2 or 3.
5. A compound according to claim 4 wherein:
each of q and w is 3.
6. A compound according to claim 5 wherein:
each of c, d, x, y and z is independently an integer from O to about 20; and X2 is 0.
7. A compound according to claim 6 wherein:
each of c, d, x, y and z is independently an integer from O to about 10; and each of Xl, X3 and X4 iS independently -C(=O)-NR5-, -NR5-C(=O)-, -C(=O)-O- or -O-C(=O)-.
8. A compound according to claim 7 wherein:
each of c, d, x, y and z is an integer from 0 to about 5.
9. A compound according to claim 8 wherein:
each of R1, R2, R3 and R4 is independently straight chain alkylene of 1 to about 10 carbons or cycloalkylene of about 4 to about 10 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 4 carbons;
each R7 is independently alkylene of 1 to about 10 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 40 carbons; and each of R9 and R10 is independently alkylene of 1 to about 10 carbons.
10. A compound according to claim 9 wherein:
each of R1, R2, R3 and R4 is independently straight chain alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons;
R5 is hydrogen;
each R7 is independently alkylene of 1 to about 4 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 20 carbons; and each of R9 and R10 is independently alkylene of 1 to about 4 carbons.
11. A compound according to claim 10 wherein:
each Y1 is independently a phosphate residue, N(R6)a- or -CO2R6;
Y2 is -N (R6)b-; and each Y3 is independently a phosphate residue, N (R6)a- or -CO2R6.
12. A compound according to claim 11 wherein:
x is 1.
13. A compound according to claim 12 wherein:
y is 2 and z is 0.
14. A compound according to claim 13 wherein:
each Y1 is independently N(R6)a- or -CO2R6; and R6 is -[R7-X3]c-R8.
15. A compound according to claim 14 wherein:
each of R1, R2, R3, R4 and R7 is independently methylene, ethylene or cyclohexylene; and each R8 is independently hydrogen or alkyl of about 1 to about 16 carbons.
16. A compound according to claim 15 wherein:

a is 3; and each c is independently 0 or 1.
17. A compound according to claim 16 wherein:
b is 1.
18. A compound according to claim 17 which is N,N'-bis(dodecyloxycarbonylmethylene)-N,N'-bis (.beta.-N,N,N-trimethylammoniumethylaminocarbonylmethylene)ethylenediamine dihalide.
19. A compound according to claim 18 wherein said halide is chloride, bromide or iodide.
20. A compound according to claim 17 which is N,N'-bis(hexadecylaminocarbonylmethylene-N,N'-bis(trimethyl-ammoniumethylaminocarbonylmethylene)ethylenediamine dihalide.
21. A compound according to claim 20 wherein said halide is chloride, bromide or iodide.
22. A compound according to claim 16 wherein:
b is 2.
23. A compound according to claim 22 which is N,N'-bis(dodecylaminocarbonylmethylene)-N,N'-bis(.beta.-N,N,N-trimethylammoniumethylaminocarbonylmethylene)-N,N'-dimethylethylenediamine tetrahalide.
24. A compound according to claim 23 wherein said halide is chloride, bromide or iodide.
25. A compound according to claim 22 which is N, N' -bis(dodecylaminocarbonylmethylene) -N, N' -bis (.beta.-N, N, N-trimethylammoniumethylaminocarbonylmethylene)-N, N' - dimethyl-cyclohexylene-1,4-diamine tetrahalide.
26. A compound according to claim 25 wherein said halide is chloride, bromide or iodide.
27. A compound according to claim 22 which is N, N' -bis(hexadecylaminocarbonylmethylene)-N,N'-bis (.beta.-N, N, N-trimethylammoniumethylaminocarbonylmethylene)-N, N' -dimethylethylenediamine tetrahalide.
28. A compound according to claim 27 wherein said halide is chloride, bromide or iodide.
29. A compound according to claim 16 wherein:
each b is independently 1 or 2.
30. A compound according to claim 29 which is N,N'-bis(hexadecyloxycarbonylmethylene) -N- (.beta.-N, N, N-trimethylammoniumethylaminocarbonylmethylene)-N-methyl-N'-(carboxymethylene)ethylenediamine dihalide.
31. A compound according to claim 30 wherein said halide is chloride, bromide or iodide.
32. A compound according to claim 12 wherein:
y is 2 and z is 0 or 1.
33. A compound according to claim 32 wherein:
a is 3;
b is 2;
each c is independently 0 or 1; and d is 1.
34. A compound according to claim 33 wherein:
R11 is -[R7-X3]c-R8
35. A compound according to claim 34 wherein:
each of R1, R2, R3, R4 and R7 is independently methylene or ethylene; and each R8 is independently hydrogen or alkyl of 1 to about 16 carbons.
36. A compound according to claim 35 which is N,N''- bis(hexadecylaminocarbonylmethylene)- N,N',N''- tris(.beta.-N, N, N- trimethylammoniumethylaminocarbonylmethylene)-N, N', N'' - trimethyldiethylenetriamine hexahalide.
37. A compound according to claim 36 wherein said halide is chloride, bromide or iodide.
38. A compound according to claim 12 wherein:
y is 3 and z is 0.
39. A compound according to claim 38 wherein:
a is 3;
b is 2;
each c is independently 0 or 1; and d is 1.
40. A compound according to claim 39 wherein:
each of R1, R2, R3, R4 and R7 is independently methylene or ethylene;
each R8 is independently hydrogen or alkyl of 1 to about 16 carbons;
each of R9 and R10 is independently methylene or ethylene; and R11 is methyl.
41. A compound according to claim 40 which is 1,1,7,7-tetra(.beta.-N,N,N,N-tetramethylammoniumethylamino-carbonylmethylene)-4-hexadecylaminocarbonylmethylene-N, N', N''-trimethyl-1,4,7-triazaheptane heptahalide.
42. A compound according to claim 41 wherein said halide is chloride, bromide or iodide.
43. A compound according to claim 12 wherein:
y is 3 and z is 0.
44. A compound according to claim 43 wherein:
a is 3;
b is 1;
c is 0; and d is 1.
45. A compound according to claim 44 wherein:
each of R1, R2 and R3 is independently methylene or ethylene;
each R8 is independently hydrogen or methyl;
each of R9 and R10 is independently methylene or ethylene; and R11 is methyl.
46. A compound according to claim 45 which is N, N, N'', N''-tetra(.beta.-N, N, N- trimethylammoniumethylamino-carbonyl-methylene)-N' - (1,2-dioleoylglycero-3-phosphoethanolaminocarbonylmethylene)diethylenetriamine tetrahalide.
47. A compound according to claim 46 wherein said halide is chloride, bromide or iodide.
48. A cationic lipid compound of the formula (II) wherein:

each Y1 is independently a phosphate residue, N(R2)a-, S(R2)a-, P(R2)a- or -CO2R2, wherein a is an integer from 1 to 3;
R1 is alkylene of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
R2 is a residue of the formula -R4- [(X1-R5)x-Y2]y-R6, wherein:
each of x and y is independently an integer from 0 to about 100;
each X1 is independently a direct bond, -O-, -S-, -NR3-, -C (=X2) -, -C (=X2) -N (R3) -, -N (R3) -C (=X2) -, -C (=X2) -O-, -O-C (=X2) - or -X2- (R3X2) P (=X2) -X2-;
each X2 is independently O or S;
each Y2 is independently -O-, -S(R2) b-, -N (R2) b- or -P (R2) b-, wherein b is an integer from 0 to 2;
each R3 is independently hydrogen or alkyl of 1 to about 10 carbons;
each of R4 and R5 is independently a direct bond or alkylene of 1 to about 30 carbons containing 0 to about 15 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups; and each R6 is independently hydrogen or alkyl of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- , -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
with the proviso that the compound of formula (II) comprises at least two quaternary salts.
49. A compound according to claim 48 wherein said quaternary salt comprises a pharmaceutically-acceptable counter ion.
50. A compound according to claim 49 wherein said counter ion is selected from the group consisting of halide, R7SO3-, R7CO2-, phosphate, sulfite, nitrate, gluconate, guluronate, galacturonate, estolate and mesylate, wherein R7 is hydrogen, alkyl of 1 to about 20 carbons or aryl of 6 to about 10 carbons.
51. A compound according to claim 48 wherein:
each of x and y is independently an integer from 0 to about 50.
52. A compound according to claim 51 wherein:
each Y1 is independently a phosphate residue, N(R2)a- or -CO2R2.
53. A compound according to claim 52 wherein:
each Y1 is independently N(R2)a- or -CO2R2.
54. A compound according to claim 53 wherein:
each of x and y is independently an integer from 0 to about 20; and X2 is O.
55. A compound according to claim 54 wherein:
each of x and y is independently an integer from 0 to about 10.
56. A compound according to claim 55 wherein:
each X1 is independently -C(=X2)-N(R3)-, -N(R3)-C(=X2)-, -C(=X2)-O- or -O-C(=X2)-.
57. A compound according to claim 56 wherein:
R1 is alkylene of 1 to about 40 carbons;
each R3 is independently hydrogen or alkyl of 1 to about 4 carbons;
each of R4 and R5 is independently a direct bond or alkylene of 1 to about 20 carbons; and each R6 is independently hydrogen or alkyl of 1 to about 40 carbons.
58. A compound according to claim 57 wherein:
R1 is alkylene of 1 to about 20 carbons;
R3 is hydrogen; and each R6 is independently hydrogen or alkyl of 1 to about 20 carbons.
59. A compound according to claim 58 wherein:
R1 is independently straight chain alkylene of 1 to about 10 carbons or cycloalkylene of about 4 to about 10 carbons; and each of R4 and R5 is independently a direct bond, straight chain alkylene of 1 to about 10 carbons or cycloalkylene of 4 to about 10 carbons.
60. A compound according to claim 59 wherein:
R1 is independently straight chain alkylene of 1 to about 4 carbons or cylcoalkylene of about 5 to about 7 carbons; and each of R4 and R5 is independently a direct bond, straight chain alkylene of 1 to about 4 carbons or cylcoalkylene of about 5 to about 7 carbons.
61. A cationic lipid compound of the formula (III) wherein:
each of x, y and z is independently an integer from 0 to about 100;
each X1 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)p(=x2)-x2-;
each X2 is independently O or S;

each Y1 is independently -O-, -N(R6)a-, -S(R6)a- or -P(R6)a-, wherein a is an integer from 0 to 2;
each Y2 is independently -N(R6)a-, -S(R6)a- or -P(R6)a-, wherein a is an integer from 0 to 2;
each Y3 is independently a phosphate residue, N(R6)b-, S(R6)b-, P(R6)b- or -CO2R6, wherein b is an integer from 1 to 3;
each of R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 10 carbons; and each R6 is independently -[R7-X3]c-R8 or -R9-[X4-R10]d-Q , wherein:
each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, -N(R11)q, -S(R11)q, -P(R11)q or -CO2R11, wherein q is an integer from 1 to 3;
each of X3 and X4 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-;
each R7 is independently alkylene of 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 60 carbons;
each of R9 and R10 is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R7-X3]c-R8 or -R9-[X4-R10] d-W, wherein:
each W is independently a phosphate residue, -N(R12)w, -S(R12)w, -P(R12)w or -CO2R12, wherein w is an integer from 1 to 3; and R12 is -[R7-X3]c-R8; with the proviso that the compound of formula (III) comprises at least two quaternary salts.
62. A compound according to claim 61 wherein said quaternary salt comprises a pharmaceutically-acceptable counter ion.
63. A compound according to claim 62 wherein said counter ion is selected from the group consisting of halide, R13SO3-, R13CO2-, phosphate, sulfite, nitrate, gluconate, guluronate, galacturonate, estolate and mesylate, wherein R13 is hydrogen, alkyl of 1 to about 20 carbons or aryl of about 6 to about 10 carbons.
64. A compound according to claim 61 wherein:
each of c, d, x, y and z is independently an integer from 0 to about 50; and each Q is independently a phosphate residue, -N(R11)q or -CO2R11, wherein q is 2 or 3; and each W is independently a phosphate residue, -N(R12)w or -CO2R12, wherein w is 2 or 3.
65. A compound according to claim 64 wherein:
each of q and w is 3.
66. A compound according to claim 65 wherein:
each of c, d, x, y and z is independently an integer from 0 to about 20; and X2 is O.
67. A compound according to claim 66 wherein:
each of c, d, x, y and z is independently an integer from 0 to about 10; and each of X1, X3 and X4 is independently -C(=O)-NR5-, -NR5-C(=O)-, -C(=O)-O- or -O-C(=O)-.
68. A compound according to claim 67 wherein:
each of c, d, x, y and z is independently an integer from 0 to about 5.
69. A compound according to claim 68 wherein:
each of R1, R2, R3 and R4 is independently straight chain alkylene of 1 to about 10 carbons or cycloalkylene of about 4 to about 10 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 4 carbons;
each R7 is independently alkylene of 1 to about 10 carbons;
each R8 is independently alkyl of 1 to about 40 carbons; and each of R9 and R10 is independently alkylene of 1 to about 10 carbons.
70. A compound according to claim 69 wherein:
each of R1, R2, R3 and R4 is independently alkylene of 1 to about 4 carbons or cycloalkylene of about 5 to about 7 carbons;
R5 is hydrogen;
R7 is alkylene of 1 to about 4 carbons;
each R8 is independently alkyl of 1 to about 20 carbons; and each of R9 and R10 is independently alkylene of 1 to about 4 carbons.
71. A compound according to claim 70 wherein:
Y1 is -N(R6)a-;
Y2 is -N (R6) a- ; and each Y3 is independently a phosphate residue, N(R6) b- or -CO2R6.-
72. A cationic lipid compound which comprises at least two cationic groups.
73. A cationic lipid composition comprising a cationic lipid compound according to claim 1.
74. A cationic lipid composition according to claim 73 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
75. A cationic lipid composition according to claim 74 further comprising an amphipathic compound for stabilizing the composition.
76. A cationic lipid composition according to claim 75 further comprising a gas, a precursor to a gas or a mixture thereof.
77. A cationic lipid composition according to claim 76 wherein said gas is selected from the group consisting of perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluorocyclobutane, and mixtures thereof.
78. A cationic lipid composition according to claim 76 which comprises a mixture of a gas and a precursor to a gas.
79. A cationic lipid composition comprising a cationic lipid compound according to claim 48.
80. A cationic lipid composition according to claim 79 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
81. A cationic lipid composition according to claim 80 further comprising an amphipathic compound for stabilizing the composition.
82. A cationic lipid composition according to claim 81 further comprising a gas, a precursor to a gas or a mixture thereof.
83. A cationic lipid composition according to claim 82 wherein said gas is selected from the group consisting of perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluorocyclobutane, and mixtures thereof.
84. A cationic lipid composition according to claim 82 which comprises a mixture of a gas and a precursor to a gas.
85. A cationic lipid composition comprising a cationic lipid compound according to claim 61.
86. A cationic lipid composition according to claim 85 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
87. A cationic lipid composition according to claim 86 further comprising an amphipathic compound for stabilizing the composition.
88.. A cationic lipid composition according to claim 87 further comprising a gas, a precursor to a gas or a mixture thereof.
89. A cationic lipid composition according to claim 88 wherein said gas is selected from the group consisting of perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluorocyclobutane, and mixtures thereof.
90. A cationic lipid composition according to claim 88 which comprises a mixture of a gas and a precursor to a gas.
91. A cationic lipid formulation for the intracellular delivery of a bioactive agent which comprises, in combination with a bioactive agent, a cationic lipid compound that comprises at least two cationic groups.
92. A lipid formulation according to claim 91 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
93. A lipid formulation according to claim 92 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
94. A cationic lipid formulation according to claim 91 wherein said bioactive agent comprises a genetic material.
95. A cationic lipid formulation for the intracellular delivery of a bioactive agent comprising, in combination with a bioactive agent, a cationic lipid composition which comprises a cationic lipid compound according to claim 1.
96. A lipid formulation according to claim 95 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
97. A lipid formulation according to claim 96 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
98. A lipid formulation according to claim 95 wherein said bioactive agent comprises genetic material.
99. A cationic lipid formulation for the intracellular delivery of a bioactive agent comprising, in combination with a bioactive agent, a cationic lipid composition which comprises a cationic lipid compound according to claim 48.
100. A lipid formulation according to claim 99 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
101. A lipid formulation according to claim 100 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
102. A lipid formulation according to claim 99 wherein said bioactive agent comprises genetic material.
103. A cationic lipid formulation for the intracellular delivery of a bioactive agent comprising, in combination with a bioactive agent, a cationic lipid composition which comprises a cationic lipid compound according to claim 61.
104. A lipid formulation according to claim 103 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
105. A lipid formulation according to claim 104 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
106. A lipid formulation according to claim 103 wherein said bioactive agent comprises genetic material.
107. A cationic lipid formulation for the intracellular delivery of a bioactive agent comprising, in combination with a bioactive agent, a cationic lipid composition according to claim 72.
108. A lipid formulation according to claim 107 which is selected from the group consisting of micelles, liposomes and mixtures thereof.
109. A lipid formulation according to claim 108 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
110. A lipid formulation according to claim 107 wherein said bioactive agent comprises genetic material.
111. A process for the preparation of a cationic lipid formulation for the intracellular delivery of a bioactive agent comprising combining together a bioactive agent and a composition which comprises a cationic lipid compound having at least two cationic groups.
112. A process according to claim 111 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
113. A process according to claim 112 comprising substantially entrapping said bioactive agent within said micelles or liposomes.
114. A process according to claim 111 wherein said bioactive agent comprises genetic material.
115. A process for the preparation of a cationic lipid formulation for the intracellular delivery of a bioactive agent comprising combining together a bioactive agent and a cationic lipid composition which comprises a cationic lipid compound according to claim 1.
116. A process according to claim 115 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
117. A process according to claim 116 comprising substantially entrapping said bioactive within said micelles or liposomes.
118. A process according to claim 115 wherein said bioactive agent comprises genetic material.
119. A process for the preparation of a cationic lipid formulation for the intracellular delivery of a bioactive agent comprising combining together a bioactive agent and a cationic lipid composition which comprises a cationic lipid compound according to claim 48.
120. A process according to claim 119 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
121. A process according to claim 120 comprising substantially entrapping said bioactive within said micelles or liposomes.
122. A process according to claim 119 wherein said bioactive agent comprises genetic material.
123. A process for the preparation of a cationic lipid formulation for the intracellular delivery of a bioactive agent comprising combining together a bioactive agent and a cationic lipid composition which comprises a cationic lipid compound according to claim 61.
124. A process according to claim 123 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
125. A process according to claim 124 comprising substantially entrapping said bioactive within said micelles or liposomes.
126. A process according to claim 123 wherein said bioactive agent comprises genetic material.
127. A method for delivering intracellularly a bioactive agent comprising contacting a cell with a cationic lipid composition which comprises a cationic lipid compound comprising at least two cationic groups and a bioactive agent.
128. A method of claim 127 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
129. A method of claim 128 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
130. A method of claim 127 wherein said bioactive agent comprises genetic material.
131. A method of claim 130 wherein said genetic material is selected from the group consisting of polynucleotide, DNA, RNA, polypeptide and mixtures thereof.
132. A method for delivering intracellularly a bioactive agent comprising contacting a cell with a cationic lipid composition which comprises a cationic lipid compound according to claim 1 and a bioactive agent.
133. A method of claim 132 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
134. A method of claim 133 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
135. A method of claim 132 wherein said bioactive agent comprises genetic material.
136. A method for delivering intracellularly a bioactive agent comprising contacting a cell with a cationic lipid composition which comprises a cationic lipid compound according to claim 48 and a bioactive agent.
137. A method of claim 136 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
138. A method of claim 137 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
139. A method of claim 136 wherein said bioactive agent comprises genetic material.
140. A method for delivering intracellularly a bioactive agent comprising contacting a cell with a cationic lipid composition which comprises a cationic lipid compound according to claim 61 and a bioactive agent.
141. A method of claim 140 wherein said composition is selected from the group consisting of micelles, liposomes and mixtures thereof.
142. A method of claim 141 wherein said bioactive agent is substantially entrapped within said micelles or liposomes.
143. A method of claim 140 wherein said bioactive agent comprises genetic material.
144. A method for the treatment of a genetic disease in a patient suffering from such disorder comprising administering to the patient a therapeutically effective amount of a formulation according to claim 91.
145. A method according to claim 144 wherein the disorder involves a pathology or condition which is associated with loss or deterioration of immune competence.
146. A method according to claim 144 wherein said administration comprises administering to the patient a catheter that is coated with said formulation.
147. A method for the treatment of a genetic disease in a patient suffering from such disorder comprising administering to the patient a therapeutically effective amount of a formulation according to claim 95.
148. A method according to claim 147 wherein the disorder involves a pathology or condition which is associated with loss or deterioration of immune competence.
149. A method for the treatment of a genetic disease in a patient suffering from such disorder comprising administering to the patient a therapeutically effective amount of a formulation according to claim 99.
150. A method according to claim 149 wherein the disorder involves a pathology or condition which is associated with loss or deterioration of immune competence.
151. A method for the treatment of a genetic disease in a patient suffering from such disorder comprising administering to the patient a therapeutically effective amount of a formulation according to claim 103.
152. A method according to claim 151 wherein the disorder involves a pathology or condition which is associated with loss or deterioration of immune competence.
153. A cationic lipid compound of the formula wherein:
each of x, y and z is independently an integer ~rom O to about 100;
each X1 is independently -O-, -S-, -NRs-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5x2)P(=x2)-x2-i each X2 is independently O or S;
each Yl is independently a phosphate residue, N(R6)a-, S(R6)a-, P(R6)a- or -C02R6, wherein a is an integer from 1 to 3;
each Y2 is independently -N(R6) b- ~ -S (R6) b- or -P(R6) b- ~ wherein b is an integer ~rom O to 2;
each Y3 is independently a phosphate residue, N(R6)a-, S(R6~a-, P(R6)a- or -C02R6, wherein a is an integer from 1 to 3;
each of Rl, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 10 carbons; and each R6 is independently -[R7-X3]C-R8 or -R9-[X4-Rlo] d- Q, wherein:

each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, -N(R11)q, -S(R11)q, -P(R11)q or -CO2R6, wherein q is an integer from 1 to 3;
each of X3 and X4 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2) -N(R5)-, -N(R5) -C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-;
each R7 is independently alkylene of 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 40 carbons;
each of R9 and R10 is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R7-X3]c-R8 or -R9-[X4-R10]d-W, wherein:
each W is independently a phosphate residue, -N(R12)w, -S(R12)w, -P(R12)w or -CO2R6, wherein w is an integer from 1 to 3; and R12 is -[R7-X3]c-R8; with the proviso that the compound of formula (I) comprises at least one quaternary salt.
154. A cationic lipid compound of the formula (II) wherein:

each Y1 is independently a phosphate residue, N(R2)a-, S(R2)a-, P(R2)a- or -CO2R2, wherein a is an integer from 1 to 3;
R1 is alkylene of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
R2 is a residue of the formula -R4-[(X1-R5)x-Y2]y-R6, wherein:
each of x and y is independently an integer from 0 to about 100;
each X1 is independently a direct bond, -O-, -S-, -NR3-, -C(=X2)-, -C(=X2)-N(R3)-, -N(R3)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R3X2)P(=X2)-X2-;
each X2 is independently O or S;
each Y2 is independently -S(R2)b-, -N(R2)b- or -P(R2)b- , wherein b is an integer from 0 to 2;
each R3 is independently hydrogen or alkyl of 1 to about 10 carbons;
each of R4 and R5 is independently a direct bond or alkylene of 1 to about 30 carbons containing 0 to about 15 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups; and each R6 is independently hydrogen or alkyl of 1 to about 60 carbons containing 0 to about 30 -O-, -S-, -NR3- or -X2-(R3X2)P(=X2)-X2- heteroatoms or heteroatom groups;
with the proviso that the compound of formula (II) comprises at least one quaternary salt.
155. A cationic lipid compound of the formula (III) wherein:
each of x, y and z is independently an integer from 0 to about 100;
each X1 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)p(=X2)-X2-;
each X2 is independently O or S;
each Y1 is independently -O-, -N(R6) a-,-S(R6) a- or -P(R6) a-, wherein a is an integer from 0 to 2;
each Y2 is independently -N(R6) a-, -S(R6) a- or -P (R6) a-, wherein a is an integer from 0 to 2;
each Y3 is independently a phosphate residue, N(R6)b-, S(R6)b-, P(R6)b- or -CO2R6, wherein b is an integer from 1 to 3;
each of R1, R2, R3 and R4 is independently alkylene of 1 to about 20 carbons;
each R5 is independently hydrogen or alkyl of 1 to about 10 carbons; and each R6 is independently -[R,-X3]c-R8 or -R9-[X4-R10]d-Q, wherein:
each of c and d is independently an integer from 0 to about 100;
each Q is independently a phosphate residue, -N(R11)q, -S(R11)q, -P(R11)q or -CO2R11, wherein q is an integer from 1 to 3;
each of X3 and X4 is independently -O-, -S-, -NR5-, -C(=X2)-, -C(=X2)-N(R5)-, -N(R5)-C(=X2)-, -C(=X2)-O-, -O-C(=X2)- or -X2-(R5X2)P(=X2)-X2-;
each R7 is independently alkylene of 1 to about 20 carbons;
each R8 is independently hydrogen or alkyl of 1 to about 60 carbons;
each of R9 and R10 is independently alkylene of 1 to about 20 carbons; and each R11 is independently -[R7-X3]c-R8 or -R9-[X4-R10]d-W, wherein:
each W is independently a phosphate residue, -N(R12)w, -S(R12)w, -P(R12)w or -CO2R12, wherein w is an integer from 1 to 3; and R12 is -[R7-X3]c-R8; with the proviso that the compound of formula (III) comprises at least one quaternary salt.
CA002213417A 1995-02-21 1996-01-29 Novel cationic lipids and the use thereof Abandoned CA2213417A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/391,938 1995-02-21
US08/391,938 US5830430A (en) 1995-02-21 1995-02-21 Cationic lipids and the use thereof

Publications (1)

Publication Number Publication Date
CA2213417A1 true CA2213417A1 (en) 1996-08-29

Family

ID=23548604

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002213417A Abandoned CA2213417A1 (en) 1995-02-21 1996-01-29 Novel cationic lipids and the use thereof

Country Status (7)

Country Link
US (2) US5830430A (en)
EP (1) EP0839125A4 (en)
JP (1) JPH11500727A (en)
CN (1) CN1082043C (en)
AU (1) AU4913896A (en)
CA (1) CA2213417A1 (en)
WO (1) WO1996026179A1 (en)

Families Citing this family (266)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6088613A (en) 1989-12-22 2000-07-11 Imarx Pharmaceutical Corp. Method of magnetic resonance focused surgical and therapeutic ultrasound
US5205290A (en) 1991-04-05 1993-04-27 Unger Evan C Low density microspheres and their use as contrast agents for computed tomography
US7323297B1 (en) * 1992-04-03 2008-01-29 The Regents Of The University Of California Stabilized polynucleotide complexes and methods
US5674908A (en) 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US6989434B1 (en) * 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
US6743779B1 (en) * 1994-11-29 2004-06-01 Imarx Pharmaceutical Corp. Methods for delivering compounds into a cell
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6521211B1 (en) 1995-06-07 2003-02-18 Bristol-Myers Squibb Medical Imaging, Inc. Methods of imaging and treatment with targeted compositions
WO2004073750A1 (en) * 1995-06-07 2004-09-02 Harald Dugstad Improvements in or relating to contrast agents
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
EP0848616A4 (en) * 1995-09-08 2000-01-19 Res Dev Foundation Glucocorticoid enhancement of gene expression
US6165442A (en) * 1996-02-19 2000-12-26 Nycomed Imaging As Thermally stabilized ultrasound contrast agent
EP0935415B1 (en) * 1996-05-01 2006-11-22 Imarx Pharmaceutical Corp. In vitro methods for delivering nucleic acids into a cell
AU728581B2 (en) * 1996-09-13 2001-01-11 Lipoxen Limited Liposomes
CA2217550A1 (en) * 1996-10-22 1998-04-22 F. Hoffmann-La Roche Ag Cationic lipids for gene therapy
US6884435B1 (en) * 1997-01-30 2005-04-26 Chiron Corporation Microparticles with adsorbent surfaces, methods of making same, and uses thereof
PT991403E (en) * 1997-01-30 2003-08-29 Chiron Corp USE OF MICROPARTICLES WITH ADSORVED ANTIGEN TO STIMULATE IMMUNE RESPONSES
US20040202680A1 (en) * 1997-01-30 2004-10-14 O'hagan Derek Microparticles with adsorbent surfaces, methods of making same, and uses thereof
US6120751A (en) * 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6143276A (en) * 1997-03-21 2000-11-07 Imarx Pharmaceutical Corp. Methods for delivering bioactive agents to regions of elevated temperatures
US6090800A (en) 1997-05-06 2000-07-18 Imarx Pharmaceutical Corp. Lipid soluble steroid prodrugs
FR2761912B1 (en) 1997-04-14 1999-07-02 Capsulis PROCESS FOR ADHERING A PRODUCT TO A SURFACE
US6416740B1 (en) 1997-05-13 2002-07-09 Bristol-Myers Squibb Medical Imaging, Inc. Acoustically active drug delivery systems
US6548047B1 (en) 1997-09-15 2003-04-15 Bristol-Myers Squibb Medical Imaging, Inc. Thermal preactivation of gaseous precursor filled compositions
US20050003008A1 (en) * 1997-09-23 2005-01-06 Natalya Rapoport Method of in vivo drug targeting to solid tumors via acoustically triggered drug delivery in polymeric micelles
US6565885B1 (en) 1997-09-29 2003-05-20 Inhale Therapeutic Systems, Inc. Methods of spray drying pharmaceutical compositions
US20060165606A1 (en) 1997-09-29 2006-07-27 Nektar Therapeutics Pulmonary delivery particles comprising water insoluble or crystalline active agents
US6433040B1 (en) * 1997-09-29 2002-08-13 Inhale Therapeutic Systems, Inc. Stabilized bioactive preparations and methods of use
US6309623B1 (en) 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
US6946117B1 (en) 1997-09-29 2005-09-20 Nektar Therapeutics Stabilized preparations for use in nebulizers
EP1032428B1 (en) 1997-11-20 2003-06-18 Vical Incorporated Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor
US20010003580A1 (en) 1998-01-14 2001-06-14 Poh K. Hui Preparation of a lipid blend and a phospholipid suspension containing the lipid blend
JP2002511446A (en) * 1998-04-08 2002-04-16 セルテック・セラピューティクス・リミテッド Fat
ES2394458T3 (en) * 1998-09-09 2013-01-31 Genzyme Corporation Methylation of plasmid vectors
EP1044679B1 (en) * 1998-11-02 2012-12-26 Terumo Kabushiki Kaisha Stable Liposomes for Targeted Drug Delivery
DK1129064T3 (en) * 1998-11-12 2008-04-28 Invitrogen Corp transfection
US6342598B1 (en) * 1998-11-26 2002-01-29 Bracco International B.V. Amphipatic polycarboxylic chelates and complexes with paramagnetic metals as MRI contrast agents
US6627421B1 (en) 1999-04-13 2003-09-30 Imarx Therapeutics, Inc. Methods and systems for applying multi-mode energy to biological samples
US6709427B1 (en) 1999-08-05 2004-03-23 Kensey Nash Corporation Systems and methods for delivering agents into targeted tissue of a living being
GB9918670D0 (en) 1999-08-06 1999-10-13 Celltech Therapeutics Ltd Biological product
JP2003511110A (en) * 1999-10-06 2003-03-25 ザ ペン ステイト リサーチ ファンデーション System and device for preventing restenosis in body vessels
US6682545B1 (en) * 1999-10-06 2004-01-27 The Penn State Research Foundation System and device for preventing restenosis in body vessels
AU1333401A (en) * 1999-10-15 2001-04-30 Ohio State University Research Foundation, The Methods for analyzing the insertion capabilities of modified group ii introns
US8029795B2 (en) * 1999-12-30 2011-10-04 Gwathmey, Inc. Targeted iron chelator delivery system
AU3366901A (en) * 1999-12-30 2001-07-16 Novartis Ag Novel colloid synthetic vectors for gene therapy
US6570196B1 (en) * 2000-03-22 2003-05-27 Max-Plank-Gesellschaft Zur Forderung Der Wissenschaften Lipid vesicles or lipid bilayers on chips
US8404217B2 (en) 2000-05-10 2013-03-26 Novartis Ag Formulation for pulmonary administration of antifungal agents, and associated methods of manufacture and use
WO2001085136A2 (en) 2000-05-10 2001-11-15 Alliance Pharmaceutical Corporation Phospholipid-based powders for drug delivery
US7871598B1 (en) 2000-05-10 2011-01-18 Novartis Ag Stable metal ion-lipid powdered pharmaceutical compositions for drug delivery and methods of use
US6696081B2 (en) 2000-06-09 2004-02-24 Duke University Carbohydrate based lipid compositions and supramolecular structures comprising same
US20040022814A1 (en) * 2000-06-15 2004-02-05 O'hagan Derek Microparticles with adsorbent surfaces, methods of making same, and uses thereof
KR100381512B1 (en) 2000-09-21 2003-04-26 굿젠 주식회사 Cationic lipids and use thereof
US6953560B1 (en) 2000-09-28 2005-10-11 Advanced Cardiovascular Systems, Inc. Barriers for polymer-coated implantable medical devices and methods for making the same
US6716444B1 (en) 2000-09-28 2004-04-06 Advanced Cardiovascular Systems, Inc. Barriers for polymer-coated implantable medical devices and methods for making the same
US6663662B2 (en) 2000-12-28 2003-12-16 Advanced Cardiovascular Systems, Inc. Diffusion barrier layer for implantable devices
EP1345629A2 (en) * 2000-12-29 2003-09-24 Advanced Inhalation Research, Inc. Particles for inhalation having sustained release properties
AU2002250034A1 (en) * 2001-02-08 2002-08-19 Sequitur, Inc. Methods of light activated release 0f ligands from endosomes
EP1383541A4 (en) * 2001-03-22 2009-11-04 Chromos Molecular Systems Inc Methods for delivering nucleic acid molecules into cells and assessment thereof
US7294511B2 (en) 2001-03-22 2007-11-13 Chromos Molecular Systems, Inc. Methods for delivering nucleic acid molecules into cells and assessment thereof
US6936469B2 (en) 2001-03-22 2005-08-30 Chromos Molecular Systems Inc. Methods for delivering nucleic acid molecules into cells and assessment thereof
US20030186390A1 (en) * 2001-03-22 2003-10-02 De Jong Gary Methods for delivering nucleic acid molecules into cells and assessment thereof
US20030096414A1 (en) * 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
US8216585B2 (en) 2001-05-25 2012-07-10 The Trustees Of The University Of Pennsylvania Targeted particles and methods of using the same
US6753071B1 (en) 2001-09-27 2004-06-22 Advanced Cardiovascular Systems, Inc. Rate-reducing membrane for release of an agent
WO2003040399A2 (en) * 2001-11-02 2003-05-15 Intradigm Corporation Therapeutic methods for nucleic acid delivery vehicles
TWI324518B (en) 2001-12-19 2010-05-11 Nektar Therapeutics Pulmonary delivery of aminoglycosides
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
EP1470148B1 (en) 2002-02-01 2012-07-18 Life Technologies Corporation Double-stranded oligonucleotides
US20030219785A1 (en) * 2002-02-01 2003-11-27 Vanderbilt University Targeted drug delivery methods
AU2003216288B2 (en) 2002-02-13 2009-09-24 Duke University Modulation of immune response by non-peptide binding stress response polypeptides
WO2003093469A2 (en) * 2002-05-01 2003-11-13 Chromos Molecular Systems, Inc. Methods for delivering nucleic acid molecules into cells and assessment thereof
EP1393719A1 (en) * 2002-08-23 2004-03-03 Munich Biotech AG Camptothecin-carboxylate formulations
EP2338478B1 (en) * 2002-06-28 2014-07-23 Protiva Biotherapeutics Inc. Method for producing liposomes
US7330851B2 (en) * 2003-08-18 2008-02-12 Eaglehawk, Limited Data security through dissembly of data elements or connections between elements
SE0302924D0 (en) * 2003-11-05 2003-11-05 Camurus Ab Pharmaceutical composition having a cationic excipient
EP1547580A1 (en) * 2003-12-23 2005-06-29 MediGene Oncology GmbH Loading of a camptothecin drug into colloidal nanoparticles
US20090142838A1 (en) * 2004-06-14 2009-06-04 Xiaoxia Cui Methods for expressing rnp particles in eukaryotic cells
WO2005123962A2 (en) * 2004-06-14 2005-12-29 The University Of Texas At Austin Gene targeting in eukaryotic cells by group ii intron ribonucleoprotein particles
ATE514776T1 (en) 2004-10-05 2011-07-15 California Inst Of Techn APTAMER-REGULATED NUCLEIC ACIDS AND USES THEREOF
MX2007008425A (en) 2005-01-13 2007-12-10 Cinv Ag Composite materials containing carbon nanoparticles.
EP1714642A1 (en) * 2005-04-18 2006-10-25 Bracco Research S.A. Pharmaceutical composition comprising gas-filled microcapsules for ultrasound mediated delivery
US9006487B2 (en) 2005-06-15 2015-04-14 Massachusetts Institute Of Technology Amine-containing lipids and uses thereof
KR20080063408A (en) * 2005-10-18 2008-07-03 신벤션 아게 Thermoset particles and methods for production thereof
MX363224B (en) 2006-10-03 2019-03-15 Alnylam Pharmaceuticals Inc Lipid containing formulations.
AU2014200910B2 (en) * 2006-10-03 2017-02-09 Arbutus Biopharma Corporation Lipid containing formulations
US8158595B2 (en) 2006-11-09 2012-04-17 California Institute Of Technology Modular aptamer-regulated ribozymes
US20080213611A1 (en) * 2007-01-19 2008-09-04 Cinvention Ag Porous, non-degradable implant made by powder molding
WO2008104586A2 (en) * 2007-02-28 2008-09-04 Cinvention Ag High surface cultivation system with surface increasing substrate
CN101652467A (en) * 2007-02-28 2010-02-17 金文申有限公司 The high surface cultivation system that contains filler
WO2008104588A1 (en) * 2007-02-28 2008-09-04 Cinvention Ag High surface cultivation system
WO2008104599A1 (en) * 2007-02-28 2008-09-04 Cinvention Ag High surface cultivation system bag
US20090082217A1 (en) * 2007-07-16 2009-03-26 California Institute Of Technology Selection of nucleic acid-based sensor domains within nucleic acid switch platform
US20120165387A1 (en) 2007-08-28 2012-06-28 Smolke Christina D General composition framework for ligand-controlled RNA regulatory systems
US8367815B2 (en) * 2007-08-28 2013-02-05 California Institute Of Technology Modular polynucleotides for ligand-controlled regulatory systems
US8865667B2 (en) 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
US9029524B2 (en) * 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
US20110045473A1 (en) * 2008-01-02 2011-02-24 De Fougerolles Antonin Liver screening method
WO2009102427A2 (en) 2008-02-11 2009-08-20 Rxi Pharmaceuticals Corp. Modified rnai polynucleotides and uses thereof
MX2010012326A (en) * 2008-05-19 2010-12-01 Univ North Carolina Methods and compositions comprising novel cationic lipids.
EP2296711A2 (en) * 2008-05-29 2011-03-23 MDRNA, Inc. Multi-arm amines and uses thereof
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
ES2646630T3 (en) 2008-11-07 2017-12-14 Massachusetts Institute Of Technology Amino alcoholic lipidoids and uses thereof
TW201019969A (en) * 2008-11-17 2010-06-01 Enzon Pharmaceuticals Inc Branched cationic lipids for nucleic acids delivery system
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
US20110008894A1 (en) * 2009-07-07 2011-01-13 Cayla Lyophilized plasmid/dna transfection reagent carrier complex
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
AU2010326132B9 (en) 2009-12-01 2014-10-02 Translate Bio, Inc. Delivery of mRNA for the augmentation of proteins and enzymes in human genetic diseases
EA201792402A3 (en) * 2010-03-08 2018-09-28 Монсанто Текнолоджи Ллс POLYNUCLEOTIDE MOLECULES FOR REGULATION OF PLANT GENES
JP6060071B2 (en) 2010-03-24 2017-01-11 アールエックスアイ ファーマシューティカルズ コーポレーション RNA interference in skin and fibrosis applications
US9095504B2 (en) 2010-03-24 2015-08-04 Rxi Pharmaceuticals Corporation RNA interference in ocular indications
EP2550000A4 (en) 2010-03-24 2014-03-26 Advirna Inc Reduced size self-delivering rnai compounds
JP5902616B2 (en) * 2010-04-28 2016-04-13 協和発酵キリン株式会社 Cationic lipid
EP2609135A4 (en) 2010-08-26 2015-05-20 Massachusetts Inst Technology Poly(beta-amino alcohols), their preparation, and uses thereof
WO2012135025A2 (en) 2011-03-28 2012-10-04 Massachusetts Institute Of Technology Conjugated lipomers and uses thereof
CN111671918A (en) 2011-06-08 2020-09-18 川斯勒佰尔公司 Lipid nanoparticle compositions and methods for MRNA delivery
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
AR088155A1 (en) 2011-09-13 2014-05-14 Monsanto Technology Llc METHODS AND COMPOSITIONS FOR WEED CONTROL
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
WO2013040057A1 (en) 2011-09-13 2013-03-21 Monsanto Technology Llc Methods and compositions for weed control
EP2755467B1 (en) 2011-09-13 2017-07-19 Monsanto Technology LLC Methods and compositions for weed control
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
AU2012308765B2 (en) 2011-09-13 2018-06-21 Monsanto Technology Llc Methods and compositions for weed control
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
EA032088B1 (en) 2011-10-27 2019-04-30 Массачусетс Инститьют Оф Текнолоджи Amino acid derivates functionalized on the n-terminal capable of forming drug incapsulating microspheres
US9737480B2 (en) 2012-02-06 2017-08-22 President And Fellows Of Harvard College ARRDC1-mediated microvesicles (ARMMs) and uses thereof
AR091143A1 (en) 2012-05-24 2015-01-14 Seeds Ltd Ab COMPOSITIONS AND METHODS TO SILENCE GENETIC EXPRESSION
WO2013185067A1 (en) 2012-06-08 2013-12-12 Shire Human Genetic Therapies, Inc. Nuclease resistant polynucleotides and uses thereof
CA2884870C (en) 2012-08-13 2022-03-29 Massachusetts Institute Of Technology Amine-containing lipidoids and uses thereof
CN102813942A (en) * 2012-08-15 2012-12-12 钟志容 Lipid ultrasound microbubble mediated adeno-associated virus gene transfection preparation and preparation technology thereof
EP2908620A4 (en) 2012-10-18 2016-07-27 Monsanto Technology Llc Methods and compositions for plant pest control
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
EP2941488B1 (en) 2013-01-01 2023-03-22 Monsanto Technology LLC Methods of introducing dsrna to plant seeds for modulating gene expression
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
MX364458B (en) 2013-03-13 2019-04-26 Monsanto Technology Llc Methods and compositions for weed control.
EP2967082A4 (en) 2013-03-13 2016-11-02 Monsanto Technology Llc Methods and compositions for weed control
IL305374A (en) 2013-03-14 2023-10-01 Ethris Gmbh Cftr mrna compositions and related methods and uses
TR201901310T4 (en) 2013-03-14 2019-02-21 Translate Bio Inc Methods of purification of messenger RNA.
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
CN105431526A (en) * 2013-03-15 2016-03-23 麦克法兰布奈特医疗研究与公共健康研究所有限公司 Immunogenic compositions and a process for producing same
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
US9315472B2 (en) 2013-05-01 2016-04-19 Massachusetts Institute Of Technology 1,3,5-triazinane-2,4,6-trione derivatives and uses thereof
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
MX359191B (en) 2013-07-19 2018-09-18 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa.
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
WO2015031536A1 (en) 2013-08-27 2015-03-05 Northeastern University Nanoparticle drug delivery system and method of treating cancer and neurotrauma
WO2016070129A1 (en) 2014-10-30 2016-05-06 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
WO2015061491A1 (en) 2013-10-22 2015-04-30 Shire Human Genetic Therapies, Inc. Mrna therapy for phenylketonuria
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
SG11201602943PA (en) 2013-10-22 2016-05-30 Shire Human Genetic Therapies Lipid formulations for delivery of messenger rna
AR098295A1 (en) 2013-11-04 2016-05-26 Monsanto Technology Llc COMPOSITIONS AND METHODS TO CONTROL INFESTATIONS OF PESTS AND PARASITES OF THE ARTHROPODS
LT3066201T (en) 2013-11-07 2018-08-10 Editas Medicine, Inc. Crispr-related methods and compositions with governing grnas
US10934550B2 (en) 2013-12-02 2021-03-02 Phio Pharmaceuticals Corp. Immunotherapy of cancer
WO2015085113A1 (en) 2013-12-04 2015-06-11 Rxi Pharmaceuticals Corporation Methods for treatment of wound healing utilizing chemically modified oligonucleotides
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
US20150166982A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting pi3k point mutations
AR099092A1 (en) 2014-01-15 2016-06-29 Monsanto Technology Llc METHODS AND COMPOSITIONS FOR WEED CONTROL USING EPSPS POLYUCLEOTIDES
US11091770B2 (en) 2014-04-01 2021-08-17 Monsanto Technology Llc Compositions and methods for controlling insect pests
CN106164248B (en) 2014-04-25 2019-10-15 川斯勒佰尔公司 The purification process of mRNA
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
CA2947619A1 (en) 2014-05-01 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treatment of disorders in the front of the eye utilizing nucleic acid molecules
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
US10988764B2 (en) 2014-06-23 2021-04-27 Monsanto Technology Llc Compositions and methods for regulating gene expression via RNA interference
KR102511554B1 (en) 2014-06-24 2023-03-16 샤이어 휴먼 지네틱 테라피즈 인크. Stereochemically enriched compositions for delivery of nucleic acids
WO2015200539A1 (en) 2014-06-25 2015-12-30 Monsanto Technology Llc Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
EP3164379A1 (en) 2014-07-02 2017-05-10 Massachusetts Institute of Technology Polyamine-fatty acid derived lipidoids and uses thereof
CN106572974B (en) 2014-07-15 2021-04-23 生命技术公司 Compositions and methods having lipid aggregates for efficient delivery of molecules to cells
US10378012B2 (en) 2014-07-29 2019-08-13 Monsanto Technology Llc Compositions and methods for controlling insect pests
CA2956224A1 (en) 2014-07-30 2016-02-11 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
KR102506169B1 (en) 2014-09-05 2023-03-08 피오 파마슈티칼스 코프. Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
US9816080B2 (en) 2014-10-31 2017-11-14 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
CA2972423A1 (en) 2014-12-31 2016-07-07 Lantheus Medical Imaging, Inc. Lipid-encapsulated gas microsphere compositions and related methods
CA2974101A1 (en) 2015-01-22 2016-07-28 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa
CN106188230B (en) * 2015-05-07 2019-10-25 内蒙古大学 A kind of cation lipid class compound and the preparation method and application thereof
CN107750125A (en) 2015-06-02 2018-03-02 孟山都技术有限公司 For by the composition and method in delivery of polynucleotides to plant
WO2016196782A1 (en) 2015-06-03 2016-12-08 Monsanto Technology Llc Methods and compositions for introducing nucleic acids into plants
CN107848988B (en) 2015-06-19 2021-10-22 麻省理工学院 Alkenyl substituted 2, 5-piperazinediones and their use in compositions for delivering an agent to a subject or cell
CA2991598A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Nucleic acid molecules targeting superoxide dismutase 1 (sod1)
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
JP6933379B2 (en) 2015-09-24 2021-09-08 ザ ユニバーシティ オブ ノース カロライナ アット チャペル ヒル Methods and compositions for reducing metastasis
JP2018531037A (en) 2015-10-19 2018-10-25 アールエックスアイ ファーマシューティカルズ コーポレーション Reduced size self-delivering nucleic acid compounds targeting long non-coding RNAs
CN108513575A (en) 2015-10-23 2018-09-07 哈佛大学的校长及成员们 Nucleobase editing machine and application thereof
TWI740937B (en) 2016-05-04 2021-10-01 美商藍瑟斯醫學影像公司 Methods and devices for preparation of ultrasound contrast agents
US9789210B1 (en) 2016-07-06 2017-10-17 Lantheus Medical Imaging, Inc. Methods for making ultrasound contrast agents
WO2018027078A1 (en) 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
KR102622411B1 (en) 2016-10-14 2024-01-10 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 AAV delivery of nucleobase editor
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
EP3592777A1 (en) 2017-03-10 2020-01-15 President and Fellows of Harvard College Cytosine to guanine base editor
WO2018176009A1 (en) 2017-03-23 2018-09-27 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
WO2018209320A1 (en) 2017-05-12 2018-11-15 President And Fellows Of Harvard College Aptazyme-embedded guide rnas for use with crispr-cas9 in genome editing and transcriptional activation
WO2018213476A1 (en) 2017-05-16 2018-11-22 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr
CN111801345A (en) 2017-07-28 2020-10-20 哈佛大学的校长及成员们 Methods and compositions using an evolved base editor for Phage Assisted Continuous Evolution (PACE)
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
WO2019057649A1 (en) 2017-09-19 2019-03-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of acute myeloid leukemia
WO2019079347A1 (en) 2017-10-16 2019-04-25 The Broad Institute, Inc. Uses of adenosine base editors
SG11202004457XA (en) 2017-11-17 2020-06-29 Iovance Biotherapeutics Inc Til expansion from fine needle aspirates and small biopsies
WO2019136459A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
SG11202006541UA (en) 2018-01-08 2020-08-28 Iovance Biotherapeutics Inc Processes for generating til products enriched for tumor antigen-specific t-cells
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
WO2019210131A1 (en) 2018-04-27 2019-10-31 Iovance Biotherapeutics, Inc. Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2020033791A1 (en) 2018-08-09 2020-02-13 Verseau Therapeutics, Inc. Oligonucleotide compositions for targeting ccr2 and csf1r and uses thereof
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US20230048010A1 (en) 2018-09-28 2023-02-16 President And Fellows Of Harvard College Cellular reprogramming to reverse aging and promote organ and tissue regeneration
KR20210099573A (en) 2018-11-05 2021-08-12 이오반스 바이오테라퓨틱스, 인크. Selection of improved tumor-reactive T-cells
JOP20210094A1 (en) 2018-11-05 2023-01-30 Iovance Biotherapeutics Inc Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2020096927A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathway inhibitors
SG11202104663PA (en) 2018-11-05 2021-06-29 Iovance Biotherapeutics Inc Treatment of nsclc patients refractory for anti-pd-1 antibody
EP3898949A1 (en) 2018-12-19 2021-10-27 Iovance Biotherapeutics, Inc. Methods of expanding tumor infiltrating lymphocytes using engineered cytokine receptor pairs and uses thereof
MX2021010288A (en) 2019-03-01 2021-09-23 Iovance Biotherapeutics Inc Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof.
AU2020240109A1 (en) 2019-03-19 2021-09-30 President And Fellows Of Harvard College Methods and compositions for editing nucleotide sequences
US20220249559A1 (en) 2019-05-13 2022-08-11 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
EP3997225A1 (en) 2019-07-10 2022-05-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of epilepsy
US11788091B2 (en) 2019-08-21 2023-10-17 University Of Virginia Patent Foundation Methods and compositions for diagnosing and treating prostate cancer based on long noncoding RNA overlapping the LCK gene that regulates prostate cancer cell growth
WO2021081378A1 (en) 2019-10-25 2021-04-29 Iovance Biotherapeutics, Inc. Gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
CN115135765A (en) 2019-11-08 2022-09-30 菲奥医药公司 Chemically modified oligonucleotides targeting bromodomain-containing protein 4(BRD4) for immunotherapy
US20230016983A1 (en) 2019-11-19 2023-01-19 lNSERM (INSTITUT NATIONAL DE LA SANTÉ ET DE LA RECHERCHE MÉDICALE) Antisense oligonucleotides and thier use for the treatment of cancer
JP2023506734A (en) 2019-12-11 2023-02-20 アイオバンス バイオセラピューティクス,インコーポレイテッド Process for the production of tumor-infiltrating lymphocytes (TIL) and methods of using same
WO2021138537A1 (en) 2019-12-31 2021-07-08 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides with improved systemic delivery
CN111208223B (en) * 2020-01-13 2023-01-17 浙江大学 Metabolite for pre-operation early warning of delayed recovery of transplanted kidney of donation receptor after cardiac death
CA3177413A1 (en) 2020-05-04 2021-11-11 Michelle SIMPSON-ABELSON Selection of improved tumor reactive t-cells
CA3176826A1 (en) 2020-05-04 2021-11-11 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
AU2021267940A1 (en) 2020-05-08 2022-12-08 President And Fellows Of Harvard College Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
JP2023546359A (en) 2020-10-06 2023-11-02 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of NSCLC patients with tumor-infiltrating lymphocyte therapy
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
CA3201818A1 (en) 2020-12-11 2022-06-16 Maria Fardis Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with braf inhibitors and/or mek inhibitors
EP4262811A1 (en) 2020-12-17 2023-10-25 Iovance Biotherapeutics, Inc. Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors
JP2024500403A (en) 2020-12-17 2024-01-09 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer with tumor-infiltrating lymphocytes
WO2022165260A1 (en) 2021-01-29 2022-08-04 Iovance Biotherapeutics, Inc. Methods of making modified tumor infiltrating lymphocytes and their use in adoptive cell therapy
JP2024510505A (en) 2021-03-19 2024-03-07 アイオバンス バイオセラピューティクス,インコーポレイテッド Methods for tumor-infiltrating lymphocyte (TIL) expansion and gene knockout in TILs associated with CD39/CD69 selection
JP2024511414A (en) 2021-03-23 2024-03-13 アイオバンス バイオセラピューティクス,インコーポレイテッド CISH gene editing of tumor-infiltrating lymphocytes and its use in immunotherapy
EP4326287A2 (en) 2021-04-19 2024-02-28 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
WO2022245754A1 (en) 2021-05-17 2022-11-24 Iovance Biotherapeutics, Inc. Pd-1 gene-edited tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2023004074A2 (en) 2021-07-22 2023-01-26 Iovance Biotherapeutics, Inc. Method for cryopreservation of solid tumor fragments
CA3226942A1 (en) 2021-07-28 2023-02-02 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
WO2023012165A1 (en) 2021-08-02 2023-02-09 Universite De Montpellier Compositions and methods for treating cmt1a or cmt1e diseases with rnai molecules targeting pmp22
WO2023015265A2 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
WO2023015264A1 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Immunotherapy of cancer utilizing natural killer cells treated with chemically modified oligonucleotides
AU2022343729A1 (en) 2021-09-09 2024-03-21 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1 talen knockdown
AR127482A1 (en) 2021-10-27 2024-01-31 Iovance Biotherapeutics Inc SYSTEMS AND METHODS TO COORDINATE THE MANUFACTURE OF CELLS FOR PATIENT-SPECIFIC IMMUNOTHERAPY
WO2023086803A1 (en) 2021-11-10 2023-05-19 Iovance Biotherapeutics, Inc. Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes
WO2023147486A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Tumor infiltrating lymphocytes engineered to express payloads
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023196877A1 (en) 2022-04-06 2023-10-12 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2023196851A1 (en) 2022-04-06 2023-10-12 President And Fellows Of Harvard College Reversing aging of the central nervous system
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
WO2023220608A1 (en) 2022-05-10 2023-11-16 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with an il-15r agonist
WO2024017990A1 (en) 2022-07-21 2024-01-25 Institut National de la Santé et de la Recherche Médicale Methods and compositions for treating chronic pain disorders
WO2024030758A1 (en) 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
WO2024055017A1 (en) 2022-09-09 2024-03-14 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1/tigit talen double knockdown
WO2024055018A1 (en) 2022-09-09 2024-03-14 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1/tigit talen double knockdown

Family Cites Families (207)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3015128A (en) * 1960-08-18 1962-01-02 Southwest Res Inst Encapsulating apparatus
NL302030A (en) 1962-12-21 1900-01-01
US3291843A (en) * 1963-10-08 1966-12-13 Du Pont Fluorinated vinyl ethers and their preparation
BE661981A (en) * 1964-04-03
US3594326A (en) * 1964-12-03 1971-07-20 Ncr Co Method of making microscopic capsules
US3968203A (en) * 1965-10-01 1976-07-06 Jerome G. Spitzer Aerosol astringent composition
US3488714A (en) * 1966-09-19 1970-01-06 Dow Chemical Co Formed laminate structure and method of preparation
US3615972A (en) * 1967-04-28 1971-10-26 Dow Chemical Co Expansible thermoplastic polymer particles containing volatile fluid foaming agent and method of foaming the same
US3532500A (en) * 1967-07-25 1970-10-06 Eastman Kodak Co Light sensitive vesicular composition comprising an azido-s-triazine compound
US3557294A (en) * 1967-10-12 1971-01-19 Allied Chem Fluorinated ethers as inhalation convulsants
US3479811A (en) * 1967-11-29 1969-11-25 Dow Chemical Co Yarn and method of making the same
US3732172A (en) * 1968-02-28 1973-05-08 Ncr Co Process for making minute capsules and prefabricated system useful therein
US3650831A (en) * 1969-03-10 1972-03-21 Armour Dial Inc Method of cleaning surfaces
US4027007A (en) * 1970-12-09 1977-05-31 Colgate-Palmolive Company Antiperspirants formulated with borax
US3873564A (en) * 1971-03-03 1975-03-25 Synvar Ass 2-Imidazolinyl-3-oxide-1-oxypropionic acid
US4108806A (en) * 1971-12-06 1978-08-22 The Dow Chemical Company Thermoplastic expandable microsphere process and product
US4179546A (en) * 1972-08-28 1979-12-18 The Dow Chemical Company Method for expanding microspheres and expandable composition
US3960583A (en) * 1974-05-02 1976-06-01 Philadelphia Quartz Company Method of preparing modified hollow, largely spherical particles by spray drying
CH588887A5 (en) * 1974-07-19 1977-06-15 Battelle Memorial Institute
US3945956A (en) * 1975-06-23 1976-03-23 The Dow Chemical Company Polymerization of styrene acrylonitrile expandable microspheres
US4138383A (en) * 1975-11-24 1979-02-06 California Institute Of Technology Preparation of small bio-compatible microspheres
GB1523965A (en) * 1976-03-19 1978-09-06 Ici Ltd Pharmaceutical compositions containing steroids
US4162282A (en) * 1976-04-22 1979-07-24 Coulter Electronics, Inc. Method for producing uniform particles
GB1599881A (en) * 1977-02-02 1981-10-07 Millington A R Preparation for diagnostic radiology
CH624011A5 (en) * 1977-08-05 1981-07-15 Battelle Memorial Institute
CH621479A5 (en) * 1977-08-05 1981-02-13 Battelle Memorial Institute
US4192859A (en) * 1978-09-29 1980-03-11 E. R. Squibb & Sons, Inc. Contrast media containing liposomes as carriers
US4310506A (en) * 1979-02-22 1982-01-12 California Institute Of Technology Means of preparation and applications of liposomes containing high concentrations of entrapped ionic species
US4276885A (en) * 1979-05-04 1981-07-07 Rasor Associates, Inc Ultrasonic image enhancement
US4265251A (en) * 1979-06-28 1981-05-05 Rasor Associates, Inc. Method of determining pressure within liquid containing vessel
US4310505A (en) * 1979-11-08 1982-01-12 California Institute Of Technology Lipid vesicles bearing carbohydrate surfaces as lymphatic directed vehicles for therapeutic and diagnostic substances
US4342826A (en) * 1980-02-04 1982-08-03 Collaborative Research, Inc. Immunoassay products and methods
US4421562A (en) * 1980-04-13 1983-12-20 Pq Corporation Manufacturing process for hollow microspheres
US4344929A (en) * 1980-04-25 1982-08-17 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4315514A (en) * 1980-05-08 1982-02-16 William Drewes Method and apparatus for selective cell destruction
US4331654A (en) * 1980-06-13 1982-05-25 Eli Lilly And Company Magnetically-localizable, biodegradable lipid microspheres
US4681119A (en) * 1980-11-17 1987-07-21 Schering Aktiengesellschaft Method of production and use of microbubble precursors
US4442843A (en) * 1980-11-17 1984-04-17 Schering, Ag Microbubble precursors and methods for their production and use
US4657756A (en) * 1980-11-17 1987-04-14 Schering Aktiengesellschaft Microbubble precursors and apparatus for their production and use
US4427330A (en) * 1981-03-03 1984-01-24 Carter Charles P Automatic fence picket stock conveyor for fence picket pointing machine
US4420442A (en) * 1981-04-13 1983-12-13 Pq Corporation Manufacturing process for hollow microspheres
US4533254A (en) * 1981-04-17 1985-08-06 Biotechnology Development Corporation Apparatus for forming emulsions
EP0068961A3 (en) * 1981-06-26 1983-02-02 Thomson-Csf Apparatus for the local heating of biological tissue
US4426330A (en) * 1981-07-20 1984-01-17 Lipid Specialties, Inc. Synthetic phospholipid compounds
US4534899A (en) * 1981-07-20 1985-08-13 Lipid Specialties, Inc. Synthetic phospholipid compounds
US4569836A (en) * 1981-08-27 1986-02-11 Gordon Robert T Cancer treatment by intracellular hyperthermia
EP0088773B1 (en) * 1981-09-23 1987-09-09 M.B. Fillers Pty. Ltd. Hollow, bilayered silicate microspheres
DE3141641A1 (en) * 1981-10-16 1983-04-28 Schering Ag, 1000 Berlin Und 4619 Bergkamen ULTRASONIC CONTRAST AGENTS AND THEIR PRODUCTION
US4540629A (en) * 1982-04-08 1985-09-10 Pq Corporation Hollow microspheres with organosilicon-silicate walls
DE3225848A1 (en) * 1982-07-07 1984-01-19 Schering AG, 1000 Berlin und 4709 Bergkamen PREPARATION OF CORTICOIDS FOR TOPICAL APPLICATION
FR2534487B1 (en) 1982-10-15 1988-06-10 Dior Christian Parfums METHOD FOR THE HOMOGENEIZATION OF HYDRATED LIPIDAL LAMELLAR PHASE DISPERSIONS, AND SUSPENSIONS OBTAINED THEREBY
EP0111386B1 (en) * 1982-10-26 1987-11-19 University Of Aberdeen Ultrasound hyperthermia unit
US4603044A (en) * 1983-01-06 1986-07-29 Technology Unlimited, Inc. Hepatocyte Directed Vesicle delivery system
US4731239A (en) * 1983-01-10 1988-03-15 Gordon Robert T Method for enhancing NMR imaging; and diagnostic use
US4572203A (en) * 1983-01-27 1986-02-25 Feinstein Steven B Contact agents for ultrasonic imaging
US4718433A (en) * 1983-01-27 1988-01-12 Feinstein Steven B Contrast agents for ultrasonic imaging
US4775522A (en) * 1983-03-04 1988-10-04 Children's Hospital Research Foundation, A Division Of Children's Hospital Medical Center NMR compositions for indirectly detecting a dissolved gas in an animal
US4981692A (en) * 1983-03-24 1991-01-01 The Liposome Company, Inc. Therapeutic treatment by intramammary infusion
US5141738A (en) 1983-04-15 1992-08-25 Schering Aktiengesellschaft Ultrasonic contrast medium comprising gas bubbles and solid lipophilic surfactant-containing microparticles and use thereof
US4900540A (en) * 1983-06-20 1990-02-13 Trustees Of The University Of Massachusetts Lipisomes containing gas for ultrasound detection
US4544545A (en) * 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4615879A (en) * 1983-11-14 1986-10-07 Vanderbilt University Particulate NMR contrast agents for gastrointestinal application
FR2563725B1 (en) * 1984-05-03 1988-07-15 Dory Jacques APPARATUS FOR EXAMINING AND LOCATING ULTRASONIC TUMORS WITH A LOCALIZED HYPERTHERMAL TREATMENT DEVICE
DE3585967D1 (en) * 1984-03-08 1992-06-11 Phares Pharma Holland LIPOSOME FORMING COMPOSITION.
GB8407557D0 (en) * 1984-03-23 1984-05-02 Hayward J A Polymeric lipsomes
US4728575A (en) * 1984-04-27 1988-03-01 Vestar, Inc. Contrast agents for NMR imaging
US5008109A (en) * 1984-05-25 1991-04-16 Vestar, Inc. Vesicle stabilization
US5008050A (en) * 1984-06-20 1991-04-16 The Liposome Company, Inc. Extrusion technique for producing unilamellar vesicles
US4620546A (en) * 1984-06-30 1986-11-04 Kabushiki Kaisha Toshiba Ultrasound hyperthermia apparatus
US4789501A (en) * 1984-11-19 1988-12-06 The Curators Of The University Of Missouri Glass microspheres
US4921706A (en) * 1984-11-20 1990-05-01 Massachusetts Institute Of Technology Unilamellar lipid vesicles and method for their formation
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
US4946787A (en) * 1985-01-07 1990-08-07 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
US4830858A (en) * 1985-02-11 1989-05-16 E. R. Squibb & Sons, Inc. Spray-drying method for preparing liposomes and products produced thereby
US4689986A (en) * 1985-03-13 1987-09-01 The University Of Michigan Variable frequency gas-bubble-manipulating apparatus and method
US5186922A (en) 1985-03-15 1993-02-16 See/Shell Biotechnology, Inc. Use of biodegradable microspheres labeled with imaging energy constrast materials
US4663161A (en) * 1985-04-22 1987-05-05 Mannino Raphael J Liposome methods and compositions
ATE78158T1 (en) * 1985-05-22 1992-08-15 Liposome Technology Inc METHOD AND SYSTEM FOR INHALATION OF LIPOSOMES.
DE3677112D1 (en) 1985-08-12 1991-02-28 Battelle Memorial Institute POROESE FILTRATION GLASS BALLS AND METHOD FOR THE PRODUCTION THEREOF.
US4684479A (en) * 1985-08-14 1987-08-04 Arrigo Joseph S D Surfactant mixtures, stable gas-in-liquid emulsions, and methods for the production of such emulsions from said mixtures
US4938947A (en) * 1985-11-01 1990-07-03 Centre National De La Recherche Scientifique (Cnrs) Aerosol composition for in vivo imaging
US4927623A (en) * 1986-01-14 1990-05-22 Alliance Pharmaceutical Corp. Dissolution of gas in a fluorocarbon liquid
US4987154A (en) * 1986-01-14 1991-01-22 Alliance Pharmaceutical Corp. Biocompatible, stable and concentrated fluorocarbon emulsions for contrast enhancement and oxygen transport in internal animal use
US4865836A (en) * 1986-01-14 1989-09-12 Fluoromed Pharmaceutical, Inc. Brominated perfluorocarbon emulsions for internal animal use for contrast enhancement and oxygen transport
DE3785054T2 (en) 1986-01-24 1993-07-08 Childrens Hosp Medical Center STABLE EMULSIONS OF STRONGLY FLUORED ORGANIC COMPOUNDS.
US4737323A (en) * 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US4834964A (en) * 1986-03-07 1989-05-30 M.R.I., Inc. Use of charged nitroxides as NMR image enhancing agents for CSF
JPH0751496B2 (en) * 1986-04-02 1995-06-05 武田薬品工業株式会社 Manufacturing method of liposome
DE3614657A1 (en) 1986-04-30 1987-11-05 Dornier Medizintechnik LIPID VESICLES CONTAINING PHARMAKA, METHOD FOR THE PRODUCTION AND INTRODUCTION THEREOF IN THE BODY OF A LIVING BEING AND RELEASE OF THE PHARMACA CONTAINING IN THE LIPID VESICLES
FR2602774B1 (en) * 1986-07-29 1990-10-19 Atta NOVEL POLYHYDROXYLATED AND PERFLUOROALKYLATED AMPHIPHILIC MOLECULES HAVING SURFACTANT PROPERTIES
IL79559A0 (en) 1986-07-29 1986-10-31 Univ Ramot Contrast agents for nmr medical imaging
US4728578A (en) * 1986-08-13 1988-03-01 The Lubrizol Corporation Compositions containing basic metal salts and/or non-Newtonian colloidal disperse systems and vinyl aromatic containing polymers
US4776991A (en) * 1986-08-29 1988-10-11 The United States Of America As Represented By The Secretary Of The Navy Scaled-up production of liposome-encapsulated hemoglobin
US4781871A (en) * 1986-09-18 1988-11-01 Liposome Technology, Inc. High-concentration liposome processing method
ZW11287A1 (en) * 1986-11-04 1989-01-25 Aeci Ltd Process for the production of an explosive
DE3637926C1 (en) 1986-11-05 1987-11-26 Schering Ag Ultrasonic manometry in a liquid using microbubbles
US5049388A (en) * 1986-11-06 1991-09-17 Research Development Foundation Small particle aerosol liposome and liposome-drug combinations for medical use
US4863717A (en) * 1986-11-10 1989-09-05 The State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of The University Of Oregon Methods for circumventing the problem of free radial reduction associated with the use of stable nitroxide free radicals as contrast agents for magnetic reasonance imaging
US4933121A (en) * 1986-12-10 1990-06-12 Ciba Corning Diagnostics Corp. Process for forming liposomes
DK175531B1 (en) 1986-12-15 2004-11-22 Nexstar Pharmaceuticals Inc Delivery vehicle with amphiphil-associated active ingredient
CA1321048C (en) * 1987-03-05 1993-08-10 Robert W. J. Lencki Microspheres and method of producing same
US5219538A (en) 1987-03-13 1993-06-15 Micro-Pak, Inc. Gas and oxygen carrying lipid vesicles
US5000960A (en) * 1987-03-13 1991-03-19 Micro-Pak, Inc. Protein coupling to lipid vesicles
IE61448B1 (en) * 1987-06-23 1994-11-02 Hafslund Nycomed Innovation Improvements in and relating to magnetic resonance imaging
US5354549A (en) 1987-07-24 1994-10-11 Nycomed Imaging As Iodinated esters
US4844882A (en) * 1987-12-29 1989-07-04 Molecular Biosystems, Inc. Concentrated stabilized microbubble-type ultrasonic imaging agent
IE61591B1 (en) 1987-12-29 1994-11-16 Molecular Biosystems Inc Concentrated stabilized microbubble-type ultrasonic imaging agent and method of production
US5425366A (en) 1988-02-05 1995-06-20 Schering Aktiengesellschaft Ultrasonic contrast agents for color Doppler imaging
JP2907911B2 (en) 1988-02-05 1999-06-21 シエーリング アクチエンゲゼルシヤフト Ultrasound contrast agent, method for producing the same, and diagnostic or therapeutic preparation comprising the ultrasound contrast agent
US4898734A (en) * 1988-02-29 1990-02-06 Massachusetts Institute Of Technology Polymer composite for controlled release or membrane formation
DE3812816A1 (en) 1988-04-16 1989-11-02 Lawaczeck Ruediger Dipl Phys P METHOD FOR SOLUBILIZING LIPOSOMES AND / OR BIOLOGICAL MEMBRANES AND THE USE THEREOF
US5171755A (en) 1988-04-29 1992-12-15 Hemagen/Pfc Emulsions of highly fluorinated organic compounds
US4893624A (en) * 1988-06-21 1990-01-16 Massachusetts Institute Of Technology Diffuse focus ultrasound hyperthermia system
US4993415A (en) * 1988-08-19 1991-02-19 Alliance Pharmaceutical Corp. Magnetic resonance imaging with perfluorocarbon hydrides
US4996041A (en) * 1988-08-19 1991-02-26 Toshiyuki Arai Method for introducing oxygen-17 into tissue for imaging in a magnetic resonance imaging system
DE3828905A1 (en) 1988-08-23 1990-03-15 Schering Ag MEDIALLY COMPOSED OF CAVITATE OR CLATHRATE MAKING HOST / GUEST COMPLEX AS A CONTRAST
US5045304A (en) * 1988-08-31 1991-09-03 Wayne State University Contras agent having an imaging agent coupled to viable granulocytes for use in magnetic resonance imaging of abscess and a method of preparing and using same
US5410516A (en) 1988-09-01 1995-04-25 Schering Aktiengesellschaft Ultrasonic processes and circuits for performing them
US4957656A (en) * 1988-09-14 1990-09-18 Molecular Biosystems, Inc. Continuous sonication method for preparing protein encapsulated microbubbles
IL91664A (en) 1988-09-28 1993-05-13 Yissum Res Dev Co Ammonium transmembrane gradient system for efficient loading of liposomes with amphipathic drugs and their controlled release
FR2637182B1 (en) 1988-10-03 1992-11-06 Lvmh Rech COMPOSITIONS BASED ON HYDRATED LIPID LAMID PHASES OR LIPOSOMES CONTAINING AN ECDYSTEROID, PREFERABLY ECDYSTERONE, OR A DERIVATIVE THEREOF; AND COSMETIC, PHARMACEUTICAL, ESPECIALLY DERMATOLOGICAL, SERICULTURE OR PHYTOSANITARY COMPOSITIONS INCORPORATING THE SAME
FR2645866B1 (en) 1989-04-17 1991-07-05 Centre Nat Rech Scient NEW LIPOPOLYAMINES, THEIR PREPARATION AND THEIR USE
US5114703A (en) * 1989-05-30 1992-05-19 Alliance Pharmaceutical Corp. Percutaneous lymphography using particulate fluorocarbon emulsions
ES2060184T3 (en) 1989-06-22 1994-11-16 Atta AMPHYPHILIC MOLECULES CONTAINING FLUORINE AND PHOSPHORUS WITH TENSOACTIVE PROPERTIES.
US5227481A (en) 1989-07-07 1993-07-13 National Starch And Chemical Investment Holding Corporation Cationic polysaccharides and reagents for their preparation
US5019370A (en) * 1989-07-10 1991-05-28 University Of Kentucky Research Foundation Biodegradable, low biological toxicity radiographic contrast medium and method of x-ray imaging
US5194266A (en) 1989-08-08 1993-03-16 Liposome Technology, Inc. Amphotericin B/cholesterol sulfate composition and method
US5100662A (en) * 1989-08-23 1992-03-31 The Liposome Company, Inc. Steroidal liposomes exhibiting enhanced stability
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5620689A (en) 1989-10-20 1997-04-15 Sequus Pharmaceuuticals, Inc. Liposomes for treatment of B-cell and T-cell disorders
US5209720A (en) 1989-12-22 1993-05-11 Unger Evan C Methods for providing localized therapeutic heat to biological tissues and fluids using gas filled liposomes
US5088499A (en) * 1989-12-22 1992-02-18 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
US5469854A (en) 1989-12-22 1995-11-28 Imarx Pharmaceutical Corp. Methods of preparing gas-filled liposomes
US5352435A (en) 1989-12-22 1994-10-04 Unger Evan C Ionophore containing liposomes for ultrasound imaging
US5149319A (en) 1990-09-11 1992-09-22 Unger Evan C Methods for providing localized therapeutic heat to biological tissues and fluids
US5542935A (en) 1989-12-22 1996-08-06 Imarx Pharmaceutical Corp. Therapeutic delivery systems related applications
US5334381A (en) 1989-12-22 1994-08-02 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
US5123414A (en) * 1989-12-22 1992-06-23 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
US5305757A (en) 1989-12-22 1994-04-26 Unger Evan C Gas filled liposomes and their use as ultrasonic contrast agents
US5230882A (en) 1989-12-22 1993-07-27 Unger Evan C Liposomes as contrast agents for ultrasonic imaging and methods for preparing the same
US5228446A (en) 1989-12-22 1993-07-20 Unger Evan C Gas filled liposomes and their use as ultrasonic contrast agents
DE4004430A1 (en) * 1990-02-09 1991-08-14 Schering Ag CONSTRUCTED POLYALDEHYDE CONSTITUENTS
US5445813A (en) 1992-11-02 1995-08-29 Bracco International B.V. Stable microbubble suspensions as enhancement agents for ultrasound echography
IN172208B (en) 1990-04-02 1993-05-01 Sint Sa
US5578292A (en) 1991-11-20 1996-11-26 Bracco International B.V. Long-lasting aqueous dispersions or suspensions of pressure-resistant gas-filled microvesicles and methods for the preparation thereof
US5556610A (en) 1992-01-24 1996-09-17 Bracco Research S.A. Gas mixtures useful as ultrasound contrast media, contrast agents containing the media and method
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5672585A (en) 1990-04-06 1997-09-30 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US5358702A (en) 1990-04-10 1994-10-25 Unger Evan C Methoxylated gel particle contrast media for improved diagnostic imaging
US5078994A (en) * 1990-04-12 1992-01-07 Eastman Kodak Company Microgel drug delivery system
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5137928A (en) 1990-04-26 1992-08-11 Hoechst Aktiengesellschaft Ultrasonic contrast agents, processes for their preparation and the use thereof as diagnostic and therapeutic agents
US5190982A (en) 1990-04-26 1993-03-02 Hoechst Aktiengesellschaft Ultrasonic contrast agents, processes for their preparation and the use thereof as diagnostic and therapeutic agents
US5205287A (en) 1990-04-26 1993-04-27 Hoechst Aktiengesellschaft Ultrasonic contrast agents, processes for their preparation and the use thereof as diagnostic and therapeutic agents
AU636481B2 (en) 1990-05-18 1993-04-29 Bracco International B.V. Polymeric gas or air filled microballoons usable as suspensions in liquid carriers for ultrasonic echography
US5196348A (en) 1990-06-11 1993-03-23 Air Products And Chemicals, Inc. Perfluoro-crown ethers in fluorine magnetic resonance spectroscopy of biopsied tissue
US5315997A (en) 1990-06-19 1994-05-31 Molecular Biosystems, Inc. Method of magnetic resonance imaging using diamagnetic contrast
US5215680A (en) 1990-07-10 1993-06-01 Cavitation-Control Technology, Inc. Method for the production of medical-grade lipid-coated microbubbles, paramagnetic labeling of such microbubbles and therapeutic uses of microbubbles
DE4028139A1 (en) 1990-09-05 1992-03-12 Hausmann Ag Labor USE OF THE COMPLEX RADIOACTIVE METALLIONS WITH ALL-CIS-1,3,5-TRIAMINO-2,4,6-CYCLOHEXANTRIOL AND ITS DERIVATIVES FOR X-RAY DIAGNOSTIC PURPOSES AND IN TUMOR THERAPY AND THE PRODUCTION OF MEDICINE AND FUTURE AND FUTURE AND FURNITURE AND FUTURE
IL95743A (en) * 1990-09-19 1993-02-21 Univ Ramot Method of measuring blood flow
AU635449B2 (en) 1990-10-05 1993-03-18 Bracco International B.V. Method for the preparation of stable suspensions of hollow gas-filled microspheres suitable for ultrasonic echography
US5487390A (en) 1990-10-05 1996-01-30 Massachusetts Institute Of Technology Gas-filled polymeric microbubbles for ultrasound imaging
US5107842A (en) 1991-02-22 1992-04-28 Molecular Biosystems, Inc. Method of ultrasound imaging of the gastrointestinal tract
CA2063529A1 (en) 1991-03-22 1992-09-23 Katsuro Tachibana Booster for therapy of diseases with ultrasound and pharmaceutical liquid composition containing the same
GB9106686D0 (en) 1991-03-28 1991-05-15 Hafslund Nycomed As Improvements in or relating to contrast agents
GB9106673D0 (en) 1991-03-28 1991-05-15 Hafslund Nycomed As Improvements in or relating to contrast agents
US5205290A (en) 1991-04-05 1993-04-27 Unger Evan C Low density microspheres and their use as contrast agents for computed tomography
US5496535A (en) 1991-04-12 1996-03-05 Alliance Pharmaceutical Corp. Fluorocarbon contrast media for use with MRI and radiographic imaging
US5147631A (en) 1991-04-30 1992-09-15 Du Pont Merck Pharmaceutical Company Porous inorganic ultrasound contrast agents
DK0586524T3 (en) 1991-06-03 1997-05-20 Nycomed Imaging As
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
MX9205298A (en) 1991-09-17 1993-05-01 Steven Carl Quay GASEOUS ULTRASOUND CONTRASTING MEDIA AND METHOD FOR SELECTING GASES TO BE USED AS ULTRASOUND CONTRASTING MEDIA
JP3231768B2 (en) 1991-09-17 2001-11-26 ソーナス ファーマシューティカルス,インコーポレイテッド Gaseous ultrasonic contrast agent and method for selecting gas to be used as ultrasonic contrast agent
US5409688A (en) 1991-09-17 1995-04-25 Sonus Pharmaceuticals, Inc. Gaseous ultrasound contrast media
US5362477A (en) 1991-10-25 1994-11-08 Mallinckrodt Medical, Inc. 19F magnetic resonance imaging agents which include a nitroxide moiety
US5196183A (en) 1991-12-04 1993-03-23 Sterling Winthrop Inc. Contrast agents for ultrasound imaging
GB9200388D0 (en) 1992-01-09 1992-02-26 Nycomed As Improvements in or relating to contrast agents
GB9200387D0 (en) 1992-01-09 1992-02-26 Nycomed As Improvements in or relating to contrast agents
IL104084A (en) 1992-01-24 1996-09-12 Bracco Int Bv Long-lasting aqueous suspensions of pressure-resistant gas-filled microvesicles their preparation and contrast agents consisting of them
WO1993015722A1 (en) 1992-02-07 1993-08-19 Syntex (Usa) Inc. Controlled delivery of pharmaceuticals from preformed porous microparticles
JP3325300B2 (en) 1992-02-28 2002-09-17 株式会社東芝 Ultrasound therapy equipment
US5247935A (en) 1992-03-19 1993-09-28 General Electric Company Magnetic resonance guided focussed ultrasound surgery
US5339814A (en) 1992-04-14 1994-08-23 Lasker Sigmund E Process for visualizing tissue metabolism using oxygen-17
US5846516A (en) 1992-06-03 1998-12-08 Alliance Pharmaceutial Corp. Perfluoroalkylated amphiphilic phosphorus compounds: preparation and biomedical applications
DE4221256C2 (en) 1992-06-26 1997-07-10 Lancaster Group Ag Galenic composition for topical use
US5334761A (en) 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5552155A (en) 1992-12-04 1996-09-03 The Liposome Company, Inc. Fusogenic lipsomes and methods for making and using same
US5558855A (en) 1993-01-25 1996-09-24 Sonus Pharmaceuticals Phase shift colloids as ultrasound contrast agents
CA2154590C (en) 1993-01-25 2001-06-12 Steven C. Quay Phase shift colloids as ultrasound contrast agents
FR2700952B1 (en) 1993-01-29 1995-03-17 Oreal New cosmetic or dermopharmaceutical compositions in the form of aqueous gels modified by the addition of expanded microspheres.
WO1994018954A1 (en) 1993-02-22 1994-09-01 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of biologics and compositions useful therefor
US5362478A (en) 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
US5716597A (en) 1993-06-04 1998-02-10 Molecular Biosystems, Inc. Emulsions as contrast agents and method of use
JP2905598B2 (en) 1993-07-02 1999-06-14 モレキュラー バイオシステムズ,インコーポレイテッド Method for producing encapsulated microspheres from heat denatured proteins
DE69434119T3 (en) 1993-07-30 2011-05-05 Imcor Pharmaceutical Co., San Diego STABILIZED MICROGAS BLOWER COMPOSITIONS FOR ECHOGRAPHY
US5433204A (en) 1993-11-16 1995-07-18 Camilla Olson Method of assessing placentation
IL113139A0 (en) 1994-03-28 1995-06-29 Nycomed Imaging As Diagnostic compositions comprising imaging agents encapsulated in liposomes
US5545396A (en) 1994-04-08 1996-08-13 The Research Foundation Of State University Of New York Magnetic resonance imaging using hyperpolarized noble gases
WO1995029705A1 (en) 1994-05-03 1995-11-09 Molecular Biosystems, Inc. Composition for ultrasonically quantitating myocardial perfusion
US5502094A (en) 1994-05-20 1996-03-26 Minnesota Mining And Manufacturing Company Physiologically acceptable emulsions containing perfluorocarbon ether hydrides and methods for use
US5562893A (en) 1994-08-02 1996-10-08 Molecular Biosystems, Inc. Gas-filled microspheres with fluorine-containing shells
US5540909A (en) 1994-09-28 1996-07-30 Alliance Pharmaceutical Corp. Harmonic ultrasound imaging with microbubbles
US5560364A (en) 1995-05-12 1996-10-01 The Board Of Regents Of The University Of Nebraska Suspended ultra-sound induced microbubble cavitation imaging
US5558092A (en) 1995-06-06 1996-09-24 Imarx Pharmaceutical Corp. Methods and apparatus for performing diagnostic and therapeutic ultrasound simultaneously
US5606973A (en) 1995-06-07 1997-03-04 Molecular Biosystems, Inc. Liquid core microdroplets for ultrasound imaging
WO1996040285A1 (en) * 1995-06-07 1996-12-19 Imarx Pharmaceutical Corp. Novel targeted compositions for diagnostic and therapeutic use

Also Published As

Publication number Publication date
AU4913896A (en) 1996-09-11
US5830430A (en) 1998-11-03
CN1082043C (en) 2002-04-03
EP0839125A4 (en) 2002-10-30
WO1996026179A1 (en) 1996-08-29
JPH11500727A (en) 1999-01-19
US6056938A (en) 2000-05-02
EP0839125A1 (en) 1998-05-06
CN1177342A (en) 1998-03-25

Similar Documents

Publication Publication Date Title
US6056938A (en) Cationic lipids and the use thereof
US5997898A (en) Stabilized compositions of fluorinated amphiphiles for methods of therapeutic delivery
CA2217494C (en) Novel compositions of lipids and stabilizing materials
US6815432B2 (en) Methods for encapsulating plasmids in lipid bilayers
US6110745A (en) Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
Shilpa et al. Niosomes as vesicular carriers for delivery of proteins and biologicals
US4971803A (en) Lamellar vesicles formed of cholesterol derivatives
EP1007002B1 (en) Lipid-containing compositions and uses thereof
JP2004511572A (en) Lipid formulations for targeted delivery
Lasic et al. Liposomes
Oku et al. Evaluation of drug targeting strategies and liposomal trafficking.
Sallam et al. Colloidal delivery of drugs: present strategies and conditions
US9962448B2 (en) Compounds and methods for targeted immune system delivery
JP6384863B2 (en) Lipid membrane structure
JPWO2005021012A1 (en) Gemcitabine encapsulated drug carrier
ES2698565B2 (en) Procedure for the elaboration of lipid nanoparticles, and lipid nanoparticles with brain macrophages as target cells
WO2023246218A1 (en) Ionizable lipid for nucleic acid delivery and composition thereof
US11833209B2 (en) Formulated and/or co-formulated liposome compositions containing PD-1 antagonist prodrugs useful in the treatment of cancer and methods thereof
Sadhu et al. Liposomes: As 12
JP2023510253A (en) Alkylated Nucleosides, and Their Compositions and Methods for Nucleic Acid Delivery
Bassous et al. Elastic Liposomes for Drug Delivery
WO2023183082A1 (en) Silyl lipids suitable for enhanced delivery of anti-viral therapeutics
Anggraeni et al. Liposome as carrier therapy for Alzheimer’s disease

Legal Events

Date Code Title Description
FZDE Discontinued