WO2001092542A2 - Vecteurs ciblant l'integrine avec activite de transfection accrue - Google Patents

Vecteurs ciblant l'integrine avec activite de transfection accrue Download PDF

Info

Publication number
WO2001092542A2
WO2001092542A2 PCT/GB2001/002394 GB0102394W WO0192542A2 WO 2001092542 A2 WO2001092542 A2 WO 2001092542A2 GB 0102394 W GB0102394 W GB 0102394W WO 0192542 A2 WO0192542 A2 WO 0192542A2
Authority
WO
WIPO (PCT)
Prior art keywords
complex
cells
integrin
binding
peptide
Prior art date
Application number
PCT/GB2001/002394
Other languages
English (en)
Other versions
WO2001092542A8 (fr
WO2001092542A3 (fr
Inventor
Stephen Lewis Hart
Original Assignee
Ich Productions Limited
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
Priority claimed from GB0013090A external-priority patent/GB0013090D0/en
Priority claimed from GB0013089A external-priority patent/GB0013089D0/en
Application filed by Ich Productions Limited filed Critical Ich Productions Limited
Priority to AU2001260467A priority Critical patent/AU2001260467A1/en
Publication of WO2001092542A2 publication Critical patent/WO2001092542A2/fr
Publication of WO2001092542A3 publication Critical patent/WO2001092542A3/fr
Publication of WO2001092542A8 publication Critical patent/WO2001092542A8/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination

Definitions

  • the present invention relates to an improved integrin-targeting vector that has enhanced transfection activity.
  • the term "transfection” is used herein to denote the introduction of a nucleic acid into a cell .
  • the nucleic acid may be of any origin and the recipient cell may be prokaryotic or eukaryotic.
  • Gene therapy and gene vaccination are techniques that offer interesting possibilities for the treatment and/or prophylaxis of a variety of conditions, as does anti-sense therapy. Such techniques require the introduction of a DNA of interest into target cells. The ability to transfer sufficient DNA to specific target cells remains one of the main limitations to the development of gene therapy, anti-sense therapy and gene vaccination. Both viral and non-viral DNA delivery systems have been proposed. In some cases RNA is used instead of DNA.
  • Receptor-mediated gene delivery is a non-viral method of gene transfer that exploits the physiological cellular process, receptor-mediated endocytosis to internalise DNA.
  • Receptor- mediated non-viral vectors have several advantages over viral vectors. In particular, they lack pathogenicity; they allow targeted gene delivery to specific cell types and they are not restricted in the size of nucleic acid molecules that can be packaged. Gene expression is achieved only if the nucleic acid component of the complex is released intact from the endosome to the cytoplasm and then crosses the nuclear membrane to access the nuclear transcription machinery.
  • transfection efficiency is generally poor relative to viral vectors owing to endosomal degradation of the nucleic acid component, failure of the nucleic acid to enter the nucleus and the exclusion of aggregates larger than about 150nm from clathrin coated vesicles .
  • Integrins are a super-family of heterodimeric membrane proteins consisting of several different and ⁇ subunits. They are important for attachment of cells to the extracellular matrix, cell-cell interactions and signal transduction. Integrin-medi- ated cell entry is exploited for cell attachment and entry by a 5 number of intracellular pathogens including Typanosoma cruzi (Fernandez et al . , 1993), adenovirus ( ickham et al . , 1993), echovirus (Bergelson et al . , 1992) and foot-and-mouth disease virus (Logan et al . , 1993) as well as the enteropathogen Y. pseudotuberculosis (Isberg, 1991) .
  • enteropathogen Y. pseudotuberculosis Isberg, 1991
  • the conserved amino acid sequence arginine-glycine-aspartic acid is an evolutionarily conserved feature of many, but not all, natural integrin-binding ligands such as extracellular 35 matrix proteins and viral capsids. Peptides, particularly those containing cyclic-RGD domains can also bind integrins. Peptides containing cyclic-RGD domains are particularly suitable ligands for vectors since they bind to integrins with higher affinities than linear peptides (Koivunen et al . 1995) . Hart et al .
  • W098/54347 relates to an invention based on the surprising 30 observation that inclusion of a lipid component in the above oligolysine/-peptide/DNA complex increases levels of transfection of DNA from about 1 to 10% to about 50 to almost 100%. Not only is the level of transfection increased dramatically but, contrary to previous experience, the increase 35 is observed in all cell lines tested, including endothelial, epithelial and tumour cell lines.
  • W098/54347 discloses a complex that comprises (i) a nucleic acid, especially a nucleic acid encoding a sequence of interest, (ii) an integrin-binding component,
  • the complex is a transfection vector.
  • the present invention relates to modifications of the integrin- binding component of W098/54347, which result in further improvements in transfection efficiency.
  • the integrin-binding component is any component that is capable of binding specifically to integrins found on the surface of cells.
  • the integrin-binding component may be a naturally occurring integrin-binding ligand, for example, an extracellular matrix protein, a viral capsid protein, the bacterial protein invasin, a snake venom disintegrin protein, or an integrin-binding fragment of any such protein.
  • integrin-binding proteins and fragments thereof may be obtained from natural sources or by recombinant techniques, but they are difficult to synthesise and purify in large amounts, they require conjugation directly to DNA or RNA or to polycationic elements for DNA or RNA binding, and are immunogenic in vivo.
  • integrin-binding peptides it is preferable to use integrin-binding peptides, in particular because of their ease of synthesis, purification and stor- age, their potential for chemical modification, and their potentially low immunogenicity in vivo.
  • Examples of integrin- binding peptides are given in Verfaille, 1994 #635; Wang, 1995 #645; Wunschz, 1991 #539; Pierschbacher, 1984 #314; Massia, 1992 #86, Clements et al . 1994 & Lu et al, 1993; and in Koivunen et al.. 1995; 1993; 1994; O'Neil et al . 1992; Healy et al 1995; and Pasqualani et al . 1995.
  • peptides containing the conserved amino acid sequence arginine-glycine-aspartic acid bind with high affinity to integrins. Accordingly, peptides comprising the RGD sequence are particularly useful .
  • the affinity between integrin and peptide ligands is influenced by the amino acid sequence flanking the RGD domain. In peptides having a cyclic region encompassing all or part of the region comprising the RGD sequence, the conformational freedom of the RGD sequence is restricted.
  • Such peptides generally have a higher affinity for integrin receptors than do their linear counterparts. Such cyclic peptides are particularly preferred.
  • Cyclic peptides may be formed by the provision of two cysteine residues flanking the RGD sequence in the peptide, thus enabling the formation of a disulphide bond.
  • a cysteine residue may be separated from the RGD sequence by one or more residues, for example, up to six residues, or may be immediately adjacent to the RGD sequence, although preferably both cysteines are not immediately adjacent to the ends of the RGD sequence. Two further cysteine residues may be present, enabling formation of two disulphide bonds.
  • a peptide that consists of or comprises the sequence CRGDMFGC may advantageously be used as an integrin- binding peptide according to the present invention.
  • Examples of peptides that comprises the sequence CRGDMFGC and that are effective integrin-binding ligands are the peptides GGCRGDMFGC [SEQ . ID.NO. : 6] , GGCRGDMFGCG [SEQ . ID.NO . : 7] , GGCRGDMFGCA [SEQ. ID.NO. :8] and GACRGDMFGCA [SEQ. ID.NO. : 9] .
  • the peptide GACDCRGDCFCA [SEQ. ID. O. : 10] has the potential to form two disulphide bonds for stabilising the RGD loop. That peptide and others having the potential to form two RGD- stabilising disulphide bonds by the presence of two or more cysteine residues, may be particularly useful as integrin- binding ligands according to the present invention.
  • integrin-binding peptides contain the conserved RGD sequence.
  • the peptides GACRRETAWACA [SEQ. ID.NO. : 11] and GACRRETAWACG [SEQ. ID.NO. : 12] are integrin- specific peptides.
  • Other peptides comprising the sequence CRRETAWAC [SEQ . ID .NO .: 13] may be used, as may other non-RGD peptides, particularly those that have the potential for disulphide bond formation by the provision of two or more cysteine residues.
  • Peptide sequences may be designed on the basis of known ligands, for example, on the basis of integrin-binding domains of naturally-occurring integrin-binding ligands, or on the basis of known peptides that bind to integrins.
  • integrins are a family of heterodimeric proteins found on the surface of cells. They consist of several different ⁇ and ⁇ subunits. Some integrins are found on may types of cells, others are more specific, for example, ⁇ 5 and o:v integrins are widespread and are found on a diverse range of cells. Integrin-binding ligands can vary in their affinity for different integrins. For example, GACRGDMFGCA [SEQ. ID.NO. : 9] (peptide 1) has affinity for ⁇ 5 and ⁇ v integrins but is no ⁇ - specific (O'Neil et al . 1992, Hart et al . 1997). GACDCRGDCFCA [SEQ. ID.NO.
  • Peptide number and Sequence SEQ. ID.NO. integrin specificity Peptide 1 ( ⁇ v, ⁇ 5 ⁇ l) GACRGDMFGCA SEQ..ID .NO. :9 Peptide 2 ( ⁇ v, ⁇ 5 ⁇ l) GACRGDMFGCGG SEQ. .ID .NO. :14 Peptide 5 ( ⁇ v) GACDCRGDCFCA SEQ. .ID .NO. :10 Peptide 6 ( ⁇ 5 ⁇ l) GACRRETAWACG SEQ. , ID, .NO. :12 Peptide 7 ( ⁇ 4 ⁇ l) GAGPEILDVPST SEQ. .ID .NO. :15 Peptide 8 ( ⁇ 4 ⁇ l) GACQIDSPCA SEQ. , ID, .NO. :16 Peptide 9 ( ⁇ 5 ⁇ l) GACRRETAWACGKGACRRETAWACG
  • the initial residues "GG” or "GC”, where present, are spacers.
  • the present invention is based on the observation that modifying the spacer can improve transfection efficiency.
  • the present invention provides a complex that comprises
  • nucleic acid especially a nucleic acid encoding a sequence of interest, (ii) an integrin-binding component,
  • a lipid component in which complex the integrin binding component comprises an integrin-binding element and a spacer element, the spacer element being longer and/or more hydrophobic than the dipeptide spacers GG (glycine-glycine) and GA (glycine-alanine) .
  • a spacer element of the present invention is generally a peptide, that is to say, it comprises amino acid residues.
  • the amino acids may be naturally occurring or non-naturally occurring. They may have L- or D-configuration.
  • a spacer of the present invention may be longer than a dipeptide. It may, for example, comprise three or more amino acids, for example, four or more, for example, five or more, for example, up to ten amino acids or more.
  • the amino acids may be the same or different, but the use of multiple lysine residues should be avoided in the spacer as oligolysine sequences are the preferred polycationic nucleic acid-binding component of a complex of the present invention.
  • the spacer may be more hydrophobic than the dipeptides GG and GA.
  • amino acids that are more hydrophobic than glycine and alanine may be used.
  • hydrophobic amino acids are well known and include €-amino hexanoic acid.
  • a spacer may be either longer or more hydrophobic than the dipeptides GG and GA, or it may be both longer and more hydrophobic .
  • S serine
  • G glycine
  • A alanine
  • X C-amino hexanoic acid. This spacer is highly hydrophobic.
  • the spacer is generally at the N-terminus of the integrin- binding peptide.
  • the non-spacer element of the integrin-binding component may be any of the integrin-binding peptides or polypeptides described above.
  • it may be a naturally occurring integrin- binding ligand, for example, an extracellular matrix protein, a viral capsid protein, the bacterial protein invasin, a snake venom disintegrin protein, or an integrin-binding fragment of any such protein.
  • integrin-binding proteins and fragments thereof may be obtained from natural sources or by recombinant techniques.
  • the integrin binding peptides described above comprise both an integrin- binding peptide element and a spacer dipeptide GG or GC, see for example, the peptides of SEQ. ID.NO: 6, which comprises the integrin-binding element CRGDMFGC and the dipeptide spacer GG.
  • the spacer element of the present invention takes the place of a GG or GA spacer in peptides described above that comprises such a GG or GA spacer .
  • integrin binding peptides include the following: CRGDMFGC [SEQ. ID.NO. : 5] ; CRGDMFGCG [SEQ. ID.NO. : 19] ; CRGDMFGCA [SEQ . ID.NO. : 20] ; CDCRGDCFCA [SEQ . ID .NO. : 21] ; CRRETAWACA [SEQ . ID .NO . : 22 ; CRRETAWACG [SEQ . ID .NO . : 23 ; CRGDMFGCGG[SEQ.ID.NO. :24; GPEILDVPST [SEQ . ID .NO. : 25 ; CQIDSPCA [SEQ. ID.NO. : 26] ; CRRETAWACGKGACRRETAWACG [SEQ. ID.NO. :27] .
  • Further suitable peptides are described, for example, in W095/14714.
  • a spacer of the present invention may be linked to any of the above peptide, preferably at the N-terminus thereof.
  • the nucleic acid, the polycationic nucleic acid-binding component and the lipid component are preferably as described in W098/54347, and as set out below.
  • the nucleic acid may be obtained from natural sources, or may be produced recombinantly or by chemical synthesis. It may be modified, for example, to comprise a molecule having a specific function, for example, a nuclear targeting molecule.
  • the nucleic acid may be DNA or RNA. DNA may be single stranded or double stranded.
  • the nucleic acid may be suitable for use in gene therapy, in gene vaccination or in an anti-sense therapy.
  • the nucleic acid may be or may relate to a gene that is the target for particular gene therapy that is to say, a gene for newer treatment by gene therapy is desired, fore example, a gene having a mutation, or another defect, or a gene that is deficient, or a gene that is deficient, or a gene that is absent or is present in insufficient amounts or that is present in excess, any of which effects causes a disease or disorder.
  • the nucleic acid may be a molecule that can function as a gene vaccine or as an anti-sense therapeutic agent.
  • the nucleic acid may be or correspond to a complete coding sequence or may be part of a coding sequence or may be a control or regulatory element or may be or correspond to a genetic sequence comprising all or some elements selected from the coding sequence of a gene and the upstream and downstream non translated sequences, including control and regulatory elements .
  • the nucleic acid may encode a protein that is commercially useful, for example industrially or scientifically useful, for example an enzyme; pharmaceutically useful, for example, a protein that can be used therapeutically or prophylactically as a medicament or vaccine; or diagnostically useful, for example, an antigen for use in an ELISA.
  • Host cells capable of producing commercially useful proteins are sometimes called "cell factories".
  • nucleic acid is a protein coding sequence.
  • the nucleic acid component is generally presented in the form of a nucleic acid insert in a plasmid or vector. In some cases, however, it is not necessary to incorporate the nucleic acid component in a vector in order to achieve expression. For example, gene vaccination and anti-sense therapy can be achieved using a naked nucleic acid.
  • the nucleic acid is generally DNA but RNA may be used in some cases, for example, in cancer vaccination.
  • the nucleic acid component may be referred to below as the plasmid component or component "D" .
  • the polycationic nucleic acid-binding component is any polycation that is capable of binding to DNA or RNA is retained.
  • from 4 to 100 cationic monomers may be present, for example, from 10 to 20, especially about 16.
  • An oligolysine is particularly preferred, for example, having from 3 to 100 lysine residues, for example, from 10 to 20, for example, from 13 to 19, for example, from 14 to 18, for example, from 15 to 17 residues, especially 16, 17 or 18 residues especially [K] ⁇ 6 , "K" denoting lysine.
  • the polycationic DNA or RNA-binding component may advantageously be linked or otherwise attached to the integrin- binding component .
  • a combined integrin-binding component/polycationic DNA or RNA-binding component may be referred to below as component "I" .
  • a polycationic DNA or RNA-binding component may be chemically bonded to an integrin-binding component, for example, by a peptide bond in the case of an oligolysine.
  • the polycationic component may be linked at any position of the integrin-binding component.
  • Preferred combinations of integrin-binding component and polycationic DNA or RNA-binding component are an oligolysine, especially [K] ⁇ 6 , linked via a peptide bond to a peptide, for example, a peptide as described above.
  • a combined integrin-binding component/polycationic DNA or RNA- binding component is a component having an integrin-binding moiety that is the integrin-binding component as defined and described herein linked or otherwise attached to a polycationic DNA or RNA binding moiety that is a polycationic DNA or RNA component as defined and described herein.
  • the polycationic DNA or RNA-binding component may advantageously be linked or otherwise attached to the integrin-binding component.
  • a combined integrin-binding component/polycationic DNA or RNA-binding component may be referred to below as component "I".
  • a polycationic DNA or RNA-binding component may be chemically bonded to an integrin-binding component, for example, by a peptide bond in the case of an oligolysine.
  • the polycationic component may be linked at any position of the integrin-binding component.
  • Preferred combinations of integrin-binding component and polycationic DNA or RNA-binding component are an oligolysine, especially [K] ⁇ 6 , linked via a peptide bond to a peptide, for example, a peptide as described above.
  • the lipid component may be or may form a cationic liposome.
  • the lipid component may be or may comprise one or more lipids selected from cationic lipids and lipids having membranae destabilising or fusogenic properties, especially a combination of a cationic lipid and a lipid that has membrane destabilising properties .
  • a preferred lipid component (“L”) is or comprises the neutral lipid dioleyl phosphatidylethanolamine, referred to herein as "DOPE” .
  • DOPE has membrane destabilising properties sometimes referred to as “fusogenic” properties (Farhood et al . 1995).
  • Other lipids, for example, neutral lipids, having membrane destabilising properties, especially membrane destabilising properties like those of DOPE may be used instead of or as well as DOPE.
  • phospholipids having at least one long chain alkyl group for example, di(long alkyl chain) phospholipids may be used.
  • the phospholipid may comprise a phosphatidyl group, for example, a phosphatidylalkanolamine group, for example, a phosphatidylethanolamine group .
  • a further preferred lipid component is or comprises the cationic lipid N- [1- (2, 3-dioleyloxy) propyl] -N,N,N-trimethyl- ammonium chloride, referred to herein, as "DOTMA" .
  • DOTMA has cationic properties.
  • Other cationic lipids may be used in addition to or as an alternative to DOTMA, in particular cationic lipids having similar properties to those of DOTMA.
  • Such lipids are, for example, quaternary ammonium salts substituted by three short chain alkyl groups, and one long chain alkyl group .
  • the short chain alkyl groups may be the same or different, and may be selected from methyl and ethyl groups . At least one and up to three of the short chain alkyl group may be a methyl group.
  • the long alkyl chain group may have a straight or branched chain, for example, a di(long chain al yl)alkyl group.
  • Another preferred lipid component is or comprises the lipid 2, 3-dioleyloxy-N- [2- (spermidinecarboxamido) ethyl] -N,N-dimethyl- 1-propanaminiumtrifluoridoacetate, referred to herein as "DOSPA" .
  • Analogous lipids may be used in addition to or as an alternative to DOSPA, in particular lipids having similar properties to those of DOSPA. Such lipids have, for example, different short chain alkyl groups from those in DOSPA.
  • a preferred lipid component comprises DOPE and one or more other lipid components, for example, as described above.
  • Such mixtures form cationic liposomes.
  • An equimolar mixture of DOPE and DOTMA is found to be particularly effective.
  • Such a mixture is known generically as “lipofectin” and is available commercially under the name “Lipofectin” .
  • the term “lipofectin” is used herein generically to denote an equimolar mixture of DOPE and DOTMA.
  • Other mixtures of lipids that are cationic liposomes having similar properties to lipofectin may be used. Lipofectin is particu- larly useful as it is effective in all cell types tested.
  • a further preferred lipid component comprises a mixture of DOPE and DOSPA. Such mixtures also form cationic liposomes.
  • a mixture of DOPE and DOSPA in a ratio by weight 3:1 DOSPA:DOPE is particularly effective.
  • Such a mixture, in membrane filtered water, is available commercially under the name "Lipo- fectamine" .
  • Mixtures comprising DOPE, DOTMA and DOSPA may be used, for example, mixtures of lipofectin and lipofectamine.
  • cationic lipids are available commercially, for example, DOTAP (Boehringer-Mannheim) and lipids in the Tfx range (Promega) .
  • DOTAP is N- [1- (2 , 3-diolyloxy) propyl] -N,N,N-tri- methylammonium methylsulphate.
  • the Tfx reagents are mixtures of a synthetic cationic lipid [N,N,N' ,N' -tetramethyl-N,N' - bis (2-hydroxyethyl) -2,3-di (oleoyloxy) -1, 4-butanediammonium iodide and DOPE. All the reagents contain the same amount of the cationic lipid component but contain different molar amounts of the fusogneic lipid, DOPE.
  • lipofectin and lipofectamine appear to be markedly more effective as the lipid component in LID complexes of the present invention than are DOTPA and Tfx agents.
  • a putative integrin-binding component polycationic DNA or RNA-binding component, or of lipid component may be determined readily using the methods described herein.
  • the efficiency of transfection using a complex of the invention is influenced by the ratio lipid component : integrin-binding component :DNA or RNA.
  • the optimal ratios can be determined simply by admixing the components in different ratios and measuring the transfection rate for that cell type, for example, as described herein.
  • a combination consisting of a pGL2 plasmid, which is a plasmid encoding luciferase (a reporter gene) under an SV40 promoter as DNA component (D) , [K] ⁇ 6 GACRGDMFGCA [SEQ. ID.NO. : 9] ( [K] ⁇ 6 -peptide 1) as a combined integrin-binding component/polycationic DNA binding component (I) , and lipofectin (DOPE:DOTMA 1:1 molar ratio) as the lipid component (L) was tested to find the optimal ratio of components.
  • a ratio L:I:D of 0.75:4:1 by weight, or the corresponding molar ratio are preferred when lipofectin is used as the lipid component.
  • the optimal ratio was found to be 12 ⁇ g lipofectamine: 4 ⁇ g [K] 16 -peptide 6 : 1 ⁇ g plasmid DNA or RNA.
  • a ratio of L:I:D of 12:4:1 by weight, or the corresponding molar ratio, is appropriate for lipofectamine-con- taining complexes.
  • Optimal ratios for other systems may be determined analogously.
  • Lipofectin and lipofectamine appear to be particularly effective in enhancing transfection. Lipofectin has the advantage that only very small amounts are required. Any side effects that may occur are therefore minimised. As indicated above, the optimal weight ratio of components L:I:D when using lipofectamine is 12:4:1. With lipofectin the optimal ratio is only 0.75:4:1.
  • the present invention provides a process for the production of a transfection complex of the present invention, which comprises admixing components (i) , (ii) , (iii) and (iv) .
  • the components may be admixed in any order, it is generally preferable that the lipid component is not added last.
  • lipid component lipid component
  • combined integrin-binding/polycationic DNA or RNA-binding component DNA or RNA component, for example, in the order: lipofectin, oligolysine-peptide component ,. DNA or RNA component .
  • the present invention also provides a mixture comprising an integrin-binding component, a polycationic nucleic acid-binding component, and a lipid component.
  • Such a mixture may be used to produce a nucleic acid-containing transfection complex of the invention by the incorporation of a nucleic acid with the mixture, for example, by admixture.
  • the mixture of the invention may be used for the production of a complex which comprises, instead of the nucleic acid component, any other component that is capable of binding to the polycationic nucleic-acid binding component, for example, a protein.
  • the present invention further provides a process for the production of a complex of the present invention, which comprises admixing a nucleic acid with a mixture of the invention.
  • the individual components of a mixture of the invention are each as described above in relation to the complex of the invention.
  • the preferred components, preferred combinations of components, preferred ratios of components and preferred order of mixing, both with regard to the mixture and to the produc- tion of a complex, are as described above in relation to the complex of the invention.
  • a mixture of the present invention preferably comprises an equimolar mixture of DOPE and DOTMA (lipofectin) as the lipid component and an oligolysine-peptide especially a [K] i ⁇ -peptide as a combined integrin-binding/nucleic acid-binding component .
  • the preferred molar ratio lipofectine: oligolysine-peptide is 0.75:4.
  • the present invention provides a method of transfecting a cell with a nucleic acid, which comprises contacting the cell in vitro or in vivo with a complex of the present invention under conditions suitable for effecting transfection, for example, as described in the Examples herein.
  • the present invention also provides a process for expressing a nucleic acid in a host cell, which comprises bringing the cell into contact with a complex of the present invention. The host cell is then cultured under conditions that enable the cell to express the nucleic acid.
  • the present invention further provides a process for the production of a protein, which comprises contacting a host cell in vitro or in vivo with a complex of the present invention under conditions suitable for effecting transfection, culturing the host cell under conditions suitable for protein expression, allowing the cell to express the protein, and obtaining the protein.
  • the host cell may be transfected in vitro with a nucleic acid by means of • a complex of the present invention and cultured, the protein being obtained either from the host cell or from the culture medium.
  • the present invention further provides a cell transfected with a complex of the present invention, and also the progeny of such a cell.
  • the present invention also provides a pharmaceutical composition which comprises a complex of the present invention in admixture or conjunction with a pharmaceutically suitable carrier.
  • the composition may be a vaccine.
  • the present invention also provides a method for the treatment or prophylaxis of a condition caused in a human or in a non- human animal by a defect and/or a deficiency in a gene, which comprises administering a complex of the present invention to the human or to the non-human animal in an amount effective for said treatment or prophylaxis.
  • the present invention also provides a method for therapeutic or prophylactic immunisation of a human or of a non-human animal, which comprises administering a complex of the present invention to the human or to the non-human animal in an amount effective for said therapeutic or prophylactic immunisation.
  • the present invention also provides a method of anti-sense therapy of a human or of a non-human animal, wherein a complex of the present invention comprising anti-sense DNA is administered to the human or to the non-human animal in an amount effective for said anti-sense therapy.
  • the present invention further provides a complex of the present invention for use as a medicament and/or vaccine, for example for the prophylaxis of a condition caused in a human or in a non-human animal by a defect and/or a deficiency in a gene, for therapeutic or prophylactic immunisation of a human or of a non-human animal, or for anti-sense therapy of a human or of a non-human animal .
  • the present invention also provides the use of a complex of the present invention for the manufacture of a medicament for the prophylaxis of a condition caused in a human or in a non-human animal by a defect and/or a deficiency in a gene, for therapeutic or prophylactic immunisation of a human or of a non- human animal, or for anti-sense therapy of a human or of a non- human animal .
  • a non-human animal is, for example, a mammal, bird or fish, and is particularly a commercially reared animal .
  • the DNA or RNA in the complex of the invention is appropriate for the intended gene therapy, gene vaccination, or anti-sense therapy.
  • the DNA or RNA and hence the complex is administered in an amount effective for the intended purpose.
  • the present invention provides a kit suitable for preparing a mixture of the present invention.
  • a kit suitable for preparing a mixture of the present invention.
  • a kit comprises the following: (i) an integrin-binding component as defined above; (ii) a polycationic nucleic acid- binding component, (iii) a lipid component and, optionally, (iv) an agent that disrupts cell-cell junctions.
  • a kit suitable for producing a complex of the present invention may comprise components (i) to (iv) above and (v) either a nucleic acid or a plasmid or vector suitable for the expression of a nucleic acid, the plasmid or vector being either empty or comprising the nucleic acid.
  • kits are, for example, as described above in relation to a complex or a mixture of the present invention. Preferred components are as described above.
  • a kit generally comprises instructions for the production of a complex or a mixture of the present invention.
  • the instruc- tions preferably indicate the preferred ratios of the components and the preferred order of admixing the components, for example, as described above.
  • a kit may be used for producing a complex suitable for gene therapy, gene vaccination or anti- sense therapy. Alternatively, it may be used for producing a complex suitable for transfecting a host cell with a nucleic acid encoding a commercially useful protein i.e. to produce a so-called "cell factory” .
  • the kit of the present invention enables the user to produce quickly and easily a highly efficient transfection complex of the present invention using any DNA or RNA of choice.
  • a kit of the invention may comprises the following components: (a) an integrin-binding component as defined above, (b) a polycationic nucleic acid-binding component, (c) a lipid component, (d) a nucleic acid and optionally (e) an agent that disrupts cell-cell junctions.
  • kit is suitable for the production of a complex for use, for example, in gene vaccination or anti-sense therapy.
  • kits of the invention the components including the preferred components are, for example, as described above in relation to a complex of the present invention.
  • the present invention also provides a lipid component as described above for use in increasing the efficiency of transfection of a cell with a nucleic acid, either DNA or RNA, the lipid component being used in combination with an integrin- binding component as defined above and a polycationic nucleic acid-binding component .
  • the present invention also provides the use of a lipid component as described above for the manufacture of a medicament comprising (i) a nucleic acid, especially a nucleic acid encoding a sequence of interest,
  • the medicament may be for gene therapy, gene vaccination, or anti-sense therapy.
  • the present invention also provides a complex that comprises (i) a nucleic acid, especially a nucleic acid encoding a sequence of interest,
  • an integrin-binding component as defined above, and (iii) a polycationic a nucleic acid-binding component, characterised in that a lipid component, for example as described above, is an additional component of the complex.
  • the present invention also provides a method for increasing the efficiency of a complex that comprises (i) a nucleic acid, especially a nucleic acid encoding a sequence of interest,
  • a polycationic a nucleic acid-binding component characterised in that a lipid component, for example as described above, is incorporated as an additional component of the complex.
  • the lipid component is, for example, a mixture of DOPE and DOSPA or, especially, a mixture of DOPE and DOTMA, in particular an equimolar mixture of DOPE and DOTMA (lipofectin) .
  • Targets for gene therapy include monogenic disorders, for example, cystic fibrosis, various cancers, and infections, for example, viral infections, for example, with HIV.
  • monogenic disorders for example, cystic fibrosis
  • various cancers for example, various cancers
  • infections for example, viral infections, for example, with HIV.
  • transfection with the p53 gene offers great potential for cancer treatment.
  • Targets for gene vaccination are also well known, and include vaccination against pathogens for which vaccines derived from natural sources are too dangerous for human use and recombinant vaccines are not always effective, for example, hepatitis B virus, HIV, HCV and herpes simplex virus.
  • Targets for anti-sense therapy are also known.
  • Complexes of the present invention have been demonstrated to transfect various different cell types, including endothelial and epithelial cells, and tumour cells. Transfection of all cell types tested including cell types that are particularly reistant to transfection with most plasmid transfection vectors, for example, neuroblastoma cells, primary smooth muscle cells and cardiac myocytes, and haematopoieic cells has been achieved with high efficiency using transfection complexes of the present invention. This enables effective gene therapy, gene vaccination and anti-sense therapy without the previous restrictions as to cell type. For example, transfection with the p53 gene for cancer therapy has great potential but is currently limited by the range of cell types in which effective transfection can be achieved.
  • the effective tranfection of neuroblastoma cells demonstrates that the complexes of the invention may be used as vaccines or for therapy of neuroblastoma, an important childhood malignancy.
  • the effective transfection of primary smooth muscle cells and cardiac myocytes, which are particularly resistant to plasmid-mediated transfection, demonstrates that diseases and other pathological conditions affecting muscles and the cardiovascular system can now be treated by gene therapy.
  • One such condition is restenosis. After balloon angioplasty plaques reform in 30-50% of cases.
  • a gene that prevents proliferation of cells in blood vessel walls may be introduced using a complex of the present invention to reduce restenosis.
  • Haematopoietic cells are another cell type that is particularly resistant to plasmid-mediated transfection.
  • the effectiveness of tranfection using a complex of the present invention which can exceed 60%, now enables gene therapy, gene vaccination and anti-sense therapy of diseases involving haematopoietic cells, including leukaemia and bone marrow stem cell disorders.
  • transfection of a cytokine gene may be used for adjuvant immunotherapy.
  • Complexes of the invention have been demonstrated to be effective vectors for intracellular transport and delivery of anti-sense oligonucleotides, which enables antiviral and cancer therapy.
  • complexes of the invention have been demonstrated to be effective for intracellular transport of very large DNA molecules, for example, DNA larger than 125kb, which is par- ticularly difficult using conventional vectors. This enables the introduction of artificial chromosomes into cells .
  • Transfection at high levels has been demonstrated in vivo, confirming the utility of the complexes of the invention for gene therapy, antisense therapy and gene vaccination.
  • Transfection of the airways, for example, the bronchial epithelium demonstrates utility for gene therapy of, for example, cystic fibrosis and asthma.
  • Transfection of corneal endothelium demonstrates utility for treatment of eye disease affecting the cornea or corneal organ transplants, for example in glaucoma.
  • the high levels of transfection make the complex of the invention particularly suitable for the production of host cells capable of producing a desired protein, so-called "cell factories".
  • cell factories For long-term production, it is desirable that the introduced nucleic acid is incorporated in the genome of the host cell, or otherwise stably maintained. That can be readily ascertained.
  • the range of proteins produced in this way is large, including enzymes for scientific and industrial use, proteins for use in therapy and prophylaxis, immunogens for use in vaccines and antigens for use in diagnosis.
  • the present invention provides a non-viral vector that is capable of high efficiency transfection.
  • the vector comprises four modular elements; an oligolysine, especially [K] ⁇ 6 , DNA or RNA-binding element; a high affinity integrin-binding peptide, for example, a peptide described herein; a DNA or RNA sequence, optionally in a plasmid, and optionally regulated by a viral promoter and an enhancing element; the cationic liposome DOTMA/DOPE (lipofectin) .
  • the combination of oligolysine-peptide/DNA or RNA complex with the cationic liposome formulation DOTMA/DOPE is a potent combination.
  • DOPE/DOSPA formulation may be used instead of or in addition to a DOTMA/DOPE formulation.
  • optimisation of variables associated with complex formation and the mode of transfection by LID complexes has been demonstrated.
  • analysis by atomic forces microscopy has been carried out to assess the structure of the complexes .
  • the level of transfection with LID (lipofectin/ [K] 16 - peptide/plasmid) complexes is three to six fold higher than that with LKD (lipofectin/ [K] ⁇ 6 /plasmid) complexes prepared with the same charge ratios, or with LD (lipofectin/plasmid) complexes. This indicates that the integrin-targeting moiety, i.e. the peptide, is a significant factor in the transfection efficiency of those complexes.
  • Optimised LID transfection complexes contain only one seventh of the amount of lipofectin required for optimal transfection with LD complexes.
  • Transfections with low-ratio LD complexes that contain the same ratio of lipofectin to [K] ⁇ 6 -peptide/- plasmid as in optimal LID complexes but no [K] i6-peptide, did not transfect cells at all. This suggests that the role of lipofectin in LID complexes is to enhance transfection mediated by the integrin receptor-binding peptide.
  • LID and ID complexes both form spherical particles of similar sizes.
  • Optimal LD complexes formed a tubular network with some tubule-associated particles, which suggests a different type of cellular interaction and transfection mechanism from LID and ID transfections .
  • transfection by LID complexes were prepared containing a range of proportions of [K] ⁇ and [K] l ⁇ integrin targeting peptide 6, [K] ⁇ e GACRRETAWACG [SEQ. ID. O. : 35] .
  • Transfection expression data indicate higher efficiencies with complexes in which increasing amounts of [K] i6 peptide 6 replace [K] ⁇ 6 and a dose-dependency on the amount of integrin-targetting (ligand- binding) domain i.e. peptide 6.
  • the ratio of components mixed together to form the optimal transfection complex is also informative as to the possible mechanism of lipofectin mediated enhancement.
  • the DOTMA element of lipofectin is cationic, which may enhance the activity of the complex, while DOPE may have the ability to destabilise the endosomal membrane (Farhood et al . , 1995) enhancing endosomal release of plasmid DNA or RNA.
  • the components of the LID complexes are mixed together in constant optimal ratios. It is assumed that the particles formed also contain these elements in the same proportions. Therefore, 3 nmol negative charge from plasmid DNA or RNA are associated with approximately 21 nmol positive charge from the [K]i6- peptide.
  • Lipofectin however, provides only a further 0.25 nmol of positive charge. This suggests that, contrary to expectations, the enhancing effect of lipofectin in LID complexes is not charge related but may relate to the membrane destabilising effect of the DOPE component.
  • the complexes are formed electrostatically by random association of lipofectin, oligolysine-peptide and plasmid DNA or RNA.
  • the relative high proportion of oligolysine-peptide ensures a high proportion of integrin-targeting ligands per plasmid molecule.
  • Particles are formed that contain one or more plasmids, associated with thousands of oligolysine-pep- tides and, therefore, a very high concentration of integrin- targeting ligands.
  • the particles due to the high density of ligands, have a high avidity for integrins on cell surfaces, bind and are internalised by a phagocytic process (Hart et al . , 1994).
  • the vesicles fuse to form endosomes where, under acid conditions, the DOPE element contained within the particles mediates destabilisation of the endosomal membrane and subsequent plasmid release into the cytoplasm. Phagocytosed particles lacking lipofectin are degraded in the endosomes. Particles lacking the integrin-targeting moiety are less efficient at cell binding and internalisation.
  • the present invention also relates to the use of the modified spacer element of the integrin-binding component in another transfection complex.
  • W096/15811 described a transfection complex that comprises an integrin-binding component, a nucleic acid binding component and a nucleic acid, that is to say, it describes a complex analogous to the LID comlex described above, but lacking the lipid component.
  • the inclusion of the lipid component greatly improves the transfection efficiency.
  • there maybe circumstances when the increased levels of transfection that result from the use of an improved spacer element as described herein may give transfection levels that are sufficient even in the absence of the lipid component.
  • the present invention also provides a complex that comprises
  • nucleic acid especially a nucleic acid encoding a sequence of interest
  • a polycationic nucleic acid-binding component in which complex the integrin binding component comprises an integrin-binding element and a spacer element, the spacer element being longer and/or more hydrophobic than the dipeptide spacers GG (glycine-glycine) and GA (glycine-alanine) .
  • the various components of the complex are as described above for the lipid-containing complex.
  • the invention also provides all the various other embodiments described above in relation to the lipid containing complex when modified by the omission of the lipid component.
  • transfection efficiency of confluent and other slowly dividing and non-dividing cells is very low, particularly when receptor targeted vectors are used.
  • transfection of confluent and other slowly dividing and non-dividing cells that are in contact with each other using an integrin-targeted vector may be greatly increased when the cells are also treated with an agent that disrupts cell-cell junctions.
  • transfection efficiency of airway epithelial cells in vi tro using two different reporter genes was increased about four-fold, and in vivo transfection of mouse lungs was also increased about four-fold.
  • the observation is particularly surprising in view of the fact that calcium ions have previously been shown to be important for transfection using polycationic vectors and, furthermore, the fact that the cells were not mitotic.
  • the use of the calcium-chelating agent EGTA would have been predicted, on the basis of the observations of Haberland, to reduce transfection efficiency, not to increase it.
  • the problem of access of the introduced nucleic acid to the nucleus still remains.
  • the major limiting step in transfection of non-mitotic cells is considered to be delivery of the transfected nucleic acid into the nucleus.
  • any of the embodiments of the present invention may be carried out in the presence of an agent that disrupts cell-cell junctions, that is to say, the transfection step involved in those embodiments may be carried out in the presence of an agent that disrupts cell-cell junctions.
  • the present invention also provides a method of transfecting confluent cells or other slowly dividing cells or non-dividing cells that are in contact with each other, that is to say, cells that are substantially non-mitotic, with a nucleic acid, which method comprises treating the cells with a complex of the present invention and with an agent that disrupts cell-cell junctions.
  • the confluent cells or other slowly dividing cells or non- dividing cells that are in contact with each other may be in the form of a culture, for example, a confluent culture in vi tro. Methods for producing such cultures are well known, and any cells that can be grown to confluence in vi tro may be transfected according to the present invention.
  • confluent cells or other slowly dividing cells or non-dividing cells that are in contact with each other may be transfected in vivo .
  • a particular example of a tissue that comprises confluent, differentiated and hence substantially non-dividing cells and that has proved recalcitrant to transfection is the airway epithelium, a target for gene therapy for cystic fibrosis and asthma.
  • Other epithelial and endothelial tissues are also particularly suitable targets for transfection according to the present invention.
  • the cell-cell junctions that are to be disrupted in the method of the present invention are junctions between adjacent cells.
  • the exact types of junction may vary from tissue to tissue to cell type to cell type, and the junctions include gap junctions and tight junctions.
  • the tight junction, found in epithelia, is also known as zonula occuldens.
  • the cell-cell junctions to be disrupted according to the present invention are not limited to junctions in any specific tissue or between any particular types of cells. Examples of junctions that may be disrupted include those found in certain types of tissues in vivo, for example, epithelial and endothelial tissues.
  • Agents that are capable of disrupting cell-cell junctions are known, for example, calcium-chelating agents and calcium-binding agents, for example, EDTA (ethylenediaminetetra-actetic acid) or, especially, EGTA (ethylene glycol-bis ( ⁇ -aminoethyl ether) - N,N,N' ,N' -tetraacetic acid) (Schmid RW & Reilly CN, Analytical Chemistry 29, 264, 1957) .
  • EGTA is generally preferred as it is more specific for calcium than is EDTA. It is also better tolerated by cells, both in vi tro and in vivo.
  • a further approach is the use of antibodies to substances involved in cell-cell adhesion, for example, at gap junctions or tight junctions, for example, cadherins.
  • Antibodies, especially monoclonal antibodies, to such substances, for example, anti-cadherins, may be used as an agent capable of disrupting cell-cell junctions.
  • the cells are treated with the vector of choice in the usual manner for transfection using that vector.
  • the agent that disrupts cell-cell junctions may be used before the cells are treated with the vector but it is preferably to treat the cells with the agent and the vector at the same time or substantially the same time.
  • the agent that disrupts cell-cell junctions is used in amount effective to disrupt the junctions.
  • the concentration of EGTA is about ImM or less, for example, from about 0.5 mM to 1 mM. Higher concentrations may be used, but care should be taken with regard to toxicity. A concentration of about 1 mM is generally preferred for use in vi tro.
  • the concentration of EGTA may be about 25 mM to 200 mM, for example, 100 mM. Concentrations greater than 200 mM may be used, but again care must be taken with regard to toxicity at high concentrations, for example, 400 mM may be lethal. Concentrations of about 100 mM are generally preferred.
  • the cells may be transfected in vi tro or in vivo .
  • Transfection in vi tro is particularly useful for transfecting non-dividing cells with genes or anti-sense DNA of interest.
  • Such cells may be harvested for use, for example, for administration to a patient or for use for protein production.
  • such cells may be used in their confluent state, for example, in si tu, as disease models for drug testing.
  • confluent cells which are generally non-dividing and may be differentiated, are often better models of tissues that are sub-confluent cells.
  • Cells may be grown to confluence in microwells, or using more sophisticated systems, for example, at an air-liquid interface.
  • Various systems for growing confluent cells for use as models for drug testing are being developed. The ability to transfect cells in such systems is a great advantage .
  • Confluent cells and other slowly dividing or non-dividing cells that are in contact with each other, including differentiated cells, may be transfected in vivo .
  • the present invention provides such a method of transfection and also provides the use of an agent that disrupts cell-cell junctions and a receptor targeted vector in the manufacture of a medicament for the transfection of cells, especially confluent, or other slowly dividing or non-dividing cells that are in contact with each other, for example, substantially non-mitotic cells.
  • Such cells include, for example, endothelial or epithelial cells, for example, cells of the any part of the airway epithelium, including bronchial and lung epithelium, and the corneal endothelium.
  • the airway epithelium is an important target for gene therapy for cystic fibrosis and asthma.
  • FIG. 1 shows the effect of different amounts of lipofectin (DOTMA:DOPE) on the enhancement of transfection of ECV304 cells using a complex consisting of lipofectin, oligolysine-peptide 1 ( [K] isGACRGDMFGCA [SEQ. ID. O. : 19] ) and plasmid pGL2.
  • DOTMA:DOPE lipofectin
  • Figure 2 shows the effect of different amounts of lipofectin on the enhancement of transfection of A375M, COS-7 and ECV-40 cells using a complex consisting of lipofectin, oligolysine- peptide 1 ( [K] leGACRGDMFGCA) and plasmid pGL2.
  • Figure 3 shows the effect of the order of mixing the components of a complex consisting of lipofectin (L) , oligolysine-peptide 1 ( [K] ⁇ 6 GACRGDMFGCA) [SEQ. ID .NO .: 31] (I) and plasmid pGL2 (D) on the enhancement of transfection of ECV40 cells.
  • Figure 4 shows a comparison of enhancement of transfection by lipofectin of complexes containing plasmid pGL2 and oligolysine-peptide 1 ( [K] isGACRGDMFGCA, [SEQ . ID.NO .: 31] pep 1), or oligolysinepeptide 5 ( [K] ⁇ 6 GACDCRGDCFCA [SEQ. ID. O. : 34] , pep 5), or oligolysine-peptide 6 ( [K] ⁇ 6 GACRRETAWACG [SEQ. ID.NO.
  • Figure 5 shows the dose-dependency of a complex containing lipofectin, oligolysine-peptide 6 ( [K] ⁇ 6 GACRRETAWACG) [SEQ. ID.NO. : 12] and plasmid pGL2 on the availability of integrin-binding ligands.
  • Figure 6 shows the structure of various complexes, as determined using atomic force microscopy, the complexes being formed with different combinations of plasmid DNA (plasmid pGL2) , oligolysine-peptide ( [K] ⁇ 6 -peptide 6) and lipofectin as follows: A: [K] i6-peptide 6 and plasmid pGL2 ; B: [K] ⁇ 6 -peptide 6, lipofectin and plasmid pGL2; C: lipofectin and plasmid pGL2, optimal ratio; D: lipofectin and plasmid pGL2 , suboptimal ratio.
  • Figure 7 shows levels of expression of IL-12 48 hours after transfection of COS-7 cells and neuroblastoma cells lines IMR- 32, KELLY and SHSY-5Y with a complex containing lipofectin, oligolysine-peptide 6 ( [K] ⁇ 6 GACRRETAWACG) [SEQ. ID. NO. : 35] and either two retroviral plasmid constructs encoding the two domains of IL-12 (MFGS-IL12) or one plasmid containing a fusion gene, Flexi-12 under a CMV promoter.
  • Figure 8 shows the effect of transfection with anti-sense oligonucleotides (AS) to the thrombin receptor (PAR-1) on thrombin induced proliferation of human foetal lung fibroblasts (HFL-1 cells) .
  • AS anti-sense oligonucleotides
  • PAR-1 thrombin receptor
  • Figure 9 shows the effect of transfection of haematopoietic cell lines HL60, PLB985, TF1 and U937 with LID complexes containing lipofectin, the reporter gene pEGFP-Nl and either [K] ⁇ 6 - peptide 6 (pep 6) or [K] ⁇ 6 -peptide 8 (GGCRGDMFGCA
  • Figure 10 shows the effect of the longer, highly hydrophobic spacer XSXGA [SEQ. ID.NO. : 18] (Pepl2) on transfection of primary porcine smooth muscle cells (VSMCs) compared with the same integrin-binding peptide having a GA spacer (Pep6) .
  • TFI cells are shown as shaded columns, Jurkat cells as solid columns.
  • Figure 11 shows the effect of the longer, highly hydrophobic spacer XSXGA [SEQ. ID.NO. : 18] (Pepl2) on transfection of Jurkat and TFI cells compared with the same integrin-binding peptide having a GA spacer (Pep6) .
  • the black columns show the results with Jurkat cells, the shaded columns the results with TFI cells.
  • Figure 12 shows flow cytometry analysis on the transfection efficiency with pEGFP in subconfluent (A, B) and confluent (C, D) IHAEo- cells (A, C) and 2CFSMEo- cells (B, D) . High efficiency was observed in subconfluent cells but it dropped dramatically when the cells became confluent.
  • Figure 13 shows luminometric assay of the effects of EGTA (A, B) and anti-E-cadherin (C, D) on transfection.
  • the confluent (A) and subconfluent (B) IHAEo- cells were transfected with LID complexes after pre-treatment with EGTA (EGTA) or in the presence of EGTA (LID+EGTA) .
  • Significant enhancement effects were observed in confluent but not subconfluent cells with LID vector incorporated with EGTA (LID+EGTA) compared with either the EGTA pretreatment (EGTA) or the control transfection without EGTA (OptiMEM) .
  • Figure 14 shows flow cytometry analysis of EGTA effects on transfection efficiency (A-C) and the rate of BrdU incorporation (D-F) in confluent IHAEo- cells.
  • Cells were transfected with pEGFP (A) or pEGFP plus EGTA (B) , or pEGFP plus EGTA and aphidicolin (C) .
  • cells were labelled with BrdU (D) or BrdU plus EGTA (E) , or BrdU plus EGTA and aphidicolin (F) .
  • D BrdU
  • E BrdU plus EGTA
  • F BrdU plus EGTA and aphidicolin
  • EGTA increased the efficiency to 31.7% (B) and this enhancement effect was still observed in the presence aphidicolin where the EGFP-positive cells accounted for 23.6% (C) . Meanwhile, EGTA showed no effects on cell proliferation with the rate of BrdU incorporation being 7.1% in confluent cells (D) , 6.5% in the presence of EGTA (E) , and 5.3% in the presence of EGTA and aphidicolin (F) .
  • Figure 15 shows sub-confluent IHAEo- cells: flow cytometry analysis of EGTA effects on transfection efficiency (A-C) and the rate of BrdU incorporation (D-F) .
  • Cells were treated in the same way as in the confluent cells shown in Figure 13.
  • Transfection efficiency was as high as 44.5% in sub-confluent cells (A) .
  • Similar efficiency of 45.6% was observed in the presence of EGTA (B) and there were still 24.8% cells being transfected in the presence of aphidicolin (C) .
  • EGTA decreased BrdU incorporation rate in sub-confluent cells as the proportion of BrdU-positive cells dropped from 43.0% (D) to about 23.9% after EGTA treatment (E) . 7.8% cells were labelled with BrdU in the presence of aphidicolin (F) .
  • Figure 16 shows double immunofluorescence for transgene ⁇ - Galactosidase (transgene expression shown by green cytoplasmic staining, irregular shapes) and BrdU labelling (shown by red nuclear staining, smaller rounder shapes) in subconfluent (A) and confluent (B, C) IHAEo- cells. Most of the subconfluent cells were labelled with BrdU and many of them expressed transgene simultaneously (irregular shape containing rounded shape) , shown in Figure 15A. Only few confluent cells were positive for ⁇ -galactosidase while few were labelled with BrdU (B) . After EGTA treatment many more cells expressed transgene see Figure 15C while their proliferation state was not affected as evident by the unchanged rate of BrdU labelling, few cells were stained red.
  • Figure 17 shows photomicrographs of confluent IHAEo- cells showing the effects of EGTA treatment.
  • the images in left-hand column (A, C, E, G) shows the cells before EGTA treatment and the right-hand column (B, D, F, H) are the cells after EGTA treatment.
  • the cell line COS-7 (monkey kidney epithelial cells) were maintained in Dulbecco's Modified Eagle Medium (DMEM; Life Technologies, Paisley, U.K.) supplemented with 10 % foetal calf serum (FCS) , L-glutamine, penicillin and streptomycin.
  • ECV304 spontaneously transformed human umbilical vein endothelial cells
  • HT1080 fibrosarcoma cells and A375M melanoma cells were maintained in DMEM and 10% FCS.
  • IMR2 neuroblastoma cells were grown in DMEM F12 Nutrient Mix (Life technologies) .
  • VSMCs Porcine primary smooth muscle cells
  • heamatopoietic cells Jurkat and TFI
  • airway epithelial cells were grown in media suitable for those cell types, for example, human airway eithelia (HAE) cells type HAEO-, were grown in Modified Eagle Medium (MEM) obtained from Life Technologies (Paisley, Scotland) supplemented with 10% foetal calf serum and 2 mM L-glutamine.
  • MEM Modified Eagle Medium
  • the sequence of peptide 6, GACRRETAWACG [SEQ. ID.NO. : 12] was based on an ⁇ 5 ⁇ l-specific peptide from a phage display library (Koivunen et al . , 1995).
  • the oligolysine-peptide was based on an ⁇ 5 ⁇ l-specific peptide from a phage display library (Koivunen et al . , 1995).
  • 9-fluorenylmethyloxy-carbonyl was used for temporary ⁇ -amino group protection.
  • Side-chain protecting groups were tert-butyloxycarbonyl for Lys and Trp, trityl for Cys, 2,2,5, 7, 8-pentamethylchroman-6sulphonyl for Arg, tert-butylester for Glu and tert-butyl ether for Thr.
  • Cleavage from the resin and deprotection of the peptide was achieved by treating the peptidyl-resin with 10 ml of a mixture containing 10 ml trifluoroacetic acid, 0.25 ml ethanedithiol, 0.25 ml triisopropylsilane at 20°C for two hours.
  • the peptide was precipitated using ice-cold diethylether and then filtered through a fine sintered glass filter funnel under light vacuum.
  • the peptide precipitate was dissolved in 10% acetic acid/water solution and freeze dried.
  • the crude peptide was analysed by reverse phase HPLC and matrix assisted laser desorption ionisation time of flight mass spectroscopy.
  • Oligolysine-peptide 1 [K] ⁇ 6 GACRGDMFGCA [SEQ. ID.NO. : 31] and oligolysine-peptide 5: [K] ⁇ 6 GACDCRGDCFCA [SEQ. ID.NO. :34] were obtained from Zinsser Analytic (Maidenhead, U.K.) .
  • Oligolysine-peptide 66 [K] ⁇ 6 GACATRWARECG [SEQ. ID.NO. :28] and oligolysine-peptide 12: [K] ⁇ 6 XSXGACRETAWACG [SEQ. ID.NO. :29] may be synthesised as described above for oligolysine-peptide 6 or obtained from Zinsser Analytic.
  • Plasmid DNA The plasmids pGL2 , which contains a luciferase reporter gene (Promega, Madison, WI, U.S.A.) and pCMV ⁇ , which contains a ⁇ - galactosidase reporter gene (Clontech, Palo Alto, California, U.S.A.) were grown in Escherichia coli DH5a and purified, after bacterial alkaline lysis, on Qiagen resin columns (Qiagen Ltd., Crawley, U.K.) by the manufacturer's instructions.
  • luciferase reporter gene Promega, Madison, WI, U.S.A.
  • pCMV ⁇ which contains a ⁇ - galactosidase reporter gene
  • Isopropanol-precipitated DNA pellets were washed with 70% ethanol then dissolved in water or TE buffer (10 mM Tris-Cl, pH 8.0 and 1 mM EDTA) .
  • Plasmid solutions were adjusted to a concentration of 1 mg/ml and stored at 4°C.
  • lipofectin an equimolar mixture of the cationic lipid N- [1- (2, 3-dioleyloxy) propyl] -N,N,N-tri- methylammonium chloride (DOTMA) and the neutral lipid dioleoyl phosphatidylethanolamine (DOPE) , obtained as "Lipofectin” from Life Technologies, Paisley, U.K.) (1 mg /ml), pGL2-control (1 mg/lOO ml) and [K] ⁇ 6 /integrin-targeting peptide 1, 5 or 6 (0.1 mg/ml) .
  • DOTMA cationic lipid N- [1- (2, 3-dioleyloxy) propyl] -N,N,N-tri- methylammonium chloride
  • DOPE neutral lipid dioleoyl phosphatidylethanolamine
  • oligolysine- peptide (I) oligolysine- peptide
  • D plasmid DNA or RNA
  • L lipofectin
  • the mixture LID was made in the same way in the optimal weight ratio 0.75:4:1 (L:I:D). Both types of mixture were left to aggregate for at least 30 min then diluted to a concentration of one microgram DNA per 0.5 ml with OptiMEM.
  • the growth medium was removed from each well then 0.5 ml of transfection complex added. The plate was then returned to the incubator for four to six hours. The transfection medium was then removed and replaced with 1 ml of complete growth-medium. Transfected cells were incubated for 48 to 72 hours then assayed for reporter gene activity.
  • the protein concentration of each sample was then determined with Protein Assay Reagent (BioRad, Hercules, CA, U.S.A.) and luciferase enzyme activity expressed in terms of relative light units per milligram of protein (RLU/mg) .
  • LacZ assays ⁇ -galactosidase activity was detected by staining with X-gal . After washing with PBS cells were fixed to the plastic plates by addition of 0.5% glutaraldehyde in PBS for 20 minutes at 40°C. Wells were washed with PBS and cells were stained with 5 X-gal at 370 C for up to six hours.
  • Transfection complexes were prepared as described above in the Materials & Methods section. The complexes were made by
  • 25 pGL2-control plasmid DNA (0.1 mg/ml) was added and mixed by repeated pipetting.
  • the ratio of mixing of each component was a constant 4 ⁇ g of oligolysine-peptide per ⁇ g of DNA, while the proportion of lipofectin varied from 1 to 10 ⁇ g per ⁇ g of DNA.
  • ECV304 cells were transfected with the complexes as described
  • Example 2 Effect of different amounts of lipofectin on transformation in three different cell lines
  • Lipofectin/- oligolysine-peptide/DNA complexes were prepared with constant amounts of [K] ⁇ 6 -peptide 1 ( [K] X6 GACRGDMFGCA) [SEQ. ID.NO. : 31] (4 ⁇ g) and pGL2 (1 ⁇ g) plasmid DNA and a range of lipofectin amounts (1 to 2.5 micrograms). Complexes were used to transfect
  • a mixing ratio L:I:D of 0.75:4:1 by weight corresponds to a molar ratio of 0.5 nmol lipofectin: 1.25 nmol oligolysine- peptide 1: 0.25 pmol pGL2-control .
  • the molar charge of each component is 0.5 moles positive charge per mole lipofectin, seventeen moles positive charge per mole [K] ⁇ 6 -peptide 1 and 12,000 moles negative charge per mole of pGL2 (6 kb) .
  • 3 nmol of negative charge from the plasmid is mixed with 21 nmol of positive charge from oligolysine-peptide 1 and 0.25 nmol positive charge from lipofectin.
  • Example 3 Effect of the order in which the components of the complex are mixed.
  • transfections were performed with complexes made by adding the components of the complexes in different orders. All combinations were prepared with the same amounts and concentrations of the components (1 ⁇ g pGL2 plasmid DNA, 0.75 ⁇ g of lipofectin and 4 ⁇ g of oligolysine- peptide 1 ( [K] ⁇ 6 GACRGDMFGCA) . Transfections were performed in ECV304 cells and luciferase activity was assessed as described above .
  • Example 4 Transfection rates Cells were transfected with optimised oligolysine-peptide/lipo- fectin/pCMV ⁇ complexes as described in Examples 1 and 2 prepared in the order of mixing LID but using pCMV ⁇ as the plasmid vector (component D) instead of pGL2. The cells were stained for ⁇ -galactosidase activity with X-gal as described above. A number of cell types, A375M, COS-7 and ECV304 displayed transfection efficiencies of 50 to 100% compared to 1 to 10% achieved with oligolysine-peptide/DNA complexes alone. This represents a very significant improvement in transfection efficiency.
  • oligolysine-peptide 1 [K] 16 GACRGDMFGCA, pep 1) [SEQ. ID.NO. : 31]
  • oligolysine-peptide 5 [K] ⁇ 6 GACDCRGDCFCA, pep 5) [SEQ. ID.NO. -.34]
  • oligolysine-peptide 6 [K] ⁇ 6 GACRRETAWACG, pep 6) [SEQ. ID.NO. :35, and [K] 16 [SEQ. ID.NO. : 30] .
  • One set of complexes also contained lipofectin (lip) , the other was without lipofectin.
  • lipofectin lipofectin
  • LID complexes were prepared with constant amounts of plasmid pGL2-control and lipofectin, and a range of combinations of [K] ⁇ 6 -peptide 6 and [K]i6 « A total of 40 ⁇ g of [K] ⁇ 6 -peptide was used, consisting of 1, 5, 10, 20, 35, 39 ⁇ g of [K] 16 -peptide 6 made up to 40 ⁇ g with [K] 1 6.
  • Transfections were performed as described in Example 1 and luciferase assays performed after 48 hours. The results are shown in Figure 5. Transfection efficiency demonstrated an apparently exponential increase with increasing amounts of oligolysine-peptide 6, and, therefore, a dose-dependent response to the amount of available integrin-binding ligands. Accordingly, while both the sixteen-lysine domain, and the lipofectin components are themselves capable of mediating transfection, both individually and in [K] i6/lipofectin combination complexes, the highest efficiency transfection is directly proportional to the amount of available integrin-binding ligand.
  • Atomic force microscopy experiments were performed to determine and compare the structures formed by mixing 4 ⁇ g [K] ⁇ 6 peptide 6 and 1 ⁇ g pGL2-control plasmid DNA (ID complexes) .
  • LID complexes were formed from [K] ⁇ 6 -peptide 6 (4 ⁇ g) /lipofectin (0.75 ⁇ g) /DNA (1 ⁇ g) in the order LID which was shown to yield optimal transfection results.
  • Lipofectin/DNA complexes (LD) were formed at two different ratios; an optimal transfection ratio of 5 ⁇ g lipofectin per microgram of pGL2 and the same ratio as used in LID complexes, 0.75 ⁇ g lipofectin per microgram of plasmid.
  • ID complexes composed of oligolysine-peptide 6 and plasmid DNA, were examined initially by AFM within fifteen minutes of mixing the two components.
  • the complexes formed particles of low polydispersity which, on the mica coverslips, had a diameter of approximately 200 nm.
  • a computer-generated contour map revealed that the particles formed were of irregular conical shape.
  • LID complexes assessed by AFM formed particles of a similar size and shape to ID complexes. The additional lipofectin did not, apparently, disrupt the particles .
  • LD complexes formed at the lower ratio (0.75:1) appeared to be short tubular structures. LD complexes formed at this lower ratio were inactive in transfection experiments.
  • LID complexes formed as above were also analysed by AFM after standing overnight. Particles were now smaller in size with diameters of approximately 50-100 nm suggesting that the particles had compacted. Computer-generated computer maps represented these particles as regular conical structures. The cones were measured and their volumes were calculated. The spheres which the particles are predicted to form when free in solution were then calculated to be 20 to 60 nm in diameter. In transfection experiments with pGL2 the compact particles formed overnight in water yielded luciferase expression results approximately twice as high as the freshly made complexes .
  • the three human neuroblastoma cell lines and COS-7 cells were then transfected using the same LID complex with, instead of the reporter gene, one of two different IL-12 expressing vectors.
  • One vector expresses a fusion protein of the two chains of IL-12, p35 and p40, (Flexi-12; Anderson et al . 1997) This fusion is regulated by a CMV promoter.
  • the second IL-12 expression system consists of two retroviral constructs MFGS- 35 and MFGS-p40, which are retroviral plasmid constructs encoding the two separate chains of interleukin-12 (IL-12) . Both genes are regulated by the retroviral long terminal repeats (LTRs) .
  • the vectors were obtained from Professor Mary Collins, UCL, London.
  • transfection system of the present invention is suitable for use in a vaccine for neuroblastoma, an important childhood malignancy, and also for vaccines against other cancers .
  • Tissue cultures of rat primary smooth muscle cells (aortic smooth muscle cells) and cardiac myocytes were prepared according to standard methods (Blank et al . 1988) .
  • An LID complex comprising lipofectin, [K] ⁇ 6 -peptide 6 and GFP as a reporter gene in the optimal LID ratio and mixing order was prepared as described in the Materials and Methods section and the Examples above.
  • the tissue cultures were transfected with the LID complex as described in the Material and Methods section above. Fluorescing imaging of GFP-expressing cells demonstrates transfection efficiency in excess of 50%.
  • transfection complex of the present invention achieved transfection efficiencies in excess of 50%, thus demonstrating the utility of the complexes for treatment of diseases affecting muscle, including smooth muscle and cardiac muscle.
  • transfection complex of the present invention Different size constructs can be delivered with the transfection complex of the present invention.
  • a fibroblast culture was transfected as described in the Materials and Methods section with an LID complex comprising [K] ⁇ 6 -peptide 6, lipofectin and a 130 kB DNA construct.
  • the complex comprising the LID components in the optimal ratio and mixing order, was prepared as described in the Methods and Materials section and Examples above. Transfection was achieved with 2-3% efficiency.
  • Cellular process associated with the enhanced integrin-mediated internalisation of DNA using a complex of the present invention are more closely related to phagocytosis than endocytosis and are thus particularly suited to the delivery of complexes containing very large DNA molecules.
  • Thrombin stimulates proliferation of human lung fibroblasts.
  • Thrombin-treated human lung fibroblasts proli- ferated 53% in response to thrombin.
  • HFL-1 cells 24 hours before treatment with thrombin, HFL-1 cells were treated with an LID complex comprising [K] ⁇ 6 -peptide 6, lipofectin and a 20-mer antisense oligonucleotide directed against the thrombin receptor PAR-1 in the optimal ratio and mixing order prepared as described in the Materials and Methods section and the Examples above.
  • the antisense oligonucleotide-containing complex was in contact with the cells for 4 hours. 24 hours after the start of the treatment with the complex, treatment with thrombin was carried out .
  • the thrombin-induced proliferation was attenuated by 76% +/- 12% by the pre-treatment with the LID complex. Cells treated with the antisense-containing complex but not with thrombin did not proliferate, see Figure 8.
  • Haematopoietic cells are particularly resistant to transfection with most plasmid-mediated vectors .
  • LID complexes were prepared as described in the Material and Methods section and Examples above using lipofectin and [K] ⁇ 6 - peptide 6, which targets ⁇ 5 ⁇ l integrins, and pEGFP-Nl (Promega) as reporter gene.
  • Complexes were prepared analogously substituting [K] ⁇ e-peptide 8 ( [K] ⁇ 6 GACQIDSPCA [SEQ. ID.NO. : 37] , which targets ⁇ 4 ⁇ l integrins, for [K] ⁇ 6 -peptide 6.
  • the complexes were prepared by mixing the components in the optimal ratio and mixing order as described in the Materials and Methods section and Examples above.
  • Oligolysine-peptide 6 [K] 16 GACRRETAWACG [SEQ. ID.NO. : 32]
  • Oligolysine-peptide 66 [K] 16 GACATRWARECG [SEQ. ID.NO. :28] Oligolysine-peptide 12: [K] ⁇ 6 XSXGACRETAWACG [SEQ. ID.NO. :29]
  • Peptides 6 and 12 contain the same K16 and ⁇ 5 ⁇ l integrin- targeting element (CRRETAWACG) [SEQ. ID.NO. : 23] .
  • the difference between the two is that peptide 6 has the dipeptide spacer GA whereas peptide 12 has the longer, highly hydrophobic spacer XSXGA, where X is ⁇ -aminohexanoic acid.
  • Peptide 66 is a non- integrin-binding control, which contains the oligolysine element followed by the same amino acids as in peptides 6 and 12, but in a scrambled order. [K] ⁇ e is the oligolysine nucleic acid binding element .
  • oligolysine-peptides were used to form a transfection complex as described in the Materials and Methods section. Luciferase was the reporter gene.
  • porcine primary smooth muscle cells VSMCs
  • haematopoietic cells Jurkat and TFI
  • airway epithelial cells were tested for transfection using the various complexes according to the protocols in the Material and Methods section.
  • peptide 12 which has the longer, highly hydrophobic spacer, performed better than peptide 6, with the GA spacer.
  • the results obtained with the porcine primary smooth muscle cells are presented in Figure 10, The results with Jurkat and TFI cells in Figure 11.
  • the results obtained with the primary smooth musce cells are particularly striking, showing an increase in transfection of about 100%.
  • Example 14 Transfection of confluent human airway epithelial cell in vi tro in the presence of EGTA
  • HAE Human airway epithelial cells
  • type HEAO- were seeded at 80% into 24 well plates.
  • the cells were grown in Modified Eagle Medium (MEM) obtained from Life Technologies (Paisley, Scotland) supplemented with 10% foetal calf serum and 2 mM L- glutamine to form cultures of confluent cells. Subconfluent cultures of the same cells were also prepared.
  • MEM Modified Eagle Medium
  • the reporter genes used were a green fluorescent protein (GFP) reporter gene and a luciferase reporter gene.
  • the GFP reporter gene was used in the form of plasmid pEGFP-Nl, obtained from Clontech.
  • the luciferase reporter gene was used in the form of construct pCILuc consisting of the firefly luciferase gene (Luc) inserted into the expresssion vector pCI (Promega, Southampton, England)
  • the oligolysine-peptide component I comprised the integrin-targeted peptide CRRETAWACG, which targets the integrin ⁇ ,5 receptor, with a spacer XSXGA (X: g amino hexanoic acid) , and the oligolysine [K] iS , giving the sequence [K] ⁇ 6 XSXGACRRETAWACG (Zinsser Analytic, Maidenhead, UK) , see Example Transfection vector/EGTA complexes
  • Transfection vector complexes were prepared as described in the Material and Methods section, see also Example 15. The complexes were prepared using 4 ⁇ g of the peptide, 0.75 ⁇ g of lipofectin and 1 ⁇ g of the reporter gene. All components and the vector complex were made up in OptiMEM (Life Technologies, Inc.) . EGTA (Sigma) at a concentration of 100 mM in water was added to the transfection vector complex preparation at one hundredth of the volume of the vector complex, giving a final EGTA concentration of 1 mM.
  • the EGTA/vector complex mixture was added to the cultures, which were then harvested two days later. As controls, cultures were treated with OptiMEM medium alone and with the transfection vector complex without EGTA.
  • transfection was carried out in the presence of aphidicolin, which inhibits DNA polymerase- ⁇ , which prevents transition of the cells from Gl to S phase.
  • aphidicolin which inhibits DNA polymerase- ⁇ , which prevents transition of the cells from Gl to S phase.
  • Aphidicolin Sigma, Poole, Dorset, England
  • Transfection levels using the luciferase reporter gene was measured as described in the Material and methods section.
  • the GFP reporter gene after harvesting the cells were analysed by fluorescence activated cell sorting (FACS) for quantification of the number of fluorescent cells.
  • Figure 10 shows clearly the influence of EGTA in the increase in the number of fluorescent cells transfected with the vector in the presence of EGTA (Figure 10B) compared with the vector alone, as shown in ( Figure 10A) .
  • Bromodeoxyuridine (BrdU) labelling enables assessment of cell proliferation.
  • BrdU kits are available from Zymed
  • BrdU was added to the cells at the same time as the transfection reagent and was maintained throughout the rest of the experiment until cell harvesting.
  • the results of BrdU labelling of sub-confluent and confluent HAE cells is given in Table 3 below.
  • Airway epi thelial cells Human airway epithelial cell lines, a normal cell line lHAEo- and a CF cell line 2CFSMEO-, were used in this study.
  • 2CFS Meo- cells are immortalised, SV-40 T antigen transformed cells obtained from Dr D. Gruenert, UCSF.
  • the IHAEo- cells retain the morphologic and functional characteristics of epithelial cells and have been used in many different studies, see (Boussat et al (2000) , Cozens et al (1992a) .
  • the 2CFSMEo- cells are ⁇ F508 heterozygous submucosal epithelial cells, see (Canonico et al (1996) , Cozens et al (1996) .
  • Both cell lines were maintained in a humidified atmosphere of 95% air and 5% C0 2 at 37°C in Eagle's minimum essential medium (MEM) supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin (Sigma, Poole, UK) in 75cm 2 culture flasks.
  • MEM Eagle's minimum essential medium
  • Cells were seeded on transwell inserts of 0.4 ⁇ m pore size and 12 mm diameter (Costar, Bucks, UK) at a density of 10 5 cells/0.5ml/insert and were left to grow until fully confluent in about 3 days . The apical medium was then removed and growth medium was added basolateraly to leave the cells growing at an air-liquid interface. Confluent cells were grown for further 10-14 days and were monitored with an ohmmeter (EVOM; World Precision Instruments, Stevenage, UK) until the transepithelial resistance was greater than 300 ⁇ -cm before transfection.
  • EVOM World Precision Instruments, Stevenage, UK
  • the cells were seeded on coverslips in 24-well plates at a density of 10 5 cells/ml/well and were left to grow for about 10-14 days until fully confluent .
  • the state of confluence and development of tight junctions were also determined by the immunofluorescent detection of occludin, the structural and functional component of tight junctions (detailed below) .
  • Sub-confluent, proliferating cells were prepared by growing the cells for less then 3 days in growth medium prior to transfection.
  • Plasmid pCI-Lux was prepared by subcloning a luciferase gene from pGL3 control (Life Technologies, Paisley, UK) into the eukaryotic expression vector pCI (Promega, Southampton, UK) .
  • Plasmid pCIK-LacZ containing cytoplasm-located bacterial ⁇ - Galactosidase gene, was obtained from Dr. Steve Hyde (Oxford) .
  • the plasmid pEGFP was commercially available (CLONTECH Laboratories UK Ltd. , Hampshire, UK) . All of these genes were driven by CMV promoter.
  • the plasmids were amplified in
  • the DNA was washed with 70% ethanol and dissolved in water.
  • the concentration of DNA was spectrophotometrically determined by its absorbance atA 2 6o and the purity was assessed with the ratio of A 2 6o/A 2 8o.
  • LID complexes and "LID vectors” hereafter, were prepared in a procedure similar to that described in Example 1. Briefly, stock solutions of the three components of the LID vector complex were prepared first. Plasmid DNA (component D) was diluted in cell culture medium OptiMEM (Life Technologies, Paisley, UK) at a concentration of 0.01 ⁇ g/ ⁇ l .
  • the oligolysine-peptide component I comprised the integrin-targeted peptide CRRETAWACG, which targets the integrin 5 receptor, with a spacer XSXGA (X: ⁇ amino hexanoic acid), and the oligolysine [K] ⁇ 6 , giving the sequence [K] l eXSXGACRRETAWACG (Zinsser Analytic, Maidenhead, UK).
  • the peptide component I was dissolved in Opti-MEM at 0.1 mg/ml.
  • DOTMA cationic lipid N- [l- (2 , 3 - dioleyloxy)propyl] -n,n,n-trimethylammonium chloride
  • DOPE neutral lipid dioleoyl phosphotid
  • the complexes were then left 30 minutes at room temperature before being diluted to a final concentration of l ⁇ g of D ⁇ A in 0.3 ml of Opti-MEM in transwells, or in 0.5 ml for 24-well plate.
  • the cells were incubated with the complexes for 4 hours at 37°C followed further culture in growth medium for 24 to 48 hours before being harvested for the assay of transgene expression.
  • Confluents cells were pre-incubated for 20-30 minutes at 37°C with 2 mM EGTA, 0.05 g of trypsin/litre-0.02 g/litre of EDTA, water, PBS or OptiMEM as control, respectively. Cells were then washed twice with OptiMEM and transfected with LID complexes or LID formulated with EGTA (detailed below) for 4 hours as described above.
  • confluent cells were pre-incubated with the antibody at increased concentrations of 2 , 10 and 50 ⁇ g/ml for 60 minutes at 4°C, followed by 4-hour transfection with LID complexes in the presence of anti-E-cadherin of same concentrations or mouse IgG as a control, respectively.
  • EGTA-formulation protocol LID+EGTA
  • OptiMEM contains 0.9 mM CaCl 2 which is of half the level of conventional mediums.
  • EGTA was first dissolved in PBS (pH7.3) to make a stock solution of 200 mM. After preparing the LID complexes, EGTA stock solution was added to the complexes to achieve a final concentration of 2 mM. The transfection incubation was performed in the presence of EGTA but was replaced with complete growth medium after 4 hours. Effects of EGTA on cell junctions were monitored by measuring the transepithelial resistance which dropped as early as about 10 minutes after exposure to EGTA and recovered at about 16-18 hours after removal of EGTA (data not shown) .
  • Luminometric assay for luciferase Twenty-four hours after transfection with pCI-luciferase, cells were washed with PBS, lysed by adding 100 ⁇ l of Reporter Lysis Buffer (Promega, Southampton, UK) and detached from the plate by manual scraping using micropipette tips. Cell-free lysates were prepared by centrifugation at 2000 rpm for 5 minutes at 4°C. The activity of luciferase was assayed with a luciferase assay kit (Promega, Southampton, UK) on a Lucy-1 plate-reading luminometer (Anthos, Salzburg, Austria) .
  • the protein concentration of each sample was determined with Protein Assay Reagent (BioRad Laboratories, Hertfordshire, UK) by measuring absorbance at 595 nm and the specific activity of luciferase was expressed as relative light unit per milligram protein (RLU/mg protein) .
  • Transfection efficiency by flow cytometry analysis of green fluorescent protein Cells transfected with pEGFP were washed twice with PBS, harvested by digesting with trypsin-EDTA and fixed in 4% paraformaldehyde . The cells were then analysed by fluorescence- activated cell sorting (FACS) with a FACS sorter EPICS XL (Beckman Coulter Inc, Buckinghamshire, UK) . Fluorescence for EGFP was measured at wavelength of 525+20 nm. Ten thousand cells were examined for the percentage of EGFP-positive cells by determining the percentage of highly fluorescent cells and subtracting the fluorescence of the untransfected control cells .
  • FACS fluorescence- activated cell sorting
  • Cell proliferation was assessed by incorporating 5-Bromo-2'- deoxy-uridine (BrdU) into cellular DNA detected by im unofluorescent staining using a monoclonal antibody.
  • a BrdU Labelling and Detection Kit (Roche, Basel, Switzerland) was used, which contains the BrdU labelling reagent and the monoclonal mouse anti-BrdU. While the confluent cells were transfected with EGTA-formulated LID containing pEGFP for analysis of transfection efficiency, a control group was transfected with the same protocol but with 10 ⁇ M BrdU in the transfection medium for analysis of cell proliferation.
  • aphidicolin a cell-proliferation inhibitor blocking the cell cycle in S-phase, was added to the cells to observe the effects on transfection efficiency. Aphidicolin was added to the growth medium (10 ⁇ g/ml) for 24 hours before transfection and maintained throughout the 4-hour transfection and the following 24 h period of incubation until the cells were harvested.
  • FITC fluorescein
  • Peptide-12 was labelled with FITC from a labelling kit (Calbiochem, San Diego, USA) and purified by dialyzing in PBS overnight, according to the manufacturer's instruction.
  • the ability of FITC-conjugated peptide-12 to form complexes with plasmid DNA was determined by retardation test, in which the electrophoresis migration of DNA is retarded by forming complexes with the cationic peptide as described previously (Hart et al . , 1995).
  • the confluent cells in transwells were first incubated with 2 mM EGTA in Opti-MEM for 30 minutes at 37°C to disrupt tight junctions and then cooled for 20 minutes at 4°C.
  • the LID complexes prepared with FITC- labelled peptide-12 were also cooled to 4°C for binding assay.
  • the cells were then transfected with apically added LID-FITC for 60 minutes at 4°C, at which the complexes would only bind to cell surface and no internalisation would occur, see(Chu et al (1999) , Cornelissen et al (1997) , Im et al (1986) .
  • the surface binding of the LID complexes was further differentiated from the internalised LID by incubating the cells with 1 ⁇ g/ml crystal violet (Sigma, Poole, UK) for 10 minutes at 4°C, to quench the fluorescence of surface-bound LID complexes, see (Ma et al (1997) , Van Amers & Van Strijp (1994) .
  • the mean numbers of fluorescent particles were then counted in 10 random microscope fields and comparison was made between the data obtained from EGTA- treated or untreated cells.
  • the cells were then incubated with a mixture of two primary antibodies, the rabbit anti- ⁇ -galactosidase at 1:200 (Chemicon International Inc, Harrow, UK) and the mouse anti- BrdU at 1:10 (BrdU Labelling and Detection Kit; Roche, Basel, Switzerland) , followed by incubation with a mixture of FITC-
  • the EGTA-treated cells were immunostained with a rabbit polyclonal antibody against tight junction protein occludin (Zymed, San Francisco, USA) diluted 1:10 with PBS.
  • the antibody is specific to the C-terminal 150 amino acids of human occludin and has been used in many other
  • the poor transfection efficiency of confluent cells could be due to 1) the lack of availability of receptors on basolateral surfaces, or 2) the low rate of cell proliferation and the
  • Cells were transfected with pEGFP (A) or pEGFP plus EGTA(B), or pEGFP plus EGTA and aphidicolin (C) .
  • cells were labelled with BrdU (D) or BrdU plus ECTA (E) , or BrdU plus EGTA and aphidicolin (F) .
  • LID transfection may involve increased cell binding of the vector to exposed receptors on the basolateral surfaces. This proposal was tested by investigating the binding of fluorescently-labelled LID complexes prepared with FITC-conjugated peptide. Incubation of
  • FIG. 16 shows photomicrographs of confluent IHAEo- cells showing the effects of EGTA treatment.
  • the images in left-hand column (A, C, E, G) shows the cells before EGTA treatment and the right-hand column (B, D, F, H) are the cells after EGTA treatment.
  • Confluent airway epithelial cells are almost entirely mitotically quiescent. As evident in this Example, the proliferating BrdU-labelled cells accounted for about 43% of subconfluent but only 7% in confluent cells. Reduced cell proliferation would result in a low efficiency of transfection as reported by Fasbender that cells in mitosis (BrdU positive) were much more likely to express transgene than BrdU-negative cells, see (Fasbender et al (1997) and, by Wilke that growth- arrested cells were less efficiently transfected, see (Wilke et al (1996) . The present study also showed the transfection was much more inefficient in confluent cells than in sub-confluent cells (8% vs. 44%).
  • EGTA was found to be highly effective for promoting access of the LID vector to basolateral receptors, enhancing transfection efficiency with low toxicity, see (Wang et al (1998) , Bals et al (1999) , Chu et al (1999) .
  • the results presented in this Example confirm that the efficient gene transfer to non-proliferating human airway epithelial cells using the synthetic nonviral vector LID, with adjuvant of a calcium chelator EGTA which transiently disrupted tight junctions to allow increased binding of LID complexes.
  • No induction of cells proliferation was observed by EGTA treatment and most of transgene-positive cells were non-proliferative. This shows the ability of LID vector to transfect non- proliferating cells and its use in gene therapy of human respiratory diseases.
  • Fernandez MA Muno-Fernandez MA, Fresno M. Involvement of ⁇ l integrins in the binding and entry of Trypanosoma cruzi into human macrophages. European J of Immunology 1993;23:552-557.
  • Kikuchi M Peptide ligands for integrin alpha v beta 3 selected from random phage display libraries. Biochemistry 1995;34:3948-3955.
  • junctional adhesion molecule a novel member of the immunoglobulin superfamily that distributes at intercellular junctions and modulates monocyte transmigration. J. Cell Biol . 25142: 117-127.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Cette invention concerne un complexe qui possède une activité de transfection accrue. Ce complexe comprend: i) un acide nucléique; ii) un composant qui se lie à l'intégrine; iii) un composant qui se lie à un acide nucléique polycationique, tel qu'une olygolysine; et iv) un composant lipidique tel qu'un liposome cationique dans lequel complexe le composant qui se lie à l'intégrine comprend un élément qui se lie à l'intégrine et un élément d'espacement, cet élément d'espacement étant plus long et/ou plus hydrophobe que les espaceurs dipeptidiques GG (glycine-glycine) et GA (glycine-alanine).
PCT/GB2001/002394 2000-05-30 2001-05-30 Vecteurs ciblant l'integrine avec activite de transfection accrue WO2001092542A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001260467A AU2001260467A1 (en) 2000-05-30 2001-05-30 Integrin-targeting vectors having enhanced transfection activity

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB0013090A GB0013090D0 (en) 2000-05-30 2000-05-30 Intergrin-targetting vectors having enhanced transfection activity
GB0013089.8 2000-05-30
GB0013089A GB0013089D0 (en) 2000-05-30 2000-05-30 Improved methods of transfection
GB0013090.6 2000-05-30
US28741001P 2001-05-01 2001-05-01
US60/287,410 2001-05-01

Publications (3)

Publication Number Publication Date
WO2001092542A2 true WO2001092542A2 (fr) 2001-12-06
WO2001092542A3 WO2001092542A3 (fr) 2003-05-30
WO2001092542A8 WO2001092542A8 (fr) 2003-12-24

Family

ID=27255741

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2001/002396 WO2001092543A2 (fr) 2000-05-30 2001-05-30 Procede ameliore de transfection
PCT/GB2001/002394 WO2001092542A2 (fr) 2000-05-30 2001-05-30 Vecteurs ciblant l'integrine avec activite de transfection accrue

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/GB2001/002396 WO2001092543A2 (fr) 2000-05-30 2001-05-30 Procede ameliore de transfection

Country Status (5)

Country Link
EP (1) EP1285081A2 (fr)
JP (1) JP2003534804A (fr)
AU (2) AU2001260467A1 (fr)
CA (1) CA2410419A1 (fr)
WO (2) WO2001092543A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051311A2 (fr) * 2001-12-17 2003-06-26 Bayer Corporation Anticorps inhibant l'activite du facteur de cellule souche et utilisation dans le traitement de l'asthme
FR2860236A1 (fr) * 2003-09-25 2005-04-01 Theraptosis Peptides possedant notamment une activite anti-angiogenique et leurs applications en therapeutique
US9255134B2 (en) 2008-01-24 2016-02-09 Esperance Pharmaceuticals, Inc. Lytic domain fusion constructs and methods of making and using same
WO2017212007A1 (fr) * 2016-06-09 2017-12-14 Curevac Ag Supports cationiques destinés à l'administration d'acides nucléiques
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2294192C2 (ru) * 2001-05-30 2007-02-27 Дзе Скриппс Рисерч Инститьют Система доставки нуклеиновых кислот

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018759A1 (fr) * 1992-03-20 1993-09-30 Baylor College Of Medicine Systeme de transport de l'adn et mode d'emploi
WO1995014714A1 (fr) * 1993-11-24 1995-06-01 La Jolla Cancer Research Foundation Nouveaux peptides fixant l'integrine
WO1996005218A1 (fr) * 1994-08-16 1996-02-22 Commonwealth Scientific And Industrial Research Organisation Apport d'acides nucleiques
WO1998007865A1 (fr) * 1996-08-21 1998-02-26 Genvec, Inc. Adenovirus de ciblage utilisant des motifs peptidiques restreints
WO1998040502A1 (fr) * 1997-03-14 1998-09-17 Life Technologies, Inc. Transfections activees par des peptides
WO1998054347A1 (fr) * 1997-05-29 1998-12-03 Institute Of Child Health Vecteurs de ciblage d'integrine presentant une activite de transfection
WO1999040214A2 (fr) * 1998-02-09 1999-08-12 Genzyme Corporation Vehicules d'apport d'acide nucleique
WO1999058694A1 (fr) * 1998-05-12 1999-11-18 The Regents Of The University Of California Procedes de formation de microparticules lipidiques liees a des proteines et leurs compositions
WO1999060110A2 (fr) * 1998-05-20 1999-11-25 The University Of Tennessee Research Corporation Proteines a enveloppe stable pour des vecteurs retroviraux

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05500504A (ja) * 1989-09-27 1993-02-04 アテナ・ニュウロサイエンスィズ・インコーポレイテッド 細胞付着阻止のための組成物及び使用方法
WO1997033605A1 (fr) * 1996-03-15 1997-09-18 Yale University Occludine humaine, ses utilisations et amelioration de l'absorption de medicaments a l'aide d'inhibiteurs d'occludine
AU9692198A (en) * 1997-10-10 1999-05-03 Kevin J. Donahue Gene delivery compositions and methods
AU4417000A (en) * 1999-04-15 2000-11-02 Glaxo Group Limited Novel pharmaceutical composition suitable for gene therapy

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018759A1 (fr) * 1992-03-20 1993-09-30 Baylor College Of Medicine Systeme de transport de l'adn et mode d'emploi
WO1995014714A1 (fr) * 1993-11-24 1995-06-01 La Jolla Cancer Research Foundation Nouveaux peptides fixant l'integrine
WO1996005218A1 (fr) * 1994-08-16 1996-02-22 Commonwealth Scientific And Industrial Research Organisation Apport d'acides nucleiques
WO1998007865A1 (fr) * 1996-08-21 1998-02-26 Genvec, Inc. Adenovirus de ciblage utilisant des motifs peptidiques restreints
WO1998040502A1 (fr) * 1997-03-14 1998-09-17 Life Technologies, Inc. Transfections activees par des peptides
WO1998054347A1 (fr) * 1997-05-29 1998-12-03 Institute Of Child Health Vecteurs de ciblage d'integrine presentant une activite de transfection
WO1999040214A2 (fr) * 1998-02-09 1999-08-12 Genzyme Corporation Vehicules d'apport d'acide nucleique
WO1999058694A1 (fr) * 1998-05-12 1999-11-18 The Regents Of The University Of California Procedes de formation de microparticules lipidiques liees a des proteines et leurs compositions
WO1999060110A2 (fr) * 1998-05-20 1999-11-25 The University Of Tennessee Research Corporation Proteines a enveloppe stable pour des vecteurs retroviraux

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HART S L: "INTEGRIN-MEDIATED VECTORS FOR GENE TRANSFER AND THERAPY" CURRENT OPINION IN MOLECULAR THERAPEUTICS, CURRENT DRUGS, LONDON,, GB, vol. 1, no. 2, April 1999 (1999-04), pages 197-203, XP001055320 ISSN: 1464-8431 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051311A2 (fr) * 2001-12-17 2003-06-26 Bayer Corporation Anticorps inhibant l'activite du facteur de cellule souche et utilisation dans le traitement de l'asthme
WO2003051311A3 (fr) * 2001-12-17 2004-09-30 Bayer Ag Anticorps inhibant l'activite du facteur de cellule souche et utilisation dans le traitement de l'asthme
FR2860236A1 (fr) * 2003-09-25 2005-04-01 Theraptosis Peptides possedant notamment une activite anti-angiogenique et leurs applications en therapeutique
WO2005030238A1 (fr) * 2003-09-25 2005-04-07 Theraptosis Peptides possedant notamment une activite anti-angiogenique et leurs applications en therapeutique
US9255134B2 (en) 2008-01-24 2016-02-09 Esperance Pharmaceuticals, Inc. Lytic domain fusion constructs and methods of making and using same
EP2252627B1 (fr) * 2008-01-24 2017-04-19 Esperance Pharmaceuticals Produits de fusion recombinants à domaine lytique et leurs procédés de fabrication et d'utilisation
WO2017212007A1 (fr) * 2016-06-09 2017-12-14 Curevac Ag Supports cationiques destinés à l'administration d'acides nucléiques
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo

Also Published As

Publication number Publication date
WO2001092542A8 (fr) 2003-12-24
AU2001258659A1 (en) 2001-12-11
WO2001092542A3 (fr) 2003-05-30
AU2001260467A1 (en) 2001-12-11
WO2001092543A3 (fr) 2002-09-12
JP2003534804A (ja) 2003-11-25
WO2001092543A2 (fr) 2001-12-06
CA2410419A1 (fr) 2001-12-06
EP1285081A2 (fr) 2003-02-26

Similar Documents

Publication Publication Date Title
EP1003898B1 (fr) Vecteurs de ciblage d'integrine presentant une activite de transfection
Perales et al. An evaluation of receptor‐mediated gene transfer using synthetic DNA‐ligand complexes
Mahato Non-viral peptide-based approaches to gene delivery
Pichon et al. Histidine-rich peptides and polymers for nucleic acids delivery
Davis et al. Vectors for airway gene delivery
van der Aa et al. The nuclear pore complex: the gateway to successful nonviral gene delivery
US20030045465A1 (en) Histidine copolymer and methods for using same
JPH10505242A (ja) キメラペントンベースタンパク質を含有する組換えアデノウイルス
AU3237995A (en) A liposomal delivery system for biologically active agents
WO1996040961A1 (fr) Transfection par lipides cationiques amelioree par des peptides
US6372720B1 (en) Liposome fusion and delivery vehicle
Welser et al. Gene delivery using ternary lipopolyplexes incorporating branched cationic peptides: the role of peptide sequence and branching
US20030162293A1 (en) Cell transfection compositions comprising genetic material, an amphipathic compound and an enzyme inhibitor and method of use
US6927278B1 (en) Peptide scaffolds for transfer of molecules into eukaryotic cells
EP1173224B1 (fr) Polyethyleneimine utilise dans des preparations d'adn administrees par aerosol
WO2001092542A2 (fr) Vecteurs ciblant l'integrine avec activite de transfection accrue
US20040014217A1 (en) Methods of transfection
Nakanishi Gene introduction into animal tissues
Lasic et al. Handbook of Nonmedical Applications of Liposomes, Vol IV From Gene Delivery and Diagnosis to Ecology
Mann et al. DNA transfer into vascular smooth muscle using fusigenic Sendai virus (HJV)-liposomes
Skalko-Basnet et al. Uptake of liposomally entrapped fluorescent antisense oligonucleotides in NG108-15 cells: conventional versus pH-sensitive
EP1007549B1 (fr) Compositions et procedes pour une transfection tres efficace
Norton et al. Methods for DNA introduction into mammalian cells
Pan et al. A RGD-containing oligopeptide (K) 16GRGDSPC: a novel vector for integrin-mediated targeted gene delivery
WO2000012114A1 (fr) Structures peptidiques permettant de transferer des molecules dans des cellules eukariotes

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
CFP Corrected version of a pamphlet front page

Free format text: PUBLISHED FIGURE DELETED

NENP Non-entry into the national phase in:

Ref country code: JP