WO1996022765A1 - Stable lipid-comprising drug delivery complexes and methods for their production - Google Patents

Stable lipid-comprising drug delivery complexes and methods for their production Download PDF

Info

Publication number
WO1996022765A1
WO1996022765A1 PCT/US1996/000816 US9600816W WO9622765A1 WO 1996022765 A1 WO1996022765 A1 WO 1996022765A1 US 9600816 W US9600816 W US 9600816W WO 9622765 A1 WO9622765 A1 WO 9622765A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
complex
drug
complexes
dna
Prior art date
Application number
PCT/US1996/000816
Other languages
French (fr)
Inventor
Xiang Gao
Leaf Huang
Original Assignee
University Of Pittsburgh
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 University Of Pittsburgh filed Critical University Of Pittsburgh
Priority to AU47037/96A priority Critical patent/AU709773B2/en
Priority to DE69624801T priority patent/DE69624801T2/en
Priority to DK96902747T priority patent/DK0814777T3/en
Priority to JP52295496A priority patent/JP4074338B2/en
Priority to CA002211118A priority patent/CA2211118C/en
Priority to AT96902747T priority patent/ATE227563T1/en
Priority to EP96902747A priority patent/EP0814777B1/en
Publication of WO1996022765A1 publication Critical patent/WO1996022765A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Dispersion Chemistry (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Novel stable, concentrated, biologically active and ready-to-use lipid-comprising drug delivery complexes and methods for their production are described. The biological activity of the complexes produced are comparable to the formulations prepared according to the prior art admixture method and upon purification, the complexes produced by the method of this invention are 50 to 500 fold more concentrated than the components of the complexes formed by admixture. The method described herein provides for the large scale production of lipid-comprising drug delivery systems useful for gene therapy and other applications.

Description

Title of Invention o
Stable Lipid-Comprising Drug Delivery Complexes And Methods For Their Production
Field of Invention
The present invention relates to cationic lipids and their use as vehicles for the transfer of nucleic acids or other macromolecules such as proteins into cells. More specifically, this invention relates to lipid-comprising drug delivery complexes which are stable, biologically active, capable of being concentrated, and to methods for their production.
BACKGROUND OF INVENTION
The development of new forms of therapeutics which use macromolecules such as proteins or nucleic acids as therapeutic agents has created a need to develop new and effective means of delivering such macromolecules to their appropriate cellular targets. Therapeutics based on either the use of specific polypeptide growth factors or specific genes to replace or supplement absent or defective genes are examples of therapeutics which may require such new delivery systems. Clinical application of such therapies depends not only on efficacy of new delivery systems but also on their safety and on the ease with which the technologies underlying these systems can be adapted for large scale pharmaceutical production, storage, and distribution of the therapeutic formulations. Gene therapy has become an increasingly important mode of treating various genetic disorders. The potential for providing effective treatments, and even cures, has stimulated an intense effort to apply this technology to diseases for which there have been no effective treatments. Recent progress in this area has indicated that gene therapy may have a significant impact not only on the treatment of single gene disorders, but also on other more complex diseases such as cancer. However, a significant obstacle in the attainment of efficient gene therapy has been the difficulty of designing new and effective means of delivering therapeutic nucleic acids to cell targets. Thus, an ideal vehicle for the delivery of exogenous genes into cells and tissues should be highly efficient in nucleic acid delivery, safe to use, easy to produce in large quantity and have sufficient stability to be practicable as a pharmaceutical.
Non-viral vehicles, which are represented mainly by the cationic liposomes, are one type of vehicle which have, for the following reasons, been considered for use in gene therapy. First, the plasmid DNA required for liposome-mediated gene therapy can be widely and routinely prepared on a large scale and is simpler and carries less risk than the use of viral vectors such as retroviruses. Second, liposome-mediated gene delivery, unlike retroviral-mediated gene delivery, can deliver either RNA or DNA. Thus, DNA, RNA, or an oligonucleotide can be introduced directly into the cell. Moreover, cationic liposomes are non-toxic, non-immunogenic and can therefore be used repeatedly in vivo as evidenced by the successful in vivo delivery of genes to catheterized blood vessels
(Nabel, E.G., et al. (1990) Science. 249: 1285-1288), lung epithelial cells (Brigham, K.L., et al. (1989) Am. J. Respir. Cell Mol. Biol.. 195-200, Stribling, R. , et al. (1992) Proc. Natl. Acad. Sci. U.S.A.. 89: 11277-11281), and other systemic uses (Zhu, N. , et al. (1993) Science.
261: 209-211, Philip, R. , et al. (1993) Science. 261: 209- 211) of cationic liposomes.
Although a variety of cationic liposome formulations, including the commercially available cationic liposome reagent DOTMA/DOPE (N-l,-(2, 3 - dioleoyloxy) propyl-N,N,N-trimethyl ammonium chloride/dioleoyl phosphatidylethanolamine) , are known in the art (Feigner, P.L. et al. (1987) Proc. Natl. Acad. Sci. U.S.A.. 84: 7413-7417), a cationic liposome formulation designated DC-Chol/DOPE (30N- (N' ,N' - dimethylaminoethane) -carbamoyl cholesterol)/(dioleoyl phosphatidylethanolamine) ) has been shown in in vitro studies (Gao, X., and Huang, L. (1991) Biochem. Biophys. Res. Commun.. 179: 280-285) to be relatively non-toxic and more efficient than DOTMA/DOPE. Moreover, following extensive in vivo studies (Plautz, G.E., et al. (1993) Proc. Natl. Acad. Sci. U.S.A.. 90: 4645-4649, Stewart, M.J., et al. (1992) Hum. Gene Ther.. 3: 267-275) in which DC-Chol/DOPE was demonstrated to be both safe and efficacious as a nucleic acid delivery system, this formulation was approved by the U.S. Food and Drug Administration (FDA) and the U.K. Medicines Control Agency (MCA) , and has been used in two separate gene therapy clinical trials (Nabel, G.J., et al. (1993) Proc. Natl. Acad. Sci. U.S.A.. 90: 11307-11311, Caplen, N.J., et al. (1995) Nature Medicine. 1: 39-46).
However, the use of DC-Chol/DOPE and other currently existing cationic liposomes as vehicles for delivering nucleic acids to cellular targets are inconvenient for large scale therapeutic applications for a number of reasons. First, the ratios of liposome to nucleic acid utilized to form nucleic acid/liposome complex in the prior art admixture method results in the formation of complexes which are large in diameter and hence, relatively low in stability. Thus, none of the presently utilized cationic liposome formulations, including DC-Chol/DOPE, are designed as stable and ready-to-use pharmaceutical formulations of nucleic acid/liposome complex. This limitation of the admixture method requires that the user prepare complex prior to each use, an inconvenience which requires special training of personnel. In addition, the preparation of complex by admixture prior to each use introduces a possible source of dosage variability which hinders evaluation of treatments utilizing these complexes due to possible over- or under-dosing of the recipient.
Second, the prior art admixture method of preparing nucleic acid/cationic liposome complexes prior to each use requires that a dilute nucleic acid solution (less than 4 μg/ml) and a dilute liposome dispersion (less than 50 μM) be used to prepare the nucleic acid/liposome complex in order to reduce the chance of forming large and less active aggregates. This limitation makes it difficult to make small biologically active complexes without using less than optimal conditions, such as reducing the amount of liposomes (which causes reduced nucleic acid transfer activity) or increasing the amount of liposome (which causes enhanced toxicity) . Moreover, the fact that the complex must be made in dilute concentrations is a significant drawback to clinical applications, particularly in the case of intratumor injection of the complex, since only a small volume of the complex can be injected in each site (Nabel, G.J., et al. (1993) Proc. Natl. Acad. Sci. U.S.A.. 90: 11307-11311).
Accordingly, an object of this invention is to provide stable, biologically active, lipid-comprising drug delivery complexes which are capable of being concentrated as well as methods of producing such complexes.
Summary of Invention
This invention provides methods for producing lipid-comprising drug delivery complexes having a net positive charge and/or a positively charged surface. By "drug" as used throughout the specification and claims is meant any molecular entity, which is either monomeric or oligomeric, and which, when complexed with lipid or with lipid and polycation, is being administered to an individual for the purpose of providing a therapeutic effect to the recipient. Thus, macromolecules having an overall net negative charge or regions of negativity would be expected to be capable of forming the delivery complexes of this invention. Macromolecules which are particularly suitable for use with the complexes of this invention are for example, DNA, RNA, oligonucleotides or negatively charged proteins. However, macromolecules having a positive charge (eg large cationic protein would also be expected to be capable of forming the complexes of this invention by sequentially complexing the cationic macromolecule with anionic lipid or polymer and then with cationic lipid.
The complexes of the present invention comprise a drug/lipid complex formed by mixing the drug to be delivered with cationic liposomes in a drug to lipid ratio such that the drug/lipid complex formed has a net positive charge and a drug/lipid/polycation complex formed by mixing drug with cationic liposomes and polycation in a drug to lipid to polycation ratio such that the drug/lipid/polycation complex formed has a net positive charge. By "net positive charge" as applied to the drug/lipid complex is meant a positive charge excess of lipid to drug. By "net positive charge" as applied to the drug/lipid/polycation complex is meant that the positive charges of the cationic lipid and the polycation exceed the negative charge of the drug. However, it is be understood that the present invention also encompasses drug/lipid and drug/lipid/polycation complexes having a positively charged surface irrespective of whether the net charge of the complex is positive, neutral or even negative. A positively charged surface of a complex may be measured by the migration of the complex in an electric field by methods known to those in the art such as by measuring zeta potential (Martin, A., Swarick, J., and Cammarata, A., Physical Pharmacy & Physical Chemical Principles in the Pharmaceutical Sciences, 3rd ed. Lea and Febiger, Philadelphia, 1983) , or by the binding affinity of the complex to cell surfaces. Complexes exhibiting a positively charged cell surface have a greater binding affinity to cell surfaces than complexes having a neutral or negatively charged surface.
The invention therefore relates to methods for producing these drug/lipid and drug/lipid/polycation complexes comprising mixing the drug to be delivered with cationic liposomes, and optionally polycation, in a ratio such that the complex formed has a net positive charge and/or a positively charged surface.
In another embodiment of this invention, the methods for producing drug lipid or drug/lipid/polycation complexes may further comprise the step of purifying said complexes from excess free components (drug, lipid, polycation) following their production.
The drug/lipid and drug/lipid/polycation complexes of this invention are generally stable, capable of being produced at relatively high concentration, and retain biological activity over time in storage. Such complexes are of utility in the delivery of nucleic acids, proteins and other macromolecules to cells and tissues.
DESCRIPTION OF FIGURES
Fig. 1 shows a typical size distribution (mean diameter) of nucleic acid/liposome complexes prepared as an admixture from DC-Chol/DOPE (3:2) liposomes and pRSVL plasmid DNA (2 μg) at the indicated lipid to DNA ratios. Figures 2A and 2B show the distribution of the liposome marker 3H-cholesteryl hexadecyl ether (O) and the 3P-DNA marker (■) among sucrose gradient fractions. The location of each fraction in the sucrose gradients of both Figures 2A and 2B is indicated at the top of Figure 2A.
Figure 2A shows the distribution of the 3H and 32P markers following ultracentrifugation of free liposomes (10 μmoles of DC-Chol/DOPE (2:3) in 2 ml volume) or free DNA (50 μg pRSVL DNA in a 2 ml volume) through a sucrose density gradient. Figure 2B shows the distribution of the 3H and 32P markers following ultracentrifugation of the DNA-lipid complex (formed via mixing of 20 μmoles DC- Chol/DOPE (2:3) liposomes and 0.4 mg pRSVL DNA in 2 ml volume) through a sucrose density gradient.
Figure 3 shows the transfection activities in CHO cells of admixture DNA/liposome complex (O) , admixture DNA/liposome/poly-L-lysine(PLL) complex (D) DNA/lipid complex (•) and DNA/lipid/PLL complex (■) . The DC- Chol/DOPE liposomes used to form the above complexes contained varying mole% of DC-Choi as indicated at the bottom of Figure 3. The DNA/lipid (•) and DNA/lipid/PLL (■) complexes were purified on a sucrose density gradient prior to being assayed for transfection activity. Transfection activity is indicated on the vertical axis as relative light units of luciferase activity.
Figure 4 shows the transfection activities of admixture DNA/liposome complex (O) and admixture DNA/liposome/PLL complex (D) compared to the transfection activities of DNA/lipid (•) and DNA/lipid/PLL complexes stored at 4°C for 130 days following their purification on a sucrose density gradient. The DC-Chol/DOPE liposomes used to form the above complexes contained varying mole% of DC-Choi as indicated at the bottom of Figure 4. Transfection activity is indicated on the vertical axis as relative light units of luciferase activity. Figure 5 shows the concentration of extractable protein from CHO cells, 36 hours after the cells were treated with admixture DNA/liposome complex (O) ; admixture DNA/liposome/PLL complex (D) ; DNA/lipid complex (•) ; or DNA/lipid/PLL complex (■) . The DNA/lipid and DNA/lipid/PLL complexes were purified on a sucrose density gradient prior to being assayed for transfection activity. The DC- Chol/DOPE liposomes used to form the above complexes contained varying mole% of DC-Choi as indicated at the bottom of Figure 5.
Figure 6 shows the results of CAT assays of tumor extracts prepared from mice having ovarian tumors. 2xl06 human ovarian carcinoma cells were subcutaneously injected into SCID mice at day 0. On day 14, 100 μl solutions containing pUCCMVCAT DNA (contains the chloramphenicol acetyl transferase gene of IjL. coli ) 30 μg) complexed with DC-Choi liposomes (30 nmoles) in the form of admixture (lanes 1 and 2; duplicate samples) or the same amount of DNA in the form of purified complex (prepared from DNA:DC-chol liposome at ratio lμg/25 nmoles, lanes 3 and 4; duplicate samples) were directly injected into tumors. 48 hours following transfection, the mice were sacrificed and tumor extract containing 100 μg protein was assayed for CAT activity. Lane 5 shows positive control CAT activity for standard E . coli CAT.
Figures 7A - 7C show the transfection activities of admixture DNA/lipid complex and purified and unpurified DNA/lipid/PLL complexes in 293 cells (Fig. 7A) C3 cells (Fig. 7B) and BL6 cells (Fig. 7C) . Transfection activity is indicated on the vertical axis of Figures 7A-7C as relative light units of luciferase activity.
Description of Invention
This invention relates to lipid-comprising drug delivery complexes having a net positive charge and/or a positively charged surface at pH 6.0-8.0. These complexes comprise cationic lipids, drugs, and optionally further comprise polycations. The invention further relates to a method for producing these complexes where the method may optionally include the step of purifying these formulations from excess individual components. For the production of the drug/lipid complexes of this invention, inclusion of the purification step is a preferred embodiment. It should be understood that where the purification step is applied to the drug/lipid/polycation complexes, the recovery of these complexes in a pure state free from excess components following purification is lower than the recovery of drug/lipid complexes following their purification since the peak containing the drug/lipid/polycation complex following sucrose purification via density centrifugation is broader than the peak containing drug/lipid complexes and hence, overlaps with the peaks of the free components.
The lipid-comprising drug delivery complexes of this invention are stable, capable of being produced at relatively high concentrations, and retain biological activity of the drug component over time in storage. The method of producing these complexes is based on a binding model between two oppositely charged polymers (e.g. negatively charged nucleic acid and positively charged lipids) in which the formation of large unstable aggregates is avoided by neutralizing the negative charge of the drug via the use of an excess amount of positive charge in the form of cationic liposomes or cationic liposomes and polycation. The complexes of this invention have been observed to retain their initial diameter and bioactivity over 4 months in storage in 10% sucrose buffer.
The "drug" which is contained in the lipid- comprising drug delivery complexes of the present invention may be nucleic acids, polyanionic proteins, polysaccharides and other macromolecules which can be complexed directly with cationic lipids. However, cationic drugs (eg large cationic protein) can be directly complexed with an anionic lipid or sequentially complexed first with anionic lipid or polymer followed by cationic lipid. The use of this process permits delivery of positive or neutral charged drug to cells by the complexes of the present invention.
To produce drug/lipid and drug/lipid/polycation complexes with a net positive charge, the positive charge excess of lipid to drug or of lipid and polycation to drug may be up to about a 30-fold positive charge excess in the complex of total lipids to drug or of lipid and polycation to drug, preferably about a 2 to 10-fold charge excess and most preferably about a 2 to 6-fold charge excess. Complexes which posses a positive charge on their surface may have similar preferred ranges of surface charge excess to drug. To produce a nucleic acid/lipid complex having a positive charge excess of lipid to nucleic acid, mole amounts of cationic liposomal lipid to be mixed with l μg of nucleic acid to produce a nucleic acid/lipid complex which has positive charge excess of lipid to nucleic acid at pH 6.0-8.0 may range from about 0.1 nmol to about 200 n ol of lipid, preferably about 5 nmol to about 100 nmol lipid, depending on the positive charge content of the cationic liposome. Of course, if the drug were a protein, the amount of lipid to be mixed with 1 μg of negatively charged protein would be at least 10- fold less than the amount of lipid to be mixed with 1 μg of DNA as shown above since proteins are less charge dense than nucleic acids. Those of ordinary skill in the art would readily understand that depending upon the positive charge content of the cationic liposomes, different mole amounts of different cationic liposomes would have to be mixed with an equivalent amount of drug to produce a positive charge excess of lipid to drug.
When a drug/lipid/polycation complex having a net positive charge and/or a positively charged surface is to be produced, the inclusion of the polycation reduces the amount of lipid which must be mixed with drug to the extent that the positive charge from the lipid may be less than the negative charge from the drug. This reduction in the amount of lipid reduces the toxicity of the polycation-containing formulations. Mole amounts of cationic liposomes to be used in formulating nucleic acid/lipid/polycation complexes may range from about 0.1 nmol to about 200 nmol lipid per 1 μg nucleic acid, more preferably from about 1 to about 25 nmoles lipid per l μg nucleic acid depending on the positive charge content of the cationic liposomes. It is to be generally understood that in producing the nucleic acid/lipid and nucleic acid/lipid/polycation complexes of the present invention, the mole amount of liposomes required to produce these complexes will increase as the concentration of nucleic acid mixed with the liposomes is increased.
Those of ordinary skill in the art would readily understand that when the complexes of the present invention are purified, the positive charge excess of cationic liposomes to drug or of cationic liposomes and polycation to drug immediately prior to mixing will be greater than the positive charge excess in the purified complexes of lipid to drug or of lipid and polycation since the purification step results in the removal of excess free lipids and/or free polycation.
In order to illustrate how the charges attributed to cationic lipid, drug and polycation may be determined at pH 6.0-8.0 the following example is provided. Assuming the drug to be delivered is DNA, one determines the negative charge of the DNA to be delivered by dividing the amount of DNA to be mixed, or the amount of DNA in the complex, by 330, the molecular weight of a single nucleotide where one nucleotide equals one negative charge. Thus, the negative charge for 1 μg of DNA is 3.3 nmols.
For 10 nmol of DC-Chol/DOPE (2:3) liposomes one calculates the effective charge of the lipid by multiplying the amount of total liposomal lipid (10 nmol) by 0.4 (40% of the total liposomal lipid is the cationic lipid DC-Choi) to yield 4 nmol DC-Choi lipid in the liposomes. Since at pH 6-8, one molecule of DC-Choi has one positive charge, the effective positive charge of liposomal lipid at the time of mixing, or in the complex, is 4.0 nmol. Of course, those of skill in the art would readily understand that other cationic lipids may have a lesser or greater amount of positive charge per molecule of cationic lipid at pH 6-8.0 than DC-Choi.
Assuming the polycation to be mixed to form the complex is a bromine salt of poly-L-lysine (PLL) , the positive charge of PLL at the time of mixing is obtained by dividing the amount of PLL to be mixed by 207, the molecular weight of one lysyl residue where one lysyl residue equals one positive charge. Thus, the positive charge for 1 μg of PLL is approximately 5.0 nmols. To calculate the positive charge contributed by lysyl residues in a formed complex, the amount of lysine present in the complex is divided by the molecular weight of one lysyl residue taking into account the weight of a counterion, if present. Application of the above calculations to data presented in Table 1 herein (see Example 3) illustrates how a positive to negative charge ratio can be calculated both at the time of mixing of DNA and liposome and, after purification of the complex produced by the mixing of DNA and liposome. In Table 1 of Example 3, 0.4 mg of DNA is mixed with 20 μmols of cationic DC-Chol/DOPE liposomes to produce DNA/lipid complex. For cationic liposomes having a DC-Chol/DOPE ratio of 4:6, the positive charge content of the liposomal lipid is calculated to be 8000 nmol and the negative charge content of the 0.4 mg DNA to be mixed with liposomes is calculated to be 1320 nmols based on the sample calculations presented in the above paragraphs. Therefore, the positive to negative charge ratio at the time of mixing is 6.06 (8000 divided by 1320) . However, after the complex was purified, the lipid to DNA ratio of this purified complex was 12.7 nmol lipid/μg DNA as shown in Table 1 (see the "4:6 row"). This 12.7 ratio translates to a positive to negative charge ratio of 1.5 thus showing that purification removed excess positive charge of free liposomes.
Also in Table 1, where DNA/lipid/PLL complex was prepared by mixing 4 μmol of liposomes (4:6 DC-Chol/DOPE) and 1 mg PLL with 0.4 mg DNA, one can calculate the positive to negative charge ratio at the time of mixing as follows. Based on the sample calculations presented in the above paragraphs, the 4 μmol liposomal lipid contributes 1600 nmol of positive charge, the 1 mg of PLL contributes 5000 nmol of positive charge and the 0.4 mg DNA contributes 1,320 nmol of negative charge. Thus, the positive to negative charge ratio at the time of mixing liposomes, PLL and DNA is 5 (1600 + 5000) .
1320
It is further to be understood by those skilled in the art that the net charge of the complex may be determined by measuring the amount of DNA, lipid and when present, polycation in the complex by the use of an appropriate analytical technique such as the use of radioisotopic labelling of each component or by elemental analysis. Once the amounts of each component (DNA, lipid and when present, polycation) in a complex at a given pH are known, one could then calculate the approximate net charge of that complex at the given pH taking into account the pK's of the components which may be known or determined analytically.
In a preferred embodiment, the drug is a nucleic acid sequence, preferably a nucleic acid sequence encoding a gene product having therapeutic utility.
In one embodiment of the invention, a method for producing nucleic acid/lipid complexes having a net positive charge and/or positively charged surface at pH 6- 8.0, comprises, combining nucleic acids with cationic liposomes in a nucleic acid to lipid ratio such that the nucleic acid/lipid complex formed has a positive charge excess of lipid to nucleic acid.
In an alternative embodiment, nucleic acid and cationic liposome may be mixed with a polycation in a nucleic acid to lipid to polycation ratio such that the nucleic acid/lipid/polycation complexes formed have a positive charge excess of lipid and polycation to nucleic acid at pH 6-8.
In a preferred embodiment, the nucleic acid/lipid and nucleic acid/lipid/polycation complexes are produced by slowly adding nucleic acid to the solution of liposome or liposome plus polycation and mixing with a stirring bar where the mixing is allowed to proceed second. Alternatively, the liposome or liposome/ polycation mix can be added into a single chamber from a first inlet at the same time the nucleic acid is added to the chamber through a second inlet. The components are then simultaneously mixed by mechanical means in a common chamber.
The cationic liposomes mixed with drug or with drug and polycation to form the complexes of the present invention may contain a cationic lipid alone or a cationic lipid in combination with a neutral phospholipid. Suitable cationic lipid species include, but are not limited to: 1, 2 bis(oleoyloxy) -3- (trimethylammonio) propane (DOTAP) ; N-l, - (2,3-dioleoyloxy) propyl -N, N, N- trimethyl ammonium chloride (DOTMA) or other N- (N, N-l- dialkoxy) -alklyl-N, N, N-trisubstituted ammonium surfactants; 1, 2 dioleoyl-3- (4' -trimethylammonio) butanoyl-sn-glycerol (DOBT) or cholesteryl (4' trimethylammonia) butanoate (ChOTB) where the trimethylammonium group is connected via a butanoyl spacer arm to either the double chain (for DOTB) or cholesteryl group (for ChOTB); DORI (DL-1, 2-dioleoyl-3- dimethylaminopropyl-B-hydroxyethylammonium) or DORIE (DL- 1, 2-0-dioleoyl-3-dimethylaminopropyl-jS- hydroxyethylammonium) (DORIE) or analogs thereof as disclosed in WO 93/03709; 1, 2-dioleoyl-3-succinyl-sn- glycerol choline ester (DOSC) ; cholesteryl hemisuccinate ester (ChOSC) ; lipopolyamines such as doctadecylamidoglycylspermine (DOGS) and dipalmitoyl phosphatidyesthanolamidospermine (DPPES) or the cationic lipids disclosed in US Patent Number 5,283,185, cholesteryl-3|S-carboxyl-amido-ethylenetrimethylammonium iodide, 1-dimethylamino-3-trimethylammonio-DL-2-propyl- cholesteryl carboxylate iodide, cholesteryl-3/3- carboxyamidoethyleneamine, cholesteryl-33-oxysuccinamido- ethylenetrimethylammonium iodide, 1-dimethylamino-3- trimethylammonio-DL-2-propyl-cholesteryl-3/5-oxysuccinate iodide, 2- (2-trimethylammonio) -ethylmethylamino ethyl- cholesteryl-3jS-oxysuccinate iodide, 33N- (N' , N' - dimethylaminoethane) carbamoyl cholesterol (DC-chol) , and 3/S-N- (polyethyleneimine) -carbamoylcholesterol.
Examples of preferred cationic lipids include choles eryl-3/3-carboxyamidoethylenetri-methylammonium iodide, 1-dimethylamino-3-trimethylammonio-DL-2-propyl- cholesteryl carboxylate iodide, cholesteryl - 3β- carboxyamidoethyleneamine, cholesteryl-3/3-oxysuccin- amidoethylenetrimethylammonium iodide, 1-dimethylamino-3- trimethylammonio-DL-2-propyl-cholesteryl-3/3-oxysuccinate iodide, 2- (2-trimethylammonio)ethylmethylamino ethyl- cholesteryl-3/3-oxysuccinateiodide, 3/3N- (N' , N'dimethyl- a inoethane) -carbamoyl-cholesterol (DC-chol) , and 3/3N- (polyethyleneimine) -carbamoyl cholesterol.
Since an attribute of the complexes of the invention is their stability during storage (i.e., their ability to maintain a small diameter and retain biological activity over time following their formation) ; it will be understood by those of ordinary skill in the art that preferred cationic lipids are those lipids in which bonds between the lipophilic group and the amino group are stable in aqueous solution. While such bonds found in cationic lipids include amide bonds, ester bonds, ether bonds and carbamoyl bonds, preferred cationic lipids are those having a carbamoyl bond. An example of a preferred cationic lipid having a carbamoyl bond is DC-Choi. Those of skill in the art would readily understand that liposomes containing more than one cationic lipid species may be used to produce the complexes of the present invention. For example, liposomes comprising two cationic lipid species, lysyl-phosphatidylethanolamine and jβ-alanyl cholesterol ester have been disclosed (Brunette, E. et al. (1992) Nucl. Acids Res.. 20:1151).
It is to be further understood that in considering cationic liposomes suitable for use in mixing with drug and optionally with polycation, to form the complexes of this invention, the methods of the invention are not restricted only to the use of the lipids recited above but rather, any lipid composition may be used so long as a cationic liposome is produced.
Thus, in addition to cationic lipids, cationic liposomes used to form the complexes of the invention may contain other lipids in addition to the cationic lipids. These lipids include, but are not limited to, lyso lipids of which lysophosphatidylcholine (1- oleoyllysophosphatidycholine) is an example, cholesterol, or neutral phospholipids including dioleoyl phosphatidyl ethanolamine (DOPE) or dioleoyl phosphatidylcholine (DOPC) . The lipid complexes of the invention may also contain negatively charged lipids as well as cationic lipids so long as the net charge of the complexes formed is positive and/or the surface of the complex is positively charged. Negatively charged lipids of the invention are those comprising at least one lipid species having a net negative charge at or near physiological pH or combinations of these. Suitable negatively charged lipid species comprise phosphatidyl glycerol and phosphatidic acid or a similar phospholipid analog.
It is further contemplated that in the cationic liposomes utilized to form the complexes of the invention, the ratio of lipids may be varied to include a majority of cationic lipids in combination with cholesterol or with mixtures of lyso or neutral lipids. When the cationic lipid of choice is to be combined with another lipid, a preferred lipid is a neutral phospholipid, most preferably DOPE.
Methods for producing the liposomes to be used in the production of the lipid-comprising drug delivery complexes of the present invention are known to those of ordinary skill in the art. A review of methodologies of liposome preparation may be found in Liposome Technology (CFC Press NY 1984) ; Liposomes by Ortro (Marcel Schher, 1987); Methods Biochem Anol. 33:337-462 (1988) and U.S. Patent 5,283,185. Such methods include freeze-thaw extrusion and sonication. Both unilamellar liposomes (less than about 200 nm in average diameter) and multilamellar liposomes (greater than about 300 nm in average diameter) may be used as starting components to produce the complexes of this invention.
In the cationic liposomes utilized to produce the drug/lipid complexes of this invention, the cationic lipid is present in the liposome at from about 10 to about 100 mole% of total liposomal lipid, preferably from about 20 to about 80 mole% and most preferably about 20 to about 60 mole%. The neutral lipid, when included in the liposome, may be present at a concentration of from about 0 to about 90 mole% of the total liposomal lipid, preferably from about 20 to about 80 mole%, and most preferably from 40 to 80 mole%. The negatively charged lipid, when included in the liposome, may be present at a concentration ranging from about 0 mole% to about 49 mole% of the total liposomal lipid, preferably from about 0 mole% to about 40 mole%. In a preferred embodiment, the liposomes contain a cationic and a neutral lipid, most preferably DC-Choi and DOPE in ratios between about 2:8 to about 6:4. It is further understood that the complexes of the present invention may contain modified lipids, protein, polycations or receptor ligands which function as a targeting factor directing the complex to a particular tissue or cell type. Examples of targeting factors include, but are not limited to, asialoglycoprotein, insulin, low density lipoprotein (LDL) , folate and monoclonal and polyclonal antibodies directed against cell surface molecules. Potential targets include, but are not limited to, liver, blood cells, endothelial cells and tumor cells.
It is to be further understood that the positive charge of the complexes of this invention may be affected not only by the lipid composition of the complex but also by the pH of the solution in which the drug/lipid complexes are formed. For example, increasing pH (more basic) will gradually neutralize the positive charge of the tertiary amine of the cationic lipid DC-Choi. In a preferred embodiment, the complexes of the present invention are produced, and stored, at a pH such that the complexes have a net positive charge and/or positively charged surface. A preferred pH range is pH 6.0 - 8.0, most preferably pH 7.0 - 7.8.
When a polycation is to be mixed with nucleic acid and cationic liposomes, the polycation may be selected from organic polycations having a molecular weight of between about 300 and about 200,000. These polycations also preferably have a valence of between about 3 and about 1000 at pH 7.0. The polycations may be natural or synthetic amino acids, peptides, proteins, polyamines, carbohydrates and any synthetic cationic polymers. Nonlimiting examples of polycations include polyarginine, polyornithine, protamines and polylysine, polybrene (hexadimethrine bromide) , histone, cationic dendrimer, spermine, spermidine and synthetic polypeptides derived from SV40 large T antigen which has excess positive charges and represents a nuclear localization signal. A preferred polycation is poly-L-lysine (PLL). In producing nucleic acid/lipid/polycation complexes of the present invention, the ratio of polycation to nucleic acid is kept fixed while varying the amount of liposome. However, those of skill in the art would recognize that the ratio of polycation to nucleic acid will be affected by the charge density of the liposome to be mixed with the nucleic acid and polycation. For example, if the charge density of liposomes is decreased as a result of changes in the lipid composition of the liposome (e^ decreasing the ratio of cationic lipid: neutral lipid in the liposome) , the amount of polycation to be mixed with nucleic acid and liposome may be increased to compensate for the decrease in positive charge contributed by the liposomes. However, when polycation is utilized, it is preferred to use subsaturating amounts of polycation (ie amounts which will not saturate all the negative charge of the nucleic acid) in order to allow the cationic lipids to complex with the nucleic acid. Thus, in a preferred embodiment of the invention, a positive charge excess of lipid to nucleic acid is used even when polycation is mixed with lipid and nucleic acid. Amounts of polycation which may be mixed with l μg of nucleic acid and varying amounts of cationic liposomes in the present invention range from about 0.01 μg to about 100 μg of polycation per μg of nucleic acid, preferably from about 0.1 μg to about 10 μg of polycation per μg of nucleic acid.
Where purification of nucleic acid/lipid and nucleic acid/lipid/polycation complexes from excess free DNA, free liposomes and free polycation is desired, purification may be accomplished by centrifugation through a sucrose density gradient or other media which is suitable to form a density gradient. However, it is understood that other methods of purification such as chromatography, filtration, phase partition, precipitation or absorption may also be utilized. In a preferred method, purification via centrifugation through a sucrose density gradient is utilized. The sucrose gradient may range from about 0% sucrose to about 60% sucrose, preferably from about 5% sucrose to about 30% sucrose. The buffer in which the sucrose gradient is made can be any aqueous buffer suitable for storage of the fraction containing the complexes and preferably, a buffer suitable for administration of the complex to cells and tissues. A preferred buffer is pH 7.0-8.0 Hepes.
It is understood that in the present invention, preferred nucleic acid sequences are those capable of directing protein expression. Such sequences may be inserted by routine methodology into plasmid expression vectors known to those of skill in the art prior to mixing with cationic liposomes or liposomes and polycation to form the lipid-comprising drug delivery complexes of the present invention. The amount of nucleic acid mixed together with cationic liposomes or with cationic liposomes and polycation may range from about 0.01 μg to about 10 mg, preferably from about 0.1 μg to about 1.0 mg. It is understood that where the nucleic acid of interest is contained in plasmid expression vectors, the amount of nucleic acid recited above refers to the plasmid containing the nucleic acid of interest.
The purification of the nucleic acid/lipid and nucleic acid/lipid/polycation complexes of the present invention serves to concentrate the nucleic acids and lipids contained in the resultant complexes from about 50 -fold to about 500 -fold such that the lipid content contained in the complexes may be as high as about 40 μmol/ml and the nucleic acid content may be as high as about 2 mg/ml.
The diameter of the complexes produced by the methods of the present invention is less than about 400 nm, preferably less than about 200 nm, and more preferably less than 150 nm.
The complexes formed by the methods of the present invention are stable for up to about one year when stored at 4° C. The complexes may be stored in 10% sucrose solution upon collection from the sucrose gradient or they may be lyophilized and then reconstituted in 10% sucrose solution prior to use. In a preferred embodiment, the complexes are stored in solution. The stability of the complexes of the present invention is measured by specific assays to determine the physical stability and biological activity of the complexes over time in storage. The physical stability of the complexes is measured by determining the diameter of the complexes by methods known to those of ordinary skill in the art, including for example, electron microscopy, gel filtration chromatography or by means of quasi-elastic light scattering using a Coulter N4SD particle sizes as described in the Examples. The physical stability of the complex is "substantially unchanged" over storage when the diameter of the stored complexes is not increased by more than 100%, preferably by not more than 50%, and most preferably by not more than 30%, over the diameter of the complexes as determined at the time the complexes were purified.
Assays utilized in determining the biological activity of the complexes vary depending on what drug is contained in the complexes. For example, if the drug is nucleic acid encoding a gene product, the biological activity can be determined by treating cells in vitro under transfection conditions utilized by those of ordinary skill in the art for the transfection of cells with admixtures of DNA and cationic liposomes. Cells which may be transfected by the complexes includes those cells which may be transfected by admixture DNA/liposome complexes. The activity of the stored complexes is then compared to the transfection activity of complexes prepared by admixture. If the drug is a protein, then activity may be determined by a bioassay suitable for that protein.
It is further understood by those of skill in the art that the complexes of the present invention may be used iri vivo as vectors in gene therapy.
Therapeutic formulations using the complexes of the invention preferably comprise the complexes in a physiologically compatible buffer such as, for example, phosphate buffered saline, isotonic saline or low ionic strength buffer such as 10% sucrose in H20 (pH 7.4-7.6) or in Hepes (pH 7-8, a more preferred pH being 7.4-7.6) . The complexes may be administered as aerosols or as liquid solutions for intratumor, intravenous, intratracheal, intraperitoneal, and intramuscular administration.
Any articles or patent referenced herein are incorporated by reference. The following examples illustrate various aspects of the invention but are intended in no way to limit the scope thereof.
EXAMPLES
Materials
DOPE was purchased from Avanti Polar Lipid, Inc. (Alabaster, AL) . pRSVL, a plasmid which encodes the luciferase gene under the control of Rous sarcoma virus long terminal repeat, (De Wet, J.R. et al. (1987) Mol. Cell. Biol.. 7: 725-737) was amplified in E. coli and purified using the standard CsCl-EtBr ultracentrifugation method (Sambrook, J. , Fritsch, E.F. and Maniatis, T., Molecular Cloning: A Laboratory Manual (2d ed) Cold Spring Harbor Laboratory Press: New York (1989)). All the tissue culture media were obtained from Gibco BRL (Gaithersburg, MD) . Human embryonic kidney 293 cells, CHO (Chinese Hamster Ovary cells) , BL6 and BHK (Baby Hamster Kidney cells) cells were from American Type Culture Collection (Rockville, MD) . Mouse lung cells (MLC) are primary culture cells originally derived from the lung of a Balb/c mouse by Dr. S. Kenned (Oak Ridge National Laboratory, TN) . 293, BL6, BHK and MLC cells were cultured with DMEM media, CHO cells were cultured with F12 media, and C3 Hela cells were cultured in RPMI-1640 medium. All media was supplemented with 10% fetal bovine serum (Hyclone
Laboratories, Inc., Logan, UT) and 100 unit/ml penicillin and 100 μg/ml streptomycin. Poly-L-lysine hydrobromide (MW 3000 and MW 25,600) and other chemicals were from Sigma (St Louis, MI) . DC-Chol was synthesized according to the method of Gao and Huang (1991) (Gao, X., and Huang, L. (1991) Biochem. Biophys. Res. Commun.. 179: 280-285) with modifications in the purification steps as follows: after the reaction, 10 ml hexane was added and the mixture was extracted three times with 10 ml water. The organic phase was collected and dried under vacuum at 4°C. The resulting solid was dissolved in a minimal amount of absolute ethanol with heat, and recrystallized in acetonitrile at 0 'C. The purity of DC-Choi was at least > 95%, as analyzed by TLC method and Η-NMR and the yield of about 70% was a significant improvement over that of the previously reported method of Gao, X., and Huang, L. ((1991) Biochem. Biophys. Res. Commun.. 179: 280-285).
Methods
Preparation and purification of complexes
Cationic liposomes at a 20 mM total lipid concentration were prepared from DC-Choi and DOPE at various ratios by a sonication method, according to a published procedure (Gao, X., and Huang, L. (1991) Biochem. Biophys. Res. Commun.. 179: 280-285). Trace amount of 3H cholesteryl hexadecyl ether (Amersham, Arlington Heights, IL) was included for quantitation purpose. The size of these liposomes was between 100 to 150 nm in diameter, as determined by quasi-elastic light scattering using a Coulter N4SD particle sizer (Coulter Electronics, Inc., Hialeah, FL) . Unless indicated otherwise in the following Examples, DNA/lipid complexes were prepared at a typical laboratory scale by adding amounts of free DC-Chol/DOPE liposomes as indicated in each Example in a volume of 1 ml of a 2 mM Hepes buffer (pH 7.6) to a 15 x 7.5 polystyrene culture tube (Baxter, McGraw Pare, ID , a micro-magnetic stirrer was placed in the tube, and the solution was well mixed. Amounts of pRSVL DNA as indicated in each Example were then added dropwise from a stock solution (0.2 mg/ml, in 2 mM Hepes buffer, pH 7.6) to the liposome solution over a period of 3 min. Trace amounts of. pRSVL labeled with 32P using a nick translation kit (Promega, Madison, WI) and 32P dCTP (Amersham, Arlington Heights, IL) was included for the purpose of quantitation.
To prepare purified lipid/PLL/DNA complexes, an amount of the above 0.2 mg/ml DNA solution as indicated in each Example was added to 1 ml PLL/liposome mixture containing amounts of liposomes and PLL as indicated in each Example. DNA/lipid complexes were loaded on the top of a sucrose step gradient composed of 0.5 ml each of 5%, 7.5%, 10% and 15% sucrose (w/w) and DNA/lipid/PLL complexes were loaded on top of a sucrose step gradient composed of 0.5 ml each of 5%, 10%, 15%, 20%, 25% and 30% sucrose (w/w) . The DNA/lipid and DNA/lipid/PLL complexes were then purified from free lipid and PLL by ultracentrifugation at 100,000 g for 30 min at 4° C. After centrifugation, fractions of 200 μl were taken from the top to the bottom of the tube. Aliquots from each fraction were assayed for both 3H and 32P radioactivity using a scintillation counter. Fractions that contained peak value of the 32P were collected and pooled. These pooled fractions were then assayed for particle size and for transfection activity.
In vitro Transfection assay
The biological activity of the above complexes were assayed by in vitro transfection of cells in the Examples as follows. Briefly, cells grown in 48 well plate were incubated with DNA/lipid complex diluted in 0.5 ml CHO-S-SFM (Gibco BRL) or with admixture DNA/liposome complex prepared according to Gao and Huang (1991) (Gao, X., and Huang, L. (1991) Biochem. Biophys. Res. Commun.. 179: 280-285). For transfection of pRSVL DNA using DC-Choi liposomes in the presence of PLL, liposomes were first mixed with PLL, then complexed with DNA. All transfections were performed for 4 hours at 37°C. After transfection, cells were further cultured for 36 hours in the appropriate media containing 10% fetal bovine serum. Cells were then washed with PBS and lysed with 100 μl of IX lysis buffer provided by a luciferase assay kit (Promega, Madison, WI) . A 4 μl sample of the lysate was assayed for luciferase activity using 100 μl substrate solution from the reconstituted luciferase assay kit and an AutoLumat LB953 luminometer (Berthold, Germany) . Protein concentration from each lysate was assayed by a Coomassie blue dye method according to the manufacturer's protocol (Pierce, Rockford, IL) .
EXAMPLE 1
The Size Of The DNA/Lipid Complex Is Determined By The Ratio Of DNA To Lipid
This experiment was conducted to show that the size of the DNA-lipid complex formed by admixture changed as the ratio of DNA mixed with liposome varied. In brief, pRSVL plasmid DNA (2 μg) was mixed with varying amounts of DC-CHOL/DOPE (3:2) liposomes in 2 mM Hepes buffer at pH 7.6 in a final volume of 500 μL and after 7 minutes, the size of the complex was determined with a Coulter N4SD laser light scattering particle sizer operating in the unimodel mode.
As shown in Figure 1, large aggregates did not form when DNA was in excess (ratios of liposome to DNA less than 7) but at ratios of lipid to DNA that were charge neutral (~10) , the size of the complex reached a maximum. In addition, when the ratio of liposome-to-DNA was kept constant at 10 nmoles/μg, the size of the complex increased as both DNA and liposome concentration increased and eventually formed precipitates. However, when the ratio of liposomes to DNA was increased, the size of the complex was progressively reduced until the size of the complex became constant (250-300 nm) when the ratio of liposome-to-DNA exceeded 25 nmoles lipid/μg DNA. This result may be due to the fact that the DNA was perhaps coated by excess liposomes and therefore aggregation between the complexes did not occur.
Based on the data presented in Figure 1, lipid/DNA complexes were prepared using a liposome-to-DNA ratio of 50 nmoles/μg by slowly adding a DNA solution of 200 μg/ml to an excess amount (10 μmols) of liposome. The size of the complex formed was about 250 nm. When the ratio of liposome-to-DNA was changed to 25 nmoles/μg, the size of the complex increased to about 350 nm. The complexes formed using either the 25 nmole/μg or 50 nmole/μg ratios appeared to be physically stable since no precipitates formed during storage for four weeks at 4° C.
EXAMPLE 2 Purification Of DNA/lipid Complexes
When DNA was mixed with liposomes at a ratio of 1 μg/50 nmoles, an excess of free liposomal lipids was observed to co-exist with the DNA/lipid complex. Since excess free lipids are toxic to cells, an experiment was conducted to determine if free liposomal lipids could be separated from the DNA/lipid complex by a density gradient ultracentrifugation method. In brief, free liposomes (10 μmoles of DC-Chol/DOPE (2:3) in a volume of 2 ml); free DNA (50 μg pRSVL in a volume of 2 ml) and DNA/lipid complex formed by mixing 20 μmoles DC-Chol/DOPE (2:3) and 0.4 mg pRSVL plasmid DNA (50 nmoles/μg) were each centrifuged for 30 minutes at 4° C at 100,000 g over a gradient consisting of 0.5 ml each of 5%, 7.5%, 10% and 15% sucrose (W/W) . Fractions of 200 μl were then collected from the top to the bottom of the tube and assayed for the distribution of DNA marker (Ϊ2P, ■) and lipid marker (3H, O) . Figures 2A and 2B show the results of typical separations of free liposomal lipid, free DNA (Fig. 2A) and DNA/lipid complex (Fig. 2B) on the sucrose gradient. The results presented in Figure 2B show that after centrifugation, the complex formed a major band at the 10% sucrose layer. By comparison, Figure 2A shows that most of the radioactivity of the free DNA or free liposomal lipids distributed at the top half of the tube and did not enter the sucrose gradient. In addition, although the peak of 3H and the peak of 32P in Figure 2B co-existed at fraction number 16, there was a significant amount of the 3H distributed in fractions 1 to 10 indicating that the excess free liposomal lipids were well separated from the DNA/lipid complex.
EXAMPLE 3
Physical Stability Of Purified Lipid/DNA And Lipid/PLL/DNA Complexes
DNA/lipid complexes were formed by mixing 20 μmoles of liposomes of various DC-Chol/DOPE compositions (see Table 1) with 0.4 mg pRSVL plasmid DNA at a ratio of 1 μg DNA/50 nmoles lipid. Lipid/PLL/DNA complexes were formed by mixing 4 μmoles of liposomes of various DC- Chol/DOPE compositions with 1 mg PLL (MW=3000) and 0.4 mg pRSVL plasmid DNA. Both complexes were then purified from free lipids, free DNA and free PLL by sucrose gradient centrifugation as described in the Methods section. The peak fractions were collected and pooled. Pooled samples were then assayed for diameter immediately after collection (0 days) or after storage in 10% sucrose at 4°C for 120 days. Table 1 shows the results of these assays.
TABLE 1. Physical stability of purified lipid/DNA and lipid/PLIJDNA pre- complexes
Liposome Purified complex Recovery composition PLL Size (nm) Ratio of lipid/DNA of DNA
(DC-chol/DOPE) (μg/μgDNA) Day 0 Day 120 (nmoles/μg) (% Total)
2:8 0 168 280 23.2 51
3:7 0 187 252 14.0 66
4:6 0 175 195 12.7 73
5:5 0 174 210 13.2 70
6:4 0 198 232 10.1 69
2:8 2.5 165 287 20.8 17
3:7 2.5 99 101 19.2 22
4:6 2.5 138 132 38.3 29
5:5 2.5 184 178 22.4 27
The data presented in Table 1 shows that purified lipid/DNA and lipid/PLL/DNA complexes were small (under 200 nm) in size at day 0 and that their size did not increase dramatically with storage. Further, the ratios of DNA-to-lipid in the purified complexes was between 10 to 23 nmoles lipid/μg DNA depending on the composition of the liposomes used and this ratio did not change after storage for 120 days. A reciprocal relationship between the concentration of DC-Choi in the liposomes and the amount of the lipid in the complex was also observed indicating that liposomes enriched with DC-Chol show stronger DNA binding or charge neutralizing activity than liposomes less enriched with DC-Choi. The far-right hand columns shows recovery of 32P-labeled DNA in the DNA/lipid and DNA/lipid/PLL complexes following their purification on the sucrose density gradient. The results show that recovery of DNA in the non-PLL containing complexes was higher than that observed for the PLL- containing complexes.
EXAMPLE 4
Biological Activity Of The Purified Complexes In Various Cells
Since the DNA/lipid complexes formed by a mixture of liposomes to DNA having a high lipid to DNA ratio were both small and stable over time, experiments were conducted to compare the transfection activity of these complexes to the activity of DNA/liposome complex prepared by the admixture method.
In one experiment, CHO cells cultured in 48 well-plates were treated for 4 hours with an admixture of either 1 μg pRSVL and 10 nmoles DC-Chol/DOPE liposomes of different lipid compositions alone (O) or together with 1 μg PLL (MW=3,000) (D) or, the cells were treated with purified DNA/lipid complex (•) or purified DNA/lipid/PLL complex (■) formed by mixing 1 μg DNA with 50 nmoles DC- Chol/DOPE liposomes (DNA/lipid complex) or with 10 nmoles DC-Chol/DOPE liposomes and 1 μg PLL (DNA/lipid/PLL complex) followed by centrifugation through a sucrose density gradient as described in the Methods section. 36 hours after treatment, cells were lysed in 100 μl lysis buffer and 4 μl of the lysate was assayed for luciferase activity using 100 μl of luciferase substrate solution. Luciferase activity was then counted over a period of 20 seconds. The results presented in Figure 3 show that the most preferred liposome composition for transfecting CHO cells was 40% DC-Choi and 60% DOPE. In addition, in the presence of additional 1 μg poly-L-lysine (PLL, MW=3,000), a 2-7 fold enhancement of the transfection activity was seen in most cases. Of particular interest, the activity of the purified DNA/lipid complex was similar to that of the admixture DNA/lipid complex when the same amount of DNA was added to cells. However, the transfection activity of the purified DNA/lipid/PLL complex was about 30% to 50% lower than that of the DNA/liposome/PLL complex prepared by the admixture procedure.
In order to determine that the results obtained in CHO cells were not cell-specific, the transfection activities of the purified DNA/lipid and DNA/lipid/PLL complexes in two other cells, BHK and mouse lung cells (MLC) , were compared to that of DNA/liposome complexes formed by admixture.
In brief, cells (either BHK or MLC) grown in 48- well plates at 60% confluency were transfected with lμg pRSVL complexed with 10 nmoles of DC-Choi liposomes (admixture complex) , with the same amount of DNA mixed with liposomes at a DNA/liposome ratio of 1 μg/50 nmols to produce purified DNA/lipid complex or with purified DNA/lipid/PLL complex prepared at a DNA/liposome/PLL ratio of 1 μg/10 nmols/2 μg. Cells were then harvested at 36 hours post-transfection, and the luciferase activity of the transfected cell lysates was determined as described in the Methods section. The results of these experiments are shown in Tables 2 and 3. TABLE 2. Expression of luciferase gene in BHK cells transfected with pRSVL
Luciferase Activity (Relative Light Units X 10"3)
Liposome Admixture Purified Purified composition DNA/liposome DNA/lipid DNA/lipid/PLL
(DC-Chol/DOPE) complex complex complex
2:8 91.8 ± 9.5 110.1 ± 5.2 214.6 ± 41.1
3:7 61.2 ± 19.9 1886.8 ± 266.7 151.7 ± 62.9
4:6 438.2 ± 14.4 1638.8 ± 63.9 446.3 ± 16.9
5:5 837.8 ± 8 1015.0 ± 41.2 234.2 ± 46.4
TABLE 3. Expression of luciferase : gene in mouse lung cells transfected with pRSVL
Luciferase Activity (Relative Light Units X 103)
Liposome Admixture Purified Purified composition DNA/liposome DNA/lipid DNA/lipid/PLL
(DC-Chol DOPE) complex complex complex
2:8 1.1 ± 0.7 0.4 ± 0.2 0.3 ± 0.1
3:7 1.5 ± 1.0 0.3 ± 0.0 4.1 ± 1.3
4:6 3.1 ± 0.2 2.0 ± 0.3 14.6 ± 3.1
5:5 0.1 ± 0.0 1.5 ± 1.2 10.1 ± 2.3
Interestingly, for the BHK cell line, the transfection activity of the purified DNA/lipid complex was significantly higher than that of the DNA/liposome complex formed by admixture. For cells such as MLC, which are difficult to transfect, purified complexes made from DNA/liposome/PLL mixtures were apparently superior to admixture complexes and to purified DNA/lipid complexes made without PLL.
In order to determine whether lipid/PLL/DNA complexes could be made using different ratios of lipid and nucleic acid and a different molecular weight PLL than that used in the previous examples, the following experiment was conducted. Lipid/poly-L-lysine/DNA complex was prepared from 20 μg pRSVL plasmid DNA, 10 μg poly-L- lysine (MW 25,600), and DC-chol/DOPE liposomes (4.5/5.5 molar ratio) at the ratios of lipid to DNA shown in Table 4. The resulting complexes were then purified by sucrose gradient ultracentrifugation as described in the methods section. An aliquot of the purified complex containing 0.5 μg of DNA was used to transfect CHO cells, and luciferase activity was then measured. The results of
10 this experiment are shown below in Table 4.
TABLE 4. Effect of lipid/DNA ratio on purified complex containing poly-L-lysine (MW 25,600)
15 Composition of Ratio purified complex Size of purified Transfection activityb (nmoles lipid/μg DNA) (nmoles lipid/μg DNA) complex (nm) (counts (SD) X10"3)
3.3 1.1 89 108 (5)
6.6 2.5 98 6,065 (604)
20 12.5 4.3 101 5,846 (668)
20.0 9.6 35 7,633 (977)
The results show that in the presence of increased amounts of polycation, lower ratios of lipid to " DNA may be used to produce DNA/lipid/polycation complexes having appreciable transfection activity.
EXAMPLE 5 Transfection Activity of Stored Complexes 0
CHO cells cultured in 48 well-plates were treated for 4 hours with admixture of 1 μg pRSVL and 10 nmoles of DC-Chol/DOPE liposomes of different DC-Chol/DOPE compositions alone (O) or together with 1 μg PLL (mw = 5 3,000) (D) , or with purified DNA/lipid (•) or DNA/lipid/PLL (■) complexes stored at 4°C for 130 days in 10% sucrose. The purified complexes had been formed by mixing 1 μg pRSVL and 50 nmoles of DC-Chol/DOPE liposomes of different DC-Chol/DOPE compositions alone (DNA/lipid) or with 10 nmole of DC-Chol/DOPE liposomes and 1 μg PLL (DNA/lipid/PLL complex) followed by centrifugation through a sucrose density gradient as described in the Methods section. The results show that the luciferase activity of cell lysates prepared from cells transfected with the stored DNA/lipid and DNA/lipid/PLL complexes was comparable with the luciferase activity observed in cell lysates of cells transfected with the corresponding complexes prepared by admixture.
EXAMPLE 6
Comparative Cytotoxicity Of DNA/liposome Complexes Prepared By Admixture To That Of Purified DNA/Lipid Complexes
Cell toxicity of the different complexes was studied in CHO cells as follows. CHO cells were treated with admixture DNA/liposome complex (O) , admixture/liposome/PLL complex (D) ; purified DNA/lipid complex (•) ; or purified DNA/lipid/PLL complex (■) . The admixture complexes were formed by mixing 1 μg pRSVL DNA with 10 nmoles DC-Chol/DOPE liposomes of different DC- Chol/DOPE compositions alone or together with 1 μg PLL (mw = 3,000) . The purified complexes were formed by mixing 1 μg pRSVL DNA with 50 nmoles DC-Chol/DOPE liposomes alone (DNA/lipid complex) or with 10 nmoles DC-Chol/DOPE liposomes and 1 μg PLL (DNA/lipid/PLL complex) followed by centrifugation through a sucrose density gradient as described in the Methods section. 36 hours after treatment, the cells were lysed, protein was extracted and then quantitated by a Coomassie blue dye method. Fig. 5 shows the results of this experiment where the amount of total extractable protein recovered at the end of the experiment serves as an indicator of the portion of the cells which survived after the indicated treatment. The data presented shows that while the purified complex appeared to be slightly more toxic to the cells than the admixture DNA/liposome complex, morphologically, the transfection did not cause any serious cytotoxic effects in cells treated with either admixture complexes or the purified complexes, except that the cells treated with purified complexes containing high mole % DC-Choi were less confluent at the end of the experiment.
EXAMPLE 7
In Vivo Transfection of Tumors By Purified DNA/Lipid Complexes
3 x 106 human ovarian carcinoma cells were injected subcutaneously into SCID mice at day 0. 14 days later, 100 μl solutions containing pUCCMVCAT DNA (30 μg) complexed with DC-Choi (3:2 DC-Choi:DOPE) liposomes (30 nmoles) in the form of admixture (lanes 1 and 2) or the same amount of DNA in the form of purified DNA/lipid complex (prepared from DNA and DC-Choi liposomes at ratios of 1 ug DNA/25 nmoles lipid were directly injected into tumors. Animals were sacrificed 2 days later and tumor extracts containing 100 μg protein were assayed for CAT activity at 37°C according to Ausubel, et al. (1991) Current Protocols in Molecular Biology (Wiley, Boston) , Vol. 1, pp. 9.6.2-9.6.5). The results show that purified complex, while prepared under non-optimal conditions, exhibited in vivo transfection activity. EXAMPLE 8
Comparative Transfection Activity of Purified and Unpurified DNA/lipid/PLL
Complexes With Admixture DNA/Lipid Complex
The transfection activities of purified and unpurified DNA/Lipid/PLL complexes and admixture DNA/lipid complex in three cell lines (293, BL6 and C3) were measured as follows:
Purified DNA/lipid/PLL complex was formed by mixing 1 μg PRSVL DNA with 10 nmoles DC-Chol/DOPE liposomes (2:3 mol/mol) and 1 μg PLL (MW = 25,600) followed by centrifugation through a sucrose density gradient as described in the Methods section.
Unpurified DNA/lipid/PLL complexes were formed by mixing 100 μg PRSVL DNA with 80 μg PLL (MW = 25,600) and 1702 nmol DC-Chol/DOPE (2:3 mol/mol) liposomes (i.e. a DNA/lipid/PLK ratio of 1 μg DNA/17 nmol lipid/0.8 μg PLL) in a final volume of 500 μl of water. 20 μl of the unpurified DNA/lipid/PLL complex (i.e. 4μg DNA, 3.2 μg PLL and 68.1 nmol lipid) was then added to 780 μl of serum free medium appropriate for the cell line to be transfected.
Admixture DNA/lipid complex was formed by mixing 1 μg PRSVL DNA with 10 nmols of DC-Chol/DOPE (3:2 mol/mol) liposomes.
293, C3 and BL6 cells grown to 80% confluence in 24-well plates were transfected for four hours at 37°C with 1 μg of DNA in the form of purified DNA/lipid/PLL complex, admixture DNA/lipid complex or unpurified DNA/lipid/PLL complex. After transfection, cells were were further cultured for 36-48 hours in the appropriate media containing 10% fetal calf serum. Luciferase activity was then measured as described in the Methods section.
The results presented in Figures 7A (293 cells) unpurified DNA/lipid/PLL complex exhibited the highest transfection activity in all three cell lines tested.
While we have hereinbefore described a number of embodiments of this invention, it is apparent that the basic constructions can be altered to provide other embodiments which utilize the methods and devices of this invention. Therefore, it will be appreciated that the scope of this invention is defined by the claims appended hereto rather than by the specific embodiments which have been presented hereinbefore by way of example.

Claims

1. A method for producing drug/lipid complexes having a positive charge excess of lipid to drug, said method comprising: mixing said drug with cationic liposomes in a drug to lipid ratio such that said drug/lipid complexes are formed.
2. The method of claim 1, wherein said ratio of drug to lipid mixed to form said complex is from about 1 μg/0.1 nmol to about 1 μg/200 nmol.
3. The method of claim 2, wherein said method further includes the step of purifying said complexes.
4. The method of claim 3, wherein said purifying step is centrifugation through a sucrose density gradient.
5. The method according to claim 1 wherein the drug is nucleic acid and the liposomes are DC-Chol/DOPE liposomes.
6. A method for producing drug/lipid/polycation complexes having a positive charge excess of lipid and polycation to drug, said method comprising: mixing said drug with cationic liposomes and at least one polycation in a ratio of drug to lipid to polycation such that said complexes are formed.
7. The method of claim 6 wherein said ratio of drug to lipid is from about 1 μg/0.1 nmol to about 1 μg/200 nmol.
8. The method of claim 6, wherein said polycation is present at from about 0.01 μg to about 100 μg- g. The method of claim 8, wherein the polycation is poly-L-lysine having a molecular weight of about 300 to about 200,000 daltons.
10. The method of claim 6, wherein the cationic liposome comprises a cationic lipid and a neutral phospholipid.
11. The method of claim 10, wherein the cationic lipid is DC-Choi.
12. The method of claim 11, wherein the neutral phospholipid is dioleoyl phosphatidylethanolamine.
13. The method of claim 12, wherein the drug is nucleic acid.
1 . The method of claims 1 or 6, wherein said complex has an average diameter less than 300 nm.
15. The method of claim 14, wherein the average diameter of said formulation remains substantially unchanged for up to one year in storage.
16. A drug/lipid complex comprising: at least one lipid species and a drug; the ratio of said lipid species to said drug being such that said complex has a positive charge excess of lipid to drug.
17. The complex of claim 16, wherein said drug is nucleic acid.
18. The complex of claim 16, wherein said ratio of drug to lipid is from about 1 μg/0.1 nmol to about 1 μg/200 mmol.
19. The complex of claim 16, wherein said lipid species is a cationic lipid.
20. The complex of claim 19, wherein said complex further comprises a neutral phospholipid species.
21. The complex of claim 16, wherein said complex is purified from excess free drug and free lipid species.
22. A drug/lipid/polycation complex comprising at least one lipid species, at least one polycation and a drug; the ratio of said drug to said lipid species to said polycation being such that said complex has a positive charge excess of lipid and polycation to drug.
23. The complex of claim 22, wherein said ratio of lipid to DNA is from about 1 μg/0.1 nmol to about 1 μg/200 nmol.
24. The complex of claim 22, wherein said polycation is present at about .Olμg to about 100 μg.
25. The complex of claim 24, wherein said polycation is poly-L-lysine having a molecular weight of about 300 to about 200,000.
26. The complexes of claim 16 and 22, wherein the net positive charge of said complexes exists at pH 6.0 - 8.0.
27. The complexes of claim 26, wherein the lipid species are DC-Choi and DOPE.
28. A method of delivering drug to an individual comprising administering the complex of any one of claims 16 or 22 in a therapeutically effective amount to an individual in need of treatment.
29. The method of claim 28, wherein the complex is a drug/lipid complex.
30. The method of claim 28, wherein the complex is a drug/lipid/polycation complex.
PCT/US1996/000816 1995-01-23 1996-01-23 Stable lipid-comprising drug delivery complexes and methods for their production WO1996022765A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU47037/96A AU709773B2 (en) 1995-01-23 1996-01-23 Stable lipid-comprising drug delivery complexes and methods for their production
DE69624801T DE69624801T2 (en) 1995-01-23 1996-01-23 STABLE DRUG DELIVERY COMPLEXES WITH LIPIDS AND METHOD FOR THE PRODUCTION THEREOF
DK96902747T DK0814777T3 (en) 1995-01-23 1996-01-23 Stable lipid-containing drug delivery complexes and methods for their preparation
JP52295496A JP4074338B2 (en) 1995-01-23 1996-01-23 Stable lipid-containing drug delivery complexes and methods for their production
CA002211118A CA2211118C (en) 1995-01-23 1996-01-23 Stable lipid-comprising drug delivery complexes and methods for their production
AT96902747T ATE227563T1 (en) 1995-01-23 1996-01-23 STABLE DRUG DELIVERY COMPLEXES WITH LIPIDS AND METHOD FOR THE PRODUCTION THEREOF
EP96902747A EP0814777B1 (en) 1995-01-23 1996-01-23 Stable lipid-comprising drug delivery complexes and methods for their production

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/376,701 US5795587A (en) 1995-01-23 1995-01-23 Stable lipid-comprising drug delivery complexes and methods for their production
US08/376,701 1995-01-23

Publications (1)

Publication Number Publication Date
WO1996022765A1 true WO1996022765A1 (en) 1996-08-01

Family

ID=23486103

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/000816 WO1996022765A1 (en) 1995-01-23 1996-01-23 Stable lipid-comprising drug delivery complexes and methods for their production

Country Status (14)

Country Link
US (5) US5795587A (en)
EP (1) EP0814777B1 (en)
JP (1) JP4074338B2 (en)
CN (1) CN1177291A (en)
AT (1) ATE227563T1 (en)
AU (1) AU709773B2 (en)
CA (1) CA2211118C (en)
DE (1) DE69624801T2 (en)
DK (1) DK0814777T3 (en)
ES (1) ES2186769T3 (en)
IL (1) IL116856A0 (en)
PT (1) PT814777E (en)
WO (1) WO1996022765A1 (en)
ZA (1) ZA96503B (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998046273A2 (en) * 1997-04-17 1998-10-22 Paola Leone Delivery system for gene therapy to the brain
US5948878A (en) * 1997-04-15 1999-09-07 Burgess; Stephen W. Cationic polymers for nucleic acid transfection and bioactive agent delivery
US5962429A (en) * 1996-11-22 1999-10-05 University Of Iowa Complexes of adenovirus with cationic molecules
US6051429A (en) * 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6107286A (en) * 1994-12-05 2000-08-22 Rhone-Poulenc Rorer S.A. Lipopolyamines as transfection agents and pharamaceutical uses thereof
US6120794A (en) * 1995-09-26 2000-09-19 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
US6251433B1 (en) 1996-08-13 2001-06-26 Chiron Corporation Polycationic polymers
US6569450B1 (en) 1997-08-13 2003-05-27 Chiron Corporation Lipid-conjugated polyamide compounds and related compositions and methods thereof
DE10322123A1 (en) * 2003-05-12 2004-12-16 Novosom Ag Stable anionic liposome depot system for prolonged release of protein or peptide drugs, e.g. insulin, comprising phosphatidyl choline, cholesterol, anionic lipid(s) and cationic polymer
US6855549B1 (en) 1998-11-23 2005-02-15 The University Of Iowa Research Foundation Methods and compositions for increasing the infectivity of gene transfer vectors
JP2008239631A (en) * 1996-11-12 2008-10-09 Regents Of The Univ Of California Preparation of stable formulation of lipid-nucleic acid complex effective for in vivo delivery
JP2010031028A (en) * 1998-05-12 2010-02-12 Regents Of The Univ Of California Method of forming protein-linked lipidic microparticle, and composition thereof
EP2569437A1 (en) * 2010-05-10 2013-03-20 Qiagen GmbH Method for transfecting a eukaryotic cell
US9295646B2 (en) 2010-07-06 2016-03-29 Novartis Ag Cationic oil-in-water emulsions
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US9636410B2 (en) 2011-07-06 2017-05-02 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
US9655845B2 (en) 2011-07-06 2017-05-23 Glaxosmithkline Biologicals, S.A. Oil-in-water emulsions that contain nucleic acids
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells

Families Citing this family (326)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6383814B1 (en) 1994-12-09 2002-05-07 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6008202A (en) * 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US6290969B1 (en) * 1995-09-01 2001-09-18 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US6458366B1 (en) 1995-09-01 2002-10-01 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
US6592877B1 (en) * 1995-09-01 2003-07-15 Corixa Corporation Compounds and methods for immunotherapy and diagnosis of tuberculosis
US6258792B1 (en) 1996-04-12 2001-07-10 University Of Pittsburgh Cationic cholesteryl derivatives containing cyclic polar groups
CA2251543A1 (en) * 1996-04-12 1997-10-23 University Of Pittsburgh Novel cationic cholesteryl derivatives containing cyclic polar groups
AU727447B2 (en) * 1996-07-03 2000-12-14 University Of Pittsburgh Emulsion formulations for hydrophilic active agents
US7083786B2 (en) * 1997-04-03 2006-08-01 Jensenius Jens Chr MASP-2, a complement-fixing enzyme, and uses for it
US7157098B1 (en) * 1998-01-06 2007-01-02 Roman Perez-Soler Gene therapy of tumors using non-viral delivery system
US20020147143A1 (en) 1998-03-18 2002-10-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US7244714B1 (en) * 1998-06-12 2007-07-17 Aradigm Corporation Methods of delivering aerosolized polynucleotides to the respiratory tract
US20030235557A1 (en) 1998-09-30 2003-12-25 Corixa Corporation Compositions and methods for WT1 specific immunotherapy
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
US6395511B1 (en) * 1998-11-27 2002-05-28 Darwin Discovery, Ltd. Nucleic acids encoding a novel family of TGF-β binding proteins from humans
AU2460900A (en) * 1999-02-15 2000-08-29 Nippon Shinyaku Co. Ltd. Shortened-chain polynucleotides and process for the preparation thereof
EP1156812A4 (en) * 1999-02-23 2004-09-29 Isis Pharmaceuticals Inc Multiparticulate formulation
CZ20013103A3 (en) * 1999-03-02 2002-01-16 The Liposome Company, Inc. Encapsulating method of biologically active complexes into liposomes
US8143386B2 (en) * 1999-04-07 2012-03-27 Corixa Corporation Fusion proteins of mycobacterium tuberculosis antigens and their uses
US20030104622A1 (en) * 1999-09-01 2003-06-05 Robbins Paul D. Identification of peptides that facilitate uptake and cytoplasmic and/or nuclear transport of proteins, DNA and viruses
WO2001024820A1 (en) * 1999-10-07 2001-04-12 Corixa Corporation Fusion proteins of mycobacterium tuberculosis
CN1433478A (en) * 1999-12-30 2003-07-30 诺瓦提斯公司 Novel colloid synthetic vectors for gene therapy
US6514523B1 (en) 2000-02-14 2003-02-04 Ottawa Heart Institute Research Corporation Carrier particles for drug delivery and process for preparation
IL151097A0 (en) 2000-02-23 2003-04-10 Smithkline Beecham Biolog Tumour-specific animal proteins
AU2001241738A1 (en) * 2000-02-25 2001-09-03 Corixa Corporation Compounds and methods for diagnosis and immunotherapy of tuberculosis
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
JP2004512010A (en) 2000-03-29 2004-04-22 ディージーアイ・バイオテクノロジーズ・エル・エル・シー Insulin and IGF-1 receptor agonists and antagonists
ATE442866T1 (en) * 2000-06-20 2009-10-15 Corixa Corp FUSION PROTEINS FROM MYCOBACTERIUM TUBERCULOSIS
JP2004513615A (en) 2000-06-28 2004-05-13 コリクサ コーポレイション Compositions and methods for treatment and diagnosis of lung cancer
US20040220100A1 (en) * 2000-07-21 2004-11-04 Essentia Biosystems, Inc. Multi-component biological transport systems
DK2364734T3 (en) * 2000-07-21 2017-10-30 Revance Therapeutics Inc Biological multicomponent transport systems
DE10049010A1 (en) * 2000-10-04 2002-04-18 Boehringer Ingelheim Int Transferrin polycation / DNA complexes for the systemic therapy of tumor diseases with cytotoxic proteins
AU9418501A (en) * 2000-10-04 2002-04-15 Kyowa Hakko Kogyo Kk Method of coating fine particle with lipid film
EP1355628A2 (en) * 2001-02-01 2003-10-29 Board of Regents, The University of Texas System Stabilised polymeric aerosols for pulmonary gene delivery
US20030096414A1 (en) * 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
US20030203865A1 (en) * 2001-04-30 2003-10-30 Pierrot Harvie Lipid-comprising drug delivery complexes and methods for their production
WO2002089747A2 (en) 2001-05-09 2002-11-14 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
AU2002309567A1 (en) * 2001-05-16 2002-12-03 The Children's Hospital Of Philadelphia Dna-antibody complexes to enhance gene transfer
AU2002348163A1 (en) * 2001-11-02 2003-05-19 Intradigm Corporation Therapeutic methods for nucleic acid delivery vehicles
CA2476755C (en) 2001-12-17 2014-08-26 Corixa Corporation Compositions and methods for the therapy and diagnosis of inflammatory bowel disease
AU2003205380A1 (en) * 2002-02-01 2003-09-02 Intradigm Corporation Cationic polymers for use in therapeutic agent delivery
US20060014695A1 (en) * 2002-02-01 2006-01-19 Hamidreza Ghandehari Hpma-polyamine conjugates and uses therefore
US7026465B2 (en) 2002-02-15 2006-04-11 Corixa Corporation Fusion proteins of Mycobacterium tuberculosis
WO2003087763A2 (en) 2002-04-03 2003-10-23 Celltech R & D, Inc. Association of polymorphisms in the sost gene region with bone mineral density
US20060193905A1 (en) * 2002-05-14 2006-08-31 University Of Louisville Research Foundation, Inc. Direct cellular energy delivery system
RU2389732C2 (en) 2003-01-06 2010-05-20 Корикса Корпорейшн Certain aminoalkyl glucosaminide phospahte derivatives and use thereof
US7960522B2 (en) 2003-01-06 2011-06-14 Corixa Corporation Certain aminoalkyl glucosaminide phosphate compounds and their use
HUE024996T2 (en) 2003-05-12 2016-01-28 Helion Biotech Aps Antibodies to masp-2
NZ544618A (en) 2003-06-16 2009-02-28 Celltech R & D Inc Antibodies specific for sclerostin and methods for increasing bone mineralization
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
PT2656859E (en) * 2004-03-03 2016-02-24 Revance Therapeutics Inc Topical application and transdermal delivery of botulinum toxins
AU2005251676B2 (en) 2004-03-03 2011-09-29 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
EP2497831B1 (en) 2004-05-25 2014-07-16 Oregon Health and Science University TB vaccination using HCMV-based vaccine vectors
PL1753456T3 (en) 2004-06-10 2017-01-31 Omeros Corporation Methods for treating conditions associated with masp-2 dependent complement activation
US7919094B2 (en) 2004-06-10 2011-04-05 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
US20060018896A1 (en) * 2004-06-10 2006-01-26 University Of Leicester Methods for treating conditions associated with lectin-dependent complement activation
US8840893B2 (en) 2004-06-10 2014-09-23 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
JP2008502622A (en) 2004-06-14 2008-01-31 サラマ,ゾーセル,ベー. An anticancer composition comprising proline or a derivative thereof and an antitumor antibody
EP1885746B1 (en) 2005-02-08 2012-01-11 Research Development Foundation Compositions related to soluble g-protein coupled receptors (sgpcrs)
US9180081B2 (en) 2005-03-03 2015-11-10 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
ATE507236T1 (en) 2005-03-03 2011-05-15 Revance Therapeutics Inc COMPOSITIONS AND METHODS FOR TOPICAL APPLICATION AND TRANSDERMAL ADMINISTRATION OF AN OLIGOPEPTIDE
WO2006104890A2 (en) 2005-03-31 2006-10-05 Glaxosmithkline Biologicals Sa Vaccines against chlamydial infection
ITMI20050739A1 (en) * 2005-04-22 2006-10-23 Effebi Spa VALVE-ACTUATOR CONNECTION PLATE
JP5164830B2 (en) 2005-04-29 2013-03-21 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム A novel method for the prevention or treatment of Mycobacterium tuberculosis infection
US8003108B2 (en) 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
US7592429B2 (en) * 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
WO2007016597A2 (en) * 2005-07-29 2007-02-08 The Regents Of The University Of California Targeting tnf-alpha converting enzyme (tace)-dependent growth factor shedding in cancer therapy
GB2430881B (en) * 2005-10-06 2010-10-13 Ntnu Technology Transfer As Oligoelectrolyte polyols for the treatment of mucosal hyperviscosity
US8568740B2 (en) * 2005-11-17 2013-10-29 Revance Therapeutics, Inc. Compositions and methods of topical application and transdermal delivery of botulinum toxins with reduced non-toxin proteins
AU2006318212C1 (en) 2005-11-23 2012-08-30 The Board Of Regents Of The University Of Texas System Oncogenic ras-specific cytotoxic compound and methods of use thereof
US20090081157A1 (en) 2006-01-09 2009-03-26 Richard Syd Kornbluth Immunostimulatory Combinations for Vaccine Adjuvants
CL2007002567A1 (en) 2006-09-08 2008-02-01 Amgen Inc ISOLATED PROTEINS FROM LINK TO ACTIVINE TO HUMAN.
US8097419B2 (en) 2006-09-12 2012-01-17 Longhorn Vaccines & Diagnostics Llc Compositions and method for rapid, real-time detection of influenza A virus (H1N1) swine 2009
US8080645B2 (en) 2007-10-01 2011-12-20 Longhorn Vaccines & Diagnostics Llc Biological specimen collection/transport compositions and methods
US9481912B2 (en) 2006-09-12 2016-11-01 Longhorn Vaccines And Diagnostics, Llc Compositions and methods for detecting and identifying nucleic acid sequences in biological samples
US20100015665A1 (en) * 2006-11-10 2010-01-21 Ucb Pharma S.A. Antibodies and diagnostics
EP2097450A2 (en) * 2006-11-10 2009-09-09 Amgen Inc. Antibody-based diagnostics and therapeutics
US20100093639A1 (en) * 2006-12-29 2010-04-15 Revance Therapeutics, Inc. Transport Molecules Using Reverse Sequence HIV-TAT Polypeptides
EP3492596A1 (en) 2007-04-09 2019-06-05 University of Florida Research Foundation, Inc. Raav vector compositions having tyrosine-modified capsid proteins and methods for use
GB0707096D0 (en) * 2007-04-12 2007-05-23 Ntnu Technology Transfer As Method
WO2009006479A2 (en) 2007-07-02 2009-01-08 Etubics Corporation Methods and compositions for producing an adenovirus vector for use with multiple vaccinations
US9683256B2 (en) 2007-10-01 2017-06-20 Longhorn Vaccines And Diagnostics, Llc Biological specimen collection and transport system
US11041215B2 (en) 2007-08-24 2021-06-22 Longhorn Vaccines And Diagnostics, Llc PCR ready compositions and methods for detecting and identifying nucleic acid sequences
EP2772267B1 (en) 2007-08-27 2016-04-27 Longhorn Vaccines and Diagnostics, LLC Immunogenic compositions and methods
US10004799B2 (en) 2007-08-27 2018-06-26 Longhorn Vaccines And Diagnostics, Llc Composite antigenic sequences and vaccines
CL2008002775A1 (en) 2007-09-17 2008-11-07 Amgen Inc Use of a sclerostin binding agent to inhibit bone resorption.
US11041216B2 (en) 2007-10-01 2021-06-22 Longhorn Vaccines And Diagnostics, Llc Compositions and methods for detecting and quantifying nucleic acid sequences in blood samples
CA2861667C (en) 2007-10-01 2017-06-13 Longhorn Vaccines And Diagnostics, Llc Biological specimen collection and transport system and methods of use
UA97559C2 (en) * 2007-11-08 2012-02-27 Оцука Фармасьютікал Ко., Лтд. Nucleic acid complex and a nucleic acid delivery composition
AU2008338464A1 (en) * 2007-12-14 2009-06-25 Amgen Inc. Method for treating bone fracture with anti-sclerostin antibodies
AU2009249403A1 (en) * 2008-05-19 2009-11-26 The University Of North Carolina At Chapel Hill Methods and compositions comprising novel cationic lipids
GB0904942D0 (en) 2009-03-23 2009-05-06 Ntnu Technology Transfer As Composition
GB0904941D0 (en) * 2009-03-23 2009-05-06 Ntnu Technology Transfer As Composition
US8753639B2 (en) 2009-03-31 2014-06-17 University Of Washington Through Its Center For Commercialization Compositions and methods for modulating the activity of complement regulatory proteins on target cells
WO2010135714A2 (en) 2009-05-22 2010-11-25 The Methodist Hospital Research Institute Methods for modulating adipocyte expression using microrna compositions
WO2010138263A2 (en) 2009-05-28 2010-12-02 University Of Massachusetts Novel aav 's and uses thereof
ES2545794T3 (en) 2009-08-28 2015-09-15 Research Development Foundation Urocortin 2 analogues and uses thereof
BR112012008970A2 (en) 2009-10-16 2019-12-10 Omeros Corp use of a composition and composition
TW201132353A (en) 2009-12-18 2011-10-01 Amgen Inc WISE binding agents and epitopes
HUE031184T2 (en) 2010-01-27 2017-06-28 Glaxosmithkline Biologicals Sa Modified tuberculosis antigens
ES2711256T3 (en) 2010-04-23 2019-04-30 Univ Florida Compositions of rAAV-guanylate cyclase and methods to treat congenital amaurosis of Leber 1 (LCA1)
JP2013533847A (en) 2010-04-23 2013-08-29 ユニバーシティ オブ マサチューセッツ AAV-based treatment of cholesterol-related disorders
WO2011133890A1 (en) 2010-04-23 2011-10-27 University Of Massachusetts Cns targeting aav vectors and methods of use thereof
EA027039B1 (en) 2010-05-14 2017-06-30 Эмджен Инк. High concentration antibody formulations
HUE037227T2 (en) 2010-05-14 2018-08-28 Univ Oregon Health & Science Recombinant HCMV and RHCMV vectors and uses thereof
EP3578205A1 (en) 2010-08-06 2019-12-11 ModernaTX, Inc. A pharmaceutical formulation comprising engineered nucleic acids and medical use thereof
US20120237975A1 (en) 2010-10-01 2012-09-20 Jason Schrum Engineered nucleic acids and methods of use thereof
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
WO2012118903A2 (en) 2011-03-01 2012-09-07 Amgen Inc. Bispecific binding agents
CN103517920B (en) 2011-03-25 2018-04-17 安进公司 Anti- hardened proteins (SCLEROSTIN) antibody crystals and its preparation
EP2691101A2 (en) 2011-03-31 2014-02-05 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
US9644035B2 (en) 2011-04-08 2017-05-09 Omeros Corporation Methods for treating conditions associated with MASP-2 dependent complement activation
MX355648B (en) 2011-04-08 2018-04-26 Univ Leicester Methods for treating conditions associated with masp-2 dependent complement activation.
WO2012145624A2 (en) 2011-04-21 2012-10-26 University Of Massachusetts Raav-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
HUE049154T2 (en) 2011-05-04 2020-09-28 Omeros Corp Compositions for inhibiting masp-2 dependent complement acitivation
WO2012177595A1 (en) 2011-06-21 2012-12-27 Oncofactor Corporation Compositions and methods for the therapy and diagnosis of cancer
EP3613852A3 (en) 2011-07-22 2020-04-22 President and Fellows of Harvard College Evaluation and improvement of nuclease cleavage specificity
PT2739311T (en) 2011-08-04 2018-03-26 Amgen Inc Method for treating bone gap defects
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
CA2850624A1 (en) 2011-10-03 2013-04-11 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9849087B2 (en) 2011-11-08 2017-12-26 The Board Of Trustees Of The University Of Arkansas Methods and compositions for X-ray induced release from pH sensitive liposomes
KR20140102759A (en) 2011-12-16 2014-08-22 모더나 세라퓨틱스, 인코포레이티드 Modified nucleoside, nucleotide, and nucleic acid compositions
EP2797953B1 (en) 2011-12-28 2020-06-03 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
CA2863083C (en) 2012-01-26 2023-09-19 Longhorn Vaccines And Diagnostics, Llc Composite antigenic sequences and vaccines
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
AU2013243949A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
CA2868398A1 (en) 2012-04-02 2013-10-10 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US20130273053A1 (en) 2012-04-06 2013-10-17 University Of Leicester Compositions and Methods of Inhibiting MASP-1 and/or MASP-2 and/or MASP-3 for the Treatment of Paroxysmal Nocturnal Hemoglobinuria
RU2709351C2 (en) 2012-06-18 2019-12-17 Омерос Корпорейшн Compositions and methods of inhibiting masp-1, and/or masp-2, and/or masp-3 for treating various diseases and disorders
AU2013285488B2 (en) 2012-07-05 2018-03-22 Ucb Pharma S.A. Treatment for bone diseases
WO2014031178A1 (en) 2012-08-24 2014-02-27 Etubics Corporation Replication defective adenovirus vector in vaccination
UY35148A (en) 2012-11-21 2014-05-30 Amgen Inc HETERODIMERIC IMMUNOGLOBULINS
JP6144355B2 (en) 2012-11-26 2017-06-07 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chemically modified mRNA
CA2897941A1 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
MX2015009901A (en) 2013-02-01 2016-04-06 Santa Maria Biotherapeutics Inc Administration of an anti-activin-a compound to a subject.
US20160024181A1 (en) 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US10258698B2 (en) 2013-03-14 2019-04-16 Modernatx, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
BR112015023502A2 (en) 2013-03-15 2017-07-18 Glaxosmithkline Biologicals Sa composition, and methods for enhancing an immune response and substantially improving or preventing an infectious disease, an autoimmune disease, or an allergic condition in an individual
JP6549554B2 (en) 2013-03-15 2019-07-24 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Peptides with reduced toxicity that stimulate cholesterol efflux
PL2970389T3 (en) * 2013-03-15 2021-03-08 Rhythm Pharmaceuticals, Inc. Pharmaceutical compositions
DK3019619T3 (en) 2013-07-11 2021-10-11 Modernatx Inc COMPOSITIONS INCLUDING SYNTHETIC POLYNUCLEOTIDES CODING CRISPR-RELATED PROTEINS, SYNTHETIC SGRNAs, AND USES OF USE
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
JP6896421B2 (en) 2013-08-21 2021-06-30 キュアバック アーゲー Respiratory syncytial virus (RSV) vaccine
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US20160194368A1 (en) 2013-09-03 2016-07-07 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
EP3052106A4 (en) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
JP2016538829A (en) 2013-10-03 2016-12-15 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. Polynucleotide encoding low density lipoprotein receptor
DK3057993T3 (en) 2013-10-17 2020-11-16 Omeros Corp Method for Treating Conditions Associated with MASP-2 Dependent Complement
DK3066201T3 (en) 2013-11-07 2018-06-06 Editas Medicine Inc CRISPR-RELATED PROCEDURES AND COMPOSITIONS WITH LEADING GRADES
CA2930973A1 (en) 2013-11-22 2015-05-28 Pal SAERTROM C/ebp alpha short activating rna compositions and methods of use
US20150166982A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting pi3k point mutations
AU2015231294B2 (en) 2014-03-18 2020-10-29 University Of Massachusetts rAAV-based compositions and methods for treating amyotrophic lateral sclerosis
CA2946751A1 (en) 2014-04-23 2015-10-29 Modernatx, Inc. Nucleic acid vaccines
WO2015164786A1 (en) 2014-04-25 2015-10-29 University Of Massachusetts Recombinant aav vectors useful for reducing immunity against transgene products
SI3766916T1 (en) 2014-06-25 2023-01-31 Acuitas Therapeutics Inc. Novel lipids and lipid nanoparticle formulations for delivery of nucleic acids
EP3169693B1 (en) 2014-07-16 2022-03-09 ModernaTX, Inc. Chimeric polynucleotides
EP3171895A1 (en) 2014-07-23 2017-05-31 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
WO2016022363A2 (en) 2014-07-30 2016-02-11 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
US10029012B2 (en) * 2014-09-05 2018-07-24 University Of Pittsuburgh—Of The Commonwealth System Of Higher Education Peptide-mediated intravesical delivery of therapeutic and diagnostic agents
US10711270B2 (en) 2014-10-03 2020-07-14 University Of Massachusetts High efficiency library-identified AAV vectors
EP3207150B1 (en) 2014-10-14 2021-04-28 Research Development Foundation Methods for generating engineered enzymes
CN107073051B (en) 2014-10-21 2021-08-24 马萨诸塞大学 Recombinant AAV variants and uses thereof
US10889812B2 (en) 2014-10-24 2021-01-12 University Of Maryland, Baltimore Short non-coding protein regulatory RNAs (sprRNAs) and methods of use
EP3212165B1 (en) 2014-10-30 2024-02-28 President and Fellows of Harvard College Delivery of negatively charged proteins using cationic lipids
US9816080B2 (en) 2014-10-31 2017-11-14 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
MA41142A (en) 2014-12-12 2017-10-17 Amgen Inc ANTI-SCLEROSTINE ANTIBODIES AND THE USE OF THEM TO TREAT BONE CONDITIONS AS PART OF THE TREATMENT PROTOCOL
WO2016100575A1 (en) 2014-12-16 2016-06-23 Board Of Regents Of The University Of Nebraska Gene therapy for juvenile batten disease
US20170369872A1 (en) 2014-12-18 2017-12-28 Alnylam Pharmaceuticals, Inc. Reversir tm compounds
US10695417B2 (en) 2015-01-09 2020-06-30 Etubics Corporation Human adenovirus serotype 5 vectors containing E1 and E2B deletions encoding the ebola virus glycoprotein
CA3019315A1 (en) 2015-04-23 2016-10-27 University Of Massachusetts Modulation of aav vector transgene expression
CA2985652C (en) 2015-05-14 2020-03-10 Gerald W. FISHER Rapid methods for the extraction of nucleic acids from biological samples
US20160346221A1 (en) 2015-06-01 2016-12-01 Autotelic Llc Phospholipid-coated therapeutic agent nanoparticles and related methods
WO2017004143A1 (en) 2015-06-29 2017-01-05 Acuitas Therapeutics Inc. Lipids and lipid nanoparticle formulations for delivery of nucleic acids
ES2937963T3 (en) 2015-07-21 2023-04-03 Modernatx Inc Infectious disease vaccines
WO2017031232A1 (en) 2015-08-17 2017-02-23 Modernatx, Inc. Methods for preparing particles and related compositions
AU2016324310B2 (en) 2015-09-17 2021-04-08 Modernatx, Inc. Compounds and compositions for intracellular delivery of therapeutic agents
EP3362102A1 (en) 2015-10-14 2018-08-22 Life Technologies Corporation Ribonucleoprotein transfection agents
MA46024A (en) 2015-10-22 2019-07-03 Modernatx Inc HERPES SIMPLEX VACCINE
CN108472354A (en) 2015-10-22 2018-08-31 摩登纳特斯有限公司 Respiratory syncytial virus vaccines
EP3364981A4 (en) 2015-10-22 2019-08-07 ModernaTX, Inc. Human cytomegalovirus vaccine
PL3718565T3 (en) 2015-10-22 2022-09-19 Modernatx, Inc. Respiratory virus vaccines
EA201891001A1 (en) 2015-10-22 2018-11-30 МОДЕРНАТиЭкс, ИНК. VACCINES ON THE BASIS OF NUCLEIC ACIDS AGAINST WINDSHEAD VIRUS (VZV)
JP7109784B2 (en) 2015-10-23 2022-08-01 プレジデント アンド フェローズ オブ ハーバード カレッジ Evolved Cas9 protein for gene editing
WO2017083371A1 (en) 2015-11-09 2017-05-18 Omeros Corporation Methods for treating conditions associated with masp-2 dependent complement activation
EP3383418B1 (en) 2015-12-04 2021-10-20 Board of Regents, The University of Texas System Slc45a2 peptides for immunotherapy
CA3007955A1 (en) 2015-12-10 2017-06-15 Modernatx, Inc. Lipid nanoparticles for delivery of therapeutic agents
PL3394030T3 (en) 2015-12-22 2022-04-11 Modernatx, Inc. Compounds and compositions for intracellular delivery of agents
DK3394093T3 (en) 2015-12-23 2022-04-19 Modernatx Inc PROCEDURES FOR USING OX40 LIGAND CODING POLYNUCLEOTIDES
SG11201805695SA (en) 2016-01-05 2018-07-30 Univ Leicester Methods for inhibiting fibrosis in a subject in need thereof
US20190241658A1 (en) 2016-01-10 2019-08-08 Modernatx, Inc. Therapeutic mRNAs encoding anti CTLA-4 antibodies
EP4094780A3 (en) 2016-02-12 2023-02-08 University of Massachusetts Anti-angiogenic mirna therapeutics for inhibiting corneal neovascularization
ES2923877T3 (en) 2016-03-01 2022-10-03 Univ Florida AAV vectors for the treatment of dominant retinitis pigmentosa
KR102336362B1 (en) 2016-03-03 2021-12-08 보이저 테라퓨틱스, 인크. Closed-ended linear duplex DNA for non-viral gene delivery
GB201604124D0 (en) 2016-03-10 2016-04-27 Ucb Biopharma Sprl Pharmaceutical formulation
CN115137816A (en) 2016-03-31 2022-10-04 奥默罗斯公司 Method of inhibiting angiogenesis in a subject in need thereof
JP7262100B2 (en) 2016-04-29 2023-04-21 アイカーン スクール オブ メディスン アット マウント サイナイ Targeting the innate immune system to induce long-term tolerance and resolve macrophage accumulation in atherosclerosis
EP3468609A1 (en) * 2016-06-09 2019-04-17 CureVac AG Cationic carriers for nucleic acid delivery
WO2017212009A1 (en) 2016-06-09 2017-12-14 Curevac Ag Hybrid carriers for nucleic acid cargo
JOP20170154B1 (en) 2016-08-01 2023-03-28 Omeros Corp Compositions and methods of inhibiting masp-3 for the treatment of various diseases and disorders
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
US20190185864A1 (en) 2016-08-23 2019-06-20 Akouos, Inc. Compositions and methods for treating non-age-associated hearing impairment in a human subject
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
AU2017341849B2 (en) 2016-10-13 2024-03-21 University Of Massachusetts AAV capsid designs
WO2018071868A1 (en) 2016-10-14 2018-04-19 President And Fellows Of Harvard College Aav delivery of nucleobase editors
WO2018089540A1 (en) 2016-11-08 2018-05-17 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
CA3043033A1 (en) * 2016-11-10 2018-05-17 Translate Bio, Inc. Improved ice-based lipid nanoparticle formulation for delivery of mrna
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
WO2018165504A1 (en) 2017-03-09 2018-09-13 President And Fellows Of Harvard College Suppression of pain by gene editing
KR20190127797A (en) 2017-03-10 2019-11-13 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Cytosine to Guanine Base Editing Agent
HUE060693T2 (en) 2017-03-15 2023-04-28 Modernatx Inc Compound and compositions for intracellular delivery of therapeutic agents
EP3596042B1 (en) 2017-03-15 2022-01-12 Modernatx, Inc. Crystal forms of amino lipids
CA3057192A1 (en) 2017-03-23 2018-09-27 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
CA3059112A1 (en) 2017-04-05 2018-10-11 University Of Massachusetts Cep290 minigene therapy
CN110799492B (en) 2017-04-28 2023-06-27 爱康泰生治疗公司 Novel carbonyl lipid and lipid nanoparticle formulations for delivery of nucleic acids
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
EP3625345B1 (en) 2017-05-18 2023-05-24 ModernaTX, Inc. Modified messenger rna comprising functional rna elements
MA49395A (en) 2017-06-14 2020-04-22 Modernatx Inc POLYNUCLEOTIDES COAGULATION FACTOR VIII CODING
WO2018232357A1 (en) 2017-06-15 2018-12-20 Modernatx, Inc. Rna formulations
US20200138921A1 (en) 2017-06-23 2020-05-07 The Trustees Of Columbia University In The City Of New York Methods of preventing and treating diseases characterized by synaptic dysfunction and neurodegeneration including alzheimer's disease
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
TW202402809A (en) 2017-08-15 2024-01-16 美商歐米諾斯公司 Methods for treating and/or preventing graft-versus-host disease and/or diffuse alveolar hemorrhage and/or veno-occlusive disease associated with hematopoietic stem cell transplant
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
JP7275111B2 (en) 2017-08-31 2023-05-17 モデルナティエックス インコーポレイテッド Method for producing lipid nanoparticles
EP4219715A3 (en) 2017-09-08 2023-09-06 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
CA3082251A1 (en) 2017-10-16 2019-04-25 The Broad Institute, Inc. Uses of adenosine base editors
US20210236644A1 (en) 2017-11-10 2021-08-05 Cocoon Biotech Inc. Ocular applications of silk-based products
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
JP7423522B2 (en) 2017-11-22 2024-01-29 モダーナティエックス・インコーポレイテッド Polynucleotide encoding ornithine transcarbamylase for the treatment of urea cycle disorders
MA50802A (en) 2017-11-22 2020-09-30 Modernatx Inc POLYNUCLEOTIDES CODING FOR ALPHA AND BETA SUB-UNITS OF PROPIONYL-COA CARBOXYLASE FOR THE TREATMENT OF PROPIONIC ACIDEMIA
EP3735270A1 (en) 2018-01-05 2020-11-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
KR20200135433A (en) 2018-03-23 2020-12-02 유니버시티 오브 매사추세츠 Gene therapy for the treatment of bone disorders
KR20200138254A (en) 2018-03-30 2020-12-09 암젠 인크 C-terminal antibody variant
EP3773745A1 (en) 2018-04-11 2021-02-17 ModernaTX, Inc. Messenger rna comprising functional rna elements
WO2019197845A1 (en) 2018-04-12 2019-10-17 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
US11904081B2 (en) 2018-05-11 2024-02-20 Lupagen, Inc. Systems and methods for closed loop, real-time modifications of patient cells
EP3794130A4 (en) 2018-05-16 2022-07-27 Synthego Corporation Methods and systems for guide rna design and use
MA52709A (en) 2018-05-23 2021-03-31 Modernatx Inc DNA ADMINISTRATION
US11584714B2 (en) 2018-05-29 2023-02-21 Omeros Corporation MASP-2 inhibitors and methods of use
US20220184185A1 (en) 2018-07-25 2022-06-16 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
JP2021534101A (en) 2018-08-09 2021-12-09 ヴェルソー セラピューティクス, インコーポレイテッド Oligonucleotide compositions for targeting CCR2 and CSF1R and their use
US20210170005A1 (en) 2018-08-15 2021-06-10 University Of Florida Research Foundation, Inc. Methods of sensitizing tumors to treatment with immune checkpoint inhibitors
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056161A1 (en) 2018-09-12 2020-03-19 University Of Florida Research Foundation, Inc. Slow-cycling cell-rna based nanoparticle vaccine to treat cancer
EP3849594A2 (en) 2018-09-13 2021-07-21 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
MA53609A (en) 2018-09-13 2021-07-21 Modernatx Inc POLYNUCLEOTIDES ENCODED GLUCOSE-6-PHOSPHATASE FOR THE TREATMENT OF GLYCOGENOSIS
AU2019339430A1 (en) 2018-09-14 2021-04-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide A1 for the treatment of Crigler-Najjar Syndrome
JP2022500543A (en) 2018-09-19 2022-01-04 モデルナティーエックス, インコーポレイテッド High-purity PEG lipids and their use
EP3852732A1 (en) 2018-09-19 2021-07-28 ModernaTX, Inc. Peg lipids and uses thereof
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
CN113164625A (en) 2018-09-28 2021-07-23 哈佛大学的校长及成员们 Mutant inverse tetracycline transactivators for gene expression
MX2021003663A (en) 2018-09-28 2021-05-28 Harvard College Cellular reprogramming to reverse aging and promote organ and tissue regeneration.
US20220001026A1 (en) 2018-11-08 2022-01-06 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US20220033824A1 (en) 2018-11-29 2022-02-03 University Of Massachusetts Modulation of sptlc1 via recombinant adeno-associated vectors
EP3908568A1 (en) 2019-01-11 2021-11-17 Acuitas Therapeutics, Inc. Lipids for lipid nanoparticle delivery of active agents
JP2022523055A (en) 2019-01-25 2022-04-21 シンテゴ コーポレイション Systems and Methods for Modulating CRISPR Activity
EP3927380A1 (en) 2019-02-22 2021-12-29 University of Massachusetts Oxr1 gene therapy
MX2021011426A (en) 2019-03-19 2022-03-11 Broad Inst Inc Methods and compositions for editing nucleotide sequences.
EP3953473A1 (en) 2019-04-12 2022-02-16 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
EP3959232A4 (en) 2019-04-24 2023-01-18 University of Massachusetts Aav capsid chimeric antigen receptors and uses thereof
EP3962521A1 (en) 2019-05-02 2022-03-09 University of Florida Research Foundation, Inc. Compositions for treatment of diffuse intrinsic pontine glioma
JP2022532078A (en) 2019-05-08 2022-07-13 アストラゼネカ アクチボラグ Compositions for skin and wounds and methods of their use
EP3987027A1 (en) 2019-06-24 2022-04-27 ModernaTX, Inc. Endonuclease-resistant messenger rna and uses thereof
US20220387628A1 (en) 2019-06-24 2022-12-08 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
US20230000997A1 (en) 2019-08-06 2023-01-05 L.E.A.F. Holdings Group Llc Processes of preparing polyglutamated antifolates and uses of their compositions
US20230097090A1 (en) 2019-08-14 2023-03-30 Acuitas Therapeutics, Inc. Improved lipid nanoparticles for delivery of nucleic acids
KR20220079857A (en) 2019-09-13 2022-06-14 루트거스, 더 스테이트 유니버시티 오브 뉴 저지 AAV-compatible laminin-linker polymeric protein
JP2022548304A (en) 2019-09-19 2022-11-17 モデルナティエックス インコーポレイテッド Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
JP2023504543A (en) 2019-12-04 2023-02-03 オメロス コーポレーション MASP-2 inhibitors and methods of use
CA3168055A1 (en) 2020-02-21 2021-08-26 Emmanuel John Simons Compositions and methods for treating non-age-associated hearing impairment in a human subject
EP4127186A1 (en) 2020-03-24 2023-02-08 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing gaucher therapeutics
US20230138409A1 (en) 2020-03-24 2023-05-04 Generation Bio Co. Non-viral dna vectors and uses thereof for expressing factor ix therapeutics
MX2022012279A (en) 2020-03-31 2023-02-23 Univ Massachusetts Aav capsids variants and uses thereof.
US20230226169A1 (en) 2020-04-01 2023-07-20 University Of Florida Research Foundation Incorporated Multilamellar rna nanoparticle vaccine against sars-cov-2
EP4150626A1 (en) 2020-04-29 2023-03-22 The Broad Institute Inc. Machine learning accelerated protein engineering through fitness prediction
US20210346416A1 (en) * 2020-05-05 2021-11-11 Ralph Lipp Pharmaceutical Compositions and Methods for their Use in Antiviral Therapy
IL297761A (en) 2020-05-08 2022-12-01 Broad Inst Inc Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
AU2021275239A1 (en) 2020-05-21 2022-12-15 Board Of Regents, The University Of Texas System T cell receptors with VGLL1 specificity and uses thereof
JP2023527875A (en) 2020-06-01 2023-06-30 モダーナティエックス・インコーポレイテッド Phenylalanine hydroxylase variants and uses thereof
IL300526A (en) 2020-08-17 2023-04-01 Massachusetts Inst Technology Shank3 gene therapy approaches
US20230406895A1 (en) 2020-11-13 2023-12-21 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
EP4255457A1 (en) 2020-12-03 2023-10-11 University of Massachusetts Development of novel gene therapeutics for fibrodysplasia ossificans progressiva
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022198101A1 (en) 2021-03-19 2022-09-22 Trained Therapeutix Discovery, Inc. Compounds for regulating trained immunity, and their methods of use
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
JP2024512026A (en) 2021-03-24 2024-03-18 モデルナティエックス インコーポレイテッド Lipid nanoparticles and polynucleotide encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
EP4314292A1 (en) 2021-03-26 2024-02-07 MiNA Therapeutics Limited Tmem173 sarna compositions and methods of use
JP2024512669A (en) 2021-03-31 2024-03-19 フラグシップ パイオニアリング イノベーションズ ブイ,インコーポレーテッド Tanotransmission polypeptides and their use in the treatment of cancer
WO2022232286A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing anti-coronavirus antibodies and uses thereof
IL308404A (en) 2021-04-27 2024-01-01 Generation Bio Co Non-viral dna vectors expressing therapeutic antibodies and uses thereof
KR20240017813A (en) 2021-05-10 2024-02-08 이슘 리서치 디벨롭먼트 컴퍼니 오브 더 히브루 유니버시티 오브 예루살렘 엘티디. Pharmaceutical composition for treating neurological diseases
EP4355882A2 (en) 2021-06-15 2024-04-24 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
CA3233097A1 (en) 2021-09-30 2023-04-06 Katherine Diane GRIBBLE Compositions and methods for treating kcnq4-associated hearing loss
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
TW202334080A (en) 2021-11-08 2023-09-01 美商歐納醫療公司 Lipid nanoparticle compositions for delivering circular polynucleotides
WO2023095874A1 (en) * 2021-11-25 2023-06-01 国立大学法人長崎大学 Lipid compound, liposome, exosome, lipid nanoparticle and drug delivery system
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
CN114306641A (en) * 2022-02-17 2022-04-12 大连民族大学 Cholesterol amino derivative-sucrose ester type cationic liposome/gene complex and preparation method thereof
WO2023164545A1 (en) 2022-02-23 2023-08-31 Massachusetts Institute Of Technology Methods for upregulating shank3 expression
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023177655A1 (en) 2022-03-14 2023-09-21 Generation Bio Co. Heterologous prime boost vaccine compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196851A1 (en) 2022-04-06 2023-10-12 President And Fellows Of Harvard College Reversing aging of the central nervous system
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression
WO2024040222A1 (en) 2022-08-19 2024-02-22 Generation Bio Co. Cleavable closed-ended dna (cedna) and methods of use thereof
WO2024077191A1 (en) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additionalpolypeptides and their use in treating cancer

Family Cites Families (143)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4244946A (en) 1979-06-11 1981-01-13 The Salk Institute For Biological Studies Water-soluble peptides affecting gonadal function
US4305872A (en) 1979-10-19 1981-12-15 Kenneth Wingrove Polypeptide derivatives
US4316891A (en) 1980-06-14 1982-02-23 The Salk Institute For Biological Studies Extended N-terminal somatostatin
US5059591B1 (en) 1983-05-26 2000-04-25 Liposome Co Inc Drug preparations of reduced toxicity
US4544545A (en) * 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US5545412A (en) 1985-01-07 1996-08-13 Syntex (U.S.A.) Inc. N-[1, (1-1)-dialkyloxy]-and N-[1, (1-1)-dialkenyloxy]-alk-1-yl-n,n,n-tetrasubstituted ammonium lipids and uses therefor
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
US5342921A (en) 1985-03-28 1994-08-30 Chiron Corporation Superoxide dismutase fusion polypeptides for expression of mammalian proteins
US5374548A (en) * 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor
US5540933A (en) * 1985-05-31 1996-07-30 La Jolla Cancer Research Foundation Isolation and use of fibronectin receptor
EP0238645A1 (en) 1985-10-03 1987-09-30 Biotechnology Research Partners, Ltd. Novel lipoprotein-based drug-delivery systems
US4959217A (en) 1986-05-22 1990-09-25 Syntex (U.S.A.) Inc. Delayed/sustained release of macromolecules
US5560929A (en) 1986-08-18 1996-10-01 The Dow Chemical Company Structured copolymers and their use as absorbents, gels and carriers of metal ions
US5166320A (en) 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
US5283634A (en) * 1987-06-18 1994-02-01 Canon Kabushiki Kaisha Luminance signal correcting method
US5008202A (en) * 1988-11-29 1991-04-16 Sequoia Turner Corporation Blood diluent for red blood cell analysis
US5196510A (en) 1988-12-29 1993-03-23 Cytogen Corporation Molecular recognition units
US5043164A (en) 1989-01-17 1991-08-27 The University Of Tennessee Research Corporation Blood-stable, cholesterol-free liposomes
DE3908047A1 (en) 1989-03-13 1990-09-20 Desitin Arzneimittel Gmbh HIGH DISPERSES PHARMACEUTICAL COMPOSITION
US5354844A (en) 1989-03-16 1994-10-11 Boehringer Ingelheim International Gmbh Protein-polycation conjugates
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US6214804B1 (en) 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5165925A (en) 1989-05-02 1992-11-24 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Oregon State University Vaccine for immunizing fish against infectious pancreatic necrosis virus
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5100662A (en) * 1989-08-23 1992-03-31 The Liposome Company, Inc. Steroidal liposomes exhibiting enhanced stability
US5286634A (en) 1989-09-28 1994-02-15 Stadler Joan K Synergistic method for host cell transformation
WO1991004753A1 (en) 1989-10-02 1991-04-18 Cetus Corporation Conjugates of antisense oligonucleotides and therapeutic uses thereof
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5670617A (en) 1989-12-21 1997-09-23 Biogen Inc Nucleic acid conjugates of tat-derived transport polypeptides
ES2091916T3 (en) 1989-12-21 1996-11-16 Whitehead Biomedical Inst METHOD FOR SUPPLYING MOLECULES INSIDE EUCARIOTIC CELLS.
US5583198A (en) 1989-12-22 1996-12-10 Commonwealth Scientific And Industrial Research Organization Amino acids, peptides or derivatives thereof coupled to fats
US5705187A (en) 1989-12-22 1998-01-06 Imarx Pharmaceutical Corp. Compositions of lipids and stabilizing materials
US5279833A (en) * 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US6011020A (en) 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US6147204A (en) 1990-06-11 2000-11-14 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
NZ239893A (en) 1990-09-25 1993-11-25 Hoechst Japan A method for introducing a foreign dna into a cell
DK0560932T3 (en) 1990-12-04 1997-02-17 Univ Texas
US5589392A (en) 1991-01-14 1996-12-31 Stratagene Nucleic acid construct encoding a nuclear transport peptide operatively linked to an inducible promoter
US5328984A (en) 1991-03-04 1994-07-12 The United States As Represented By The Department Of Health & Human Services Recombinant chimeric proteins deliverable across cellular membranes into cytosol of target cells
AU667680B2 (en) * 1991-08-07 1996-04-04 Albert Einstein College Of Medicine Of Yeshiva University Proliferation of hepatocyte precursors
US5283185A (en) * 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
NZ244306A (en) 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5298422A (en) * 1991-11-06 1994-03-29 Baylor College Of Medicine Myogenic vector systems
DE4139001A1 (en) 1991-11-27 1993-06-03 Boehringer Mannheim Gmbh PROCESS FOR INJECTION OF NUCLEAR ACIDS IN CELLS
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
US5858784A (en) 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US5922859A (en) * 1992-02-01 1999-07-13 Boehringer Ingelheim International Gmbh Complexes containing nucleic acid which can be taken-up by endocytosis into higher eukaryotic cells
EP0643835B1 (en) 1992-02-24 2001-04-18 Genelabs Technologies, Inc. Htlv-i/htlv-ii assay and method
IL101241A (en) 1992-03-16 1997-11-20 Yissum Res Dev Co Pharmaceutical or cosmetic composition comprising stabilized oil-in-water type emulsion as carrier
JPH07505283A (en) 1992-03-20 1995-06-15 ベイラー・カレッジ・オブ・メディシン DNA transporter system and method of use
US6033884A (en) * 1992-03-20 2000-03-07 Baylor College Of Medicine Nucleic acid transporter systems and methods of use
EP1236473A3 (en) 1992-04-03 2003-01-15 The Regents Of The University Of California Self-assembling polynucleotide delivery system
US6113946A (en) 1992-04-03 2000-09-05 The Regents Of The University Of California Self-assembling polynucleotide delivery system comprising dendrimer polycations
IL105914A0 (en) 1992-06-04 1993-10-20 Univ California Methods and compositions for in vivo gene therapy
US5334761A (en) * 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5266106A (en) 1992-10-22 1993-11-30 Xerox Corporation Ink compositions with dendrimer grafts
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5674977A (en) 1993-02-05 1997-10-07 The Ontario Cancer Institute Branched synthetic peptide conjugate
AU6133394A (en) 1993-02-09 1994-08-29 Scripps Research Institute, The Targeting and delivery of genes and antiviral agents into cells by the adenovirus penton
US5532142A (en) 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
WO1994018834A1 (en) 1993-02-16 1994-09-01 Virginia Tech Intellectual Properties, Inc. Polyelectrolyte dna conjugation and genetic transformation of an animal
US5491074A (en) 1993-04-01 1996-02-13 Affymax Technologies Nv Association peptides
DE4311651A1 (en) 1993-04-08 1994-10-13 Boehringer Ingelheim Int Virus for the transport of foreign DNA into higher eukaryotic cells
WO1994023751A1 (en) 1993-04-14 1994-10-27 Boehringer Mannheim Gmbh Nucleic acid tranfer peptides and their use for injecting nucleic acids into eucaryotic cells
US5554382A (en) 1993-05-28 1996-09-10 Aphios Corporation Methods and apparatus for making liposomes
US5776486A (en) 1993-05-28 1998-07-07 Aphios Corporation Methods and apparatus for making liposomes containing hydrophobic drugs
WO1994027435A1 (en) 1993-06-01 1994-12-08 Life Technologies, Inc. Genetic immunization with cationic lipids
WO1995002698A1 (en) 1993-07-12 1995-01-26 Life Technologies, Inc. Composition and methods for transfecting eukaryotic cells
EP0708637B1 (en) 1993-07-14 2004-01-28 The Regents of the University of California Self-assembling polynucleotide delivery system comprising dendrimer polycations
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
AU688428B2 (en) 1993-11-09 1998-03-12 Johns Hopkins University, The Generation of high titers of recombinant AAV vectors
US5674908A (en) 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
ATE190663T1 (en) 1994-01-28 2000-04-15 Targeted Genetics Corp CD69 - TRANSCRIPTION REGULATORY ELEMENTS
US5928944A (en) 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US6075012A (en) 1994-02-11 2000-06-13 Life Technologies, Inc. Reagents for intracellular delivery of macromolecules
US5844107A (en) * 1994-03-23 1998-12-01 Case Western Reserve University Compacted nucleic acids and their delivery to cells
ES2316148T3 (en) 1994-03-23 2009-04-01 Ohio University COMPACTED NUCLEIC ACIDS AND ITS SUPPLY TO CELLS.
US5972901A (en) 1994-03-23 1999-10-26 Case Western Reserve University Serpin enzyme complex receptor--mediated gene transfer
US6077835A (en) 1994-03-23 2000-06-20 Case Western Reserve University Delivery of compacted nucleic acid to cells
FR2719316B1 (en) * 1994-04-28 1996-05-31 Idm New nucleic acid and polymer complexes, their preparation process and their use for cell transfection.
US5670347A (en) 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
US5807746A (en) 1994-06-13 1998-09-15 Vanderbilt University Method for importing biologically active molecules into cells
US5908635A (en) 1994-08-05 1999-06-01 The United States Of America As Represented By The Department Of Health And Human Services Method for the liposomal delivery of nucleic acids
AUPM747694A0 (en) 1994-08-16 1994-09-08 Commonwealth Scientific And Industrial Research Organisation Delivery of nucleic acids and peptides
US5837533A (en) 1994-09-28 1998-11-17 American Home Products Corporation Complexes comprising a nucleic acid bound to a cationic polyamine having an endosome disruption agent
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5744355A (en) * 1994-10-18 1998-04-28 Mayo Foundation For Medical Education And Research cDNA cloning and expression of human liver estrogen sulfotransferase
US5627159A (en) 1994-10-27 1997-05-06 Life Technologies, Inc. Enhancement of lipid cationic transfections in the presence of serum
US5587441A (en) 1994-11-08 1996-12-24 Cornell Research Foundation, Inc. Hyperbranched polymers from AB monomers
US5719131A (en) 1994-12-09 1998-02-17 Genzyme Corporation Cationic amphiphiles containing dialkylamine lipophilic groups for intracellular delivery of therapeutic molecules
US5939401A (en) 1994-12-09 1999-08-17 Genzyme Corporation Cationic amphiphile compositions for intracellular delivery of therapeutic molecules
US5840710A (en) 1994-12-09 1998-11-24 Genzyme Corporation Cationic amphiphiles containing ester or ether-linked lipophilic groups for intracellular delivery of therapeutic molecules
US5948767A (en) 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5650096A (en) 1994-12-09 1997-07-22 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US5635487A (en) 1994-12-29 1997-06-03 Wolff; Jon A. Amphipathic, micellar delivery systems for biologically active polyions
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US6008202A (en) 1995-01-23 1999-12-28 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5830430A (en) 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
JP4335310B2 (en) 1995-06-07 2009-09-30 ザ ユニバーシティ オブ ブリティッシュ コロンビア Lipid-nucleic acid particles prepared through hydrophobic lipid-nucleic acid complex intermediates and use for gene transfer
EP0874910A4 (en) 1995-06-07 1999-04-21 Life Technologies Inc Peptide-enhanced cationic lipid transfections
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5705385A (en) 1995-06-07 1998-01-06 Inex Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US20030069173A1 (en) 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US5753262A (en) 1995-06-07 1998-05-19 Aronex Pharmaceuticals, Inc. Cationic lipid acid salt of 3beta N- (N', N'-dimethylaminoethane) - carbamoyl!cholestrol and halogenated solvent-free preliposomal lyophilate thereof
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US5908777A (en) 1995-06-23 1999-06-01 University Of Pittsburgh Lipidic vector for nucleic acid delivery
US5672662A (en) 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
US6033848A (en) 1995-08-01 2000-03-07 Incyte Pharmaceuticals, Inc. Human ice homolog
US5744335A (en) 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US6086913A (en) 1995-11-01 2000-07-11 University Of British Columbia Liposomal delivery of AAV vectors
US6126964A (en) 1996-01-04 2000-10-03 Mirus Corporation Process of making a compound by forming a polymer from a template drug
AUPN741696A0 (en) 1996-01-05 1996-01-25 Commonwealth Scientific And Industrial Research Organisation Delivery of nucleic acids ii
US5994316A (en) 1996-02-21 1999-11-30 The Immune Response Corporation Method of preparing polynucleotide-carrier complexes for delivery to cells
US5811297A (en) 1996-03-07 1998-09-22 Amba Biosciences, Llc Immortalized hematopoietic cell lines, cell system thereof with stromal cells, in vitro, ex vivo and in vivo uses, & in vitro generation of dendritic cells and macrophages
JPH09268195A (en) * 1996-04-02 1997-10-14 Mitsubishi Chem Corp Production of sucrose fatty acid ester
CA2264140A1 (en) 1996-08-26 1998-03-05 Transgene S.A. Cationic lipid-nucleic acid complexes
US5905777A (en) * 1996-09-27 1999-05-18 At&T Corp. E-mail paging system
US5861397A (en) 1996-10-03 1999-01-19 Vical Incorporated Piperazine based cytofectins
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
US6224903B1 (en) 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
JP2001510457A (en) 1996-11-12 2001-07-31 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Preparation of stable lipid-nucleic acid complex formulations effective for in vivo delivery
US6017735A (en) 1997-01-23 2000-01-25 Marie Curie Cancer Care Materials and methods for intracellular transport and their uses
US5948925A (en) 1997-05-06 1999-09-07 Genzyme Corporation Cationic amphiphiles containing linkers derived from neutral or positively charged amino acids
US6835395B1 (en) 1997-05-14 2004-12-28 The University Of British Columbia Composition containing small multilamellar oligodeoxynucleotide-containing lipid vesicles
EP2138191A1 (en) 1998-01-05 2009-12-30 University Of Washington Enhanced transport using membrane disruptive agents
US6271209B1 (en) 1998-04-03 2001-08-07 Valentis, Inc. Cationic lipid formulation delivering nucleic acid to peritoneal tumors
US6043390A (en) 1998-04-03 2000-03-28 The Regents Of The University Of California Pentaerythritol lipid derivatives and nucleic-acid complexes
US6011202A (en) * 1998-07-28 2000-01-04 Delta And Pine Land Company Soybean cultivar 95-00981
JP4854853B2 (en) 1998-11-12 2012-01-18 ライフ テクノロジーズ コーポレーション Transfection drug
US6773920B1 (en) 1999-03-31 2004-08-10 Invitrogen Corporation Delivery of functional protein sequences by translocating polypeptides
US6037176A (en) 1999-06-25 2000-03-14 Isis Pharmaceuticals Inc. Antisense inhibition of integrin beta 3 expression
AU2002232387A1 (en) 2000-10-27 2002-05-06 Invitrogen Corporation Method for introducing antisense oligonucleotides into eucaryotic cells
US20040102606A1 (en) 2001-04-24 2004-05-27 Danuta Balicki Histone H2A -derived peptides useful in gene delivery
US20030203865A1 (en) 2001-04-30 2003-10-30 Pierrot Harvie Lipid-comprising drug delivery complexes and methods for their production
EP1587908A4 (en) 2003-01-09 2008-02-20 Invitrogen Corp Cellular delivery and activation polypeptide-nucleic acid complexes

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
B.A. DEMENEIX: "TEMPORAL AND SPATIAL EXPRESSION OF LIPOSPERMINE-COMPACTED GENES TRANSFERRED INTO CHICK EMBRYOS IN VIVO", BIOTECHNIQUES, vol. 16, no. 3, March 1994 (1994-03-01), NACICK (US), pages 496 - 501, XP002006209 *
J.-P. BEHR ET AL.: "EFFICIENT GENE TRANSFER INTO MAMMALIAN PRIMARY ENDOCRINE CELLS WITH LIPOPOLYAMINE-COATED DNA", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 86, September 1989 (1989-09-01), WASHINGTON (US), pages 6982 - 6986, XP002006207 *
K.M. HUI ET AL.: "INDUCTION OF ALLOREACTIVE CYTOTOXIC T LYMPHOCYTES BY INTRA-SPLENIC IMMUNIZATION WITH ALLOGENIC CLASS I MAJOR HISTOCOMPATIBILITY COMPLEX DNA AND DC-CHOL CATIONIC LIPOSOMES", JOURNAL OF LIPOSOME RESEARCH, vol. 4, no. 3, November 1994 (1994-11-01), NEW YORK (US), pages 1075 - 1090, XP000476607 *
M.J. STEWART ET AL.: "GENE TRANSFER IN VIVO WITH DNA-LIPOSOME COMPLEXES: SAFETY AND ACUTE TOXICITY IN MICE", HUMAN GENE THERAPY, vol. 3, no. 3, June 1992 (1992-06-01), pages 267 - 275, XP002006208 *

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6107286A (en) * 1994-12-05 2000-08-22 Rhone-Poulenc Rorer S.A. Lipopolyamines as transfection agents and pharamaceutical uses thereof
US6051429A (en) * 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6586003B2 (en) 1995-09-26 2003-07-01 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
US6120794A (en) * 1995-09-26 2000-09-19 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
US6468986B1 (en) 1996-08-13 2002-10-22 Chiron Corporation Compositions and methods for polynucleotide delivery
US7462592B2 (en) 1996-08-13 2008-12-09 Novartis Vaccines And Diagnostics, Inc. Compositions and methods for polynucleotide delivery
US6251433B1 (en) 1996-08-13 2001-06-26 Chiron Corporation Polycationic polymers
JP2008239631A (en) * 1996-11-12 2008-10-09 Regents Of The Univ Of California Preparation of stable formulation of lipid-nucleic acid complex effective for in vivo delivery
JP4551464B2 (en) * 1996-11-12 2010-09-29 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Preparation of a stable formulation of lipid-nucleic acid complex effective for in vivo delivery
US5962429A (en) * 1996-11-22 1999-10-05 University Of Iowa Complexes of adenovirus with cationic molecules
US6376248B1 (en) 1997-03-14 2002-04-23 Life Technologies, Inc. Peptide-enhanced transfections
US5948878A (en) * 1997-04-15 1999-09-07 Burgess; Stephen W. Cationic polymers for nucleic acid transfection and bioactive agent delivery
US6436708B1 (en) 1997-04-17 2002-08-20 Paola Leone Delivery system for gene therapy to the brain
WO1998046273A2 (en) * 1997-04-17 1998-10-22 Paola Leone Delivery system for gene therapy to the brain
WO1998046273A3 (en) * 1997-04-17 1999-01-21 Paola Leone Delivery system for gene therapy to the brain
US7214384B2 (en) 1997-08-13 2007-05-08 Novartis Vaccines And Diagnostics, Inc. Lipid-conjugated polyamide compounds
US6572881B1 (en) 1997-08-13 2003-06-03 Chiron Corporation Lipid-conjugated polyamide compounds and related compositions and methods thereof
US6569450B1 (en) 1997-08-13 2003-05-27 Chiron Corporation Lipid-conjugated polyamide compounds and related compositions and methods thereof
JP2010031028A (en) * 1998-05-12 2010-02-12 Regents Of The Univ Of California Method of forming protein-linked lipidic microparticle, and composition thereof
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US6855549B1 (en) 1998-11-23 2005-02-15 The University Of Iowa Research Foundation Methods and compositions for increasing the infectivity of gene transfer vectors
DE10322123A1 (en) * 2003-05-12 2004-12-16 Novosom Ag Stable anionic liposome depot system for prolonged release of protein or peptide drugs, e.g. insulin, comprising phosphatidyl choline, cholesterol, anionic lipid(s) and cationic polymer
EP2569437A1 (en) * 2010-05-10 2013-03-20 Qiagen GmbH Method for transfecting a eukaryotic cell
US10238733B2 (en) 2010-07-06 2019-03-26 Glaxosmithkline Biologicals S.A. Cationic oil-in-water emulsions
US9295646B2 (en) 2010-07-06 2016-03-29 Novartis Ag Cationic oil-in-water emulsions
US11135287B2 (en) 2010-07-06 2021-10-05 Glaxosmithkline Biologicals Sa Method for preparing composition comprising a cationic oil-in-water emulsion
US9636410B2 (en) 2011-07-06 2017-05-02 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
US9655845B2 (en) 2011-07-06 2017-05-23 Glaxosmithkline Biologicals, S.A. Oil-in-water emulsions that contain nucleic acids
US10183074B2 (en) 2011-07-06 2019-01-22 Glaxosmithkline Biologicals S.A. Cationic oil-in-water emulsions
US10307374B2 (en) 2011-07-06 2019-06-04 Glaxosmithkline Biologicals S.A. Oil-in-water emulsions that contain nucleic acids
US11026890B2 (en) 2011-07-06 2021-06-08 Glaxosmithkline Biologicals Sa Oil-in-water emulsions that contain nucleic acids
US11167028B2 (en) 2011-07-06 2021-11-09 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10792362B2 (en) 2014-07-15 2020-10-06 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US11872285B2 (en) 2014-07-15 2024-01-16 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells

Also Published As

Publication number Publication date
US5795587A (en) 1998-08-18
US20130281382A1 (en) 2013-10-24
CA2211118C (en) 2009-06-30
CN1177291A (en) 1998-03-25
JP4074338B2 (en) 2008-04-09
DE69624801D1 (en) 2002-12-19
US20120178702A1 (en) 2012-07-12
US20100184953A1 (en) 2010-07-22
MX9705572A (en) 1998-06-28
US7655468B2 (en) 2010-02-02
AU709773B2 (en) 1999-09-09
US8771728B2 (en) 2014-07-08
US7993672B2 (en) 2011-08-09
CA2211118A1 (en) 1996-08-01
DK0814777T3 (en) 2003-03-03
EP0814777A1 (en) 1998-01-07
JPH10512882A (en) 1998-12-08
IL116856A0 (en) 1996-07-23
ES2186769T3 (en) 2003-05-16
EP0814777B1 (en) 2002-11-13
ZA96503B (en) 1996-08-07
DE69624801T2 (en) 2003-04-10
ATE227563T1 (en) 2002-11-15
AU4703796A (en) 1996-08-14
PT814777E (en) 2003-03-31
US20080153166A1 (en) 2008-06-26

Similar Documents

Publication Publication Date Title
EP0814777B1 (en) Stable lipid-comprising drug delivery complexes and methods for their production
US7361640B2 (en) Stable lipid-comprising drug delivery complexes and methods for their production
US5908777A (en) Lipidic vector for nucleic acid delivery
US5705385A (en) Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5990089A (en) Self-assembling polynucleotide delivery system comprising dendrimer polycations
US6210708B1 (en) Cationic virosomes as transfer system for genetic material
KR19980702200A (en) NUCLEIC ACID CONTAINING COMPOSITIONS AND METHODS AND USES
WO1997028817A1 (en) Receptor ligand-facilitated delivery of biologically active molecules
US20010048940A1 (en) Cationic amphiphile micellar complexes
EP0904282B1 (en) Novel cationic cholesteryl derivatives containing cyclic polar groups
US6583131B2 (en) Cationic cholesteryl derivatives containing cyclic polar groups
AU4698499A (en) Cationic amphiphile micellar complexes
MXPA97005572A (en) Stable complexes for the supply of pharmacosque contain lipids and methods for your production
AU715376C (en) Novel cationic cholesteryl derivatives containing cyclic polar groups
MXPA97006016A (en) Composition containing nucleic acids, preparation and use

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 96192287.7

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AZ BY KG KZ RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2211118

Country of ref document: CA

Ref document number: 2211118

Country of ref document: CA

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 1996 522954

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/1997/005572

Country of ref document: MX

Ref document number: 1019970705016

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1996902747

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1996902747

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1019970705016

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1019970705016

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1996902747

Country of ref document: EP