WO2011056682A1 - Lipides à têtes polaires inversées, compositions particulaires lipidiques comprenant les lipides à têtes polaires inversées, et procédés d'administration d'acides nucléiques - Google Patents

Lipides à têtes polaires inversées, compositions particulaires lipidiques comprenant les lipides à têtes polaires inversées, et procédés d'administration d'acides nucléiques Download PDF

Info

Publication number
WO2011056682A1
WO2011056682A1 PCT/US2010/054351 US2010054351W WO2011056682A1 WO 2011056682 A1 WO2011056682 A1 WO 2011056682A1 US 2010054351 W US2010054351 W US 2010054351W WO 2011056682 A1 WO2011056682 A1 WO 2011056682A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
lipids
headgroup
acid
dofab
Prior art date
Application number
PCT/US2010/054351
Other languages
English (en)
Inventor
Alex K. K. Leung
Srinivasulu Masuna
Marco Ciufolini
Pieter R. Cullis
Original Assignee
The University Of British Columbia
Laherty, Carol, D.
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 The University Of British Columbia, Laherty, Carol, D. filed Critical The University Of British Columbia
Publication of WO2011056682A1 publication Critical patent/WO2011056682A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/06Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton
    • C07C229/10Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings
    • C07C229/12Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of acyclic carbon skeletons
    • 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
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • 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

Definitions

  • the present invention relates to lipids and lipid particles useful in the delivery of agents, including nucleic acids, to cells, as well as related methods of use in the treatment or prevention of diseases and disorders.
  • Lipid particle delivery systems are currently the leading systems for the delivery of oligonucleotides (OGN) such as small interfering RNA (siRNA) in vivo.
  • OGN oligonucleotides
  • siRNA small interfering RNA
  • the cationic lipids contained in current lipid particle formulations designed for nucleic acid delivery perform two functions. First, they are required to encapsulate the negatively charged OGN in the lipid nanoparticles. Second, cationic lipids enhance intracellular delivery by combining with anionic lipids in target cell membranes to form non-lamellar structures, thus allowing macromolecules such as siRNA to penetrate to intracellular sites of action.
  • the permanently charged cationic lipids commonly used for transfection in vitro are not suitable for systemic use, since they are rapidly cleared from the circulation and also give rise to serious toxic effects, lonizable cationic lipids that are positively charged at low pH values and uncharged at physiological pH values have been used to at least partially overcome this problem.
  • the maximum amount of these lipids that can currently be present in lipid particles is in the range of 50 mol% due to stability issues, and at least 10% of a bilayer stabilizing lipid such as DSPC must be incorporated to maintain lipid particle stability.
  • the present invention provides novel lipids, and lipid particles comprising the same, as well as methods of using such particles for the delivery of agents to cells, including but not limited to the delivery of therapeutic nucleic acids to cells.
  • the present invention provides a zwitterionic lipid comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the headgroup.
  • the lipid has the following structure (A):
  • R 1 and R 2 are either the same or different and are each, independently, C6-C32alkyl;
  • R 3 is either hydrogen or d-C 6 alkyl
  • n and m are independently either 1 , 2, 3, or 4.
  • the headgroup of the lipid comprises
  • the lipid has the following structure (I):
  • Ri and R2 are each, independently, C6-C32alkyl.
  • the lipid has the following structure (I):
  • Ri and R2 are each, independently, C6-C32alkyl.
  • the lipid has one of the following structures:
  • the present invention provides a lipid particle comprising one or more zwitterionic lipids comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the headgroup.
  • the headgroup comprises 4-amino butyric acid (FAB).
  • the one or more zwitterionic lipids have a structure shown above.
  • the lipids particle further comprises cholesterol.
  • the lipid particle further comprises a PEG-lipid.
  • the lipid particle comprises one or more zwitterionic lipids comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the headgroup, cholesterol, and a PEG-lipid.
  • the lipid particle consists of one or more zwitterionic lipids comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the headgroup, cholesterol, and a PEG-lipid.
  • the one or more zwitterionic lipids have a headgroup comprising 4-amino butyric acid (FAB) or is selected from the structures shown above.
  • FAB 4-amino butyric acid
  • the lipid particles of the present invention comprise one or more encapsulated agent.
  • the one or more agent is a therapeutic agent.
  • the therapeutic agent is a nucleic acid, e.g., an interfering RNA.
  • the interfering RNA is a siRNA.
  • the present invention includes a method for delivering an agent to a cell comprising contacting a cell with a lipid particle of the present invention.
  • said contacting occurs in vitro or in vivo.
  • said cell is a mammalian cell, e.g., a human cell.
  • the present invention includes a method of treating or preventing a disease or disorder in a subject, comprising providing to the subject a lipid particle of the present invention comprising a therapeutic agent.
  • the subject is a mammal, e.g., a human.
  • the therapeutic agent is an interfering RNA.
  • the interfering RNA is an antisense RNA or a siRNA.
  • Figure 1 provides results from a TNS assay to determine the apparent pKa values of FABs incorporated in equimolar DLinFAB/DPPC/Cholesterol (-o-) and DOFAB/DPPC/Cholesterol (- ⁇ -) multilamellar vesicles.
  • An increase in TNS fluorescence correlates with an increase of positive charge.
  • pKa is defined as the point at which 50% of the molecules are charged.
  • Figure 2 provides results from a TNS assay to determine the apparent pKa values of DOFAB incorporated in DOFAB/PEG-c-DOMG (90:10 mol%) multilamellar vesicles. An increase in fluorescence correlates with an increase of positive surface charge. Data points plotted represents replicate trials.
  • Figure 3 demonstrates the influence of FAB lipids on the polymorphic phase properties of DOPC.
  • the 31 P NMR spectra shown were obtained for pure DOPC and equimolar mixtures of DPFAB/DOPC, DOFAB/DOPC and DLinFAB/DOPC in HEPES buffered saline, pH 7.0, at 25°C, 37°C and 50°C.
  • the axis shows PPM from 80 to -80.
  • the axis indicates PPM from 80 to -80.
  • Figure 4 demonstrates the influence of FAB lipids on the polymorphic phase properties of POPC.
  • 31 P NMR spectra corresponding to equimolar mixtures of DPFAB/POPC, DOFAB/POPC and DLinFAB/POPC in HEPES buffered saline at 25°C, 37°C and 50°C are shown.
  • the axis indicates PPM from 80 to -80.
  • Figure 5 shows the influence of DOFAB on the polymorphic phase properties of DOPC at 37°C.
  • 31 P NMR spectra were obtained for pure DOPC (top) and equimolar mixture of DOFAB/DOPC (bottom) in HEPES buffered saline, pH 7.0. The axis indicates PPM from 80 to -80.
  • Figure 6 shows the influence of DOFAB on the polymorphic phase properties of various phospholipids in equimolar mixtures.
  • 31 P NMR spectra were obtained for (A) DOFAB/milk sphingomyelin, (B) DOFAB/diPoPE, (C) DOFAB/DOPS, (D) DOFAB/DOPG and (E) DOFAB/DOPA. The axis indicates PPM from 80 to -80.
  • Figure 7 demonstrates the effect of DOPE on the phase properties of DOPC and POPC.
  • the 31 P NMR spectra shown were obtained for equimolar mixtures of DOPE/DOPC and DOPE/POPC in HEPES buffered saline at 25°C, 37°C and 50°C.
  • the axis indicates PPM from 80 to -80.
  • Figure 8 demonstrates the effect of cholesterol on the polymorphic phase properties of DOPC.
  • the 31 P NMR spectra shown were obtained for equimolar mixtures of DPFAB/DOPC/cholesterol, DOFAB/DOPC/cholesterol and DLinFAB/DOPC/cholesterol in HEPES buffered saline at 25°C, 37°C and 50°C.
  • the axis indicates PPM from 80 to -80.
  • Figure 9 demonstrates the effect of cholesterol on the polymorphic phase properties of POPC.
  • 31 P NMR spectra for equimolar mixtures of DPFAB/POPC/cholesterol, DOFAB/POPC/cholesterol and DLinFAB/POPC/cholesterol in HEPES buffered saline at 25°C, 37°C and 50°C are shown.
  • the axis indicates PPM from 80 to -80.
  • Figure 10 provides 31 P NMR spectra for equimolar mixtures of
  • FIG. 10A shows 31 P NMR spectra of diPoPE, indicating lamellar-to- hexagonal (HII) phase transition.
  • Figure 10B demonstrates that FAB lipids caused non-lamella phase transition in diPoPE LNs. The axis indicates PPM from 80 to -80.
  • Figure 1 1 provides immunoblots from cells treated for 24 or 48 hours with the indicated amounts of siRNA against the androgen receptor (siAR) or low-GC content control siRNA (loGC) encapsulated with DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%).
  • siRNA against the androgen receptor (siAR) or low-GC content control siRNA (loGC) encapsulated with DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%).
  • Figure 12 provides immunofluorescence images of LNCap cells treated with 10 g/mL of AR siRNA encapsulated with DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%) for 24 and 48 hours. Nuclei were stained blue with Hoescht's, and SPDiO fluorescence was observed in green. SPDiO fluorescence appears as light spots in the panels on the right, but was not observed in the panels to the left.
  • Figure 13 provides the chemical structures of DOFAB (top) and
  • Figure 14 depicts the proposed mechanisms of interaction between "reverse headgroup” and cellular phospholipids in generating membrane disruptove hexagonal phase structures.
  • Lipid nanoparticle (LN or LNP) delivery systems are currently the leading systems for the delivery of oligonucleotides (OGN) such as small interfering RNA (siRNA) in vivo.
  • OGN oligonucleotides
  • siRNA small interfering RNA
  • the cationic lipids contained in current LN formulations perform two functions. First, they are required to encapsulate the negatively charged OGN in the lipid nanoparticles. Second, cationic lipids enhance intracellular delivery by combining with anionic lipids in target cell membranes to form non-lamellar structures, thus allowing macromolecules such as siRNA to penetrate into intracellular sites of action.
  • the present invention provides a new class of zwitterionic lipids that combine with naturally occurring lipids, including zwitterionic lipids such as phosphatidylcholine (PC), to result in non-lamellar structures (such as the hexagonal ( ⁇ ⁇ ) phase), and which also have the ability to exhibit a positive charge for the encapsulation of nucleic acids, including oligonucleotides, such as siRNA.
  • the zwitterionic lipids of the present invention contain a "reversed" headgroup in which the positive charge is located near the acyl chain region, and the negative charge is located at the distal end of the headgroup.
  • FAB lipids which contain 4-amino butyric acid (FAB) headgroups in particular embodiments (FAB lipids), exhibit strong bilayer destabilizing characteristics in mixture with naturally occurring zwitterionic lipids such as PC.
  • FAB lipids are stronger bilayer destabilizing agents than dioleoylphosphatidylethanolamine (DOPE), which is commonly used as a "helper” lipid in nucleic acid-lipid particle formulations to assist with intracellular delivery properties.
  • DOPE dioleoylphosphatidylethanolamine
  • the reverse headgroup lipids of the present invention provide considerable advantages over previously used cationic lipids in lipid particles for the delivery of nucleic acids, such as siRNA.
  • these lipids may combine with phospholipids such as PC and PE in such a way that the positive and negative charges are apposed, leading to local charge neutralization (see Figure 14).
  • non-lamellar structures such as the hexagonal (Hn) phase.
  • anionic lipids results in formation of non-lamellar phase structure for all anionic and cationic lipids investigated (Lewis and McElhaney 2000; Hafez et al. 2001 ; Hafez and Cullis, 2001 ; Heyes et al. 2005; Koynova et al. 2006). It is believed that successful induction of non-lamellar structures by these reverse headgroup or FAB lipids may lead to efficient fusion of LN with the plasma membrane of target cells, resulting in delivery of encapsulated cargo to the cell interior.
  • a second point is that reverse headgroup lipids, such as FAB lipids, may exhibit unusual pH-dependent properties.
  • the FAB lipid is expected to exhibit a net positive charge, whereas a net negative charge is expected at pH values above the amino pKa of the FAB.
  • a net neutral lipid is expected.
  • These physical properties, particularly the phase behaviour of the FAB lipids, should therefore be sensitive to the pH of the aqueous medium.
  • unsaturated phosphatidyserines preferentially adopt the H N phase at pH values below the pKa of the carboxyl function (Hope and Cullis, 1980), where the headgroup changes from being negatively charged to being net neutral.
  • reverse headgroup lipids have potential for encapsulation and delivery of anionic macromolecules such as siRNA.
  • the reverse headgroup lipids such as the FAB lipids, are expected to exhibit a positive charge at pH values below the pKa of the carboxyl function and should therefore be able to be used to load negatively charged nucleic acid polymers such as siRNA into lipid-based nanoparticles (Maurer et al. 2001 , 2007).
  • LNs comprising reverse headgroup lipids have a low surface charge, which is required for long circulating LN to access target tissues other than the fixed and free macrophages of the reticuloendothelial system.
  • the present prevention further include a novel LN system that may induce fusion with the plasma membrane and, therefore, deliver nucleic acids, such as siRNA, directly into the cytoplasm of cells. This may bypass the endocytic pathway in certain embodiments. Electrostatic interactions between the cationic lipids of the LN carrier and the anionic lipids of the endosome can result in membrane-disruptive lipid structures. This disrupts the endosomal membrane, promoting the release of the siRNA payload.
  • a novel LN system may induce fusion with the plasma membrane and, therefore, deliver nucleic acids, such as siRNA, directly into the cytoplasm of cells. This may bypass the endocytic pathway in certain embodiments. Electrostatic interactions between the cationic lipids of the LN carrier and the anionic lipids of the endosome can result in membrane-disruptive lipid structures. This disrupts the endosomal membrane, promoting the release of the siRNA payload.
  • this new class of lipids may form similar electrostatic interactions with zwitterionic lipids (such as phosphatidylcholine, PC) of the plasma membrane.
  • zwitterionic lipids such as phosphatidylcholine, PC
  • These novel lipids are specifically designed to induce charge interaction with zwitterionic lipids such as PC by having a reversed charge orientation in the headgroup, hence the name reverse headgroup (RH) lipids. Reversal of charge orientation in the headgroup allows RH lipids to form electrostatic interactions with PC in a similar fashion as the interaction between cationic lipids and anionic endosomal lipids ( Figure 1 1 ).
  • LN formulated with RH lipids are anticipated to form membrane-disruptive structures with PC, resulting in fusion with the plasma membrane and delivery of their nucleic acid, e.g., siRNA, cargo directly into the cytoplasm.
  • nucleic acid e.g., siRNA
  • the present invention includes a zwitterionic lipid comprising a headgroup wherein the positive charge is located near the acyl chain region and the negative charge is located at the distal end of the headgroup.
  • lipids are referred to herein as reverse headgroup lipids, which are defined as zwitterionic lipids comprising a headgroup and acyl chains ⁇ e.g., two acyl chains), wherein the headgroup comprises a positive charge and a negative charge under certain pHs, and where the positive charge is located in a region of the headgroup near or proximal to the acyl chains as compared to the negative charge, which is located in a region of the headgroup distal to the acyl chains as compared to the positive charge.
  • a reverse headgroup lipid of the present invention has the followin structure (A):
  • R 1 and R 2 are either the same or different and are each, independently, C6-C32alkyl
  • R 3 is either hydrogen or Ci-C6alkyl
  • n and m are independently either 1 , 2, 3, or 4.
  • alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e. , contains one or more double and/or triple bonds), having from one to thirty-two carbon atoms, preferably one to six carbon atoms (Ci-C6alkyl), six to 32 carbon atoms (C6-C32alkyl), eight to twenty- four carbon atoms (C 8 -C 24 alkyl), or eight to twenty carbon atoms (C 8 -C 2 oalkyl), and which is attached to the rest of the molecule by a single bond.
  • Ri and F3 ⁇ 4 are each, independently, a saturated C6-C32alkyl, a saturated Ci2-C2 4 alkyl, a C12- C 24 alkenyl, or a Ci 2 -C 24 alkynyl.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4- acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1 ,2-disulfonic acid, ethanesulfonic acid, 2- hydroxyethanes
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, /V-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are is
  • the headgroup of a reverse headgroup lipids of the present invention comprises 4-amino butyric acid (FAB).
  • FAB 4-amino butyric acid
  • Reverse headgroup lipids comprising FAB headgroups are also referred to herein as FAB lipids.
  • the lipid has the following structure I):
  • Ri and R 2 are each, independently, C6-C 32 alkyl.
  • the lipid has the following structure (I):
  • Ri and F3 ⁇ 4 are each, independently, C6-C32alkyl.
  • alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e. , contains one or more double and/or triple bonds), having from one to thirty-two carbon atoms, preferably one to six carbon atoms (Ci-C6alkyl), six to 32 carbon atoms (C6-C32alkyl), eight to twenty- four carbon atoms (C 8 -C 24 alkyl), or eight to twenty carbon atoms (C 8 -C 2 oalkyl), and which is attached to the rest of the molecule by a single bond.
  • lipids of structure (I), Ri and R 2 are each, independently, a saturated C6-C32alkyl, a saturated Ci2-C2 4 alkyl, a C12- C2 4 alkenyl, or a Ci2-C2 4 alkynyl.
  • a RH lipid of the present invention has one of the following structures:
  • lipids of the present invention may be synthesized as described in the accompanying Synthetic Examples. In addition, it is understood that one skilled in the art may be able to make these lipids by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other lipids of structures (A) and (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in herein.
  • the present invention further includes lipid particles, e.g., LNs or liposomes, comprising one or more reverse headgroup lipids of the present invention.
  • lipid particles e.g., LNs or liposomes, comprising one or more reverse headgroup lipids of the present invention.
  • lipid particles comprise one or more reverse headgroup li ids having the following structure (A):
  • R 1 and R 2 are either the same or different and are each, independently, C 6 -C 32 alkyl;
  • n and m are independently either 1 , 2, 3, or 4.
  • lipid particles of the present invention comprise a reverse head roup lipid having the following structure (I):
  • Ri and F3 ⁇ 4 are each, independently, C6-C32alkyl.
  • lipid particles of the present invention comprise a reverse headgroup lipid having the following structure (I):
  • Ri and F3 ⁇ 4 are each, independently, C6-C32alkyl.
  • the reverse headgroup lipid is DLinFAB, DOFAB, or DPFAB, or a combination thereof.
  • the one or more reverse headgroup lipids of the present invention comprise at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the total lipid present in the lipid particle (mol%). In one embodiment, the one or more reverse headgroup lipids of the present invention comprise between 50% and 90% of the total lipid (mol%).
  • lipid particles of the present invention which comprise one of more reverse headgroup lipids of the present invention, further comprise one or more phosphatidylcholine or phosphatidylethanolamine lipids.
  • the phosphatidylcholine lipid is selected from DOPC and POPC.
  • the phosphatidylethanolamine lipid is 1 ,2-dipalmitoleoyl-sn-glycero-3-phophoethanolamin (diPoPE).
  • lipid particles of the present invention which comprise one or more one of more reverse headgroup lipids of the present invention, further comprise cholesterol .
  • lipid particles of the present invention comprise: one of more reverse headgroup lipids of the present invention, one or more phosphatidylcholine or phosphatidylethanolamine lipids, and cholesterol.
  • the molar amount of reverse headgroup lipid: PE or PC lipid: cholesterol is about equimolar or about 1 :1 :1 .
  • lipid particles of the present invention comprise or consist of one of more reverse headgroup lipids of the present invention, cholesterol, and a PEG-lipid.
  • the reverse headgroup lipid comprises greater than 50%, greater than 60%, greater than 70%, greater than 80%, or greater than 90% of the total lipid (mol%). In one embodiment, the one or more reverse headgroup lipids of the present invention comprise between 50% and 90% or between 50% and 95% of the total lipid (mol%).
  • PEG-lipid conjugates include, but are not limited to, PEG coupled to dialkyloxypropyls, PEG coupled to diacylglycerols (DAG), PEG coupled to cholesterol, PEG coupled to phosphatidylethanolamines, PEG conjugated to ceramides (see, e.g., U.S. Patent No. 5,885,613); cationic PEG lipids; cationic- polymer-lipid conjugates (CPLs).
  • DAG diacylglycerols
  • CPLs cationic- polymer-lipid conjugates
  • PEG-modified lipids examples include PEG-modified phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates ⁇ e.g., PEG-CerC14 or PEG-CerC20) which are described in copending USSN 08/486,214, incorporated herein by reference, PEG-modified dialkylamines and PEG-modified 1 ,2-diacyloxypropan-3-amines. Particularly preferred are PEG-modified diacylglycerols and dialkylglycerols.
  • the PEG-lipid conjugate is 3-N-[( -methoxypoly(ethylene glycol)2000)carbamoyl]-1 ,2-dimyristyloxy-propylamine (PEG-DMG).
  • PEG-DMG 3-N-[( -methoxypoly(ethylene glycol)2000)carbamoyl]-1 ,2-dimyristyloxy-propylamine
  • a PEG-lipid is selected from:
  • lipid particle of the present invention comprises between 1 to 20 mol% PEG-lipid of total lipid, or between 1 to 15 mol% PEG-lipid of total lipid.
  • the term "between" in this context is inclusive of the two values defining the range.
  • lipid particles of the present invention comprises between 50 to 95 mol% reverse headgroup lipid of the present invention, between 0 and 20 mol% cholesterol, and between 0 and 20 mol% PEG-lipid, where these values indicate mol% of total lipid.
  • lipid particles of the present invention comprises between 75 to 95 mol% reverse headgroup lipid of the present invention, between 1 and 10 mol% cholesterol, and between 1 and 10 mol% PEG-lipid, where these values indicate mol% of total lipid.
  • lipid particles of the present invention comprises about 90 mol% reverse headgroup lipid of the present invention, about 5 mol% cholesterol, and about 5 mol% PEG-lipid, where these values indicate mol% of total lipid.
  • the reverse headgroup lipid is DOFAB.
  • lipid particles of the present invention comprises about 90 mol% DOFAB, about 5 mol% cholesterol, and about 5 mol% PEG-S-DMG.
  • lipid particles of the present invention further comprise or encapsulate a nucleic acid, such as, e.g., an siRNA, which may target a therapeutic target, such as the androgen receptor, and may be used e.g., for the treatment or prevention of a disease or disorder in a subject, such as, e.g., prostate cancer.
  • a nucleic acid such as, e.g., an siRNA
  • a therapeutic target such as the androgen receptor
  • lipids may be present in a lipid particle of the present invention, including, e.g., one or more cationic lipids or non-cationic lipids.
  • cationic lipid refers to any of a number of lipid species that carry a net positive charge at a selected pH, such as physiological pH.
  • cationic lipids that may be used in the lipid particles of the present invention include, but are not limited to, DLinDMA, DLin-K-DMA, N,N-dioleyl- ⁇ , ⁇ -dimethylammonium chloride (DODAC), dioctadecyldimethylammonium (DODMA), distearyldimethylammonium (DSDMA), N-(1 -(2,3-dioleyloxy)propyl)- ⁇ , ⁇ , ⁇ -trimethylammonium chloride (DOTMA), N,N-distearyl-N,N- dimethylammonium bromide (DDAB), N-(1 -(2,3-dioleoyloxy)propyl)-N,N,N- trimethylammonium chloride (DOTAP), 3-(N-(N',N'-dimethylaminoethane)- carbamoyl)cholesterol (DC-Choi), N-(1
  • non-cationic lipid refers to any amphipathic lipid as well as any other neutral lipid or anionic lipid.
  • Non-cationic lipids used in the lipid particles, e.g., SNALP, of the present invention can be any of a variety of neutral uncharged, zwitterionic, or anionic lipids capable of producing a stable complex.
  • neutral lipid refers to any of a number of lipid species that exist either in an uncharged or neutral zwitterionic form at a selected pH.
  • such lipids include, for example, diacylphosphatidylcholine, diacylphosphatidylethanolamine, ceramide, sphingomyelin, cephalin, cholesterol, cerebrosides, and diacylglycerols.
  • anionic lipid refers to any lipid that is negatively charged at physiological pH. These lipids include, but are not limited to, phosphatidylglycerols, cardiolipins, diacylphosphatidylserines, diacylphosphatidic acids, N-dodecanoyl phosphatidylethanolamines, N-succinyl phosphatidylethanolamines, N-glutarylphosphatidylethanolamines, lysylphosphatidylglycerols, palmitoyloleyolphosphatidylglycerol (POPG), and other anionic modifying groups joined to neutral lipids.
  • phosphatidylglycerols cardiolipins
  • diacylphosphatidylserines diacylphosphatidic acids
  • N-dodecanoyl phosphatidylethanolamines N-succinyl phosphatidylethanolamines
  • Non-limiting examples of non-cationic lipids include phospholipids such as lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebrosides, dicetylphosphate, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyl
  • acyl groups in these lipids are preferably acyl groups derived from fatty acids having Ci 0 -C 24 carbon chains, e.g., lauroyl, myristoyl, palmitoyl, stearoyl, or oleoyl.
  • non-cationic lipids suitable for use in the present invention include, but are not limited to, nonphosphorous containing lipids such as, e.g., stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerolncinoleate, hexadecyl stereate, isopropyl myristate, amphoteric acrylic polymers, triethanolamine-lauryl sulfate, alkyl-aryl sulfate polyethyloxylated fatty acid amides, dioctadecyldimethyl ammonium bromide, ceramide, sphingomyelin, and the like.
  • nonphosphorous containing lipids such as, e.g., stearylamine, dodecylamine, hexadecylamine, acetyl palmitate, glycerolncinoleate, hexadecyl
  • lipid particles of the present invention comprise a therapeutic agent. While any agent, e.g., antibodies, polypeptides, toxins, or small molecules, may be delivered to a cell or tissue using the lipid particles of the present invention, in particular embodiments, the lipid particles of the present invention comprise a nucleic acid.
  • the nucleic acid may comprise DNA, RNA, or both, including modified forms of DNA and/or RNA. In certain embodiments, the nucleic acid is single-stranded or double-stranded.
  • the lipid particle of the present invention comprises an interfering RNA capable of mediating knockdown (i.e., reduced expression) of a target gene ⁇ e.g., a siRNA, microRNA (miRNA), short hairpin RNA (shRNA), including plasmids from which an interfering RNA is transcribed may be encapsulated within the lipid particle.
  • a target gene e.g., a siRNA, microRNA (miRNA), short hairpin RNA (shRNA), including plasmids from which an interfering RNA is transcribed
  • lipid encapsulated refers to a lipid formulation that provides a compound, such as a nucleic acid ⁇ e.g., a siRNA), with full encapsulation, partial encapsulation, or both.
  • the nucleic acid is fully encapsulated in the lipid particle. In both instances, the nucleic acid is protected from nuclease degradation.
  • RNA interference methods using interfering RNAi molecules may be used to disrupt the expression of a gene or polynucleotide of interest, such as a gene associated with an inflammatory or immune disease or disorder, e.g., a gene overexpressed in such as disease or disorder.
  • the interfering RNA is a small interfering RNA (siRNA).
  • siRNAs are RNA duplexes typically 19-30 nucleotides long that can associate with a cytoplasmic multi-protein complex known as RNAi-induced silencing complex (RISC). RISC loaded with siRNA mediates the degradation of homologous mRNA transcripts. Therefore, siRNA can be designed to knock down protein expression with high specificity.
  • RNAi molecules While the first described RNAi molecules were RNA:RNA hybrids comprising both an RNA sense and an RNA antisense strand, it has now been demonstrated that DNA sense:RNA antisense hybrids, RNA sense:DNA antisense hybrids, and DNA:DNA hybrids are capable of mediating RNAi (Lamberton, J.S. and Christian, AT., (2003) Molecular Biotechnology 24:1 1 1 -1 19). Thus, the invention includes the use of RNAi molecules comprising any of these different types of double-stranded molecules. In addition, it is understood that RNAi molecules may be used and introduced to cells in a variety of forms.
  • RNAi molecules encompasses any and all molecules capable of inducing an RNAi response in cells, including, but not limited to, double-stranded polynucleotides comprising two separate strands, i.e. a sense strand and an antisense strand, e.g., small interfering RNA (siRNA); polynucleotides comprising a hairpin loop of complementary sequences, which forms a double-stranded region, e.g., small hairpin RNA (shRNA) molecules, and expression vectors that express one or more polynucleotides capable of forming a double-stranded polynucleotide alone or in combination with another polynucleotide.
  • siRNA small interfering RNA
  • shRNA small hairpin RNA
  • RNAi molecules targeting specific polynucleotides can be readily prepared according to procedures known in the art. Structural characteristics of effective siRNA molecules have been identified. Elshabir, S.M. et al. (2001 ) Nature 41 1 :494-498 and Elshabir, S.M. et al. (2001 ), EMBO 20:6877-6888. Accordingly, one of skill in the art would understand that a wide variety of different siRNA molecules may be used to target a specific gene or transcript.
  • siRNA molecules according to the invention are typically double-stranded and 16-30 or 18 - 25 nucleotides in length, including each integer in between. In one embodiment, a siRNA is about 21 nucleotides in length.
  • siRNAs have 0-7 nucleotide 3' overhangs or 0-4 nucleotide 5' overhangs. In one embodiment, a siRNA molecule has a two nucleotide 3' overhang. In one embodiment, a siRNA is 21 nucleotides in length with two nucleotide 3' overhangs (i.e. they contain a 19 nucleotide complementary region between the sense and antisense strands). In certain embodiments, the overhangs are UU or dTdT 3' overhangs.
  • siRNA target sites are selected by scanning the target mRNA transcript sequence for the occurrence of AA dinucleotide sequences. Each AA dinucleotide sequence in combination with the 3' adjacent approximately 19 nucleotides are potential siRNA target sites.
  • siRNA target sites are preferentially not located within the 5' and 3' untranslated regions (UTRs) or regions near the start codon (within approximately 75 bases), since proteins that bind regulatory regions may interfere with the binding of the siRNP endonudease complex (Elshabir, S. et al. Nature 41 1 :494-498 (2001 ); Elshabir, S. et al. EMBO J. 20:6877-6888 (2001 )).
  • potential target sites may be compared to an appropriate genome database, such as BLASTN 2.0.5, available on the NCBI server at www.ncbi.nlm, and potential target sequences with significant homology to other coding sequences eliminated.
  • short hairpin RNAs constitute the nucleic acid component of nucleic acid-lipid particles of the present invention.
  • ShRNAs contain a stem loop structure. In certain embodiments, they may contain variable stem lengths, typically from 19 to 29 nucleotides in length, or any number in between. In certain embodiments, hairpins contain 19 to 21 nucleotide stems, while in other embodiments, hairpins contain 27 to 29 nucleotide stems. In certain embodiments, loop size is between 4 to 23 nucleotides in length, although the loop size may be larger than 23 nucleotides without significantly affecting silencing activity. ShRNA molecules may contain mismatches, for example G-U mismatches between the two strands of the shRNA stem without decreasing potency.
  • shRNAs are designed to include one or several G-U pairings in the hairpin stem to stabilize hairpins during propagation in bacteria, for example.
  • complementarity between the portion of the stem that binds to the target mRNA (antisense strand) and the mRNA is typically required, and even a single base pair mismatch is this region may abolish silencing. 5' and 3' overhangs are not required, although they may be present.
  • an interfering RNA or siRNA comprises one or more modification, such as a modified nucleoside or a modified phosphate linkage.
  • a siRNA comprises at least one modified nucleotide in the double-stranded region.
  • the modified siRNA contains at least one 2'OMe purine or pyrimidine nucleotide such as a 2'OMe-guanosine, 2'OMe-uhdine, 2'OMe-adenosine, and/or 2'OMe-cytosine nucleotide.
  • modified nucleotides suitable for use in the present invention include, but are not limited to, ribonucleotides having a 2'-O-methyl (2'OMe), 2'-deoxy-2'-fluoro (2'F), 2'-deoxy, 5-C-methyl, 2'-O-(2-methoxyethyl) (MOE), 4'-thio, 2'-amino, or 2'-C-allyl group.
  • Non-limiting examples of phosphate backbone modifications that may be present in interfering RNA of the present invention include phosphorothioate, phosphorodithioate, methylphosphonate, phosphotriester, morpholino, amidate, carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and alkylsilyl substitutions (see, e.g., Hunziker et al., Nucleic Acid Analogues: Synthesis and Properties, in Modern Synthetic Methods, VCH, 331 -417 (1995); Mesmaeker et ai, Novel Backbone Replacements for Oligonucleotides, in Carbohydrate Modifications in Antisense Research, ACS, 24-39 (1994)).
  • Such chemical modifications can occur at the 5'-end and/or 3'-end
  • cells contacted with lipid particle of the present invention comprising an interfering RNA under conditions and for a time sufficient for RNA interference to occur express less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, or less than 10% the amount of the targeted gene as expressed by the same cell type not contacted with the lipid particle.
  • Expression may be measured as either protein expression or mRNA expression, or as microRNA expression.
  • Levels of the protein expressed by the target gene may be readily determined using routine procedures, e.g., such as Western blotting or FACS.
  • Levels of RNA expressed by a targeted gene may be readily determined using routine procedures such as RT-PCR.
  • An interfering RNA used in the present invention comprises a region corresponding to or complementary to a region of a target gene.
  • this complementary region is completely complementary, while in other embodiments, it may comprise one or more mismatches.
  • the complementary region is between 19 and 25 bases in length.
  • the target gene is the androgen receptor
  • lipid particles comprise an siRNA directed against the androgen receptor mRNA.
  • the interfering RNA comprises a region that is complementary to an mRNA expressed by an androgen gene. In preferred embodiments, this complementary region is completely complementary, while in other embodiments, it may comprise one or more mismatches. In certain embodiments, the complementary region is between 19 and 25 bases in length.
  • the interfering RNA is a siRNA comprising the following two strands: 5 ' -AGCACUGCUACUCUUCAGCAUdTdT-3 ' (AR sense) and 5 ' -AUGCUGAAGAGUAGCAGUGCdTdT-3 ' (AR anti-sense). This 23-mer AR siRNA may be purchased from Thermo Scientific (Dharmacon).
  • the coding sequences and mRNA sequences of various mammalian androgen receptor genes are known in the art.
  • Lipid particles of the present invention may be prepared by procedures described in the art, including those described in the accompanying Example and those described in WO 96/40964, WO 01/05374, U.S. Patent No. 5,981 ,501 , U.S. Patent No. 6,1 10,745, WO 1999/18933, and WO 1998/51278.
  • a mixture of lipids is combined with a buffered aqueous solution of nucleic acid to produce an intermediate mixture containing nucleic acid encapsulated in lipid particles, e.g., wherein the encapsulated nucleic acids are present in a nucleic acid/lipid ratio of about 10 wt% to about 20 wt%.
  • the intermediate mixture may optionally be sized to obtain lipid-encapsulated nucleic acid particles wherein the lipid portions are unilamellar vesicles, preferably having a diameter of 30 to 150 nm, more preferably about 40 to 90 nm.
  • the pH may then be raised to neutralize at least a portion of the surface charges on the lipid particles, thus providing an at least partially surface-neutralized lipid particle composition.
  • the mixture of lipids may be a solution of lipids in an organic solvent. This mixture of lipids can then be dried to form a thin film or lyophilized to form a powder before being hydrated with an aqueous buffer to form liposomes.
  • the lipid mixture can be solubilized in a water miscible alcohol, such as ethanol, and this ethanolic solution added to an aqueous buffer resulting in spontaneous liposome formation.
  • the alcohol is used in the form in which it is commercially available.
  • ethanol can be used as absolute ethanol (100%), or as 95% ethanol, the remainder being water. This method is described in more detail in US Patent 5,976,567.
  • the mixture of lipids is a mixture of a reverse headgroup lipid of the present invention, cholesterol, and a PEG-lipid in an alcohol solvent.
  • the lipid mixture consists essentially of a cationic lipid, a non-cationic lipid, cholesterol and a PEG- modified lipid in alcohol, more preferably ethanol.
  • the lipid mixture is combined with a buffered aqueous solution that may contain the nucleic acids.
  • the buffered aqueous solution is typically a solution in which the buffer has a pH of less than the pK a of the protonatable lipid in the lipid mixture. Examples of suitable buffers include citrate, phosphate, acetate, and MES.
  • a particularly preferred buffer is citrate buffer.
  • Preferred buffers will be in the range of 1 -1000 mM of the anion, depending on the chemistry of the nucleic acid being encapsulated, and optimization of buffer concentration may be significant to achieving high loading levels (see, e.g., US Patent 6,287,591 and US Patent 6,858,225).
  • pure water acidified to pH 5-6 with chloride, sulfate or the like may be useful.
  • it may be suitable to add 5% glucose, or another non- ionic solute which will balance the osmotic potential across the particle membrane when the particles are dialyzed to remove ethanol, increase the pH, or mixed with a pharmaceutically acceptable carrier such as normal saline.
  • the amount of nucleic acid in buffer can vary, but will typically be from about 0.01 mg/mL to about 200 mg/mL, more preferably from about 0.5 mg/mL to about 50 mg/mL.
  • the mixture of lipids and the buffered aqueous solution of nucleic acids is combined to provide an intermediate mixture.
  • the intermediate mixture is typically a mixture of lipid particles having encapsulated nucleic acids. Additionally, the intermediate mixture may also contain some portion of nucleic acids which are attached to the surface of the lipid particles (liposomes or lipid vesicles) due to the ionic attraction of the negatively-charged nucleic acids and positively-charged lipids on the lipid particle surface (the cationic lipids or other lipid making up the protonatable first lipid component are positively charged in a buffer having a pH of less than the pK a of the protonatable group on the lipid).
  • the mixture of lipids is an alcohol solution of lipids and the volumes of each of the solutions is adjusted so that upon combination, the resulting alcohol content is from about 20% by volume to about 45% by volume.
  • the method of combining the mixtures can include any of a variety of processes, often depending upon the scale of formulation produced. For example, when the total volume is about 10-20 ml_ or less, the solutions can be combined in a test tube and stirred together using a vortex mixer. Large-scale processes can be carried out in suitable production scale glassware.
  • the lipid particles that are produced by combining the lipid mixture and the buffered aqueous solution of nucleic acids can be sized to achieve a desired size range and relatively narrow distribution of lipid particle sizes.
  • the compositions provided herein will be sized to a mean diameter of from about 70 to about 200 nm, more preferably about 90 to about 130 nm.
  • Sonicating a liposome suspension either by bath or probe sonication produces a progressive size reduction down to small unilamellar vesicles (SUVs) less than about 0.05 microns in size.
  • Homogenization is another method which relies on shearing energy to fragment large liposomes into smaller ones.
  • multilamellar vesicles are recirculated through a standard emulsion homogenizer until selected liposome sizes, typically between about 0.1 and 0.5 microns, are observed.
  • the particle size distribution can be monitored by conventional laser-beam particle size determination.
  • extrusion is used to obtain a uniform vesicle size.
  • lipid particles Extrusion of lipid particles through a small-pore polycarbonate membrane or an asymmetric ceramic membrane results in a relatively well- defined size distribution. Typically, the suspension is cycled through the membrane one or more times until the desired liposome complex size distribution is achieved. The liposomes may be extruded through successively smaller-pore membranes, to achieve a gradual reduction in lipid particle siz. In some instances, the lipid particles which are formed can be used without any sizing.
  • methods of the present invention further comprise a step of neutralizing at least some of the surface charges on the lipid portions of the lipid-nucleic acid compositions.
  • unencapsulated nucleic acid is freed from the lipid particle surface and can be removed from the composition using conventional techniques.
  • unencapsulated and surface adsorbed nucleic acids are removed from the resulting compositions through exchange of buffer solutions.
  • buffer solutions For example, replacement of a citrate buffer (pH about 4.0, used for forming the compositions) with a HEPES-buffered saline (HBS pH about 7.5) solution, results in the neutralization of liposome surface and nucleic acid release from the surface.
  • the released nucleic acid can then be removed via chromatography using standard methods, and then switched into a buffer with a pH above the pKa of the lipid used.
  • the lipid vesicles (i.e., lipid particles) can be formed by hydration in an aqueous buffer and sized using any of the methods described above prior to addition of the nucleic acid.
  • the aqueous buffer may be of a pH below the pKa of the reverse headgroup lipid.
  • a solution of the nucleic acids can then be added to these sized, preformed vesicles.
  • the mixture should contain an alcohol, such as ethanol. In the case of ethanol, it should be present at a concentration of about 20% (w/w) to about 45% (w/w).
  • the aqueous buffer-ethanol mixture it may be necessary to warm the mixture of pre-formed vesicles and nucleic acid in the aqueous buffer-ethanol mixture to a temperature of about 25° C to about 50° C depending on the composition of the lipid vesicles and the nature of the nucleic acid. It will be apparent to one of ordinary skill in the art that optimization of the encapsulation process to achieve a desired level of therapeutic agent, e.g., nucleic acid, in the lipid vesicles will require manipulation of variable such as ethanol concentration and temperature.
  • the therapeutic agents e.g., nucleic acids
  • the external pH can be increased to at least partially neutralize the surface charge. Unencapsulated and surface adsorbed therapeutic agent, e.g., nucleic acids, can then be removed as described above.
  • the lipid particles of present invention may be formulated as a pharmaceutical composition, e.g., which further comprises a pharmaceutically acceptable diluent, excipient, or carrier, such as physiological saline or phosphate buffer, selected in accordance with the route of administration and standard pharmaceutical practice.
  • a pharmaceutically acceptable diluent, excipient, or carrier such as physiological saline or phosphate buffer, selected in accordance with the route of administration and standard pharmaceutical practice.
  • the present invention includes a kit comprising a lipid particle of the present invention.
  • Suitable carriers include, e.g., physiological saline, water, buffered water, 0.9% saline, 0.3% glycine, and the like, including glycoproteins for enhanced stability, such as albumin, lipoprotein, globulin, etc.
  • the carrier is preferably added following lipid particle formation.
  • the compositions can be diluted into pharmaceutically acceptable carriers, such as normal saline.
  • the resulting pharmaceutical preparations may be sterilized by conventional, well known sterilization techniques.
  • the aqueous solutions can then be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile aqueous solution prior to administration.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, etc.
  • the lipidic suspension may include lipid- protective agents which protect lipids against free-radical and lipid-peroxidative damages on storage. Lipophilic free-radical quenchers, such as a-tocopherol and water-soluble iron-specific chelators, such as ferrioxamine, are suitable.
  • the concentration of lipid particle in the pharmaceutical formulations can vary widely, i.e., from less than about 0.01 %, usually at or at least about 0.05-5% to as much as 10 to 30% by weight and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • the present invention further includes methods of delivering an agent, e.g., a therapeutic agent, to a cell, comprising contacting a cell with a lipid particle of the present invention comprising the agent.
  • an agent e.g., a therapeutic agent
  • contacting occurs in vitro or in vivo.
  • the cell is a mammalian cell, e.g., a human cell.
  • the present invention provides a method of treating or preventing a disease or disorder in a subject, comprising providing to the subject a lipid particle of the present invention comprising a therapeutic agent.
  • the subject is provided with an effective amount or a therapeutically effective amount of the lipid particle.
  • the subject is a mammal, e.g., a human.
  • the disease or disorder is a tumor ⁇ e.g., prostate cancer), an inflammatory disease or disorder, a metabolic disease or disorder, a neurological disease or disorder, or a cardiac disease or disorder.
  • the present methods may be used to deliver an encapsulated agent to a variety of different cells and subcellular locations. Accordingly, the methods of the invention may be used to modulate the expression of a variety of different genes, modulate an immune response, and treat or prevent various related diseases and disorders, including tumors, inflammatory or immune-related diseases and disorders.
  • treatment indicates an approach for obtaining beneficial or desired results, including and preferably clinical results.
  • Treatment can involve optionally either the amelioration of symptoms of the disease or condition, or the delaying of the progression of the disease or condition.
  • prevention indicates an approach for preventing, inhibiting, or reducing the likelihood of, the onset or recurrence of a disease or condition. It also refers to preventing, inhibiting, or reducing the likelihood of, the occurrence or recurrence of the symptoms of a disease or condition, or optionally an approach for delaying the onset or recurrence of a disease or condition or delaying the occurrence or recurrence of the symptoms of a disease or condition. As used herein, "prevention” and similar words also includes reducing the intensity, effect, symptoms and/or burden of a disease or condition prior to onset or recurrence of the disease or condition.
  • an “effective amount” or a “therapeutically effective amount” of a substance is that amount sufficient to affect a desired biological effect, such as beneficial results, including clinical results.
  • the lipid particle comprises a therapeutic agent ⁇ e.g., a nucleic acid such as an siRNA) and one or more reverse headgroup lipids ⁇ e.g., a reverse headgroup lipid of structure (I), DLinFAB, DOFAB, or DPFAB).
  • a therapeutic agent e.g., a nucleic acid such as an siRNA
  • one or more reverse headgroup lipids e.g., a reverse headgroup lipid of structure (I), DLinFAB, DOFAB, or DPFAB.
  • it further comprises cholesterol and a PEG-lipid.
  • the concentration of lipid particles in the medium can vary widely depending on the particular application, but is generally between about 1 ⁇ and about 10 mmol.
  • treatment of the cells with the lipid particles will generally be carried out at physiological temperatures (about 37°C) for periods of time from about 1 to 24 hours, preferably from about 2 to 8 hours.
  • the cell may be grown or maintained in culture, whether of plant or animal origin, vertebrate or invertebrate, and of any tissue or type.
  • the cells will be animal cells, more preferably mammalian cells, and most preferably human cells.
  • compositions comprising lipid particles of the present invention may be administered by any means available in the art.
  • they may be administered parenterally, i.e., intraarticularly, intravenously, intraperitoneally, subcutaneously, or intramuscularly.
  • the pharmaceutical compositions are administered intravenously or intraperitoneally by a bolus injection.
  • Stadler, et al., U.S. Patent No. 5,286,634 which is incorporated herein by reference.
  • Intracellular nucleic acid delivery has also been discussed in Straubringer, et al., METHODS IN ENZYMOLOGY, Academic Press, New York.
  • the pharmaceutical preparations may be contacted with a desired tissue by direct application of the preparation to the tissue.
  • the lipid particles can also be administered in an aerosol inhaled into the lungs (see, Brigham, et al., Am. J. Sci. 298(4):278-281 (1989)) or by direct injection at the site of disease (Culver, Human Gene Therapy, MaryAnn Liebert, Inc., Publishers, New York. pp.70-71 (1994)).
  • the methods of the present invention may be practiced in a variety of subjects or hosts.
  • Preferred subjects or hosts include mammalian species, such as humans, non-human primates, dogs, cats, cattle, horses, sheep, and the like.
  • the subject is a mammal, such as a human, in need of treatment or prevention of a disease or disorder, e.g., a subject diagnosed with or considered at risk for a disease or disorder.
  • Dosages for the lipid particles of the present invention will depend on the ratio of therapeutic agent, e.g., nucleic acid, to lipid and the administrating physician's opinion based on age, weight, and condition of the patient.
  • the reverse headgroup lipids described herein were prepared by O-alkylation of 1 -(2,3-dihydroxypropyl)pyrrolidin-2-one (2), obtained as depicted and described below, with the commercially available mesylates of linoleyl (3), oleyl (4), or palmitoyl (5) alcohol, followed by base hydrolysis of the lactam ring.
  • DLinFAB was prepared as described above and depicted in the synthetic scheme shown below, as described in further detail below.
  • DOFAB was prepared as described above and depicted in the synthetic scheme shown below, as described in further detail below.
  • DPFAB (DPFAB, 1 1 ) DPFAB was prepared as described above and depicted in the synthetic scheme shown below, as described in further detail below.
  • PEG-lipids such as mPEG2000-1 ,2-Di-O-Alkyl-sn3- Carbomoylglyceride (PEG-C-DOMG) are synthesized as shown in the schematic diagram and described below. "OH
  • the reaction mixture was diluted with DCM (400 mL) and the organic layer was washed with water (2X500 mL), aqueous NaHCO3 solution (500 mL) followed by standard work-up. The residue obtained was dried at ambient temperature under high vacuum overnight. After drying, the crude carbonate Ma thus obtained was dissolved in dichloromethane (500 mL) and stirred over an ice bath. To the stirring solution, mPEG 2 ooo-NH 2 (III, 103.00 g, 47.20 mmol, purchased from NOF Corporation, Japan) and anhydrous pyridine (Py, 80 mL, excess) were added under argon.
  • the x in compound III has a value of 45-49, preferably 47-49, and more preferably 49.
  • the reaction mixture was then allowed to stir at ambient temperature overnight. Solvents and volatiles were removed under vacuum and the residue was dissolved in DCM (200 mL) and charged on a column of silica gel packed in ethyl acetate. The column was initially eluted with ethyl acetate and subsequently with gradient of 5-10 % methanol in dichloromethane to afford the desired PEG-Lipid IVa as a white solid (105.30g, 83%).
  • MPEG2000-NH2 III (1 .50g, 0.687 mmol, purchased from NOF Corporation, Japan) and lie (0.760g, 1 .5eq) were dissolved in dichloromethane (20 mL) under argon.
  • the x in compound III has a value of 45- 49, preferably 47-49, and more preferably 49.
  • the reaction was cooled to 0°C. Pyridine (1 mL, excess) was added and the reaction was stirred overnight. The reaction was monitored by TLC.
  • Reverse headgroup lipids comprisig FAB headgroups were characterized with respect to the pKa values exhibited by the FAB headgroups and the abilities of these FAB lipids to combine with phosphatidylcholine (PC) and phosphatidylethanolamine (PE) to induce non-lamellar structures.
  • PC phosphatidylcholine
  • PE phosphatidylethanolamine
  • Bovine milk sphingomyelin (SM),1 ,2-dioleoyl-sn-glycero-3- phosphocholine (DOPC), cholesterol, 1 ,2-dioleoyl-sn-glycero-3- phosphoethanolamine (DOPE), 1 ,2-dioleoyl-sn-glycero-3-phosphate sodium salt (DOPA), 1 ,2-dioleoyl-sn-glycero-3-phosphoserim sodium salt (DOPS), 1 ,2- dioleoyl-sn-glycero-3-phospho-(1 '-rac-glycerol) sodium salt (DOPG), 1 ,2- dipalmitoleoyl-sn-glycero-3-phosphoethanolamine (diPoPE), 1 ,2-dipalmitoyl-sn- glycero-3-phosphocholine (DPPC) and 1 -Palmitoyl-2-oleoyl-sn
  • the pKa's of various FAB lipids were determined using the fluorescence probe, TNS, which increases in fluorescence quantum yield upon binding to lipid membrane.
  • Multilamellar vesicles were formed by rehydration of lipid film in HEPES buffered saline (100 mM HEPES, 50 mM NaCI, pH 7.0) followed by vigorous vortexing.
  • MLVs composed of equimolar FAB/DPPC/cholesterol were diluted to 200 ⁇ in buffered solution at various pH containing 10 mM acetate, 10 mM MES, 10 mM HEPES, 10 mM CAPS, 10 mM CHES and 100 mM of NaCI. Ammonium acetate was added to a final concentration of 10 mM to abolish any pH gradients in the liposomes.
  • DOFAB and PEG-c-DOMG lipid stocks were dissolved in chloroform.
  • DOFAB and PEG-c-DOMG stock solutions were mixed at appropriate molar ratio, and solvent was removed by drying under a stream of nitrogen gas and subsequently by vacuum overnight.
  • Multilamellar vesicles were formed by rehydration of lipid film in a poly-buffer solution containing 10mM ammonium acetate, 10mM HEPES, 10mM MES, and 130mM NaCI at pH 7.0 followed by vigorous vortexing.
  • MLVs were diluted to 100 ⁇ in buffered solution at various pH containing 10 mM ammonium acetate, 10 mM MES, 10 mM HEPES and 130 mM of NaCI.
  • Lipid films were hydrated in the NMR tube with 100 mM HEPES,
  • the FAB headqroup exhibited two pKa values
  • the TNS assay was used to determine the pKa values of FAB lipids.
  • the TNS assay was conducted on MLVs consisting of equimolar mixture of a FAB lipid, DPPC and cholesterol.
  • the FAB lipids tested were DLinFAB, DOFAB, and DPFAB.
  • DPPC was used because it is a saturated lipid that does not readily adopt non-lamellar structures such as the H ii phase and is expected to maintain lamellar structure despite the presence of equimolar quantities of FAB lipids.
  • the phosphate group in DPPC exhibits a pKa of approximately 2 (Boggs, 1987), which is considerably lower than that expected for the carboxyl group of FAB. Therefore, interference by the phosphate moiety in the TNS assay was expected to be minimal.
  • TNS fluorescence of different FAB liposomes was determined at different pH values. Since the quantum yield of TNS increases when it binds to membranes, comparing the TNS fluorescence of different formulations can provide an estimate of the relative affinity of TNS for each type of liposome.
  • Table 1 shows the fluorescence intensity of 2 ⁇ TNS with various FAB/DPPC/cholesterol (1 :1 :1 ) liposomes at different pH. It was expected that increasing the pH decreases the fluorescence of TNS, as it loses affinity to the liposomes when FAB is deprotonated.
  • TNS fluorescence of DOFAB and DLinFAB liposomes decreased from approximately 871 .5 to 27.8 units and 861 .1 to 50.3 units, respectively, with increasing pH (Table 1 ).
  • the fluorescence intensity of DPFAB-containing liposomes was approximately 7 regardless of the pH and was much lower than that of the other two formulations. This suggests that the affinity of TNS was significantly higher for both DOFAB and DLinFAB-containing systems than for DPFAB-ontaining systems.
  • the lower affinity of TNS for DPFAB may result from phase separation of the DPFAB in mixtures with DPPC or could arise if the lipid mixture is in a gel phase, which would limit the ability of TNS to partition into the lipid bilayer.
  • DOFAB was zwitterionic at physiological pH
  • the TNS assay was also used to determine the pKa values of DOFAB in MLVs consisting of mixture of DOFAB with 10% PEG-c-DOMG.
  • PEG-c-DOMG was incorporated into the particle to prevent aggregation as pure DOFAB does not hydrate readily.
  • PEG-c-DOMG does not contain any ionizable groups; therefore interference of the TNS assay was expected to be minimal.
  • the titration curve of DOFAB/PEG-c-DOMG liposomes against pH showed the isoelectric point (pi) of the particle to be at pH 6.7 ( Figure 2).
  • the apparent pKa of the amino and carboxyl groups in DOFAB can be estimated by using the half-way point between the maximal fluorescence and the pi (carboxyl moiety) and between the pi and minimal fluorescence (amino moiety).
  • the apparent pKa for the carboxyl and amino moieties were estimated to be 4.5 and 8.0, respectively. This suggests that DOFAB is zwitterionic and does not carry a net charge at physiological pH, an important requirement for increasing circulation lifetime.
  • FAB lipids exhibited strong bilayer destabilizing capabilities in mixtures with DOPC, POPC and other phospholipids
  • DOFAB could act as a strong bilayer destabilizing agent capable of inducing non-lamellar structures when mixed with the bilayer-preferring lipid DOPC. It is unclear why the DLinFAB/DOPC system did not form Hn phase, even though DLinFAB lipid would be expected to be the most effective bilayer destabilizing agent due to the very unsaturated nature of its acyl chains. It is possible that the DLinFAB did not mix well with DOPC, resulting in reduced bilayer-destabiling capabilities.
  • FAB lipids such as DOFAB
  • DOFAB can act as strong bilayer destabilizing agents in mixture with phosphatidylcholines such as DOPC and phosphatidylethanolamines such as DOPE. It was of interest to extend these observations to mixtures with other phospholipids such as, sphingomyelin (SM), phosphatidylserine (PS) phosphatidylglycerol (PG) and phosphatidic acid (PA).
  • SM sphingomyelin
  • PS phosphatidylserine
  • PG phosphatidylglycerol
  • PA phosphatidic acid
  • FAB lipids such as DOFAB
  • SM zwitterionic lipids
  • anionic phospholipids such as PS, PG and PA.
  • DOFAB/milk sphingomyelin Figure 6A
  • DOFAB/diPoPE Figure 6B
  • DOFAB/DOPS Figure 6C
  • DOFAB/DOPG Figure 6D
  • DOFAB/DOPA Figure 6E
  • DOPE is commonly used as a "helper" lipid in lipid nanoparticle carrier systems to aid in destabilizing cellular membranes.
  • the effect of DOPE on phase behaviour of DOPC and POPC was examined ( Figure 7).
  • Equimolar mixtures of DOPE/DOPC and DOPE/POPC remained lamellar at all temperatures tested, indicating that the DOPE was not able to destabilize bilayer structure for DOPC in our experimental conditions.
  • the FAB lipids were able to induce non-lamellar structures in mixture with DOPC and POPC ( Figures 2 and 3), they were much more potent bilayer-destabilizing agents than DOPE.
  • FAB lipids exhibited strong bilayer destabilizing properties in mixtures with PE
  • negatively charged lipids such as phosphatidylserine (PS), phosphatidylglycerol (PG) and phosphatidylinositol (PI) all adopt bilayer structure on hydration and are all converted into the ⁇ ⁇ organization in the presence of cationic lipids such as 3 ⁇ - [N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC- Chol), N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N-[l-(2,3- dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) and 1 ,2- dioleoyloxy-3-(trimethylammonio) propane (DOTAP) (Hafez et al. 2001 ).
  • DC- Chol N,N-dioleyl-N,N-dimethylammonium chloride
  • FAB lipids encourage non-lamellar structure by associating directly via ionic interactions with PC or PE headgroups to reduce headgroup hydration.
  • the presence of FAB lipids directly affects PC and PE headgroup orientation.
  • the unsaturated FAB lipids could act solely via their influence in the acyl chain region, acting to increase the effective cross-sectional area.
  • the apparent failure to adopt the ⁇ ⁇ phase is likely to be caused by highly saturated acyl chains found in milk sphingomyelin, which effectively protects it against any disruption from the bilayer conformation despite charge neutralization in the headgroup.
  • DOFAB can induce non-lamellar structures in anionic phospholipids such as DOPS, DOPG and DOPA.
  • lipid nanoparticles comprising reverse headgroup lipids to deliver siRNA to cells and mediate gene knockdown was determined. Specifically, knockdown of the androgen receptor (AR) in LNCap cells with DOFAB-encapsulated siRNA was measured.
  • the siRNA targeting AR comprised two strands: 5 ' - AGCACUGCUACUCUUCAGCAUdTdT-3 ' (AR sense) and 5 ' - AUGCUGAAGAGUAGCAGUGCdTdT-3 ' (AR anti-sense).
  • This 23-mer AR siRNA was purchased from Thermo Scientific (Dharmacon).
  • Negative control sequence was purchased from Invitrogen, StealthTM RNAi Negative Control Duplexes, Lo GC duplexs (Cat. No. 12935 - 200).
  • lipid stock solutions were dissolved in ethanol and were mixed at appropriate molar ratios (e.g., 85 mol% DOFAB, 4.8 mol% cholesterol, 10% PEG-s-DMG, 0.2 mol% SPDiO).
  • the lipid/ethanol mixtures were added dropwise into a vortexing buffer solution containing 150 mM acetate at pH 4.0.
  • the resulting multilamellar vesicles (MLVs) were extruded 5 times through two Nuclepore polycarbonate filters with a pore size of 80nm at -400 psi.
  • siRNA was hydrated in citrate buffer (10 mM citrate, 30 mM NaCI, pH 6.0) and quantified by measuring absorbance at 260 nm.
  • siRNA solution was added into the lipid mixture to make a total lipid:siRNA ratio of 10:1 and incubated at room temperature for 30 minutes.
  • the resulting vesicles were dialyzed with citrate buffer (50mM citrate, pH 4.0) for 4 hours and then overnight in PBS (pH 7.4).
  • the mean diameter of the vesicles was determined by dynamic light scattering using a NICOMP 370 particle sizer (Nicomp Particle Sizing Inc., Santa Barbara, CA) and was measured to be 403.3 ⁇ 267.8 nm.
  • Lipid concentrations were determined by measuring cholesterol content using the Cholesterol E Enzymatic Assay Kit (Wako Chemicals USA, Richmond, VA). siRNA content was verified by measuring absorbance at 260 nm in 70% to lyze the vesicles.
  • LNCap cells were seeded in 12-well plates (-2.0 x 10 5 cells/well) with RPMI media supplemented with 5% FBS overnight prior to siRNA treatment. Cells were treated continuously for either 24 or 48 hours with DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%) LNPs comprising siRNA against the androgen receptor (siAR) or low-GC content control siRNA (loGC) was encapsulated with and protein expression was analyzed with immunoblotting.
  • siRNA against the androgen receptor (siAR) or low-GC content control siRNA (loGC) was encapsulated with and protein expression was analyzed with immunoblotting.
  • LNCap cells were plated in 12-well plates (-2.0 x 10 5 cells/well) with a L-lysine coated coverslip (BD Biosciences, Mississauga, ON) and treated with 10 g/mL of encapsulated siRNA for 24 and 48 hours. Cells were fixed with 3% parformaldehyde in the prescence of Hoescht for 15 minutes. Coverslips were rinsed with PBS, followed by water and were mounted onto slides with SlowFade (Invitrogen, Eugene, OR). Slides were imaged and analyzed with confocal microscopy (Olympus FV1000). Fluorochromes were excited with lasers operating at 488 nm (DiO), and 350 nm (Hoescht). Images were collected with a 60 x oil immersion objective lens.
  • DOFAB encapsulated siRNA was able to transfect LNCap cells and mediate the knockdown of the androgen receptor.
  • DOFAB exhibited a positive charge at lower pH values ⁇ e.g., pH values below 7) and becomes progressively net neutral and then negatively charged at higher pH values. At lower pH values, DOFAB should therefore behave as a cationic lipid. Cationic lipids form complexes characterized as "lipoplexes" with RNA and DNA-based macromolecules, and can act as effective transfection agents to introduce these macromolecules into cells.
  • lipoplexes formed of DOFAB with siRNA against the androgen receptor or control siRNA were expressed in LNCaP prostate cancer cells.
  • the formulation comprised DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%) and was approximately 400 nm in diameter.
  • Cells were treated continuously with the DOFAB lipoplex for both 24 and 48 hours. Immunoblots were used to determine the expression of the androgen receptor and actin (control).
  • DOFAB lipoplex was labeled with SPDiO, and distribution of the lipid was examined with confocal microscopy. It was expected that direct plasma membrane fusion would distribute the SPDiO fluorescence around the plasma membrane, while entry via the endocytic pathway would lead to punctate fluorescence caused by the entrapment of the lipoplex in endocytic vesicles. Cellular uptake of DOFAB lipoplex was examined by confocal microscopy.
  • LNCap cells were treated with 10 g/mL of AR siRNA encapsulated with DOFAB/cholesterol/PEG-s-DMG/SPDiO (90:4.8:5:0.2 mol%) for 24 and 48 hours. Nuclei were stained blue with Hoescht's, and SPDiO fluorescence was observed in green. Figure 12 shows that the cellular uptake of DOFAB lipoplex was mainly through the endocytic pathway, as suggested by the punctate fluorescence pattern from SPDiO. However, several cells had green fluorescence around the plasma membrane at the 24 hours time-point (data not shown). Therefore, uptake via plasma membrane fusion could not be completely ruled out.
  • reverse headgroup lipids such as FAB lipids for delivery of macromolecules such as siRNA stems from three factors.
  • LN systems in order for LN systems to exhibit the long circulation lifetimes required to reach tissues other than the liver, they should be small (i.e., diameter ⁇ 100 nm) and should have neutral or near-neutral surface charge.
  • permanently charged cationic lipids commonly used for transfection in vitro are not suitable for systemic use, as they are rapidly cleared from the circulation and also give rise to serious toxic side effects.
  • ionizable cationic lipids that are positively charged at low pH values (e.g., pH 4), thereby allowing anionic polymers such as siRNA to be loaded into the LN, but relatively uncharged at physiological pH values.
  • the FAB lipids of the present invention would be expected to have similar properties but with one important advantage.
  • pH values at or below the pKa of the carboxyl function pKa ⁇ 4.5
  • the FAB lipids exhibit a net positive charge and should therefore be expected to act as efficient agents for loading oligonucleotides in lipid nanoparticles (LN).
  • LN lipid nanoparticles
  • the LNs exhibit net neutral surface charge and, hence, should exhibit long circulation lifetimes.
  • LN containing siRNA where the only lipid components are the FAB lipid, cholesterol, and a limited amount of PEG-lipid for stability.
  • the maximum amount of bilayer-destabilizing cationic lipid that can currently be associated with LN systems is in the range of 50 mol% due to stability issues.
  • at least 10% of a bilayer- stabilizing lipid such as DSPC must be incorporated to maintain LN stability.
  • LN systems containing FAB could contain substantially more of the bilayer- destabilizing lipid without compromising LN stability or circulation lifetimes, with the additional feature that it could act on PC and PE lipid species which are the majority lipids in target membranes.

Abstract

La présente invention concerne une nouvelle classe de lipides zwitterioniques qui sont utiles dans la préparation de particules lipidiques appropriées pour administrer des acides nucléiques encapsulés à des cellules.
PCT/US2010/054351 2009-10-27 2010-10-27 Lipides à têtes polaires inversées, compositions particulaires lipidiques comprenant les lipides à têtes polaires inversées, et procédés d'administration d'acides nucléiques WO2011056682A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US25540109P 2009-10-27 2009-10-27
US61/255,401 2009-10-27
US40076410P 2010-07-30 2010-07-30
US61/400,764 2010-07-30

Publications (1)

Publication Number Publication Date
WO2011056682A1 true WO2011056682A1 (fr) 2011-05-12

Family

ID=43559081

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/054351 WO2011056682A1 (fr) 2009-10-27 2010-10-27 Lipides à têtes polaires inversées, compositions particulaires lipidiques comprenant les lipides à têtes polaires inversées, et procédés d'administration d'acides nucléiques

Country Status (1)

Country Link
WO (1) WO2011056682A1 (fr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015075557A2 (fr) 2013-11-22 2015-05-28 Mina Alpha Limited Compositions c/ebp alpha et méthodes d'utilisation
WO2015093588A1 (fr) * 2013-12-21 2015-06-25 Ktnバイオテック株式会社 Agent anticancéreux
WO2018064350A1 (fr) * 2016-09-30 2018-04-05 Eriochem Usa, Llc Nanoparticules lipidiques modifiées par apo-e pour administrer des médicaments à des tissus ciblés et méthodes thérapeutiques
WO2019048631A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions de petits arn activateurs de hnf4a et procédés d'utilisation
WO2019048645A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions stabilisées de petits arn activateurs (parna) de cebpa et procédés d'utilisation
WO2019197845A1 (fr) 2018-04-12 2019-10-17 Mina Therapeutics Limited Compositions de sirt1-sarna et procédés d'utilisation
CN110746354A (zh) * 2019-10-29 2020-02-04 山东瑞博龙化工科技股份有限公司 一种丙烯酸酯类化合物及制备方法与应用
US10562849B2 (en) 2016-05-16 2020-02-18 The Board Of Regents Of The University Of Texas System Cationic sulfonamide amino lipids and amphiphilic zwitterionic amino lipids
WO2020208361A1 (fr) 2019-04-12 2020-10-15 Mina Therapeutics Limited Compositions de sirt1-sarna et procédés d'utilisation
US11246933B1 (en) 2011-12-07 2022-02-15 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
WO2022122872A1 (fr) 2020-12-09 2022-06-16 Ucl Business Ltd Agents thérapeutiques pour le traitement des troubles neurodégénératifs
WO2022200810A1 (fr) 2021-03-26 2022-09-29 Mina Therapeutics Limited Compositions de petits arn activateurs de tmem173 et procédés d'utilisation
WO2023099884A1 (fr) 2021-12-01 2023-06-08 Mina Therapeutics Limited Compositions d'arnsa de pax6 et procédés d'utilisation
WO2023104964A1 (fr) 2021-12-09 2023-06-15 Ucl Business Ltd Agents thérapeutiques pour le traitement de troubles neurodégénératifs
WO2023170435A1 (fr) 2022-03-07 2023-09-14 Mina Therapeutics Limited Compositions de petits arn activateurs d'il10 et procédés d'utilisation

Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3993754A (en) 1974-10-09 1976-11-23 The United States Of America As Represented By The United States Energy Research And Development Administration Liposome-encapsulated actinomycin for cancer chemotherapy
US4145410A (en) 1976-10-12 1979-03-20 Sears Barry D Method of preparing a controlled-release pharmaceutical preparation, and resulting composition
US4224179A (en) 1977-08-05 1980-09-23 Battelle Memorial Institute Process for the preparation of liposomes in aqueous solution
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4522803A (en) 1983-02-04 1985-06-11 The Liposome Company, Inc. Stable plurilamellar vesicles, their preparation and use
US4588578A (en) 1983-08-08 1986-05-13 The Liposome Company, Inc. Lipid vesicles prepared in a monophase
US4737323A (en) 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US5208036A (en) 1985-01-07 1993-05-04 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
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5286634A (en) 1989-09-28 1994-02-15 Stadler Joan K Synergistic method for host cell transformation
WO1996010390A1 (fr) 1994-09-30 1996-04-11 Inex Pharmaceuticals Corp. Nouvelles compositions d'introduction de substances polyanioniques dans des cellules
US5534499A (en) 1994-05-19 1996-07-09 The University Of British Columbia Lipophilic drug derivatives for use in liposomes
WO1996040964A2 (fr) 1995-06-07 1996-12-19 Inex Pharmaceuticals Corporation Particules d'acides nucleiques et de lipides preparees au moyen d'un intermediaire de complexe hydrophobe d'acides nucleiques et de lipides et utilisation pour transferer des genes
US5785992A (en) 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
WO1998051278A2 (fr) 1997-05-14 1998-11-19 Inex Pharmaceuticals Corporation Encapsulation hautement efficace d'agents therapeutiques charges dans des vesicules lipidiques
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
WO1999018933A2 (fr) 1997-10-10 1999-04-22 Inex Pharmaceuticals Corporation Methodes d'encapsulation d'acides nucleiques dans des bicouches lipidiques
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US6110745A (en) 1997-07-24 2000-08-29 Inex Pharmaceuticals Corp. Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
WO2001005374A1 (fr) 1999-07-15 2001-01-25 Inex Pharmaceuticals Corp. Preparation d'agents therapeutiques a encapsulation lipidique
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060240554A1 (en) 2005-02-14 2006-10-26 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
US20080088676A1 (en) 2002-11-23 2008-04-17 Silverbrook Research Pty Ltd Ink Jet Printhead With Suspended Heater Element
WO2009047006A2 (fr) * 2007-10-12 2009-04-16 Novosom Ag Perfectionnements aux ou se rapportant aux liposomes amphotères comprenant des lipides neutres
WO2009086558A1 (fr) * 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Compositions et procédés améliorés pour la délivrance d'acides nucléiques

Patent Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3993754A (en) 1974-10-09 1976-11-23 The United States Of America As Represented By The United States Energy Research And Development Administration Liposome-encapsulated actinomycin for cancer chemotherapy
US4145410A (en) 1976-10-12 1979-03-20 Sears Barry D Method of preparing a controlled-release pharmaceutical preparation, and resulting composition
US4224179A (en) 1977-08-05 1980-09-23 Battelle Memorial Institute Process for the preparation of liposomes in aqueous solution
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4522803A (en) 1983-02-04 1985-06-11 The Liposome Company, Inc. Stable plurilamellar vesicles, their preparation and use
US4588578A (en) 1983-08-08 1986-05-13 The Liposome Company, Inc. Lipid vesicles prepared in a monophase
US5208036A (en) 1985-01-07 1993-05-04 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
US4737323A (en) 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US5286634A (en) 1989-09-28 1994-02-15 Stadler Joan K Synergistic method for host cell transformation
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
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5534499A (en) 1994-05-19 1996-07-09 The University Of British Columbia Lipophilic drug derivatives for use in liposomes
US5785992A (en) 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5753613A (en) 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
WO1996010390A1 (fr) 1994-09-30 1996-04-11 Inex Pharmaceuticals Corp. Nouvelles compositions d'introduction de substances polyanioniques dans des cellules
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5976567A (en) 1995-06-07 1999-11-02 Inex Pharmaceuticals Corp. Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
WO1996040964A2 (fr) 1995-06-07 1996-12-19 Inex Pharmaceuticals Corporation Particules d'acides nucleiques et de lipides preparees au moyen d'un intermediaire de complexe hydrophobe d'acides nucleiques et de lipides et utilisation pour transferer des genes
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
WO1998051278A2 (fr) 1997-05-14 1998-11-19 Inex Pharmaceuticals Corporation Encapsulation hautement efficace d'agents therapeutiques charges dans des vesicules lipidiques
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
US6858225B2 (en) 1997-05-14 2005-02-22 Inex Pharmaceuticals Corporation Lipid-encapsulated polyanionic nucleic acid
US6110745A (en) 1997-07-24 2000-08-29 Inex Pharmaceuticals Corp. Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
WO1999018933A2 (fr) 1997-10-10 1999-04-22 Inex Pharmaceuticals Corporation Methodes d'encapsulation d'acides nucleiques dans des bicouches lipidiques
WO2001005374A1 (fr) 1999-07-15 2001-01-25 Inex Pharmaceuticals Corp. Preparation d'agents therapeutiques a encapsulation lipidique
US20080088676A1 (en) 2002-11-23 2008-04-17 Silverbrook Research Pty Ltd Ink Jet Printhead With Suspended Heater Element
US20060083780A1 (en) 2004-06-07 2006-04-20 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
US20060240554A1 (en) 2005-02-14 2006-10-26 Sirna Therapeutics, Inc. Lipid nanoparticle based compositions and methods for the delivery of biologically active molecules
WO2009047006A2 (fr) * 2007-10-12 2009-04-16 Novosom Ag Perfectionnements aux ou se rapportant aux liposomes amphotères comprenant des lipides neutres
WO2009086558A1 (fr) * 2008-01-02 2009-07-09 Tekmira Pharmaceuticals Corporation Compositions et procédés améliorés pour la délivrance d'acides nucléiques

Non-Patent Citations (36)

* Cited by examiner, † Cited by third party
Title
"Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition", December 2000, WILEY
AKINC, A.; A, ZUMBUEHL; M. GOLDBERG; E.S. LESHCHINER; V. BUSINI; N. HOSSAIN; S.A. BACALLADO; D.N. NGUYEN; J. FULLER; R. ALVAREZ: "A combinatorial library of lipid-like materials for delivery of RNAi therapeutics", NAT. BIOTECH., vol. 26, 2008, pages 561 - 569
BEHR, ACC. CHEM. RES., vol. 26, 1993, pages 274 - 278
BOGGS, J.M.: "Lipid intermolecular hydrogen bonding: influence on structural organization and membrane function", BIOCHIM. BIOPHYS. ACTA, vol. 906, 1987, pages 353 - 404, XP025854891, DOI: doi:10.1016/0304-4157(87)90017-7
BRIGHAM ET AL., AM. J. SCI., vol. 298, no. 4, 1989, pages 278 - 281
CULLIS, P.R.; A.J. VERKLEIJ; P.H.J.TH. VERVERGAERT: "Polymorphic phase behavior of cardiolipin as detected by 31p NMR and freeze- fracture techniques. Effects of calcium, dibucaine and chlorpromazine", BIOCHIM. BIOPHYS. ACTA, vol. 513, 1978, pages 11 - 20
CULLIS, P.R.; M.J. HOPE; C.P.S. TILCOCK: "Lipid polymorphism and the roles of lipids in membranes", CHEM. PHYS. LIPIDS, vol. 40, 1986, pages 127 - 144
CULVER: "Human Gene Therapy", 1994, MARYANN LIEBERT, INC., pages: 70 - 71
ELSHABIR, S. ET AL., EMBO J., vol. 20, 2001, pages 6877 - 6888
ELSHABIR, S. ET AL., NATURE, vol. 411, 2001, pages 494 - 498
ELSHABIR, S.M. ET AL., EMBO, vol. 20, 2001, pages 6877 - 6888
ELSHABIR, S.M. ET AL., NATURE, vol. 411, 2001, pages 494 - 498
HAFEZ, I.M.; N. MAURER; P.R. CULLIS: "On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids", GENE THER., vol. 8, 2001, pages 1188 - 1196, XP002358998, DOI: doi:10.1038/sj.gt.3301506
HAFEZ, I.M.; P.R. CULLIS.: "Roles of lipid polymorphism in intracellular delivery", ADV. DRUG DELIVERY REV., vol. 47, 2001, pages 139 - 148
HEYES, J.; L. PALMER; K. BREMNER; I. MACLACHLAN: "Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids", J. CONTROL. RELEASE, vol. 107, 2005, pages 276 - 287, XP008157522, DOI: doi:10.1016/j.jconrel.2005.06.014
HOLTHUIS, J.M.C.; T.P. LEVINE.: "Lipid traffic: floppy drives and a superhighway", NAT. REV. MOL. CELL BIOL., vol. 6, 2005, pages 209
HOPE, M.J.; P.R. CULLIS.: "Effects of divalent cations and pH on phosphatidylserine model membranes: A 3'P NMR study", BIOCHEM. BIOPH. RES. CO., vol. 92, 1980, pages 846 - 852, XP024775899, DOI: doi:10.1016/0006-291X(80)90780-9
HSIEH, C.H.; S.C. SUE; P.C. LYU; W.G. WU: "Membrane packing geometry of diphytanoylphosphatidylcholine is highly sensitive to hydration: phopholipid polymorphism induced by molecular rearrangement in the headgroup region", BIOPHYS. J., vol. 73, 1997, pages 870 - 877
HUNZIKER ET AL.: "Nucleic Acid Analogues: Synthesis and Properties, in Modem Synthetic Methods", 1995, VCH, pages: 331 - 417
IKNONE, E.: "Cellular cholesterol trafficking and compartmentalization", NAT. REV. MOL. CELL BIOL., vol. 9, 2008, pages 125 - 138
KOYNOVA, R.; L. WANG; R.C. MACDONALD: "An intracellular lamellar-nonlamellar phase transition rationalizes the superior performance of some cationic lipid transfection agents", PROC. NATL. ACAD. SCI. U.S.A., vol. 103, 2006, pages 14373 - 14378
KULIG, K.; U. HOLZGRABE; B. MALAWSKA: "Stereocontrolled synthesis of the enantiomers of 1-[2-hydroxy-3-(4-phenyl-1-piperazinyl)-propyl]-pyrrolidin-2-one", TETRAHEDRON: ASYM., vol. 12, 2001, pages 2533 - 2536, XP004323479, DOI: doi:10.1016/S0957-4166(01)00390-1
LAMBERTON, J.S.; CHRISTIAN, A.T., MOLECULAR BIOTECHNOLOGY, vol. 24, 2003, pages 111 - 119
LEWIS, R.N.A.H.; R.N. MCELHANEY: "Surface charge markedly attenuates the nonlamellar phase-forming propensities of lipid bilayer membranes: calorimetric and 31 P-nuclear magnetic resonance studies of mixtures of cationic, anionic, and zwitterionic lipids", BIOPHYS. J., vol. 79, 2000, pages 1455 - 1464, XP055198322, DOI: doi:10.1016/S0006-3495(00)76397-1
LUZZATI, V.; T. GULIK-KRZY WICKI; A. TARDIEU: "Polymorphism of lecithins", NATURE, vol. 218, 1968, pages 1031 - 1034
MANNINO ET AL., BIOTECHNIQUES, vol. 6, 1988, pages 682 - 690
MAURER, N.; K.F. WONG; H. STARK; L. LOUIE; D. MCLNTOSH; T. WONG; P. SCHERRER; S.C. SEMPLE; P.R. CULLIS: "Spontaneous entrapment of polynucleotides upon electrostatic interactions with ethanol-destabilized cationic liposomes", BIOPHYS. J., vol. 80, 2001, pages 2310 - 2326, XP002589197
MAURER, N.; ZHIGALTSEV; P.R. CULLIS: "Lipsome Technology", 2007, article "Encapsulation of nucleic acid-based therapeutics", pages: 131 - 147
MESMAEKER ET AL.: "Novel Backbone Replacements for Oligonucleotides, in Carbohydrate Modifications in Antisense Research", 1994, ACS, pages: 24 - 39
NICOLAU ET AL., CRIT. REV. THER. DRUG CARRIER SYST., vol. 6, 1989, pages 239 - 271
SEMPLE, S.C.; S.K. KLIMUK; T.O. HARASYM; N. DOS SANTOS; S.M. ANSELL; K.F. WONG; N. MAURER; H. STARK; P.R. CULLIS; M.J. HOPE: "Efficient encapsulation of antisense oligonucleotides in lipid vesicles using ionisable aminolipids: formation of novel small multilamellar vesicle structures", BIOCHIM. BIOPHYS. ACTA, vol. 1510, 2001, pages 152 - 166
SILVIUS, J.R.; D. DEL GIUDICE; M. LAFLEUR: "Cholesterol at different bilayer concentrations can promote or antagonize lateral segregation of phospholipids of differing acyl chain length", BIOCHEMISTY, vol. 35, 1996, pages 15198 - 15208
STRAUBRINGER ET AL.: "METHODS IN ENZYMOLOGY", vol. 101, 1983, ACADEMIC PRESS, pages: 512 - 527
VEPKHISHVILI, N.G.; L.M. KHANANASHVILI; D.S. AKHOBADZE; Z.S. LOMTATIDZE; N.G. MAKHARADZE; N.G. GIORGOBIANI: "Synthesis and bactericidal properties of N-allyllactams", KHIM.-FARM. ZH., vol. 25, 1991, pages 35 - 36
VOELKER, D.R.: "New perspective on the regulation of intermembrane glycerophospholipid traffic", J. LIPID RES., vol. 44, 2003, pages 441 - 449
WANG, L.; R.C. MACDONALD.: "New strategy for transfection: mixtures of medium-chain and long chain cationic lipids synergistically enhance transfection", GENE THERA., vol. 11, 2004, pages 1358 - 1362

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11612657B2 (en) 2011-12-07 2023-03-28 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11246933B1 (en) 2011-12-07 2022-02-15 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11633480B2 (en) 2011-12-07 2023-04-25 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11633479B2 (en) 2011-12-07 2023-04-25 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11382979B2 (en) 2011-12-07 2022-07-12 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11679158B2 (en) 2011-12-07 2023-06-20 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11400158B2 (en) 2011-12-07 2022-08-02 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
US11590229B2 (en) 2011-12-07 2023-02-28 Alnylam Pharmaceuticals, Inc. Biodegradable lipids for the delivery of active agents
EP3594348A1 (fr) 2013-11-22 2020-01-15 Mina Therapeutics Limited Compositions d'arn à activation courte c/ebp alpha et leurs procédés d'utilisation
WO2015075557A2 (fr) 2013-11-22 2015-05-28 Mina Alpha Limited Compositions c/ebp alpha et méthodes d'utilisation
EP3985118A1 (fr) 2013-11-22 2022-04-20 MiNA Therapeutics Limited Compositions d'arn à activation courte c/ebp alpha et leurs procédés d'utilisation
JPWO2015093588A1 (ja) * 2013-12-21 2017-03-23 Ktnバイオテック株式会社 抗癌剤
US9770458B2 (en) 2013-12-21 2017-09-26 Nishizaki Bioinformation Research Institute Anti-cancer agent containing a diacylphosphatidylethanolamine as an active ingredient
WO2015093588A1 (fr) * 2013-12-21 2015-06-25 Ktnバイオテック株式会社 Agent anticancéreux
US11542229B2 (en) 2016-05-16 2023-01-03 The Board Of Regents Of The University Of Texas System Cationic sulfonamide amino lipids and amphiphilic zwitterionic amino lipids
US10562849B2 (en) 2016-05-16 2020-02-18 The Board Of Regents Of The University Of Texas System Cationic sulfonamide amino lipids and amphiphilic zwitterionic amino lipids
US11685710B2 (en) 2016-05-16 2023-06-27 The Board Of Regents Of The University Of Texas System Cationic sulfonamide amino lipids and amphiphilic zwitterionic amino lipids
WO2018064350A1 (fr) * 2016-09-30 2018-04-05 Eriochem Usa, Llc Nanoparticules lipidiques modifiées par apo-e pour administrer des médicaments à des tissus ciblés et méthodes thérapeutiques
EP4183882A1 (fr) 2017-09-08 2023-05-24 MiNA Therapeutics Limited Compositions stabilisées de petits arn activateurs (sarn) de hnf4a et procédés d'utilisation
WO2019048632A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions stabilisées de petits arn activateurs (parna) de hnf4a et procédés d'utilisation
EP4233880A2 (fr) 2017-09-08 2023-08-30 MiNA Therapeutics Limited Compositions de petits arn activateurs de hnf4a et procédés d'utilisation
WO2019048645A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions stabilisées de petits arn activateurs (parna) de cebpa et procédés d'utilisation
EP4219715A2 (fr) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Compositions stabilisées de saarn cebpa et procédés d'utilisation
WO2019048631A1 (fr) 2017-09-08 2019-03-14 Mina Therapeutics Limited Compositions de petits arn activateurs de hnf4a et procédés d'utilisation
EP4242307A2 (fr) 2018-04-12 2023-09-13 MiNA Therapeutics Limited Compositions
WO2019197845A1 (fr) 2018-04-12 2019-10-17 Mina Therapeutics Limited Compositions de sirt1-sarna et procédés d'utilisation
WO2020208361A1 (fr) 2019-04-12 2020-10-15 Mina Therapeutics Limited Compositions de sirt1-sarna et procédés d'utilisation
CN110746354A (zh) * 2019-10-29 2020-02-04 山东瑞博龙化工科技股份有限公司 一种丙烯酸酯类化合物及制备方法与应用
WO2022122872A1 (fr) 2020-12-09 2022-06-16 Ucl Business Ltd Agents thérapeutiques pour le traitement des troubles neurodégénératifs
WO2022200810A1 (fr) 2021-03-26 2022-09-29 Mina Therapeutics Limited Compositions de petits arn activateurs de tmem173 et procédés d'utilisation
WO2023099884A1 (fr) 2021-12-01 2023-06-08 Mina Therapeutics Limited Compositions d'arnsa de pax6 et procédés d'utilisation
WO2023104964A1 (fr) 2021-12-09 2023-06-15 Ucl Business Ltd Agents thérapeutiques pour le traitement de troubles neurodégénératifs
WO2023170435A1 (fr) 2022-03-07 2023-09-14 Mina Therapeutics Limited Compositions de petits arn activateurs d'il10 et procédés d'utilisation

Similar Documents

Publication Publication Date Title
WO2011056682A1 (fr) Lipides à têtes polaires inversées, compositions particulaires lipidiques comprenant les lipides à têtes polaires inversées, et procédés d'administration d'acides nucléiques
JP6409011B2 (ja) 活性剤の送達のための生分解性脂質
US10155945B2 (en) Method of producing lipid nanoparticles for drug delivery
JP5977394B2 (ja) 脂質に封入された干渉rna
JP2023071947A (ja) Rna送達のためのイオン化可能なカチオン性脂質
KR101762466B1 (ko) 지질, 지질 조성물 및 이의 사용 방법
EP3673898A1 (fr) Procédé de production de nanoparticules lipidiques pour l'administration de médicaments
US20120264810A1 (en) Compositions and methods for enhancing cellular uptake and intracellular delivery of lipid particles
EP2125031A2 (fr) Lipides et ensembles lipidiques comprenant des éléments d'amélioration de la transfection
JP7153109B2 (ja) 生分解性化合物および活性剤を含む脂質粒子を用いた、活性剤送達方法
JP2023537609A (ja) 脂質ナノ粒子を凍結乾燥する方法
AU2001254548B2 (en) Enhanced stabilised plasmid-lipid particle-mediated transfection using endosomal membrane
EP1938843A1 (fr) Lipides et ensembles lipidiques comprenant des éléments d'amélioration de la transfection
CA3194951A1 (fr) Encapsulation de grands rna dans des nanoparticules lipidiques
JP7043411B2 (ja) カチオン性脂質としての化合物
JP2024518375A (ja) Rna送達のためのイオン化可能なカチオン性脂質
WO2023225621A2 (fr) Lipides pour l'administration d'agents thérapeutiques
NZ583623A (en) Lipid encapsulated interfering RNA

Legal Events

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

Ref document number: 10776226

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10776226

Country of ref document: EP

Kind code of ref document: A1