US20020151070A1 - Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy - Google Patents

Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy Download PDF

Info

Publication number
US20020151070A1
US20020151070A1 US10/021,421 US2142101A US2002151070A1 US 20020151070 A1 US20020151070 A1 US 20020151070A1 US 2142101 A US2142101 A US 2142101A US 2002151070 A1 US2002151070 A1 US 2002151070A1
Authority
US
United States
Prior art keywords
cell
complex
compound
active substance
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/021,421
Inventor
Rainer Bischoff
Abdesslame Nazih
Yves Cordier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/021,421 priority Critical patent/US20020151070A1/en
Publication of US20020151070A1 publication Critical patent/US20020151070A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • 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

Definitions

  • the present invention relates to new lipid compounds and new compositions containing them. More particularly, the present invention relates to the use of said compounds or of said compositions to prepare a vector for transferring an active substance, in particular a therapeutically active substance comprising negative charges, in particular a polynucleotide, into a target cell, particularly a vertebrate cell, and more particularly a mammalian cell.
  • an active substance in particular a therapeutically active substance comprising negative charges, in particular a polynucleotide
  • the first category relates to physical techniques such as microinjection, electroporation or particle bombardment which, although effective, are greatly limited to applications in vitro and whose implementation is cumbersome and delicate.
  • the second category involves techniques relating to molecular and cell biology in which the gene to be transferred is combined with a vector of a biological or synthetic nature which promotes the introduction of said material.
  • vectors are viral, in particular adenoviral or retroviral, vectors.
  • the techniques developed are based on the natural properties which these viruses have to cross the cell membranes, to escape degradation of their genetic material and to cause their genome to penetrate into the nucleus.
  • These viruses have already been the subject of numerous studies and some of them are already used experimentally as vectors for genes in humans for the purpose, for example, of a vaccination, an immunotherapy or a therapy intended to make up for a genetic deficiency.
  • cationic lipids have been presented as molecules which are advantageous for promoting the introduction of large anionic molecules, such as nucleic acids, into certain cells. These cationic lipids are capable of complexing anionic molecules, thus tending to neutralize the negative charges on said molecules and to promote their coming close to the cells.
  • DOTMA (Felgner et al., 1987, PNAS, 84, 7413-7417), DOGS or TransfectamTM (Behr et al., 1989, PNAS, 86, 6982-6986), DMRIE and DORIE (Felgner et al., 1993, Methods 5, 67-75), DC-CHOL (Gao and Huang, 1991, BBRC, 179, 280-285), DOTAP ⁇ (McLachlan et al., 1995, Gene Therapy, 2,674-622) or Lipofectamine ⁇ , as well as those described in Patent Applications WO9116024 or WO9514651
  • Patent Application WO-A-9116024 describes cationic lipids of formula:
  • R 1 and R 2 are in particular alkyl or alkenyl radicals
  • Y 1 and Y 2 are radicals —OCH 2 —, —OC( ⁇ O)— or —O—;
  • R 3 and R 4 are alkyl or alkenyl radicals
  • R 5 is an alkylene chain
  • R 6 is C( ⁇ O)—(CH 2 ) m —NH—, a diaminocarboxylic acid or —C( ⁇ O)—(CH 2 ) m —NH— bound to said diaminocarboxylic acid;
  • R 7 is H, spermine, spermidine, histone, a protein, an amino acid or a polypeptide.
  • the Applicant has now identified new lipid compounds, which can be provided in cationic form, useful in particular for transferring an active substance, in particular a therapeutically active substance, comprising negative charges, in particular a polynucleotide, into a target cell, whose use may be envisaged in particular in vivo in the context of a gene therapy.
  • the subject of the present invention is first of all a lipid compound of formula:
  • R residues are, independently of each other, a hydrogen atom or a group of formula II:
  • R 1 and R 2 are, independently of each other, C 6 -C 23 alkyl or alkenyl radicals, which are linear or branched, or radicals —C( ⁇ O)—(C 6 -C 23 ) alkyl or —C( ⁇ O)—(C 6 -C 23 ) alkenyl, which are linear or branched, aryl radicals, cycloalkyl radicals, fluoroalkyl radicals, polyethylene glycol groups, oxyethylene or oxymethylene groups which are optionally repeated, linear or branched, optionally substituted,
  • [0021] is a positive integer from 1 to 4,
  • n is a positive integer from 1 to 6
  • m is a positive integer from 1 to 6 which may
  • alkenyl is understood to mean that the carbon chain may comprise one or more double bond(s) along said chain.
  • the invention relates to a compound chosen from the compounds of formulae:
  • R is a group of formula II as defined above and
  • R has one of the meanings indicated for the formula I provided that at least one R is a group of formula II and for each of the formulae:
  • n is a positive integer from 1 to 6
  • m is a positive integer from 1 to 6 which may be different for each motif —(CH 2 ) m , and more particularly for each motif —(CH 2 ) m —NR— when n>1.
  • the compounds of formula IIIb contain one or two R groups of formula II.
  • R 1 and R 2 are, independently of each other, linear —C( ⁇ O)-alkyl or linear —C( ⁇ O)-alkenyl radicals,
  • R 1 and R 2 are, independently o each other, —C( ⁇ O)-alkyl or —C( ⁇ O)-alkenyl radicals comprising from 12 to 20 carbon atoms, preferably 12, 16 or 18 carbon atoms, when the lipid comprises 1 or 2 R groups of formula II,
  • n is an integer chosen from the numbers 2, 3 or 4,
  • m is an integer chosen from the numbers 2, 3 or 4.
  • R 1 and R 2 are radicals having 6 to 10 carbon atoms, but in this case compounds for which the number of R groups of formula II is equal to 2, 3 or 4 will be preferably chosen.
  • the lipids according to the invention are chosen from the group consisting of the compounds of the following formulae: IV IVa
  • R 1 and R 2 are identical and are chosen from the stearoyl and oleoyl radicals.
  • R 3 and R 4 are protecting groups, in particular Fmoc (Grandas et al., 1989, Int. Journal pept. prot. Res. vol 33, 386-390), with an amine of formula:
  • R 5 being a protecting group, in particular t-butoxycarbonyl (BOC) or a hydrogen atom, at least one of the R 5 radicals and at most four of the R 5 radicals corresponding to the hydrogen atom.
  • BOC t-butoxycarbonyl
  • the compounds according to the invention may, in addition, be substituted.
  • substitutions may in particular consist-of a labeling molecule (see labeling molecules in U.S. Pat. No. 4,711,955) which makes it possible, for example, to visualize the distribution of the compounds or of the complexes containing them after administration in vitro or in vivo, a cell targeting molecule or an anchoring molecule.
  • the invention consequently also relates to a compound as presented above, conjugated with one or more targeting components, also called ligands of interest, via the intermediacy of at least a) one of the carbon atoms, in particular chosen from those present on the groups R 1 and/or R 2 , or b) one of the secondary or primary nitrogen atoms of the polyamine chain or of the diaminocarboxylic acid.
  • targeting components also called ligands of interest
  • oligonucleotides may be, for example, all or part of sugars, peptides (GRP peptide, Gastrin Releasing Peptide, for example), oligonucleotides, lipids, hormones, vitamins, antigens, antibodies, ligands specific for membrane receptors, ligands capable of reacting with an anti-ligand, fusogenic peptides, nuclear localization peptides, or a combination of such compounds.
  • galactosyl residues which make it possible to target the asyaloglycoprotein receptor at the surface of hepatic cells, the fusogenic peptide INF-7 derived from the influenza virus hemagglutinin subunit HA-2 (Plank et al., 1994, J.
  • Such conjugates can be easily obtained by techniques widely described in the literature, and more particularly by chemical coupling, in particular using protecting groups such as trifluoroacetyl or Fmoc or Boc; onto the polyamine and more particularly using one or more orthogonal protecting groups such as those described in Protective Groups in Organic Synthesis (p. 309-406, 1991, eds. T. W. Greene, P. G. M. Wuts, Wiley) onto the polyamine or the diaminocarboxylic acid.
  • the selective deprotection of a protecting group then makes it possible to couple the targeting component, and the lipid is then deprotected.
  • said compound is in a cationic form, that is to say that it is in a form which is protonated by binding of a proton onto one or more nitrogen atoms present on the polyamine chain.
  • said cationic lipid is combined with one or more biologically acceptable anions, such as for example the trifluoroacetate, halide, monomethylsulfate, acetate or phosphate, iodide, chloride, or bromide anion and the like. It is also possible to obtain compounds in cationic form by substitution of the amines, for example, with a methyl or ethyl radical, and the like.
  • the invention also relates to a composition
  • a composition comprising at least one compound as described above and optionally at least one adjuvant capable of enhancing the formation of the complex between a said compound and an active substance, or of enhancing the function of these complexes toward the cell.
  • such an adjuvant will be a neutral or zwitterionic lipid, such as for example a lipid which is or is derived from a triglyceride, a diglyceride, cholesterol (see for example U.S. Pat. No. 5,438,044), in particular, a neutral or zwitterionic lipid which is or is derived from a phosphatidylethanolamine (PE), phosphatidylcholine, phosphocholine, sphyngomyelin, ceramide or cerebroside.
  • PE phosphatidylethanolamine
  • DOPE dioleoylphosphatidylethanolamine
  • the weight ratio between the compound of the invention and the neutral or zwitterionic lipid is generally between 0.1 and 10, it being understood that this ratio may vary depending on the nature of the components considered. Persons skilled in the art have sufficient knowledge to allow these minor adaptations. It is also possible to use a mixture of neutral and/or zwitterionic lipids or alternatively a mixture of cationic lipids and neutral and/or zwitterionic lipids.
  • the invention relates, in addition, to a complex comprising at least one compound or at least one composition as described above and at least one active substance, in particular a therapeutically active substance, comprising at least one negative charge.
  • said complex may, in addition, contain one or more cationic amphiphilic agents such as those described in the literature of which examples were provided above.
  • said active substance is chosen from nucleic acids and proteins.
  • the active substance of the complex according to the invention is a polynucleotide, said compound or said composition then making it possible to enhance the transfecting power of the polynucleotide in a cell.
  • Polynucleotide is understood to designate a DNA and/or RNA fragment which is double-stranded or single-stranded, linear or circular, natural, isolated or synthetic, designating a precise succession of nucleotides, which are modified or otherwise (see by way of example U.S. Pat. No. 5,525,711), labeled or otherwise (see for example U.S. Pat. No. 4,711,955 or EP 302175), making it possible to define a fragment or a region of a nucleic acid without size limitation.
  • Polynucleotide is understood to designate in particular a cDNA, a genomic DNA, a plasmid DNA, a messenger RNA, an antisense RNA, a ribozyme, a transfer RNA, a ribosomal RNA or a DNA encoding such RNAs.
  • Polynucleotide or “nucleic acid” are synonymous terms in the context of the present application.
  • Antisense is understood to designate a nucleic acid having a sequence complementary to a target sequence, for example an mRNA sequence for which it is sought to block the expression by hybridization with the target sequence; “sense” is understood to designate a nucleic acid having a sequence homologous or identical to a target sequence, for example a sequence which binds to a proteinaceous transcription factor and which is involved in the expression of a given gene.
  • said polynucleotide comprises a gene of interest and components allowing the expression of said gene of interest.
  • said poly-nucleotide is advantageously in the form of a plasmid.
  • the components allowing expression are all the components allowing the transcription of said DNA fragment into RNA (antisense RNA or mRNA) and the translation of the mRNA into a polypeptide. They are in particular promoter sequences and/or regulatory sequences which are effective in said cell, and optionally the sequences required to allow excretion or expression of said polypeptide at the surface of the target cells.
  • promoters such as the promoters of the viruses RSV, MPSV, SV40, CMV or 7.5 k, of the vaccinia virus, the promoters of the gene encoding muscle creatine kinase, actin, or pulmonary surfactant. It is, in addition, possible to choose a promoter sequence specific for a given cell type or which can be activated under defined conditions.
  • the literature provides a large amount of information relating to such promoter sequences.
  • said polynucleotide may comprise at least two sequences, which are identical or different, exhibiting a transcriptional promoter activity and/or at least two coding DNA sequences, which are identical or different, situated, relative to each other, contiguously, far apart, in the same direction or in the opposite direction, as long as the transcriptional promoter function or the transcription of said sequences is not affected.
  • Said polynucleotide may also contain sequences required for intracellular transport, for replication and/or for integration.
  • polynucleotides according to the present invention may also be polynucleotides which are modified such that it is not possible for them to become integrated into the genome of the target cell or polynucleotides which are stabilized with the aid of agents such as, for example, spermine.
  • the polynucleotide may be homologous or heterologous to the target cell. It may be advantageous to use a polynucleotide which encodes all or part of a polypeptide, in particular a polypeptide having a therapeutic or prophylactic activity, and more particularly an immunogenic activity of the cellular or humoral type.
  • polypeptide is understood without restriction as to its size or its degree of modification (for example glycosylation).
  • HSV-1 Herpes Simplex
  • the complexes according to the invention are small in size (less than 500 nm, advantageously less than 200 nm and preferably less than 100 nm).
  • the transfection experiments carried out show that advantageously the weight ratio of the lipid compound according to the invention to said polynucleotide is 0.01 to 100.
  • the optimum ratio is between 0.05 and 10.
  • the invention also relates to a process for preparing the complexes cationic compounds/anionic active substances, said process being characterized in that one or more lipids in cationic form or a composition according to the invention whose lipid is in cationic form are brought into contact with one or more active substances comprising at least one negative charge and in that said complex is recovered, optionally after a purification step. It also relates to the kits for preparing such complexes comprising one or more lipids or one or more compositions according to the invention.
  • one or more cationic compounds are dissolved with an appropriate quantity of solvent or mixture of solvents which are miscible in water, in particular ethanol, dimethylsulfoxide (DMSO), or preferably a 1:1 (v:v) ethanol/DMSO mixture, so as to form lipid aggregates according to a known method described, for example, in Patent Application WO-A-9603977, or according to a second variant, are suspended with an appropriate quantity of a solution of detergent such as an octylglucoside such as n-octyl- ⁇ -D-glucopyranoside, or 6-O-(N-heptylcarbomoyl)-methyl- ⁇ -D-glucopyranoside.
  • a solution of detergent such as an octylglucoside such as n-octyl- ⁇ -D-glucopyranoside, or 6-O-(N-heptylcarbomoyl)-methyl- ⁇ -D-glucopyranoside.
  • the suspension may then be placed in a buffer medium and mixed with a solution of active substance comprising negative charges.
  • a film is formed, in the known manner, prior to the dissolution in the solvent which is miscible with water or in the solution of detergent, with a mixture containing a said cationic compound and a said neutral or zwitterionic lipid, such as for example DOPE.
  • One of the important characteristics of the process consists in the choice of the ratio between the positive charges of the cationic lipid and the negative charges of the active substance.
  • quantities of the different charges will be chosen so that the ratio between the number of positive charges of the compound or of the cationic composition and the number of negative charges of the active substance is between 0.05 and 20, in particular between 0.1 and 15, and preferably between 5 and 10.
  • This ratio between the number of positive charges of the cationic compound(s) and/or composition(s) and the number of negative charges of said active substance also constitutes an advantageous characteristic of the complex according to the invention.
  • subsequent dialysis may be carried out in order to reduce the detergent and to recover the complexes.
  • the principle of such a method is for example described by Hofland et al. (1996, PNAS 93, p 7305-7309) and in chapter II of the Philippot et al. document (G. Gregoriadis, 81-89, CRC Press 1993).
  • one or more cationic compositions or compounds are suspended in a buffer and then the suspension is subjected to sonication until visual homogeneity is obtained.
  • the lipid suspension is then extruded through two microporous membranes under appropriate pressure.
  • the lipid suspension is then mixed with a solution of active substance comprising negative charges.
  • a film of the mixture of cationic lipid and neutral lipid such as DOPE is formed in a known manner prior to the preparation as a suspension.
  • This so-called sonication-extrusion technique is well known in the art.
  • the characteristics of the complexes formed may be evaluated by several means which make it possible to determine, for example:
  • the object of the present invention is also the complexes obtained using the processes listed above.
  • the invention also relates to the use of a compound, of a composition or of a complex according to the invention to transfer at least one active substance, especially a therapeutically active substance, more particularly a nucleic acid, into target cells, in vitro, ex vivo or in vivo, more particularly in vivo.
  • “Target cells” is understood to mean prokaryotic cells, yeast cells and eukaryotic cells, plant cells, human or animal cells, and in particular mammalian cells. Cancer cells should, moreover, be mentioned.
  • the invention may be applied at the level of the interstitial or luminal space of tissues such as the lungs, trachea, skin, muscle, brain, liver, heart, spleen, bone marrow, thymus, bladder, lymph, blood, pancreas, stomach, kidney, ovaries, testicles, rectum, peripheral or central nervous system, eyes, lymphoid organs, cartilages and endothelium.
  • the target cell will be a muscle cell, a hematopoietic stem cell or alternatively a cell of the airways, more particularly a tracheal or pulmonary cell, and advantageously a cell of the respiratory epithelium.
  • the complexes according to the invention can be used as a medicament for curative, preventive or vaccinal purposes.
  • the subject of the invention is also the complexes of the invention as a medicament for curative, preventive or vaccinal purposes.
  • Such complexes may be used in a method of therapeutic treatment which consists in transferring at least one therapeutically active substance, in particular a polynucleotide, into target cells, in particular a mammalian cell, and more precisely a muscle cell, a hematopoietic stem cell, a cell of the airways, more particularly a tracheal or pulmonary cell, a cell of the respiratory epithelium.
  • a compound according to the invention is most particularly advantageous for transferring a nucleic acid into a muscle cell or a pulmonary cell. It is the compound noted pcTG37 (see example).
  • the present invention also relates to a process for introducing an active substance comprising negative charges into a cell in particular in vitro, characterized in that cells, in particular cultured on an appropriate medium are brought into contact with a complex cationic compound/active substance comprising at least one negative charge according to the invention, in particular in the form of a suspension of complexes. After a certain incubation time, the cells are washed and recovered. The introduction of the active substance may be checked (optionally after lysis of the cell) by any appropriate means.
  • introduction is understood to mean that the active substance comprising negative charges is transferred into the cell and is located, at the end of the process, inside said cell or at the level of the membrane thereof.
  • the active substance is a nucleic acid
  • transfection in this case, the verification of the transfection of the nucleic acid can be carried out by any appropriate means, for example by measuring the expression of the gene considered or the concentration of the expressed protein.
  • the invention relates more particularly to the use of a compound, of a composition or of a complex according to the invention for the preparation of a medicament for curative, preventive or vaccinal purposes, intended for the treatment of the human or animal body, in particular by gene therapy.
  • the medicament may be administered directly in vivo (for example into a muscle, into the lungs by aerosol and the like) . It is also possible to adopt the ex vivo approach which consists in collecting cells from the patient (bone marrow stem cells, peripheral blood lymphocytes, muscle cells and the like), transfecting them in vitro according to the present invention and readministering them to the patient.
  • the complexes according to the invention may be administered by the intramuscular, intratracheal, intranasal, intracerebral, intrapleural, intratumoral, intracardiac, intragastric, intraperitoneal, epidermal, intravenous or intraarterial route by a syringe or by any other equivalent means, systems suitable for the treatment of the airways or of the mucous membranes such as inhalation, instillation or aerosolization.
  • modes of administration by application of a cream, by oral administration or any other means perfectly known to the person skilled in the art and applicable to the present invention.
  • the invention also relates to a method of gene therapy consisting in administering to a patient an appropriate quantity of a composition according to the invention.
  • a method of gene therapy consisting in administering to a patient an appropriate quantity of a composition according to the invention.
  • the administration may take place in a single dose or repeated once or several times after a certain time interval.
  • the repeated administration would make it possible to reduce the quantity of therapeutically active substance, more particularly of DNA, to be administered for a given dose.
  • the appropriate route of administration and dosage vary according to various parameters, for example the individual or disease to be treated or alternatively the polynucleotide to be transferred.
  • the invention relates more particularly to a pharmaceutical preparation comprising at least one complex as described above, optionally containing, in addition, at least one adjuvant capable of stabilizing said pharmaceutical preparation for the purpose of its storage for example and/or of enhancing the transfecting power of said complex.
  • Such an adjuvant could, for example, be chosen from the group consisting of chloroquine, a protic polar compound chosen in particular from propylene glycol, polyethylene glycol, glycerol, ethanol, 1-methyl-L-2-pyrrolidone or derivatives thereof, or an aprotic polar compound chosen in particular from dimethyl sulfoxide (DMSO), diethyl sulfoxide, di-n-propyl sulfoxide, dimethyl sulfone, sulfolane, dimethylformamide, dimethylacetamide, tetramethylurea, acetonitrile or derivatives thereof.
  • DMSO dimethyl sulfoxide
  • said preparation may contain a pharmaceutically acceptable carrier allowing its administration to humans or animals.
  • the invention relates to a cell transfected with a complex as defined above, particularly a prokaryotic cell, a yeast cell or eukaryotic cell, especially an animal cell, in particular a mammalian cell, and more particularly a cancer cell.
  • said cell is a cell of the airways, more particularly a tracheal or pulmonary cell, and advantageously a cell of the respiratory epithelium.
  • FIG. 1 An explanatory diagram of synthesis which forms an integral part of the description is appended (FIG. 1).
  • FIG. 2 indicates the results observed after intravenous injection of complexes according to the invention, the luciferase activity is indicated in RLU/mg of protein).
  • the Fmoc groups are removed from the product in a solution of 20% piperidine, 20% tetrahydrofuran and 60% CH 2 Cl 2 for 1 hour.
  • another purification is carried out on a silica column in a 1/1 CH 2 Cl 2 /CH 3 OH solvent.
  • the fractions containing the product are determined by thin-layer chromatography.
  • the product is dried before the last coupling. 510 ⁇ mol of the N-hydroxysuccinimide ester of oleic acid (Sigma 0 9506) diluted in 5 ml of anhydrous CH 2 Cl 2 are added to di-boc-spermidine-diaminopropionamide.
  • the product is purified on a silica column (ether/ethyl acetate; 1/1).
  • the final product, taken up in 1 ml of CH 2 Cl 2 is Boc deprotected in 10 ml of trifluoroacetic acid (Fluka 91699).
  • evaporation is carried out under reduced pressure after dilution in 50 ml of n-hexane.
  • the compound pcTG39 is prepared in an amount of 140 ⁇ mol in the same manner using stearic acid and dicyclohexylcarbodiimide or (benzotriazol-1-yloxy) tri-pyrrolidinophosphonium hexafluorophosphate (PyBOP) as coupling agent.
  • the quantities of lipids are calculated based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the chosen cationic lipid.
  • concentration of final DNA 0.1 mg/ml for the tests in vitro
  • desired charge ratio the concentration of the chosen cationic lipid.
  • molar mass the concentration of the chosen cationic lipid.
  • the different ingredients are mixed according to the following calculation:
  • 0.1 mg of DNA/ml that is to say (0.1/330) mmol of negative charges (330 Da is the average molecular weight of a nucleotide) per ml correspond to 0.30 ⁇ mol/ml of negative charges.
  • a concentration of 3.0 ⁇ mol/ml of positive charges provided by the cationic lipid is required.
  • the molar mass of pcTG37 in trifluoroacetate form is 988 g/mol and the molecule contains 2 positive charges. Therefore, 1.5 ⁇ mol/ml of pcTG37 is required, which corresponds to 1.49 mg/ml.
  • the lipids are taken up in chloroform, dried by evaporation and then solubilized in chloroform/methanol (v:v) and again dried.
  • the cationic lipids are weighed and the quantity of DOPE is added from a stock solution of 10 or 20 mg/ml in chloroform to a glass tube sterilized with alcohol and with UV in order to obtain a cationic lipid concentration of 2 mM.
  • the solvents are evaporated under vacuum (0.2 ⁇ 105 Pa (200 mbar)) for 45 min at 45° C. using a vortex of 40 revolutions per minute (Labconco, Rapidvap, Uniequip, Martinsried, Germany).
  • the lipid film is taken up in ethanol so as to be at the cationic lipid concentration of 50 mg/ml.
  • the plasmid DNA is prepared from a stock solution at 1 mg/ml (10 mM Tris, 1 mM EDTA, pH 7.5).
  • the lipids are added to the DNA.
  • the suspension is mixed by aspiration/discharge using a pipette (10 times).
  • the complexes are stored at +4° C.
  • the quantities of lipids are calculated as described above based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the cationic lipid chosen.
  • the lipids are mixed in a glass tube, sterilized with alcohol and with UV, in order to obtain a 2 mM cationic lipid solution (see above).
  • the solvents are evaporated and the lipid film is taken up in a solution of n-octyl, ⁇ -D-glucopyranoside (octylglucoside, Sigma, O 9882) according to a cationic lipid/detergent ratio of 1:5 (mol:mol).
  • 375 ⁇ l of a 20 mM octylglucoside solution in 20 mM HEPES pH 7.5 are collected and used to take up the film of pcTG37/DOPE lipid mixture.
  • the plasmid DNA is prepared from a stock solution of plasmid DNA at 1 mg/ml of which 50 ⁇ l are removed for a final volume of 0.5 ml (0.1 mg/ml final) to which 262.5 ⁇ l of 20 mM HEPES pH 7.5 are added. 187.5 ⁇ l of the lipid suspension are added to the DNA by aspirating and discharging 10 times using a pipette in order to obtain the final suspension at 0.1 mg/ml of DNA and a +/ ⁇ charge ratio of 10.
  • a dialysis of 3 times 4 hours at room temperature against 20 mM HEPES pH 7.5 is carried out in dialysis microbags (cut-off of 13.2 kD; Sartorius, Göttingen, Germany).
  • the dialyzed DNA/lipid complexes are stored at +4° C.
  • the preparation is carried out in a laminar flow cabinet.
  • the quantities of lipids are calculated as described above based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the cationic lipid chosen.
  • the lipids are mixed in a glass tube, sterilized with alcohol and with UV, in order to obtain a 2 mM cationic lipid solution, as indicated above.
  • the solvents are evaporated and the lipid film is taken up in 900 ⁇ l of 20 mM HEPES pH 7.5 at 4° C. for about 16 h.
  • the suspension is sonicated in a sonication bath (Bransonic 221) to visual homogeneity.
  • the lipid suspension is extruded through two membranes with a pore diameter of 0.2 ⁇ m (Nucleopore, Costar, Cambridge, Mass., USA) and rinsed with 20 mM HEPES pH 7.5 (extruder from Lipex Biomembranes, Vancouver, Canada) at a maximum pressure of 50 bars.
  • the lipid suspension is kept at room temperature for 1 hour.
  • 450 ⁇ l of the lipid suspension are added to 50 ⁇ l of a stock solution of plasmid DNA (1 mg/ml) and mixed by aspirating/discharging 10 times using a pipette.
  • the lipid/DNA complexes are stored at +4° C.
  • the preparations are carried out under a laminar flow cabinet.
  • a 1% (w:v) agarose gel is prepared in a TAE buffer (TAE: Tris 4.86 g/l+sodium acetate 0.68 g/l+EDTA 0.336 g/l pH 7.8. If necessary, the sample is diluted in TAE and then the sample buffer 0.083% bromophenol blue, 0.083% cyanol xylene FF, 10% glycerol in water) is added so as to have 50 ng DNA/ ⁇ l. The sample is briefly subjected to a vortex and left for 30 min at room temperature. As a control, the non-complexed plasmid prepared at the same concentration is used.
  • the analyses are carried out on a Coulter N4Plus (Coultronics France S.A., Margency, France) at 25° C. after equilibration of the sample for 20 min. An aliquot of the sample is aspirated and discharged several times before being pipetted. The sample is diluted in the measuring tank and homogenized. The measurement of the light diffracted at 90° is carried out for 180 sec after a 180 sec wait. The range used goes from 3 nm to 10 000 nm using 31 bins. To be valid, the sample should give between 50 000 and 1 000 000 counts/sec.
  • the cells are inoculated 24 h to 48 h before the transfection into a 96-well culture plate with about 5 ⁇ 10 3 to 10 4 cells per well, at about 30% confluence, and kept at 37° C. under a 5% CO 2 and 95% air atmosphere.
  • the transfections are carried out with mixtures of variable quantities of lipids and plasmid DNA in order to determine the charge ratios and the optimum DNA concentrations per well.
  • the complexes used are prepared 24 h to 48 h before the transfection and diluted in HamF 14 medium containing 40 ⁇ g/ml of gentamycin and 2 mM glutamine.
  • All the transfection media are then adjusted to 10% FCS, 10 ⁇ g/ml of insulin (Sigma), 10 ng/ml of EGF (Sigma) and of FGF (Pepro Tech Inc., Rocky Hill, N.J.), 2 mM glutamine (bioMérieux), and 40 ⁇ g/ml of gentamycin (Schering Plough) for a final volume of 250 ⁇ l.
  • the cultures are incubated for 48 h and then the cells are recovered and tested for their capacity to express the luciferase gene.
  • the protein concentrations are determined by the system for testing quantity of protein (Promega).
  • the A549 cells are cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal calf serum (Gibco BRL) 24 hours before the start of the transfection in 96-well plates (2 ⁇ 10 4 cells per well) in a humid atmosphere at 37° C. and 5% CO 2 /95% air. For the transfection in the absence of serum, the medium is removed and replaced with serum-free medium.
  • DMEM Dulbecco's modified Eagle's medium
  • Gibco BRL 10% fetal calf serum
  • lipid/DNA complexes In another microplate, the following suspensions of lipid/DNA complexes are prepared (lipid/DNA complexes at 0.1 mg/ml of DNA and at the indicated charge ratio):44 ⁇ l (4.4 ⁇ g DNA), 22 ⁇ l (2.2 ⁇ g DNA), 5.5 ⁇ l (0.55 ⁇ g DNA) of stock solution in the first 3 wells, and 11 ⁇ l (0.11 ⁇ g DNA) of the stock solution diluted 10-fold in the next well. The volume is adjusted to 110 ⁇ l with DMEM and 100 ⁇ l are transferred over the A549 cells. The incubation is carried out with 4, 2, 0.5 and 0.1 ⁇ g of DNA per well for 4 hours.
  • DMEM+30% fetal calf serum 50 ⁇ l of DMEM+30% fetal calf serum are added 4 hours after the start of transfection and then 100 ⁇ l of DMEM+10% FCS 24 hours after the start of transfection.
  • the transfections in the presence of 10% fetal calf serum are carried out in an identical manner except that the transfection occurs in medium with serum.
  • the medium is removed and the cells are washed with 100 ⁇ l of PBS phosphate solution and lyzed with 50 ⁇ l of lysis buffer (Promega).
  • the lysates are frozen at ⁇ 80° C. until the luciferase activity is measured. The latter is carried out on 20 ⁇ l of mixture for one minute using the Luciferase” determination system (Promega) (LB96P Berthold luminometer) in 96-well plates in kinetic mode.
  • N-N′-(di-boc)-N,N′-(dipropio-nitrile)-1,4-diaminobutane is obtained by reacting 37.6 mmol of diaminobutane (Fluka; reference 32790) diluted in 3.8 ml of dichloromethane+2 ml of methanol at 0° C. with 75.2.mmol of acrylonitrile (Fluka; reference 01710), for 1 hour at 0° C.
  • the product of the reaction is blocked with 100 mmol of BOC-ON [(2-boc-oxyimino)-2-phenylacetonitrile] (Fluka; reference 15475) in 50 ml of 2/1 CH 2 Cl 2 /CH 3 OH solvent.
  • the blocking reaction is placed overnight at room temperature.
  • the solvents are evaporated and the product is taken up in 200 ml of ethyl acetate and then subjected to 3 extractions with 200 ml of 1 M NaOH.
  • the organic phase is again extracted with 5% citric acid, dried and filtered over Na 2 SO 4 before evaporation to dryness.
  • a step of reducing the nitrites is then carried out in ether at 0° C.
  • the solvents are evaporated and the product is taken up in 300 ml of ethyl acetate for 3 extractions with 300 ml of 1 M NaOH.
  • the organic phase is again extracted with 5% citric acid, dried and filtered over Na 2 SO 4 before evaporation to dryness.
  • the step of reduction of the nitrites is then carried out in ether at 0° C. in the presence of 140 mmol of LiAlH4 (Sigma; L0260). After 15 hours, the reaction is stopped by addition of 100 ml of 1 M sodium hydroxide at 0° C., with evolution of hydrogen.
  • the mixture is filtered and extracted three times with a 20% NaCl solution. After drying the ethereal fraction, the product is ready to be deposited on a silica column for the final purification.
  • the activated ester precipitates upon the addition and the reaction takes place in a heterogeneous phase for 15 hours, with stirring, at room temperature. And the same purification protocol (see N- ⁇ -N- ⁇ -di[oleylamido]-propionic acid) is applied. Weighing: 0.14 g (0.15 mmol), yield: 15%.
  • mice used are 9- to 11-week-old C57BL/6 female mice.
  • the mice are pretreated by injecting 50 ⁇ l of a preparation of chlodronate encapsulated in a liposome (see Van Rooijen and Sanders, 1994, J. Immunol. Methods, 174, 83-93) incorporated in a total volume of 200 ⁇ l (+150 ⁇ l of PBS buffer).
  • the intravenous injections are carried out into the tail after disinfecting the skin with 70% ethanol.
  • the volume injected is 250 ⁇ l and the quantity of DNA is 60 ⁇ g, the quantity of lipid is 345 ⁇ g.
  • mice Two days after the injections, the mice are sacrificed. After extraction, the tissues are frozen in liquid nitrogen and stored at ⁇ 80° C. To measure the luciferase activity, the tissues are mechanically ground with the aid of a pestle in a mortar placed on dry ice. 500 ⁇ l of a lysis buffer (Promega) are added to the tissue debris obtained from the lungs and subjected to three freeze/thaw steps. The cellular debris is removed by centrifugation and the luciferase activity (in RLU/min, relative light unit per minute) is measured on 20 ⁇ l of supernatant in accordance with the manufacturer's instructions (Promega) by adding 100 ⁇ l of reagent and by measuring the activity by luminescence.
  • a lysis buffer Promega
  • the luciferase activity measured is standardized relative to the protein quantity with the aid of a calibration series prepared by commercially available luciferase (Promega).
  • the total protein quantity is, moreover, determined by the bicinchoninic acid BCA colorimetric method (Smith et al., 1985, Anal. Biochem., 150, 76-85 Pierce) using an aliquot of supernatant. This makes it possible to express the luciferase activity in RLU per milligram of protein extracted from the tissues. Controls are carried out for which the mice were not injected or were injected with DNA alone (60 ⁇ g) in a 20 mM HEPES buffer pH 7.5. The results (FIG.

Landscapes

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

Abstract

The invention relates to new lipid compounds of formula:
R—HN—[—(CH2)m—NR—]n−1—(CH2)m—NH—R  I
in which:
the R residues are, independently of each other, a hydrogen atom or a group of formula II:
Figure US20020151070A1-20021017-C00001
 for which:
R1 and R2 are, independently of each other, C6-C23 alkyl or alkenyl radicals, which are linear or branched, or radicals —C(═O)—(C6-C23) alkyl or —C(═O)—(C6-C23) alkenyl, which are linear or branched, aryl radicals, cycloalkyl radicals, fluoroalkyl radicals, polyethylene glycol groups, oxyethylene or oxymethylene groups which are optionally repeated, linear or branched, optionally substituted,
p is a positive integer from 1 to 4,
n is a positive integer from 1 to 6,
m is a positive integer from 1 to 6 which may be different for each motif —(CH2)m, and more particularly for each motif —(CH2)m—NR— when n>1,
the number of R groups of formula II being between 1 and 4
said compounds being optionally in a cationic form and being combined with one or more biologically acceptable anions.
It also relates to new complexes comprising at least one said cationic compound and an active substance comprising negative charges allowing the introduction of said active substances into cells. It relates in particular to new complexes, in which the active substance consists of one or more nucleic acids, useful for transfecting cells.

Description

  • The present invention relates to new lipid compounds and new compositions containing them. More particularly, the present invention relates to the use of said compounds or of said compositions to prepare a vector for transferring an active substance, in particular a therapeutically active substance comprising negative charges, in particular a polynucleotide, into a target cell, particularly a vertebrate cell, and more particularly a mammalian cell. [0001]
  • The transfer of a gene into a given cell is the very basis of gene therapy. This new technology, whose field of application is vast, makes it possible to envisage the treatment of serious diseases for which the conventional therapeutic alternatives are not very effective, or are even inexistent, and applies to diseases which are either of genetic origin (hemophilia, cystic fibrosis, myopathy and the like) or acquired (cancer, AIDS and the like). [0002]
  • During the past 30 years, numerous tools have been developed which allow the introduction of various heterologous genes into cells, in particular mammalian cells. These different techniques may be divided into two categories. The first category relates to physical techniques such as microinjection, electroporation or particle bombardment which, although effective, are greatly limited to applications in vitro and whose implementation is cumbersome and delicate. The second category involves techniques relating to molecular and cell biology in which the gene to be transferred is combined with a vector of a biological or synthetic nature which promotes the introduction of said material. [0003]
  • Currently, the most effective vectors are viral, in particular adenoviral or retroviral, vectors. The techniques developed are based on the natural properties which these viruses have to cross the cell membranes, to escape degradation of their genetic material and to cause their genome to penetrate into the nucleus. These viruses have already been the subject of numerous studies and some of them are already used experimentally as vectors for genes in humans for the purpose, for example, of a vaccination, an immunotherapy or a therapy intended to make up for a genetic deficiency. However, this viral approach has many limitations, in particular because of the limited capacity for cloning into the viral genome, the risks of spreading in the host organism and in the environment the infectious viral particles produced, the risk of artefactual mutagenesis by insertion into the host cell in the case of retroviral vectors, and the high induction of immune and inflammatory responses in vivo during the therapeutic treatment, considerably limiting the number of administrations which can be envisaged (McCoy et al., 1995, Human Gene Therapy, 6, 1553-1560; Yang et al., 1996, Immunity, 1, 433-442)1 These numerous disadvantages, in particular in the context of a use in humans, have led several teams to develop alternative systems of transferring poly-nucleotides. [0004]
  • Several nonviral methods are currently available. By way of example, there may be mentioned coprecipitation with calcium phosphate, the use of receptors mimicking viral systems (for a review see Cotten and Wagner, 1993, Current Opinion in Bio-technology, 4, 705-710), or the use of polymers such as polyamidoamine (Haensler and Szoka, 1993, Bioconjugate Chem., 4, 372-379) or of polymer such as those presented in WO 95/24221 describing the use of dendritic polymers, the document WO 96/02655 describing the use of polyethyleneimine, or of polypropyleneimine and the documents U.S. Pat. No. 5,595,897 and FR 2,719,316 describing the use of conjugates of polylysine. Other non-viral techniques are based on the use of liposomes whose value as agent allowing the introduction, into cells, of certain biological macromolecules, such as for example DNA, RNA, proteins or certain pharmaceutically active substances, has been widely described in the literature. To this end, several teams have already proposed the use of cationic lipids which have a high affinity for the cell membranes and/or the nucleic acids. Indeed, although it has been shown, in the case of nucleic acids, that this type of macromolecule is capable of crossing the plasma membrane of some cells in vivo (WO 90/11092), it is nevertheless the case that the observed transfection efficiency is still highly limited, because of in particular the polyanionic nature of the nucleic acids which prevent their passage across the cell membrane, which itself has a negative net apparent charge. Since 1989 (Felgner et al., Nature, 337, 387-388), cationic lipids have been presented as molecules which are advantageous for promoting the introduction of large anionic molecules, such as nucleic acids, into certain cells. These cationic lipids are capable of complexing anionic molecules, thus tending to neutralize the negative charges on said molecules and to promote their coming close to the cells. Many teams have already developed various cationic lipids. By way of example, there may be mentioned DOTMA (Felgner et al., 1987, PNAS, 84, 7413-7417), DOGS or Transfectam™ (Behr et al., 1989, PNAS, 86, 6982-6986), DMRIE and DORIE (Felgner et al., 1993, [0005] Methods 5, 67-75), DC-CHOL (Gao and Huang, 1991, BBRC, 179, 280-285), DOTAP□ (McLachlan et al., 1995, Gene Therapy, 2,674-622) or Lipofectamine□, as well as those described in Patent Applications WO9116024 or WO9514651
  • More particularly, Patent Application WO-A-9116024 describes cationic lipids of formula: [0006]
    Figure US20020151070A1-20021017-C00002
  • in which: [0007]
  • R[0008] 1 and R2 are in particular alkyl or alkenyl radicals;
  • Y[0009] 1 and Y2 are radicals —OCH2—, —OC(═O)— or —O—;
  • R[0010] 3 and R4 are alkyl or alkenyl radicals;
  • R[0011] 5 is an alkylene chain;
  • R[0012] 6 is C(═O)—(CH2)m—NH—, a diaminocarboxylic acid or —C(═O)—(CH2)m—NH— bound to said diaminocarboxylic acid;
  • R[0013] 7 is H, spermine, spermidine, histone, a protein, an amino acid or a polypeptide.
  • However, several studies (by way of examples, see Mahato et al., J. Pharm. Sci., 1995, 84, 1267-1271, Thierry et al., 1995, P.N.A.S., 92, 9742-9746) have demonstrated that the efficiency of transferring the anionic macromolecule into cells could vary depending in particular on the interaction between the complexes and the cell membranes, the cell considered, the lipid composition of the cationic compounds, the size of the complexes formed with the anionic molecules and more particularly the ratio between the positive and negative charges on the different components of said complex. The mechanisms which allow in particular the interaction of the complexes with the cell membranes and the transfer of the complexes into the cell are still to a large extent poorly understood and researchers proceed in their studies based on a highly empirical approach. Other factors such as, for example, the formation of the complexes, the stability, the behavior in vivo, or possibly their toxicity make, in addition, the choice of the lipids to priori non-obvious. It is consequently desirable to provide other cationic lipids possibly having improved properties or properties which are different from the cationic lipids already described. [0014]
  • The Applicant has now identified new lipid compounds, which can be provided in cationic form, useful in particular for transferring an active substance, in particular a therapeutically active substance, comprising negative charges, in particular a polynucleotide, into a target cell, whose use may be envisaged in particular in vivo in the context of a gene therapy. [0015]
  • Accordingly, the subject of the present invention is first of all a lipid compound of formula:[0016]
  • R—HN—[—(CH2)m—NR—]n−1—(CH2)m—NH—R  I
  • in which: [0017]
  • the R residues are, independently of each other, a hydrogen atom or a group of formula II: [0018]
    Figure US20020151070A1-20021017-C00003
  • in which: [0019]
  • R[0020] 1 and R2 are, independently of each other, C6-C23 alkyl or alkenyl radicals, which are linear or branched, or radicals —C(═O)—(C6-C23) alkyl or —C(═O)—(C6-C23) alkenyl, which are linear or branched, aryl radicals, cycloalkyl radicals, fluoroalkyl radicals, polyethylene glycol groups, oxyethylene or oxymethylene groups which are optionally repeated, linear or branched, optionally substituted,
  • is a positive integer from 1 to 4, [0021]
  • n is a positive integer from 1 to 6, [0022]
  • m is a positive integer from 1 to 6 which may [0023]
  • be different for each motif —(CH[0024] 2)m, and more
  • particularly for each motif —(CH[0025] 2)m—NR— when n>1,
  • the number of R groups of formula II being between 1 and 4. [0026]
  • The expression “alkenyl” is understood to mean that the carbon chain may comprise one or more double bond(s) along said chain. [0027]
  • According to a specific case, the invention relates to a compound chosen from the compounds of formulae:[0028]
  • H2N—[—(CH2)m—NH—]nR  III
  • RNH—[—(CH2)m—NH]nH  IIIa
  • in which: [0029]
  • R is a group of formula II as defined above and[0030]
  • H2N—[—(CH2)m—NR]n−1—(CH2)m—NH2  IIIb
  • in which: [0031]
  • R has one of the meanings indicated for the formula I provided that at least one R is a group of formula II and for each of the formulae: [0032]
  • n is a positive integer from 1 to 6, [0033]
  • m is a positive integer from 1 to 6 which may be different for each motif —(CH[0034] 2)m, and more particularly for each motif —(CH2)m—NR— when n>1.
  • According to a preferred case, the compounds of formula IIIb contain one or two R groups of formula II. [0035]
  • According to preferred embodiments of the invention, the variations presented below, taken in combination with each other or otherwise, will be chosen: [0036]
  • R[0037] 1 and R2 are, independently of each other, linear —C(═O)-alkyl or linear —C(═O)-alkenyl radicals,
  • R[0038] 1 and R2 are, independently o each other, —C(═O)-alkyl or —C(═O)-alkenyl radicals comprising from 12 to 20 carbon atoms, preferably 12, 16 or 18 carbon atoms, when the lipid comprises 1 or 2 R groups of formula II,
  • n is an integer chosen from the numbers 2, 3 or 4, [0039]
  • m is an integer chosen from the numbers 2, 3 or 4. [0040]
  • According to a specific case, it is possible to reduce the length of the alkyl or alkenyl chain so that R[0041] 1 and R2 are radicals having 6 to 10 carbon atoms, but in this case compounds for which the number of R groups of formula II is equal to 2, 3 or 4 will be preferably chosen.
  • Preferably, the lipids according to the invention are chosen from the group consisting of the compounds of the following formulae: [0042]
    IV
    Figure US20020151070A1-20021017-C00004
    IVa
    Figure US20020151070A1-20021017-C00005
  • for which R[0043] 1 and R2 are identical and are chosen from the stearoyl and oleoyl radicals.
  • The compounds according to the invention are prepared by reacting a compound or formula: [0044]
    Figure US20020151070A1-20021017-C00006
  • in which: [0045]
  • R[0046] 3 and R4 are protecting groups, in particular Fmoc (Grandas et al., 1989, Int. Journal pept. prot. Res. vol 33, 386-390), with an amine of formula:
  • R5NH[(—CH2)m—NR5]n−1—(CH2)mNHR5  VI
  • m and n having the same meaning as for the formula I, [0047]
  • R[0048] 5 being a protecting group, in particular t-butoxycarbonyl (BOC) or a hydrogen atom, at least one of the R5 radicals and at most four of the R5 radicals corresponding to the hydrogen atom.
  • The functional groups N—R[0049] 3 and —N—R4 are then deprotected so as to bind, by amidation or alkylation, the radicals R1 and R2 in a known manner, in particular by the action of the corresponding —N-hydroxy-succinimide ester.
  • The compound obtained is deprotected in the presence of trifluoroacetic acid. [0050]
  • The amines of formula VI are prepared in a known manner. [0051]
  • In the case where the compound of formula VI is 1-4-di-boc-spermidine, reference will be made to the examples indicated below in order to know the practical modalities for the synthesis. The processes described are applicable in general to the syntheses of the compounds according to the invention subject to adaptations within the capability of persons skilled in the art. [0052]
  • However, the compounds of the invention cannot be limited to those obtained by the modes of preparation described above. [0053]
  • The compounds according to the invention may, in addition, be substituted. Such substitutions may in particular consist-of a labeling molecule (see labeling molecules in U.S. Pat. No. 4,711,955) which makes it possible, for example, to visualize the distribution of the compounds or of the complexes containing them after administration in vitro or in vivo, a cell targeting molecule or an anchoring molecule. The invention consequently also relates to a compound as presented above, conjugated with one or more targeting components, also called ligands of interest, via the intermediacy of at least a) one of the carbon atoms, in particular chosen from those present on the groups R[0054] 1 and/or R2, or b) one of the secondary or primary nitrogen atoms of the polyamine chain or of the diaminocarboxylic acid. Such components may allow targeting to a specific cell type, facilitate penetration into the cell, lysis of the endosomes or alternatively intracellular transport and are widely described in the literature. They may be, for example, all or part of sugars, peptides (GRP peptide, Gastrin Releasing Peptide, for example), oligonucleotides, lipids, hormones, vitamins, antigens, antibodies, ligands specific for membrane receptors, ligands capable of reacting with an anti-ligand, fusogenic peptides, nuclear localization peptides, or a combination of such compounds. There may be mentioned more particularly the galactosyl residues which make it possible to target the asyaloglycoprotein receptor at the surface of hepatic cells, the fusogenic peptide INF-7 derived from the influenza virus hemagglutinin subunit HA-2 (Plank et al., 1994, J. Biol. Chem. 269, 12918-12924) or a nuclear localization signal derived from the SV40 virus T antigen (Lanford and Butel, 1984, Cell 37, 801-813) or the Epstein Barr virus EBNA-1 protein (Ambinder et al., 1991, J. Virol. 65, 1466-1478).
  • Such conjugates can be easily obtained by techniques widely described in the literature, and more particularly by chemical coupling, in particular using protecting groups such as trifluoroacetyl or Fmoc or Boc; onto the polyamine and more particularly using one or more orthogonal protecting groups such as those described in Protective Groups in Organic Synthesis (p. 309-406, 1991, eds. T. W. Greene, P. G. M. Wuts, Wiley) onto the polyamine or the diaminocarboxylic acid. The selective deprotection of a protecting group then makes it possible to couple the targeting component, and the lipid is then deprotected. It should be stated, however, that the substitution of the nonreactive groups such as the carbon atoms in the CH or CH2 groups will be carried out during synthesis of the compounds of the invention by methods known to a person skilled in the art, whereas the reactive groups, such as the primary or secondary amines, may be the subject of substitutions on the neosynthesized lipids of the invention. [0055]
  • According to an advantageous case of the invention, said compound is in a cationic form, that is to say that it is in a form which is protonated by binding of a proton onto one or more nitrogen atoms present on the polyamine chain. In this case, said cationic lipid is combined with one or more biologically acceptable anions, such as for example the trifluoroacetate, halide, monomethylsulfate, acetate or phosphate, iodide, chloride, or bromide anion and the like. It is also possible to obtain compounds in cationic form by substitution of the amines, for example, with a methyl or ethyl radical, and the like. [0056]
  • According to another aspect, the invention also relates to a composition comprising at least one compound as described above and optionally at least one adjuvant capable of enhancing the formation of the complex between a said compound and an active substance, or of enhancing the function of these complexes toward the cell. [0057]
  • Preferably, such an adjuvant will be a neutral or zwitterionic lipid, such as for example a lipid which is or is derived from a triglyceride, a diglyceride, cholesterol (see for example U.S. Pat. No. 5,438,044), in particular, a neutral or zwitterionic lipid which is or is derived from a phosphatidylethanolamine (PE), phosphatidylcholine, phosphocholine, sphyngomyelin, ceramide or cerebroside. Advantageously, dioleoylphosphatidylethanolamine (DOPE) will be chosen. [0058]
  • The weight ratio between the compound of the invention and the neutral or zwitterionic lipid is generally between 0.1 and 10, it being understood that this ratio may vary depending on the nature of the components considered. Persons skilled in the art have sufficient knowledge to allow these minor adaptations. It is also possible to use a mixture of neutral and/or zwitterionic lipids or alternatively a mixture of cationic lipids and neutral and/or zwitterionic lipids. [0059]
  • The invention relates, in addition, to a complex comprising at least one compound or at least one composition as described above and at least one active substance, in particular a therapeutically active substance, comprising at least one negative charge. According to a variant of the invention, said complex may, in addition, contain one or more cationic amphiphilic agents such as those described in the literature of which examples were provided above. [0060]
  • According to a specific embodiment, said active substance is chosen from nucleic acids and proteins. Preferably, the active substance of the complex according to the invention is a polynucleotide, said compound or said composition then making it possible to enhance the transfecting power of the polynucleotide in a cell. [0061]
  • “Polynucleotide” is understood to designate a DNA and/or RNA fragment which is double-stranded or single-stranded, linear or circular, natural, isolated or synthetic, designating a precise succession of nucleotides, which are modified or otherwise (see by way of example U.S. Pat. No. 5,525,711), labeled or otherwise (see for example U.S. Pat. No. 4,711,955 or EP 302175), making it possible to define a fragment or a region of a nucleic acid without size limitation. Polynucleotide is understood to designate in particular a cDNA, a genomic DNA, a plasmid DNA, a messenger RNA, an antisense RNA, a ribozyme, a transfer RNA, a ribosomal RNA or a DNA encoding such RNAs. “Polynucleotide” or “nucleic acid” are synonymous terms in the context of the present application. “Antisense” is understood to designate a nucleic acid having a sequence complementary to a target sequence, for example an mRNA sequence for which it is sought to block the expression by hybridization with the target sequence; “sense” is understood to designate a nucleic acid having a sequence homologous or identical to a target sequence, for example a sequence which binds to a proteinaceous transcription factor and which is involved in the expression of a given gene. [0062]
  • According to a specific embodiment of the invention, said polynucleotide comprises a gene of interest and components allowing the expression of said gene of interest. In this embodiment, said poly-nucleotide is advantageously in the form of a plasmid. The components allowing expression are all the components allowing the transcription of said DNA fragment into RNA (antisense RNA or mRNA) and the translation of the mRNA into a polypeptide. They are in particular promoter sequences and/or regulatory sequences which are effective in said cell, and optionally the sequences required to allow excretion or expression of said polypeptide at the surface of the target cells. By way of example, there may be mentioned promoters such as the promoters of the viruses RSV, MPSV, SV40, CMV or 7.5 k, of the vaccinia virus, the promoters of the gene encoding muscle creatine kinase, actin, or pulmonary surfactant. It is, in addition, possible to choose a promoter sequence specific for a given cell type or which can be activated under defined conditions. The literature provides a large amount of information relating to such promoter sequences. Moreover, said polynucleotide may comprise at least two sequences, which are identical or different, exhibiting a transcriptional promoter activity and/or at least two coding DNA sequences, which are identical or different, situated, relative to each other, contiguously, far apart, in the same direction or in the opposite direction, as long as the transcriptional promoter function or the transcription of said sequences is not affected. Likewise, it is possible to introduce into this type of nucleic acid construct “neutral” nucleic sequences or introns which do not affect transcription and are spliced before the translation step. Such sequences and their uses are described in the literature. Said polynucleotide may also contain sequences required for intracellular transport, for replication and/or for integration. Such sequences are well known to persons skilled in the art. Moreover, the polynucleotides according to the present invention may also be polynucleotides which are modified such that it is not possible for them to become integrated into the genome of the target cell or polynucleotides which are stabilized with the aid of agents such as, for example, spermine. [0063]
  • In the context of the present invention, the polynucleotide may be homologous or heterologous to the target cell. It may be advantageous to use a polynucleotide which encodes all or part of a polypeptide, in particular a polypeptide having a therapeutic or prophylactic activity, and more particularly an immunogenic activity of the cellular or humoral type. The term polypeptide is understood without restriction as to its size or its degree of modification (for example glycosylation). There may be mentioned, by way of examples, the genes encoding an enzyme, a hormone, a cytokine, a membrane receptor, a structural polypeptide, a polypeptide forming a membrane channel, a transport polypeptide, an adhesion molecule, a ligand, a factor for regulation of transcription, of translation, of replication, or of the stabilization of the transcripts, or an antibody, such as for example the gene encoding the CFTR protein, dystrophin, factor VIII or IX, E6/E7 of HPV, MUC1, BRAC1, β-interferon, γ-interferon, interleukin (IL)2, IL-4, IL-6, IL-7, IL-12, tumor necrosis factor (TNF) type alpha, GM-CSF (Granulocyte Macrophage Colony Stimulating Factor), the Herpes Simplex virus type 1 (HSV-1) tk gene, the gene associated with retinoblastoma or p53 or all or part of immunoglobulins, such as the fragments F(ab)[0064] 2, Fab′, Fab or the anti-idiotypes (U.S. Pat. No. 4,699,880). This list is of course not limiting and other genes may be used.
  • According to a preferred embodiment, the complexes according to the invention are small in size (less than 500 nm, advantageously less than 200 nm and preferably less than 100 nm). [0065]
  • Moreover, the transfection experiments carried out show that advantageously the weight ratio of the lipid compound according to the invention to said polynucleotide is 0.01 to 100. The optimum ratio is between 0.05 and 10. [0066]
  • The invention also relates to a process for preparing the complexes cationic compounds/anionic active substances, said process being characterized in that one or more lipids in cationic form or a composition according to the invention whose lipid is in cationic form are brought into contact with one or more active substances comprising at least one negative charge and in that said complex is recovered, optionally after a purification step. It also relates to the kits for preparing such complexes comprising one or more lipids or one or more compositions according to the invention. [0067]
  • In a first instance, according to a first variant, one or more cationic compounds are dissolved with an appropriate quantity of solvent or mixture of solvents which are miscible in water, in particular ethanol, dimethylsulfoxide (DMSO), or preferably a 1:1 (v:v) ethanol/DMSO mixture, so as to form lipid aggregates according to a known method described, for example, in Patent Application WO-A-9603977, or according to a second variant, are suspended with an appropriate quantity of a solution of detergent such as an octylglucoside such as n-octyl-β-D-glucopyranoside, or 6-O-(N-heptylcarbomoyl)-methyl-α-D-glucopyranoside. [0068]
  • The suspension may then be placed in a buffer medium and mixed with a solution of active substance comprising negative charges. [0069]
  • In the case where it is desirable that a neutral or zwitterionic lipid is present in the final complex, a film is formed, in the known manner, prior to the dissolution in the solvent which is miscible with water or in the solution of detergent, with a mixture containing a said cationic compound and a said neutral or zwitterionic lipid, such as for example DOPE. [0070]
  • One of the important characteristics of the process consists in the choice of the ratio between the positive charges of the cationic lipid and the negative charges of the active substance. [0071]
  • Without wishing to be limited by a specific ratio, quantities of the different charges will be chosen so that the ratio between the number of positive charges of the compound or of the cationic composition and the number of negative charges of the active substance is between 0.05 and 20, in particular between 0.1 and 15, and preferably between 5 and 10. [0072]
  • This ratio between the number of positive charges of the cationic compound(s) and/or composition(s) and the number of negative charges of said active substance also constitutes an advantageous characteristic of the complex according to the invention. [0073]
  • The calculation to arrive at such a ratio will take into consideration the negative charges carried by the active substance and the quantity of compound necessary to satisfy the ratio indicated above will be adjusted. The quantities and the concentrations for the other components are adjusted according to their respective molar masses and the number of their positive and/or negative charges. [0074]
  • In the case of the second variant and optionally, subsequent dialysis may be carried out in order to reduce the detergent and to recover the complexes. The principle of such a method is for example described by Hofland et al. (1996, PNAS 93, p 7305-7309) and in chapter II of the Philippot et al. document (G. Gregoriadis, 81-89, CRC Press 1993). [0075]
  • It has been shown that the first variant leads to excellent results in terms of the size of the complexes obtained. [0076]
  • According to a third variant, one or more cationic compositions or compounds are suspended in a buffer and then the suspension is subjected to sonication until visual homogeneity is obtained. The lipid suspension is then extruded through two microporous membranes under appropriate pressure. The lipid suspension is then mixed with a solution of active substance comprising negative charges. In the case where a neutral lipid is present in the complex, a film of the mixture of cationic lipid and neutral lipid such as DOPE is formed in a known manner prior to the preparation as a suspension. The same remarks relating to the ratio between the positive charges of the cationic lipid and the negative charges of the active substance as those indicated in the first variant are applicable to this third variant. This so-called sonication-extrusion technique is well known in the art. [0077]
  • The characteristics of the complexes formed may be evaluated by several means which make it possible to determine, for example: [0078]
  • the state of complex formation with the active substance, in particular by identification of the free nucleic acids by agarose gel electrophoresis in the case where the substances are nucleic acids, [0079]
  • the size of the particles by a quasi-elastic scattering of light, [0080]
  • the absence of precipitation over the long term. [0081]
  • The object of the present invention is also the complexes obtained using the processes listed above. [0082]
  • The invention also relates to the use of a compound, of a composition or of a complex according to the invention to transfer at least one active substance, especially a therapeutically active substance, more particularly a nucleic acid, into target cells, in vitro, ex vivo or in vivo, more particularly in vivo. [0083]
  • “Target cells” according to the invention is understood to mean prokaryotic cells, yeast cells and eukaryotic cells, plant cells, human or animal cells, and in particular mammalian cells. Cancer cells should, moreover, be mentioned. In vivo, the invention may be applied at the level of the interstitial or luminal space of tissues such as the lungs, trachea, skin, muscle, brain, liver, heart, spleen, bone marrow, thymus, bladder, lymph, blood, pancreas, stomach, kidney, ovaries, testicles, rectum, peripheral or central nervous system, eyes, lymphoid organs, cartilages and endothelium. According to an advantageous choice of the invention, the target cell will be a muscle cell, a hematopoietic stem cell or alternatively a cell of the airways, more particularly a tracheal or pulmonary cell, and advantageously a cell of the respiratory epithelium. [0084]
  • The complexes according to the invention can be used as a medicament for curative, preventive or vaccinal purposes. Accordingly, the subject of the invention is also the complexes of the invention as a medicament for curative, preventive or vaccinal purposes. Such complexes may be used in a method of therapeutic treatment which consists in transferring at least one therapeutically active substance, in particular a polynucleotide, into target cells, in particular a mammalian cell, and more precisely a muscle cell, a hematopoietic stem cell, a cell of the airways, more particularly a tracheal or pulmonary cell, a cell of the respiratory epithelium. [0085]
  • A compound according to the invention is most particularly advantageous for transferring a nucleic acid into a muscle cell or a pulmonary cell. It is the compound noted pcTG37 (see example). [0086]
  • More widely, the present invention also relates to a process for introducing an active substance comprising negative charges into a cell in particular in vitro, characterized in that cells, in particular cultured on an appropriate medium are brought into contact with a complex cationic compound/active substance comprising at least one negative charge according to the invention, in particular in the form of a suspension of complexes. After a certain incubation time, the cells are washed and recovered. The introduction of the active substance may be checked (optionally after lysis of the cell) by any appropriate means. [0087]
  • The process of introduction is well known per se. The term “introduction” is understood to mean that the active substance comprising negative charges is transferred into the cell and is located, at the end of the process, inside said cell or at the level of the membrane thereof. In the case where the active substance is a nucleic acid, reference will be made more particularly to “transfection”. in this case, the verification of the transfection of the nucleic acid can be carried out by any appropriate means, for example by measuring the expression of the gene considered or the concentration of the expressed protein. [0088]
  • The invention relates more particularly to the use of a compound, of a composition or of a complex according to the invention for the preparation of a medicament for curative, preventive or vaccinal purposes, intended for the treatment of the human or animal body, in particular by gene therapy. [0089]
  • According to a first possibility, the medicament may be administered directly in vivo (for example into a muscle, into the lungs by aerosol and the like) . It is also possible to adopt the ex vivo approach which consists in collecting cells from the patient (bone marrow stem cells, peripheral blood lymphocytes, muscle cells and the like), transfecting them in vitro according to the present invention and readministering them to the patient. [0090]
  • The complexes according to the invention may be administered by the intramuscular, intratracheal, intranasal, intracerebral, intrapleural, intratumoral, intracardiac, intragastric, intraperitoneal, epidermal, intravenous or intraarterial route by a syringe or by any other equivalent means, systems suitable for the treatment of the airways or of the mucous membranes such as inhalation, instillation or aerosolization. There may also be mentioned the modes of administration by application of a cream, by oral administration or any other means perfectly known to the person skilled in the art and applicable to the present invention. [0091]
  • It is also within the scope of the invention to target specific organs or tissues by administration, in particular by the intravenous route, of a complex according to the invention prepared so as to adjust the ratio compound or composition/therapeutically active substance in said complex, the apparent charge of the complex (see in particular Liu et al., 1997, Gene Therapy, 4, 517-523; Thierry et al., 1995, P.N.A.S., 92, 9742-9746). [0092]
  • The invention also relates to a method of gene therapy consisting in administering to a patient an appropriate quantity of a composition according to the invention. According to the present invention and in the context of gene therapy in vivo, it is possible to repeat several times, in a given patient, the method as proposed without any major immune reaction being elicited against one of the compounds administered. The administration may take place in a single dose or repeated once or several times after a certain time interval. The repeated administration would make it possible to reduce the quantity of therapeutically active substance, more particularly of DNA, to be administered for a given dose. The appropriate route of administration and dosage vary according to various parameters, for example the individual or disease to be treated or alternatively the polynucleotide to be transferred. [0093]
  • The invention relates more particularly to a pharmaceutical preparation comprising at least one complex as described above, optionally containing, in addition, at least one adjuvant capable of stabilizing said pharmaceutical preparation for the purpose of its storage for example and/or of enhancing the transfecting power of said complex. Such an adjuvant could, for example, be chosen from the group consisting of chloroquine, a protic polar compound chosen in particular from propylene glycol, polyethylene glycol, glycerol, ethanol, 1-methyl-L-2-pyrrolidone or derivatives thereof, or an aprotic polar compound chosen in particular from dimethyl sulfoxide (DMSO), diethyl sulfoxide, di-n-propyl sulfoxide, dimethyl sulfone, sulfolane, dimethylformamide, dimethylacetamide, tetramethylurea, acetonitrile or derivatives thereof. Likewise, said preparation may contain a pharmaceutically acceptable carrier allowing its administration to humans or animals. [0094]
  • In the context of the use of a method of treatment in vivo according to the present invention, it is, in addition, possible to carry out, before the administration of a pharmaceutical preparation as described above, a treatment of the patient designed to observe a temporary depletion of the macrophages making it possible to enhance the transfection rate. Such a technique is described in the literature; see in particular Van Rooijen et al., 1997, TibTech, 15, 178-184. [0095]
  • Finally, the invention relates to a cell transfected with a complex as defined above, particularly a prokaryotic cell, a yeast cell or eukaryotic cell, especially an animal cell, in particular a mammalian cell, and more particularly a cancer cell. According to a preferred case of the invention, said cell is a cell of the airways, more particularly a tracheal or pulmonary cell, and advantageously a cell of the respiratory epithelium.[0096]
  • The examples below illustrate the invention without limiting it in any manner. An explanatory diagram of synthesis which forms an integral part of the description is appended (FIG. 1). FIG. 2 indicates the results observed after intravenous injection of complexes according to the invention, the luciferase activity is indicated in RLU/mg of protein).[0097]
  • EXAMPLES
  • The preparation of the cationic lipid of formula I in which R[0098] 1 and R2, which are identical, are oleoyl or stearoyl radicals, n=2; mα=3, mβ=4 is described below with reference to the diagram of synthesis represented in FIG. 1. α and β are the positions of the —(CH2)m—NH— motifs from the carbonyl.
  • Synthesis of the Cationic Lipid of Formula I in which n=2, mα=3, mβ=4, R=oleoyl (pcTG37) (see FIG. 1) Synthesis of the Intermediate 1-4-di-boc-spermidine
  • The protocol applied in order to obtain 1,4-di-boc-spermidine is published by Goodnow et al. (1990, Tetrahedron 46, 3267-3286). In practice, the intermediate N-propionitrile-1,4-diaminobutane is obtained by reacting 37.6 mmol of diaminobutane (Fluka; reference 32790) diluted in 3.8 ml of dichloromethane +2 ml of methanol at 0° C. with 18.8 mmol of acrylonitrile (Fluka; reference 01710) for 1 hour at 20° C. The amino groups of the product of the reaction are protected with 99 mmol of BOC-ON [(2-boc-oxyimino)-2-phenylacetonitrile] (Fluka; reference 15475) in 50 ml of 2/1 CH[0099] 2Cl2/CH3OH solvent. The protecting reaction is left overnight at room temperature. The solvents are evaporated off and the product is taken up in 75 ml of ethyl acetate for 3 extractions with 75 ml of 1 M NaOH. The organic phase is again extracted with 5% citric acid and then dried and filtered over Na2SO4 before evaporation to dryness. The step of reducing the nitriles is then carried out in ether at 0° C. in the presence of 20 mmol of LiAlH4 (Sigma; L0260). After 1 hour 30 min, the reaction is stopped by addition of 20 ml of 1 M NaOH at 0° C., with evolution of hydrogen. The mixture is filtered and extracted 3 times with a 20% NaCl solution in water. After drying the ethereal fraction, the product is deposited on a silica column in a 2/1 CH2Cl2/CH3OH solvent and then eluted in a 15/5/1 CH2Cl2/CH3OH/(C2H5)3N solvent. The fractions containing the di-boc-spermidine are determined by thin-layer chromatography.
  • NMR data: 1H NMR (200 MHz, CDC13): 1.43-1.48 ppm (s, 18 H, Boc), 1.48-1.72 ppm (m, 6H, NH(Boc)—CH[0100] 2—CH2—CH2—CH2—N(Boc)—CH2CH2 —CH2—NH2), 2.69 ppm (t, 2H, J=6.7 Hz, —CH2—NH2), 3.07-3.25 ppm (m, 6H, NH(Boc)—CH2 —CH2—CH2CH2 —N(Boc)—CH2 —CH2—CH2—NH2), 4.59 ppm (b, 1H, NH(Boc)). Quantity synthesized (1.45 g; 5.1 mmol)—27% yield relative to the quantity of acrylonitrile.
  • Synthesis of pcTG37
  • 140 μmol of N-hydroxybenzotriazole (Sigma; H2006), then 140 μmol of di-boc-spermidine in 1 ml of anhydrous tetrahydrofuran, and finally 180 μmol of dicyclohexylcarbodiimide (Sigma; D3128) diluted in 2 ml CH[0101] 2Cl2 are added to 140 μmol of N-α N-β, di-Fmoc-diaminopropionic (Bachem B2265). After 2 hours of reaction, the dicyclohexylurea formed is filtered and the product is purified on a silica column in a 99/1 CH2Cl2/CH3OH solvent. After evaporation, the Fmoc groups are removed from the product in a solution of 20% piperidine, 20% tetrahydrofuran and 60% CH2Cl2 for 1 hour. After evaporation, another purification is carried out on a silica column in a 1/1 CH2Cl2/CH3OH solvent. The fractions containing the product are determined by thin-layer chromatography. The product is dried before the last coupling. 510 μmol of the N-hydroxysuccinimide ester of oleic acid (Sigma 0 9506) diluted in 5 ml of anhydrous CH2Cl2 are added to di-boc-spermidine-diaminopropionamide. After evaporation, the product is purified on a silica column (ether/ethyl acetate; 1/1). The final product, taken up in 1 ml of CH2Cl2, is Boc deprotected in 10 ml of trifluoroacetic acid (Fluka 91699). To remove the trifluoroacetic acid, evaporation is carried out under reduced pressure after dilution in 50 ml of n-hexane.
  • Overall yield of synthesis: 64% (89 mg; 90 μmol). [0102] 1H NMR (200 MHz, CDCl3/CD3OD: 0.81 ppm (t, j=6.4 Hz, 6H —CH3), 1.22-1.3 ppm (m, 44 H, —CH2—), 1.51 ppm (m, 4 H, —CH2 —CH2—CO—NH—), 1.72-1.90 ppm (m, 6 H, NH3 +—CH2CH2 —CH2—CH2—NH2 +—CH2CH2 —CH2—NH—), 1.92 ppm (m, 8 H, CH2—CH═), 2.07-2.2 ppm (m, 4 H, CH2—CO—NH—), 2.89 ppm (m: 6 H, NH3 +CH2 —CH2—CH2CH2 —NH2 +CH2 —CH2—CH2—NH—), 4.22 ppm (t, 1 H, NH—CO—CHR—NH—), 5.27 ppm (m, 4H, —CH═). Mass spectrometry: measured 759.7 Da (calculated 760.2 Da).
  • Synthesis of the Cationic Lipid of Formula I in which n=2, mα3, mp=4; R=stearoyl (lipid pcTG39)
  • The compound pcTG39 is prepared in an amount of 140 μmol in the same manner using stearic acid and dicyclohexylcarbodiimide or (benzotriazol-1-yloxy) tri-pyrrolidinophosphonium hexafluorophosphate (PyBOP) as coupling agent. [0103]
  • Overall yield of synthesis: 22% (30 mg; 30 μmol). [0104] 1H NMR (200 MHz, CDCl3/CD3OD/D2O): 0.68 ppm (t, j=6.8 Hz, 6H, —CH3), 1.06 ppm (m, 48 H, —CH2—), 1.37 ppm (m, 4 H, —CH2 —CH2—CO—NH—), 1.53-1.75 ppm (m, 6 H, NH3 +—CH2CH2 CH2 —CH2—NH2 +—CH2CH2 —CH2—NH—), 1.90-2.30 ppm (m, 4 H, CH2—CO—NH═), 2.74 ppm (m, 6H, NH3 +CH2 —CH2—CH2CH2 —NH2 +CH2 —CH2—CH2—NH—), 3.25-3.55 ppm (m, 4 H, —CH2—NH—CO—). Mass spectrometry: measured 764.5 Da (calculated 764.3 Da).
  • Preparation of the Lipid-DNA Complexes by Suspension in Ethanol
  • 1. Preparation of the Complexes Lipid pcTG37, Optionally DOPE,-DNA [0105]
  • The quantities of lipids are calculated based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the chosen cationic lipid. To obtain a complex between [0106] pcTG37/DOPE and the plasmid DNA in a ratio of 10 between positive charges provided by the cationic lipid and negative charges provided by the DNA at a final DNA concentration of 0.1 mg/ml, the different ingredients are mixed according to the following calculation:
  • 0.1 mg of DNA/ml, that is to say (0.1/330) mmol of negative charges (330 Da is the average molecular weight of a nucleotide) per ml correspond to 0.30 μmol/ml of negative charges. To obtain 10 times more positive charges, a concentration of 3.0 μmol/ml of positive charges provided by the cationic lipid is required. The molar mass of pcTG37 in trifluoroacetate form is 988 g/mol and the molecule contains 2 positive charges. Therefore, 1.5 μmol/ml of pcTG37 is required, which corresponds to 1.49 mg/ml. [0107]
  • To obtain an equimolar concentration of L-α-dioleoyl-phosphatidylethanolamine (DOPE, 744 g/mol, Sigma; P0510), 1.12 mg/ml is required in the lipid preparation. The quantities and the concentrations for the other compounds are adjusted according to their respective molar masses and the number of their positive charges. [0108]
  • The lipids are taken up in chloroform, dried by evaporation and then solubilized in chloroform/methanol (v:v) and again dried. The cationic lipids are weighed and the quantity of DOPE is added from a stock solution of 10 or 20 mg/ml in chloroform to a glass tube sterilized with alcohol and with UV in order to obtain a cationic lipid concentration of 2 mM. The solvents are evaporated under vacuum (0.2×105 Pa (200 mbar)) for 45 min at 45° C. using a vortex of 40 revolutions per minute (Labconco, Rapidvap, Uniequip, Martinsried, Germany). The lipid film is taken up in ethanol so as to be at the cationic lipid concentration of 50 mg/ml. [0109]
  • pcTG37/DOPE 1.49 mg+1.12 mg=2.61 mg in 30 μl of ethanol. This solution is adjusted to 270 μl with 20 mM HEPES pH 7.5 (adjusted with NaOH) in order to prepare a solution at 5 mg/ml final. [0110]
  • The plasmid DNA is prepared from a stock solution at 1 mg/ml (10 mM Tris, 1 mM EDTA, pH 7.5). [0111]
  • For a solution of 0.5 ml final, 50 μl of the stock solution (50 μg DNA) are collected to which 300 μl of 20 mM HEPES pH 7.5 are added. [0112]
  • To complex the DNA with the lipid preparations, the lipids are added to the DNA. The suspension is mixed by aspiration/discharge using a pipette (10 times). The complexes are stored at +4° C. [0113]
  • 150 μl of pcTG37/DOPE are added to 350 μl of the DNA solution in order to obtain 0.5 ml of complex at 0.1 mg/ml DNA and a charge ratio of 10. [0114]
  • The preparation of the complexes is carried out under a laminar flow cabinet. [0115]
  • The complexes are obtained whose characteristics are indicated in Table 1 below. [0116]
  • 2. Preparation of the Complexes Lipids pcTG39, Optionally DOPE,-DNA [0117]
  • The complexes are obtained according to the same protocol as above. [0118]
  • Preparation of the Lipid-DNA Complexes by Suspension in a Detergent Solution
  • 1. Preparation of the Complexes Lipids pcTG37, Optionally DOPE, DNA [0119]
  • The quantities of lipids are calculated as described above based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the cationic lipid chosen. The lipids are mixed in a glass tube, sterilized with alcohol and with UV, in order to obtain a 2 mM cationic lipid solution (see above). The solvents are evaporated and the lipid film is taken up in a solution of n-octyl, β-D-glucopyranoside (octylglucoside, Sigma, O 9882) according to a cationic lipid/detergent ratio of 1:5 (mol:mol). [0120]
  • 375 μl of a 20 mM octylglucoside solution in 20 mM HEPES pH 7.5 are collected and used to take up the film of pcTG37/DOPE lipid mixture. The plasmid DNA is prepared from a stock solution of plasmid DNA at 1 mg/ml of which 50 μl are removed for a final volume of 0.5 ml (0.1 mg/ml final) to which 262.5 μl of 20 mM HEPES pH 7.5 are added. 187.5 μl of the lipid suspension are added to the DNA by aspirating and discharging 10 times using a pipette in order to obtain the final suspension at 0.1 mg/ml of DNA and a +/− charge ratio of 10. To remove the detergent, a dialysis of 3 times 4 hours at room temperature against 20 mM HEPES pH 7.5 is carried out in dialysis microbags (cut-off of 13.2 kD; Sartorius, Göttingen, Germany). The dialyzed DNA/lipid complexes are stored at +4° C. The preparation is carried out in a laminar flow cabinet. [0121]
  • The complexes are obtained whose characteristics are indicated in Table 1 below. [0122]
  • 2. Preparation of the Complexes Lipids pcTG39, Optionally DOPE,-DNA [0123]
  • Based on the same protocol, the complexes whose characteristics are indicated in Table 1 below are obtained. [0124]
  • Preparation of the Lipid-DNA Complexes by Sonication Extrusion
  • The quantities of lipids are calculated as described above based on the concentration of final DNA (0.1 mg/ml for the tests in vitro), the desired charge ratio, the molar mass and the number of positive charges of the cationic lipid chosen. The lipids are mixed in a glass tube, sterilized with alcohol and with UV, in order to obtain a 2 mM cationic lipid solution, as indicated above. The solvents are evaporated and the lipid film is taken up in 900 μl of 20 mM HEPES pH 7.5 at 4° C. for about 16 h. The suspension is sonicated in a sonication bath (Bransonic 221) to visual homogeneity. The lipid suspension is extruded through two membranes with a pore diameter of 0.2 μm (Nucleopore, Costar, Cambridge, Mass., USA) and rinsed with 20 mM HEPES pH 7.5 (extruder from Lipex Biomembranes, Vancouver, Canada) at a maximum pressure of 50 bars. The lipid suspension is kept at room temperature for 1 hour. 450 μl of the lipid suspension are added to 50 μl of a stock solution of plasmid DNA (1 mg/ml) and mixed by aspirating/discharging 10 times using a pipette. The lipid/DNA complexes are stored at +4° C. The preparations are carried out under a laminar flow cabinet. [0125]
  • Protocol for Evaluation of the Complexing of the DNA by the Lipids
  • A 1% (w:v) agarose gel is prepared in a TAE buffer (TAE: Tris 4.86 g/l+sodium acetate 0.68 g/l+EDTA 0.336 g/l pH 7.8. If necessary, the sample is diluted in TAE and then the sample buffer 0.083% bromophenol blue, 0.083% cyanol xylene FF, 10% glycerol in water) is added so as to have 50 ng DNA/μl. The sample is briefly subjected to a vortex and left for 30 min at room temperature. As a control, the non-complexed plasmid prepared at the same concentration is used. 10 μl (500 ng of DNA) are deposited on the gel and the migration is carried out at 60 mV for 3 hours. The gel is developed in TAE containing 0.006% (v:v) of ethidium bromide at 10 mg/ml for at least 30 min. Next, the gel is rinsed in TAE and analyzed under UV. [0126]
  • Protocol for Measuring the Size of the Particles by Quasi-elastic Scattering of Light
  • The analyses are carried out on a Coulter N4Plus (Coultronics France S.A., Margency, France) at 25° C. after equilibration of the sample for 20 min. An aliquot of the sample is aspirated and discharged several times before being pipetted. The sample is diluted in the measuring tank and homogenized. The measurement of the light diffracted at 90° is carried out for 180 sec after a 180 sec wait. The range used goes from 3 nm to 10 000 nm using 31 bins. To be valid, the sample should give between 50 000 and 1 000 000 counts/sec. [0127]
  • Physicochemical Characteristics
  • The three methods of formulation “injection of ethanol”, “dialysis of detergent” and “sonication/extrusion” are applied to the cationic lipids according to the invention with or without equimolar quantities of DOPE at charge ratios of about 10, or 5. The formulations are considered to be appropriate when the DNA is completely complexed (no migration in the agarose gel) and when the complexes have diameters, determined by quasi-elastic scattering of light, less than 500 nm. The table 1 summarizes the results of these analyses. All the DNA/lipid complexes indicated in the table complex the DNA completely. [0128]
    TABLE 1
    Lipid Ratio1 Size (nm)2 Formulation
    pcTG37 10  77 ethanol
    pcTG37
     5  80 ethanol
    pcTG37/DOPE 10  79 ethanol
    pcTG37/DOPE  5  64 ethanol
    pcTG37 10 240 detergent
    pcTG37
     5  59 detergent
    pcTG37/DOPE 10 312 detergent
    pcTG39/DOPE 10 312 detergent
    pcTG37/DOPE  5 280 sonication
  • These analyses show that the formulations meet the necessary requirements. The “injection of ethanol” method gives the best results, the different preparations tested being less than 100 nm in size. The method by dialysis of detergent or sonication/extrusion also makes it possible to obtain complexes meeting the criteria of the present invention. [0129]
  • In vitro Transfection of Satellite Cells
  • Cultures of dog muscle and human muscle [lacuna] are carried out in an HamF 14 medium (Life Technologies) supplemented with 10% fetal calf serum (FCS, Hyclone, Logan, Utah), 10 μg/ml of insulin (Sigma), 10 ng/ml of EGF (Sigma) and of FGF (Pepro Tech Inc., Rocky Hill, N.J.) 2 mM of glutamine (bioMérieux), and 40 μg/ml of gentamycin (Schering Plough). [0130]
  • The cells are inoculated 24 h to 48 h before the transfection into a 96-well culture plate with about 5×10[0131] 3 to 104 cells per well, at about 30% confluence, and kept at 37° C. under a 5% CO2 and 95% air atmosphere.
  • The transfections are carried out with mixtures of variable quantities of lipids and plasmid DNA in order to determine the charge ratios and the optimum DNA concentrations per well. [0132]
  • The complexes used are prepared 24 h to 48 h before the transfection and diluted in HamF 14 medium containing 40 μg/ml of gentamycin and 2 mM glutamine. [0133]
  • After removing the culture medium, 100 μl of transfection mixtures with or without 10% FCS are transferred into each of the wells and the plates are incubated for 4 h at 37° C. [0134]
  • All the transfection media are then adjusted to 10% FCS, 10 μg/ml of insulin (Sigma), 10 ng/ml of EGF (Sigma) and of FGF (Pepro Tech Inc., Rocky Hill, N.J.), 2 mM glutamine (bioMérieux), and 40 μg/ml of gentamycin (Schering Plough) for a final volume of 250 μl. The cultures are incubated for 48 h and then the cells are recovered and tested for their capacity to express the luciferase gene. The protein concentrations are determined by the system for testing quantity of protein (Promega). [0135]
  • Transfection of A549 Cells with Lipid Complexes
  • The A549 cells (epithelial cells derived from human pulmonary carcinoma) are cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal calf serum (Gibco BRL) 24 hours before the start of the transfection in 96-well plates (2×10[0136] 4 cells per well) in a humid atmosphere at 37° C. and 5% CO2/95% air. For the transfection in the absence of serum, the medium is removed and replaced with serum-free medium. In another microplate, the following suspensions of lipid/DNA complexes are prepared (lipid/DNA complexes at 0.1 mg/ml of DNA and at the indicated charge ratio):44 μl (4.4 μg DNA), 22 μl (2.2 μg DNA), 5.5 μl (0.55 μg DNA) of stock solution in the first 3 wells, and 11 μl (0.11 μg DNA) of the stock solution diluted 10-fold in the next well. The volume is adjusted to 110 μl with DMEM and 100 μl are transferred over the A549 cells. The incubation is carried out with 4, 2, 0.5 and 0.1 μg of DNA per well for 4 hours. 50 μl of DMEM+30% fetal calf serum are added 4 hours after the start of transfection and then 100 μl of DMEM+10% FCS 24 hours after the start of transfection. The transfections in the presence of 10% fetal calf serum are carried out in an identical manner except that the transfection occurs in medium with serum.
  • Analysis of the Transfection
  • 48 h after the transfection, the medium is removed and the cells are washed with 100 μl of PBS phosphate solution and lyzed with 50 μl of lysis buffer (Promega). The lysates are frozen at −80° C. until the luciferase activity is measured. The latter is carried out on 20 μl of mixture for one minute using the Luciferase” determination system (Promega) (LB96P Berthold luminometer) in 96-well plates in kinetic mode. [0137]
  • Transfection in vitro
  • The preparations of cationic lipids pcTG37 were evaluated in transfection in vitro using the A549 cells and the dog primary myoblasts. [0138]
  • The results are summarized in Table 2 below and show the relative light units (RLU) per well. The values given are obtained with 0.5 μg of DNA per well. All the complexes are prepared using the injection of ethanol method. The total protein concentration per well is determined by the conventional techniques (BCA test, Pierce) . As a guide, a well contains about 20 to 30 μg of protein. [0139]
    TABLE 2
    Lipid Ratio1 A5492 A549 + serum myoblasts myoblast + serum
    pcTG37 10 1.7 × 106 3.2 × 104 0 1.2 × 105
    (4.0 × 108
    RLU/mg/min)
    pcTG37 5 9.9 × 106 3.9 × 103 8.5 × 107 8.6 × 103
    (1.4 × 109
    RLU/mg/min)
    pcTG37/DOPE 10 3.5 × 106 1.2 × 106 1.5 × 104 4.9 × 105
    pcTG37/DOPE 5 5.0 × 104 6.7 × 106 1.9 × 106 4.9 × 104
  • Another Mode of Synthesis of the Compounds According to the Invention has been Developed
  • 1. Synthesis of the Polyamine Part [0140]
  • 1.1. In order to be able to synthesize the “T-branched” compounds for which the group R of formula II is bound to the secondary amines, an N,N′-di-boc-spermine was synthesized. The protocol applied in order to obtain the 1,4-diboc-spermidine is published by Goodnow et al. (1990, Tetrahedron 46, 3267-3286). [0141]
  • The intermediate N-N′-(di-boc)-N,N′-(dipropio-nitrile)-1,4-diaminobutane is obtained by reacting 37.6 mmol of diaminobutane (Fluka; reference 32790) diluted in 3.8 ml of dichloromethane+2 ml of methanol at 0° C. with 75.2.mmol of acrylonitrile (Fluka; reference 01710), for 1 hour at 0° C. The product of the reaction is blocked with 100 mmol of BOC-ON [(2-boc-oxyimino)-2-phenylacetonitrile] (Fluka; reference 15475) in 50 ml of 2/1 CH[0142] 2Cl2/CH3OH solvent. The blocking reaction is placed overnight at room temperature. The solvents are evaporated and the product is taken up in 200 ml of ethyl acetate and then subjected to 3 extractions with 200 ml of 1 M NaOH. The organic phase is again extracted with 5% citric acid, dried and filtered over Na2SO4 before evaporation to dryness. A step of reducing the nitrites is then carried out in ether at 0° C. in the presence of 260 mmol of LiAlH4 (Sigma; L0260). After 15 hours, the reaction is stopped by addition of 100 ml of 1 M sodium hydroxide at 0° C., with evolution of hydrogen. The mixture is filtered and extracted three times with a 20% NaCl solution. After drying the ethereal fraction, the product is deposited on a silica column in a 1/1 CH2Cl2/CH3OH solvent and then eluted in a 15/5/1 CH2Cl2/CH3OH/(C2H5)3N solvent. The fractions containing the di-boc-spermine are determined by thin-layer chromatography. Weighing of the final product: 2.45 g (5.8 mmol). Overall yield: 15.5%.
  • 1.2. Synthesis of N,N′-di-boc-spermidine from spermidine. 20 mmol of spermidine (Sigma S2626) diluted in 20 ml of dichloromethane added to 20 ml of methanol, 11 mmol (2 ml) of diisopropylethylamine at 0° C. and 40 mmol of BOC-O-BOC [di-(tert-butyl)-dicarbonate] (Fluka; reference 34660) in 80 ml of 1/1 CH[0143] 2Cl2/CH3OH solvent. The blocking reaction is placed overnight at room temperature. The solvents are evaporated and then the product is deposited on a silica column and eluted in steps of 9/1 to 8/2 CH2Cl2/CH3OH solvents. The fractions containing the N,N′-di-boc-spermidine are determined by RP-HPLC chromatography. A pure portion of N,N′-di-boc-spermidine is thus isolated from the other position isomers of di-boc-spermidine. Weighing of the final product: 0.58 g (1.68 mmol) Overall yield: 8.5%.
  • 1.3. New process for the synthesis of 1-4-di-boc-spermidine: 50 mmol of diaminobutane (Fluka; reference 32790) diluted in 50 ml of dichloromethane are added to 20 ml of methanol at 0° C. with 58 mmol of acrylonitrile (Fluka; reference 01710) and placed for 1 hour at 0° C. The product of the reaction is blocked with 109 mmol of BOC-ON [(2-boc-oxyimino)-2-phenylacetonitrile] (Fluka; reference 15475) in 50 ml of 2/1 CH[0144] 2Cl2/CH3OH solvent. The blocking reaction is placed overnight at room temperature. The solvents are evaporated and the product is taken up in 300 ml of ethyl acetate for 3 extractions with 300 ml of 1 M NaOH. The organic phase is again extracted with 5% citric acid, dried and filtered over Na2SO4 before evaporation to dryness. The step of reduction of the nitrites is then carried out in ether at 0° C. in the presence of 140 mmol of LiAlH4 (Sigma; L0260). After 15 hours, the reaction is stopped by addition of 100 ml of 1 M sodium hydroxide at 0° C., with evolution of hydrogen. The mixture is filtered and extracted three times with a 20% NaCl solution. After drying the ethereal fraction, the product is ready to be deposited on a silica column for the final purification.
  • 2. Synthesis of the Hydrophobic Part [0145]
  • Synthesis of N-α-N-β-di[oleylamido]propionic acid. 9.04 mmol (3.43 g) of the N-hydroxysuccinimide ester of oleic acid (Sigma O 9506) diluted in 10 ml of dioxane are added to 4.1 mmol (0.58 g) of N-α-N-β-diaminopropionic acid,HCl (Bachem F3040) in a mixture of 22 ml of water, 44 ml of dioxane and 20.5 mmol (2.65 ml) of diisopropylethylamine. After 2 hours of reaction, 2 extractions with ether/[0146] HCl 5% are carried out, followed by saturated NaCl washing and drying over Na2SO4 before evaporating the ethereal fraction to dryness. The product is deposited on a silica column in a 95/5 CH2Cl2/CH3OH solvent, and then eluted in steps of 95/5, then 92.5/7.5, and finally 90/10 CH2Cl2/CH3OH solvents. The fractions containing n-α-N-β-di[oleyl-amido]propionic acid are determined by thin-layer chromatography. Control on TLC (CH2Cl2/CH3OH 90/10) and NMR. Drying. Weighing: 0.916 g (1.45 mmol), yield: 35.4%.
  • Synthesis of N-α-N-β-di-[laurylamido]propionic acid. 2.2 mmol (0.65 g) of the N-hydroxysuccinimide ester of lauric acid (Sigma L3900) diluted in 10 ml of dioxane are added to 1 mmol (0.14 g of N-α-N-β-diaminopropionic acid,HCl (Bachem F3040) in a mixture of 10 ml of water, 20 ml of dioxane and 5 mmol (0.65 ml) of diisopropylethylamine. The same purification protocol (see N-α-N-β-di[oleylamido]-propionic acid) is applied. Weighing: 0.4 g (0.5 mmol), yield: 50%. [0147]
  • Synthesis of N-α-N-β-di[palmitylamido]propionic acid. 2.2 mmol (0.79 g) of the N-hydroxysuccinimide ester of palmitic acid (Sigma P1162) diluted in 10 ml of dioxane are added to 1 mmol (0.14 g) of N-α-N-β-diaminopropionic acid,HCl (Bachem F3040) in a mixture of 10 ml of water, 20 ml of dioxane and 5 mmol (0.65 ml) of diisopropylethylamine. The activated ester precipitates upon the addition and the reaction takes place in a heterogeneous phase for 15 hours, with stirring, at room temperature. And the same purification protocol (see N-α-N-β-di[oleylamido]-propionic acid) is applied. Weighing: 0.14 g (0.15 mmol), yield: 15%. [0148]
  • 3. New synthesis of pcTG37 [0149]
  • 1.45 mmol 916 mg of N-α-N-β-di[oleylamido]-propionic acid, 1.74 mmol (235 mg) of N-hydroxybenzo-triazole (Sigma; H2006), 2 mmol (265 μl) of diisopropylethylamine, and finally 2.175 mmol (449 mg) of dicyclohexylcarbodiimide (Sigma; D3128) diluted in 10 ml of CHCl[0150] 3 are added to 1.45 mmol (500 mg) of 1,4-di-boc-spermidine diluted in 30 ml of anhydrous CHCl3. After 2 hours of reaction, the medium is filtered and after evaporation, rediluted in 50/50 ether/hexane, and another filtration-evaporation is carried out. This product is purified on a silica column deposited in ether and eluted in steps of ether/ethyl acetate solvent: 75/25, 50/50, 25/75 and 100% ethyl acetate. Control on TLC (ethyl acetate) and NMR. The final product, taken up in 10 ml of CH2Cl2, is Boc deprotected by adding dropwise at 0° C. 50 ml of trifluoroacetic acid (Fluka 91699) redistilled at 72° C. and diluted in 50 ml of CH2Cl2. After 4 hours, the medium is evaporated twice at reduced pressure after dilution in 50 ml of n-hexane. The product is taken up in 10 ml of ether, precipitated in hexane and filtered on filter paper. The product is taken up by washing the paper in a 1/1 CH2Cl2/CH3OH solvent. Controls on TLC (ethyl acetate), HPLC and NMR. Evaporation and weighing: 670 mg (0.68 mmol) yield: 47%.
  • 4. Synthesis of pcTG89 (equivalent of pcTG37 but in a T-branched form) [0151]
  • The same protocol is applied as for pcTG37 but by replacing 1,4-di-boc-spermidine with N,N′-di-boc-spermidine. Weighing of the protected product: 170 mg (0.18 mmol), yield: 12.2%. [0152]
  • Intravenous Injection of Complexes According to the Invention
  • The results are summarized in FIG. 2. Complexes according to the invention were synthesized according to the methods described above from the compound pcTG337, in the presence or in the absence of DOPE (2:1, mol:mol), at a fixed charge ratio of 5, using a plasmid containing the gene for luciferase pTG11033 (French Patent Application No. 97/08267). [0153]
  • The mice used are 9- to 11-week-old C57BL/6 female mice. One day before the injection with the complex according to the invention, the mice are pretreated by injecting 50 μl of a preparation of chlodronate encapsulated in a liposome (see Van Rooijen and Sanders, 1994, J. Immunol. Methods, 174, 83-93) incorporated in a total volume of 200 μl (+150 μl of PBS buffer). The intravenous injections are carried out into the tail after disinfecting the skin with 70% ethanol. The volume injected is 250 μl and the quantity of DNA is 60 μg, the quantity of lipid is 345 μg. [0154]
  • Two days after the injections, the mice are sacrificed. After extraction, the tissues are frozen in liquid nitrogen and stored at −80° C. To measure the luciferase activity, the tissues are mechanically ground with the aid of a pestle in a mortar placed on dry ice. 500 μl of a lysis buffer (Promega) are added to the tissue debris obtained from the lungs and subjected to three freeze/thaw steps. The cellular debris is removed by centrifugation and the luciferase activity (in RLU/min, relative light unit per minute) is measured on 20 μl of supernatant in accordance with the manufacturer's instructions (Promega) by adding 100 μl of reagent and by measuring the activity by luminescence. The luciferase activity measured is standardized relative to the protein quantity with the aid of a calibration series prepared by commercially available luciferase (Promega). The total protein quantity is, moreover, determined by the bicinchoninic acid BCA colorimetric method (Smith et al., 1985, Anal. Biochem., 150, 76-85 Pierce) using an aliquot of supernatant. This makes it possible to express the luciferase activity in RLU per milligram of protein extracted from the tissues. Controls are carried out for which the mice were not injected or were injected with DNA alone (60 μg) in a 20 mM HEPES buffer pH 7.5. The results (FIG. 2) show that the expression of the luciferase reporter gene in the lungs after intravenous injection of complexes containing the compound according to the invention at a charge ratio of 5, in the presence of DOPE is markedly enhanced relative to the injection of DNA alone. The values indicated are the mean values obtained from 3 mice injected. [0155]

Claims (44)

1. Lipid compound of formula:
R—HN—[—(CH2)0—NR—]n−1—(CH2)m—NH—R  I
in which:
the R residues are, independently of each other, a hydrogen atom or a group of formula II:
Figure US20020151070A1-20021017-C00007
 for which:
R1 and R2 are, independently of each other, C6-C23 alkyl or alkenyl radicals, which are linear or branched, or radicals —C(═O)—(C6-C23) alkyl or —C(═O)—(C6-C23) alkenyl, which are linear or branched, aryl radicals, cycloalkyl radicals, fluoroalkyl radicals, polyethylene glycol groups, oxyethylene or oxymethylene groups which are optionally repeated, linear or branched, optionally substituted,
p is a positive integer from 1 to 4,
n is a positive integer from 1 to 6,
m is a positive integer from 1 to 6 which may be different for each motif —(CH2)m, and more particularly for each motif —(CH2)m—NR— when n>1,
the number of R groups of formula II being between 1 and 4.
2. Compound according to claim 1, chosen from the compounds of formulae:
H2N—[—(CH2)m—NH—]nR  IIIRNH—[—(CH2)m—NH]nH  IIIa
in which:
R is a group of formula II as defined in claim 1,
n is a positive integer from 1 to 6,
m is a positive integer from 1 to 6 which may be different for each motif —(CH2)m.
3. Compound according to claim 1 of formula:
H2N—[—(CH2)m—NR]n−1—(CH2)m—NH2  IIIb
in which:
R has one of the meanings indicated for the formula I of claim 1 provided that at least one R group is of formula II,
n is a positive integer from 1 to 6,
m is a positive integer from 1 to 6 which may be different for each motif —(CH2)m, and more particularly for each motif —(CH2)m—NR— when n>1.
4. Compound according to claim 3, characterized in that it contains one or two R groups of formula II.
5. Compound according to claims 1 to 4, characterized in that R1 and R2 are, independently of each other, linear —C(═O)-alkyl or linear —C(═O)-alkenyl radicals.
6. Compound according to claim 5, characterized in that said alkyl or alkenyl comprises from 12 to 20 carbon atoms and in that said compound comprises 1 or 2 R groups of formula II.
7. Compound according to claim 6, characterized in that said alkyl or alkenyl comprises 12, 16 or 18 carbon atoms.
8. Compound according to claims 1 to 7, characterized in that n is an integer chosen from the numbers 2, 3 or 4.
9. Compound according to claims 1 to 8, characterized in that m is an integer chosen from the numbers 2, 3 or 4.
10. Compound according to claim 1, characterized in that it is chosen from the group consisting of the compounds of the following formulae:
IV
Figure US20020151070A1-20021017-C00008
IVa
Figure US20020151070A1-20021017-C00009
in which R1 and R2 are identical and are chosen from the stearoyl and oleoyl radicals.
11. Compound according to one of claims 1 to 10, characterized in that it is conjugated with one or more ligands of interest via one of the secondary or primary nitrogen atoms of the polyamine chain or of the diaminocarboxylic acid.
12. Compound according to claim 11, characterized in that said targeting component is chosen from the group consisting of all or part of sugars, peptides, oligonucleotides, lipids, hormones, vitamins, antigens, antibodies, ligands specific for membrane receptors, ligands capable of reacting with an anti-ligand, fusogenic peptides, nuclear localization peptides, or a combination of such compounds.
13. Compound according to one of claims 1 to 12, characterized in that it is in a cationic form.
14. Composition characterized in that it comprises at least one compound according to one of the preceding claims and optionally at least one adjuvant capable in particular of enhancing the formation of the complex between said compound and an active substance.
15. Composition according to claim 14, characterized in that said adjuvant is a neutral or zwitterionic lipid.
16. Composition according to claim 15, characterized in that said neutral or zwitterionic lipid is or is derived from a triglyceride, a diglyceride, cholesterol, a phosphatidylethanolamine (PE), phosphatidylcholine, phosphocholine, sphyngo-myelin, ceramide or cerebroside.
17. Composition according to claim 16, characterized, in that said neutral or zwitterionic lipid is dioleylphosphatidylethanolamine (DOPE).
18. Composition according to claims 14 to 17, characterized in that the compound/adjuvant weight ratio is between 0.1 and 10.
19. Complex comprising at least one compound according to claim 13 or at least one composition according to one of claims 14 to 18 and at least one active substance, in particular a therapeutically active substance, comprising at least one negative charge.
20. Complex according to claim 19, characterized in that said active substance is chosen from nucleic acids and proteins.
21. Complex according to claim 20, characterized in that said active substance is a nucleic acid chosen from the group consisting of a cDNA, a genomic DNA, a plasmid DNA, an antisense polynucleotide, a messenger RNA, a ribosomal RNA, a ribozyme, a transfer RNA, or a DNA encoding such RNAs.
22. Complex according to claim 21, characterized in that said nucleic acid comprises a gene of interest and components allowing the expression of said gene of interest.
23. Complex according to one of claims 19 to 22, characterized in that it has a size of less than 500 nm, advantageously less than 200 nm.
24. Complex according to one of claims 23, characterized in that it has a size of less than 100 nm.
25. Complex according to one of claims 19 to 24, characterized in that the ratio between the number of positive charges of the cationic compound(s) and/or composition(s) and the number of negative charges of said active substance varies from 0.05 to 20, more particularly from 0.1 to 15, and preferably from 5 to 10.
26. Process for preparing a complex according to claims 19 to 25, characterized in that one or more compounds according to claim 13 and/or at least one composition according to one of claims 14 to 18 are brought into contact with one or more active substances comprising at least one negative charge and in that said complex is recovered, optionally after a purification step.
27. Process of preparation according to claim 26, characterized in that said compounds and/or compositions are dissolved beforehand in a solvent which is miscible with water, in particular ethanol or dimethyl sulfoxide or a mixture of both.
28. Process of preparation according to claim 26, characterized in that said compounds and/or compositions are suspended beforehand in a detergent solution.
29. Process of preparation according to claim 28, characterized in that, in addition, a step of purification of said complex by dialysis is carried out.
30. Use of a compound according to any one of claims 1 to 13, of a composition according to any one of claims 14 to 18, of a complex according to any one of claims 19 to 25 to transfer at least one active substance, especially a therapeutically active substance, in particular a nucleic acid, into target cells in vitro, ex vivo or in vivo, more particularly in vivo.
31. Use according to claim 30, characterized in that said target cell is a mammalian cell.
32. Use according to claim 31, characterized in that said target cell is selected from a muscle cell, a hematopoietic stem cell, a cell of the airways, more particularly a tracheal or pulmonary cell.
33. Process for transferring an active substance comprising at least one negative charge, into a cell, characterized in that said cell is brought into contact with a complex according to any one of claims 19 to 25.
34. Complex according to one of claims 19 to 25, as a medicament for curative, preventive or vaccinal purposes.
35. Complex according to one of claims 19 to 25 for carrying out a method of therapeutic treatment which consists in transferring at least one therapeutically active substance, in particular a nucleic acid, into target cells.
36. Complex according to claim 35, characterized in that said target cell is a mammalian cell.
37. Complex according to claim 36, characterized in that said target cell is selected from a muscle cell, a hematopoietic stem cell, a cell of the airways, more particularly a tracheal or pulmonary cell, a cell of the respiratory epithelium.
38. Use of a compound according to any one of claims 1 to 13, of a composition according to any one of claims 14 to 18, of a complex according to any one of claims 19 to 25 for the preparation of a medicament for curative, preventive or vaccinal purposes, intended for the treatment of the human or animal body, in particular by gene therapy.
39. Use according to claim 38, characterized in that the medicament is intended to be administered by intramuscular injection, by inhalation, by intratracheal injection, by instillation, by aerosolization, by the topical route or by the oral route.
40. Pharmaceutical preparation characterized in that it comprises at least one complex according to any one of claims 19 to 25.
41. Preparation according to claim 40, characterized in that it comprises, in addition, at least one adjuvant capable of enhancing the transfecting power of said complex.
42. Preparation according to claim 41, characterized in that said adjuvant is chosen from the group consisting of chloroquine, a protic polar compound chosen in particular from propylene glycol, polyethylene glycol, glycerol, ethanol, 1-methyl-L-2-pyrrolidone or derivatives thereof, or an aprotic polar compound chosen in particular from dimethyl sulfoxide (DMSO), diethyl sulfoxide, di-n-propyl sulfoxide, dimethyl sulfone, sulfolane, dimethylformamide, dimethylacetamide, tetramethylurea, acetonitrile or derivatives thereof.
43. Preparation according to one of claims 40 to 42, characterized in that it comprises, in addition, a pharmaceutically acceptable carrier allowing its administration to humans or animals.
44. Cell transfected with a complex according to any one of claims 19 to 25.
US10/021,421 1997-02-28 2001-12-19 Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy Abandoned US20020151070A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/021,421 US20020151070A1 (en) 1997-02-28 2001-12-19 Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
FR9702420A FR2760193B1 (en) 1997-02-28 1997-02-28 LIPIDS AND COMPLEXES OF CATIONIC LIPIDS AND ACTIVE SUBSTANCES, IN PARTICULAR FOR THE TRANSFECTION OF CELLS
FR9702420 1997-02-28
US09/171,845 US6335199B1 (en) 1997-02-28 1998-02-27 Lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use
US10/021,421 US20020151070A1 (en) 1997-02-28 2001-12-19 Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US09/171,845 Continuation US6335199B1 (en) 1997-02-28 1998-02-27 Lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use
PCT/FR1998/000389 Continuation WO1998037916A1 (en) 1997-02-28 1998-02-27 Novel lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use

Publications (1)

Publication Number Publication Date
US20020151070A1 true US20020151070A1 (en) 2002-10-17

Family

ID=9504295

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/171,845 Expired - Fee Related US6335199B1 (en) 1997-02-28 1998-02-27 Lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use
US10/021,421 Abandoned US20020151070A1 (en) 1997-02-28 2001-12-19 Lipid compounds and compositions containing them which can be used for the transfer of at least one active substance, in particular a polynucleotide, into a target cell and use in gene therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/171,845 Expired - Fee Related US6335199B1 (en) 1997-02-28 1998-02-27 Lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use

Country Status (7)

Country Link
US (2) US6335199B1 (en)
EP (1) EP0948360A1 (en)
JP (1) JP2000510871A (en)
AU (1) AU727040B2 (en)
CA (1) CA2252942A1 (en)
FR (1) FR2760193B1 (en)
WO (1) WO1998037916A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7008791B1 (en) * 1999-10-01 2006-03-07 Lipoxen Technologies Limited Liposome-entrapped DNA oral vaccines

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
DE69937964T2 (en) * 1998-11-12 2009-01-02 Invitrogen Corp., Carlsbad TRANSPORTREAGENTIEN
US6184035B1 (en) * 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
AU2637500A (en) * 1999-12-10 2001-06-18 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
US20020142304A1 (en) * 2001-03-09 2002-10-03 Anderson Daniel G. Uses and methods of making microarrays of polymeric biomaterials
GB0222824D0 (en) * 2002-10-02 2002-11-06 Moredun Res Inst Bacteriophage-mediated immunisation II
US9006487B2 (en) 2005-06-15 2015-04-14 Massachusetts Institute Of Technology Amine-containing lipids and uses thereof
US8357531B2 (en) 2007-07-03 2013-01-22 Transgene S.A. Immortalized avian cell lines
US8969353B2 (en) 2008-11-07 2015-03-03 Massachusetts Institute Of Technology Aminoalcohol lipidoids and uses thereof
CN102548396A (en) * 2009-04-30 2012-07-04 贝克特里弗里特公司 Composition for sterilizing surfaces
DK3338765T3 (en) 2009-12-01 2019-03-04 Translate Bio Inc STEROID DERIVATIVE FOR THE SUPPLY OF MRNA IN HUMANGENETIC DISEASES
WO2012027675A2 (en) 2010-08-26 2012-03-01 Massachusetts Institute Of Technology Poly(beta-amino alcohols), their preparation, and uses thereof
CA2831392C (en) 2011-03-28 2020-04-28 Massachusetts Institute Of Technology Conjugated lipomers and uses thereof
PL2717893T3 (en) 2011-06-08 2019-12-31 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mrna delivery
WO2013185067A1 (en) 2012-06-08 2013-12-12 Shire Human Genetic Therapies, Inc. Nuclease resistant polynucleotides and uses thereof
CA2884870C (en) 2012-08-13 2022-03-29 Massachusetts Institute Of Technology Amine-containing lipidoids and uses thereof
BR112015022660A2 (en) 2013-03-14 2017-10-31 Shire Human Genetic Therapies Methods for purification of messenger rna
ES2689523T3 (en) 2013-03-14 2018-11-14 Translate Bio, Inc. MRNA-based compositions of the CFTR gene and related methods and uses
US9315472B2 (en) 2013-05-01 2016-04-19 Massachusetts Institute Of Technology 1,3,5-triazinane-2,4,6-trione derivatives and uses thereof
CN105813656B (en) 2013-10-22 2021-01-15 夏尔人类遗传性治疗公司 Lipid formulations for delivery of messenger RNA
EP3501605B1 (en) 2013-10-22 2023-06-28 Translate Bio, Inc. Mrna therapy for argininosuccinate synthetase deficiency
EA201992208A1 (en) 2013-10-22 2020-07-31 Транслейт Био, Инк. TREATMENT OF PHENYLKETONURIA USING mRNA
CN110511927A (en) 2014-04-25 2019-11-29 川斯勒佰尔公司 The purification process of mRNA
US10022455B2 (en) 2014-05-30 2018-07-17 Translate Bio, Inc. Biodegradable lipids for delivery of nucleic acids
EP3160959B1 (en) 2014-06-24 2023-08-30 Translate Bio, Inc. Stereochemically enriched compositions for delivery of nucleic acids
US9840479B2 (en) 2014-07-02 2017-12-12 Massachusetts Institute Of Technology Polyamine-fatty acid derived lipidoids and uses thereof
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
CN110418650A (en) 2016-11-16 2019-11-05 免疫治疗有限公司 For treating the nucleic acid of allergy
EA201991747A1 (en) 2017-02-27 2020-06-04 Транслейт Био, Инк. NEW CODON-OPTIMIZED CFTR mRNA
JP2020517271A (en) 2017-04-22 2020-06-18 イミュノミック セラピューティックス, インコーポレイテッドImmunomic Therapeutics, Inc. Improved LAMP construct
JP7222915B2 (en) 2017-05-02 2023-02-15 イミュノミック セラピューティックス, インコーポレイテッド Improved LAMP constructs containing cancer antigens
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
JP2021523185A (en) 2018-05-15 2021-09-02 イミュノミック セラピューティックス, インコーポレイテッドImmunomic Therapeutics, Inc. Improved LAMP constructs containing allergens
AU2019325702A1 (en) 2018-08-24 2021-02-25 Translate Bio, Inc. Methods for purification of messenger RNA
TW202043256A (en) 2019-01-10 2020-12-01 美商健生生物科技公司 Prostate neoantigens and their uses
US20240108703A1 (en) 2019-10-18 2024-04-04 Immunomic Therapeutics, Inc. Improved LAMP Constructs Comprising Cancer Antigens
PE20221182A1 (en) 2019-11-18 2022-08-05 Janssen Biotech Inc VACCINES BASED ON CALR AND JAK2 MUTANTS AND THEIR USES
WO2022009051A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. A method for determining responsiveness to prostate cancer treatment
US20230029453A1 (en) 2020-07-06 2023-02-02 Janssen Biotech, Inc. Prostate Neoantigens And Their Uses
EP4175721A1 (en) 2020-07-06 2023-05-10 Janssen Biotech, Inc. Prostate neoantigens and their uses
WO2023201201A1 (en) 2022-04-10 2023-10-19 Immunomic Therapeutics, Inc. Bicistronic lamp constructs comprising immune response enhancing genes and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030946A (en) * 1995-06-07 2000-02-29 Axys Pharmaceuticals, Inc. Reversible cysteine protease inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5334761A (en) * 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
AU701106B2 (en) 1995-06-07 1999-01-21 Promega Biosciences, Inc. Novel carbamate-based cationic lipids
JPH11510489A (en) * 1995-07-21 1999-09-14 ジンタ・インコーポレイテッド New amide-based cationic lipid
DE19607686A1 (en) * 1996-02-29 1997-09-04 Chemicon Lab Gmbh New metabolizable lipopolyamines, their presentation and application

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030946A (en) * 1995-06-07 2000-02-29 Axys Pharmaceuticals, Inc. Reversible cysteine protease inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7008791B1 (en) * 1999-10-01 2006-03-07 Lipoxen Technologies Limited Liposome-entrapped DNA oral vaccines

Also Published As

Publication number Publication date
JP2000510871A (en) 2000-08-22
AU6735298A (en) 1998-09-18
FR2760193B1 (en) 1999-05-28
WO1998037916A1 (en) 1998-09-03
FR2760193A1 (en) 1998-09-04
US6335199B1 (en) 2002-01-01
EP0948360A1 (en) 1999-10-13
CA2252942A1 (en) 1998-09-03
AU727040B2 (en) 2000-11-30

Similar Documents

Publication Publication Date Title
US6335199B1 (en) Lipid compounds and compositions containing same used for the transfer of at least an active substance, in particular a polynucleotide, in a target cell and therapeutic use
US6218370B1 (en) Glycerolipidic compounds used for the transfer of an active substance into a target cell
JP4176831B2 (en) Lipopolyamine and its pharmaceutical use as transfection agent
JP4467084B2 (en) Compounds for introducing nucleic acids into cells, their production and their use
US6638529B2 (en) Amide-based cationic lipids
US6291423B1 (en) Lipid complexes for transferring at least a therapeutically active substance, in particular a polynucleotide into a target cell and use in gene therapy
US6407178B1 (en) Cationic polymers, complexes associating said cationic polymers with therapeutically active substances comprising at least a negative charge, in particular nucleic acids, and their use in gene therapy
EP2035566B1 (en) Materials and complexes for the delivery of biologically-active material to cells
SK283575B6 (en) Compounds related to the amidinium family, pharmaceutical compositions containing same and use thereof
EP0987029B1 (en) Use of a catonic polymer for the preparation of a complex with nucleic acid and related compositions
EP1052288A1 (en) Complex for transferring an anionic substance of interest into a cell
EP0834572A2 (en) Alpha, gamma-diaminobutyric acid (DAB) containing oligopeptide derivatives
EP1052287A2 (en) Complex for transferring an anionic substance of interest into a cell
EP0965583A1 (en) Polyamine compounds and compositions containing them useful for the transfer of active substances into a cell

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE