EP1144012A1 - Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery - Google Patents

Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery

Info

Publication number
EP1144012A1
EP1144012A1 EP00904440A EP00904440A EP1144012A1 EP 1144012 A1 EP1144012 A1 EP 1144012A1 EP 00904440 A EP00904440 A EP 00904440A EP 00904440 A EP00904440 A EP 00904440A EP 1144012 A1 EP1144012 A1 EP 1144012A1
Authority
EP
European Patent Office
Prior art keywords
ligand
peg
polymer
cationic
post
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00904440A
Other languages
German (de)
French (fr)
Inventor
Liang Xu
Esther H. Chang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Georgetown University
Original Assignee
Georgetown University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Georgetown University filed Critical Georgetown University
Publication of EP1144012A1 publication Critical patent/EP1144012A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6935Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention relates to the methods to prepare ligand-PEG post-coating stabilized liposome-DNA complex (Lipoplex) or polymer-DNA complex (Polyplex) for targeted gene delivery in vivo.
  • DNA have a defined size with the ligand-PEG coating outside and have the capability of targeted gene delivery with systemic administration.
  • Cationic liposomes are composed of positively charged lipid bilayers and can be complexed to negatively charged, naked DNA by simple mixing of lipids and DNA such that the resulting complex has a net positive charge.
  • the liposome-DNA complex (lipoplex) is easily bound and taken up by cells with a relatively high transfection efficiency (8).
  • Features of cationic liposomes that make them versatile and attractive for DNA delivery include: simplicity of preparation; the ability to complex large amounts of DNA; versatility in use with any type and size of DNA or RNA; the ability to transfect many different types of cells, including non-dividing cells; and lack of immunogenicity or biohazardous activity (reviewed in 4, 9).
  • cationic liposomes have been proven to be safe and efficient for in vivo gene delivery (8, 12). More than tv/enty clinical trials are now underway using cationic liposomes for gene delivery, and liposomes for delivery of small molecule therapeutics (e.g., chemotherapeutic and antifungal agents) are already on the market
  • cationic liposomes lack tumor specificity and have relatively low transfection efficiencies as compared to viral vectors. However, this can be dramatically increased when the liposomes bear a ligand recognized by a cell surface receptor. Receptor-mediated endocytosis represents a highly efficient internalization pathway in eukaryotic cells (13-15). The presence of a ligand on a liposome facilitates the entry of DNA into cells through initial binding of ligand by its receptor on the cell surface followed by internalization of the bound complex. Once internalized, sufficient DNA escapes the endocytic pathway to be expressed in the cell nucleus.
  • ligands have been examined for their liposome- targeting ability, including folic acid, a vitamin necessary for DNA synthesis (13, 16).
  • Folate receptor levels are found elevated in various types of cancer including breast cancer (17-22), and most significantly, the folate receptor recycles during the internalization of folate in rapidly dividing cells such as cancer cells (23).
  • Folate-conjugated macromolecules and liposomes have also been shown to be specifically taken up in vitro by receptor-bearing tumor cells (24, 25).
  • the folate receptor is considered to be useful as a prognostic tumor marker for cancer and as a potential target for drug delivery in the therapy of malignant cell growth (16, 22, 24, 26).
  • a report by Zalipsky et al. (72) uses a different chemistry and a different methodology than the one described herein to attach a ligand-PEG-lipid conjugate to a liposome. Their methodology employs a lipid anchor (DSPE) to attach a ligand to a liposome vesicle via a PEG linking moiety.
  • DSPE lipid anchor
  • Polyethylenimine is the organic macromolecule with the highest cationic-charge- density potential, and a versatile vector for gene and oligonucleotide transfer in vitro and in vivo, as first reported by Boussif et al. (66). Since then, there has been a flurry of research aimed at this polycation and its role in gene therapy (73).
  • Cell-binding ligands can be introduced to the polycation to 1) target specific cell types and 2) enhance intracellular uptake after binding the target cell (13).
  • Erbacher et al. conjugated the integrin-binding peptide 9-mer RGD via a disulfide bridge and showed physical properties of interest for systemic gene delivery. Ogris et al.
  • PEG-SPA interfering with or blocking binding to its receptor, or even completely inactivating the ligand.
  • the present invention discloses a novel strategy for preparing the ligand-PEG-liposome- DNA or ligand-PEG-PEI-DNA complexes.
  • This is referred to as a ligand-PEG post-coating method.
  • ligand-PEG is linked after the DNA has been condensed inside the liposomes (or PEI).
  • the PEG layer will coat only the outside of the DNA-liposome complex (lipoplex) or DNA-PEI complex (polyplex), and will not interfere with the internal structure of the complexes. Therefore, the complex has a condensed DNA-cationic lipid or PEI structure inside and a PEG coating outside, with ligand at the distal end of the coating PEG.
  • ligand-PEG post-coating to distinguish it from previous methods (24, 29, 33, 68, 72).
  • the ligand-PEG post-coating method takes advantage of the protective and long- circulating properties of the reported PEG-liposomes such as "sterically stabilized” liposomes or "StealthTM” liposomes, while keeping the unique characteristics of ligand-cationic liposome- DNA complexes.
  • the ligand-PEG post-coated lipoplexes have low cytotoxicity and the capability of SCCNH tumor-targeted gene delivery after systemic administration. This strategy is a very useful and promising way to design and develop a targeted gene delivery system for systemic gene therapy.
  • FIGURES Figure 1 shows the scheme for preparation of a folate-liposome-DNA complex by a
  • Figure 2 shows the folate-PEG-liposome-mediated transfection of JSQ-3 cells and the effect of serum on their transfection.
  • the LipA-PEG-F complexed with plasmid pSVb DNA (27) was prepared by the "Post-coating Method".
  • LipA consists of equal molar DOTAP and DOPE plus 5% molar DOPE-MB.
  • LipA-PEG complexed with pSVb was prepared similarly by post- coating PEG without folate.
  • JSQ-3 cells in a 96-well plate (lxlO 4 cells/well) were transfected by 1 ⁇ g DNA/well of LipA-PEG-F(pSVb) or LipA-PEG(pSVb) with or without 10% fetal bovine serum.
  • FIG. 3 shows the specificity of folate-PEG-liposome-mediated gene transfection of JSQ-3 cells.
  • LipA-PEGl-F and LipA-PEG2-F were prepared by the "Post-coating Method", SPDP method and 2-iminothiolane method, respectively .
  • JSQ-3 cells in A 96-well plate (lxlO 4 cells/well) were transfected with serially diluted LipA-PEGl-F(pSVb) or LipA-PEG2-F(pSVb), starting from 0.32 ⁇ g DNA/well.
  • Figures 4A-I show the results of "post-coated" folate-PEG-liposome-mediated systemic gene transfection in vivo.
  • LipA-PEGl-F was prepared by the SPDP method and LipA-PEG2-F by the 2-iminothiolane method.
  • Figures 5A-D show spectrophotograms of purified F-PEG-PDP and Gal-PEG-PDP, before and after the addition of DTT, using a Beckman DU640 spectrophotometer from 200 nm to 700 nm.
  • the ligand-PEG post-coating method takes advantage of the protective and long- circulating properties of the reported PEG-liposomes such as "sterically stabilized” liposomes or "StealthTM” liposomes, while keeping the unique characteristics of ligand-cationic liposome- DNA complexes.
  • the ligand-PEG post-coated lipoplexes have low cytotoxicity and the capability of SCCNH tumor-targeted gene delivery after systemic administration. This strategy is a very useful and promising way to design and develop a targeted gene delivery system for systemic gene therapy.
  • the methodology described herein does not employ a lipid anchor, nor is PEG used as a "linking moiety".
  • the liposome used by Zalipsky et al. (72) is an empty vesicle, not one encapsulating DNA.
  • the conjugate described by Zalipsky et al. (72) would not work to deliver the DNA-liposome complex produced by our method which has demonstrated small size and demonstrated efficacy. Attachment of the DSPE-lipid anchor to this liposome-DNA complex would effectively destroy its structure and therefore its potential as a deliver ⁇ - system for gene therapy.
  • Folate is used as an example of a ligand for lipoplex formation, as illustrated in Figure
  • DNA is complexed and condensed with cationic liposomes, containing 1-10%, preferably 5%, molar of DOPE-Maleimidophenyl butyrate (DOPE-MPB), or any other sulfhydryl-reacting molecule- lipid conjugate, in their formulation.
  • DOPE-MPB DOPE-Maleimidophenyl butyrate
  • the sulfhydryl group is necessary for subsequent conjugation.
  • a dithiopyridine (PDP) group is first introduced at one end of PEG-bis-amine and then folate is introduced at the other end. The folate-PEG-PDP is then reduced by dithiothreitol
  • a free sulfhydryl (-SH) group is then introduced directly to the other end by reacting it with 2- iminothiolane.
  • the resulting folate-PEG-SH is then post-coated onto DNA-liposomes.
  • This method is called the 2-iminothiolane method, although other thiol-introducing reagents can also be used.
  • the advantage of the 2-iminothiolane method is that the whole process can be carried out in aqueous phase, which is especially suitable for ligands unstable in organic phase, e.g., peptide or protein ligands such as RGD, Fas-ligands, EGF, FGF, antibodies or their fragments, etc.
  • Cationic polymers e.g., polylysine, protamine or polyethylenimine can also be used to replace the cationic liposome.
  • PEI is used as an example, similar to that of liposomes.
  • the maleimide groups of MB, or other sulfhydryl-reacting molecules, are introduced to PEI, in the range of 0.1-25% modification of amines in the PEI molecule.
  • DNA is complexed and condensed first with PEI-MB, at a N/P ratio (amine nitrogen of PEI vs. phosphate of DNA, molar ratio) of 1-50, preferably 5-15, as described (66, 67).
  • the freshly reduced ligand-PEG-SH is added to the polyplex and reacted for 2-6 hours at room temperature, or overnight at 4°C.
  • the post- coated polyplex can be used directly, or purified by Sepharose CL-4B chromatography to remove the unconjugated ligand-PEG.
  • a galactose-PEG-SH post-coated PEI-DNA complex prepared as described above and detailed below in Example 8 showed enhanced stability, increased resistance to serum, and reduced toxicity both in vitro and in vivo.
  • conjugation strategies can also be used to link a ligand at one end of PEG and an active group, e g , a thiol group (-SH), at the other end for the subsequent "post-coating"
  • an active group e g , a thiol group (-SH)
  • an active group (with protection) can be linked first, followed by a ligand at the other end After deprotection (like the reduction step in the SPDP method), the free active group is ready for "post-coatmg"
  • An alternative strategy is to make gand-PEG-succinimidyl (active) ester, then react this with the primary amino groups in lipoplex (from DOPE) or polyplex (from PEI) for post-coating In this case, no prior modification of lipids or PEI to introduce a reactive group (e g , maleimide group of MB) is required.
  • a reactive group e g , maleimide group of MB
  • the PEG layer will coat the outside of the DNA-liposome complex and will not interfere with the internal structure of the DNA-liposome complex Therefore, the complex has a condensed DNA- cationic hpid structure or acentric onion-like core structure (see Example 6 below, detailing an electron microscopy analysis) inside and a PEG coating outside, with folate at the distal end of PEG
  • the ligand-PEG "post-coating" method takes advantage of the protective and long- circulating properties of the reported PEG-hposomes such as “sterically stabilized” liposomes or “StealthTM” liposomes, while keeping the unique characteristics of hgand-cationic liposome- DNA complexes
  • the ligand-PEG post-coated liposomes have the capability of tumor-targeted gene delivery after systemic administration and are suitable for systemic gene therapy in vivo
  • the invention is not limited to the use of any specific targeting ligand Folate is used as an example of a targeting ligand Other ligands can easily be used in the invention with minor modifications
  • the ligand can be any ligand the receptor for which is differentially expressed on the target cell Examples include other vitamins, EGF, insulin, FGF, Heregulm, RGD peptides or other polypeptides reactive to lnteg ⁇ n receptors, antibodies or their fragments
  • the ligand is folate Foi lig
  • the invention is not limited to the use of any specific coating polymer for "post-coating".
  • the coating polymer can be any polymer with inert chemical activity within the polymer chain and active groups at the two ends for conjugation. Zalipsky S et al., US Patent No. 5,395,619
  • the polymer is PEG with primary amine groups at the two ends as active groups.
  • the invention is not limited to the delivery of any specific type of DNA.
  • Any polynucleotides that can be complexed by cationic liposomes or polymers can be used in the invention.
  • the polynucleotides include but are not limited to: plasmid DNA, DNA fragments, oligonucleotides, oligodeoxynucleotides, chimeric RNA/DNA oligonucleotides, RNA, ribozymes, etc.
  • Anionic peptides, polymers, synthetic or natural molecules can also be used in the invention so long as they can be complexed by cationic liposomes or polymers.
  • molecules which can be delivered via ligand-PEG post coated cationic liposomes include a gene, high molecular weight DNA, plasmid DNA, an antisense oligonucleotide, peptide nucleic acids, a chemical agent such as a chemotherapeutic molecule or any large molecule including, but not limited to DNA, RNA, viral particles, growth factors, cytokines, immunomodulating agents and other proteins, including proteins which when expressed present an antigen which stimulates or suppresses the immune system.
  • Example 1 Preparation of ligand-PEG-SH for post-coating by SPDP method This example discloses the SPDP reaction procedure for preparation of ligand-PEG-SH for post-coating. This is illustrated in Figure 1 by the solid line flowchart.
  • N-Hydroxysuccinimide ester of folic acid was prepared by reacting 1 gram folic acid (F, Sigma), in 30 ml dry dimethyl sulfoxide plus 0.5 ml triethylamine (TEA, Sigma), with 0.52 g N-hydroxysuccinimide (NHS, Sigma) in the presence of 0.94 g dicyclohexylcarbodiimide (DCC, Fluka) overnight at room temperature (75). The solution was filtered and purified as described by Lee RJ and Low PS (75). PDP-PEG-NH2 was reacted with 2-5 molar excess of folate-NHS in dry chloroform plus 2 molar of TEA overnight at room temperature.
  • EDTA may be added to a 1 mM final concentration to help stabilize the folate-PEG-SH.
  • the purified folate-PEG-SH can be frozen at -20°C under inert gas for later use.
  • the free -SH group was confirmed by DTNB (5,5'-dithiobis-(2-nitrobenzoic acid)) titration (76). Folate was measured by absorption at 363 nm. The molar ratio of folate/-SH in the purified product was approximately 0.9-1.1.
  • Example 2 Preparation of ligand-PEG-SH for post-coating by direct thiolation (2-iminothiolane method)
  • This example discloses the reaction procedure of preparing ligand-PEG-SH for post- coating by direct thiolation. This is illustrated in Figure 1 by the dashed line flowchart. Folate is linked to one end of PEG-bis(amine) first. 30 ⁇ mol Polyoxyethylene bis(amine)
  • the product was purified to remove folate-PEG-folate or unreacted NH2-PEG-NH2 as described (48).
  • the product was confirmed by thin layer chromatography (TLC) (chloroform/me hanol/ acetic acid 100/30/2) with iodine staining.
  • Free -SH is introduced to the other end of folate-PEG directly by reacting with 2- iminothiolane.
  • the reaction is performed in aqueous solution.
  • Folate-PEG-NH2 was dissolved in 100 mM HEPES, pH 8.0. 2-5 molar excess of 2-iminothiolane HCl (Sigma) in PBS was added and stirred for 0.5-2 hours at room temperature.
  • the resulting folate-PEG-SH was purified by desalting column chromatography (10DG, BioRad) or by Centricon 3 ultrafiltration to obtain reduced folate-PEG-SH.
  • the free -SH group was confirmed by DTNB titration and folate was measured by absorption at 363 nm. The molar ratio of folate/-SH in the purified product was approximately 0.9-1.1.
  • Example 3 Post-coating " of cationic liposome-DNA complex (Lipoplex ) This example discloses the procedure of ligand-PEG-SH post-coating onto DNA- liposomes. A. Preparation of cationic liposomes
  • DOPE-MPB was prepared by reacting DOPE with succinimidyl-4-(/ maleimidophenyl)butyrate (SMPB, Sigma) in dry chloroform in the presence of TEA (65).
  • the maleimide group of MPB is reactive to the sulfhydryl group and serves as the conjugating molecule on the liposomal surface.
  • Other thiol-reactive groups can also be used here, such as other maleimido-containing molecules, etc.
  • DOPE-MPB is also available from Avanti Polar Lipids, Inc., Alabaster, AL.
  • Cationic liposomes LipA were prepared as follows: a chloroform solution of 5 ⁇ mol dioleoyltrimethylammonium-propane (DOTAP), 5 ⁇ mol DOPE (Avanti Polar Lipids, Inc., Alabaster, AL) and 0.1-1 ⁇ mol DOPE-MPB were mixed together in a round-bottom flask, and the chloroform was evaporated under reduced pressure. 10 ml pure water, was added to the flask to suspend the lipids, then sonicated for 10 minutes in a bath-type sonicator at 4°C. The final concentration of the liposome was 1-2 nmol/ ⁇ l total lipids. Other cationic liposomes were also prepared in similar fashion with the compositions shown below:
  • LipD and LipE were sonicated at 65 °C instead of 4°C.
  • liposome preparation methods can also be used to prepare the cationic liposomes.
  • the ethanol injection method modified from that described by Campbell (80) was used successfully in the present invention.
  • all lipids were solubilized in ethanol and mixed, injected into vortexing pure water of 50-60°C with a Hamilton syringe. The solution was vortexed for a further 10-15 minutes. The final concentration was 1 -2 ⁇ M total lipids.
  • the ethanol injection method is faster, easier and more robust. Since we have found that the maleimide group is not stable in aqueous solution with pH >7, the liposomes should be prepared in water (pH 5-6.5).
  • the pH can be adjusted to 7.5-8.0 before “post-coating” with 1 M HEPES buffer, pH 7.5-8.0, to facilitate the post-coating reaction.
  • B. "Post-coating" of DNA-liposomes complex Plasmid DNA was complexed with MPB-liposomes in water or 10 mM HEPES buffer, pH 7.0, as described (34) at DNA/lipid ratios 1/6-1/26 ( ⁇ g/nmol), preferably 1/10-1/20. Before “post-coating", 1 M HEPES buffer, pH 7.5-8.0, was added to the MPB-liposome-DNA complexes to a final concentration of 10-20 mM.
  • Folate-PEG-SH was added at 0.5-5 molar excess to MPB-DOPE, preferably 1 -2 molar excess and stirred several hours to overnight at room temperature in the dark.
  • the resulting folate-PEG-LipA-DNA is used directly with 5% dextrose for in vivo gene transfection with pSVb as a reporter gene.
  • Example 4 In vitro gene transfection by "post-coated" folate-PEG-Lip-pSVb This example describes the in vitro gene transfection efficiencies of "post-coated” folate - PEG-liposomes using a reporter gene.
  • the medium used here was folate-free medium (RPMI-1640 folate-free, Gibco).
  • 1 x 10 4 cells were plated in each well of a 96-well plate or 5 x 10 4 cells/well in a 24-well plate. 24 hours later, the cells were washed once with medium without serum or antibiotics and 100 ⁇ l transfection solution containing various amounts of folate-PEG-Lip-pSVb or PEG-Lip-pSVb as well as pSVb alone were added to each well. After 5 hours of transfection at 37°C, an equal amount of medium containing 20% fetal bovine serum were added to each well. 48 hours later, the cells were washed once with PBS and lysed in IX reporter lysis buffer (Promega).
  • the cell lysates were treated with 100 ⁇ l 150 ⁇ M O-nitrophenyl- ⁇ -galactopyranoside in 20 mM Tris (pH 7.5) containing 1 mM MgCL and 450 mM ⁇ -mercaptoethanol at 37°C for 0.5 hour. The reaction was stopped by the addition of 150 ⁇ l/well of 1 M Na 2 C0 3 . The absorbance was determined at
  • FIG. 2 shows the in vitro folate-PEG-liposome-mediated transfection of JSQ-3 cells and the effect of serum on the transfection efficiency.
  • PEG "post-coated" liposomes show resistance to serum and folate-PEG-LipA resulted in two-fold higher reporter gene expression than LipA- PEG without folate ligand.
  • Figure 3 shows the specificity of folate-PEG-liposome transfection. Free folic acid can block the transfection activity of folate-PEG-liposomes to the level of the LipA-PEG, demonstrating that the transfection of the folate-PEG-liposomes is mediated by the folate through the folate-receptor.
  • the tumors were allowed to develop to a size of 100 mnr ⁇
  • the "post-coated" folate-PEG-Lip-pSVb were prepared as described in Examples 1 -3, wherein LipA-PEG 1 -F indicates the "post-coated” folate-PEG-Lip-pSVb prepared by SPDP method (Examples 1 and 3) and LipA-PEG2-F indicates that prepared by the direct thiolation method (Examples 2 and 3).
  • LipA-PEG 1-F, LipA-PEG2-F or pSVb plasmid alone in
  • 5% dextrose was injected intravenously via the tail vein, at 50 ⁇ g of plasmid DNA/300 ⁇ l/animal.
  • the tumors as well as mouse organs were excised, cut into 1 mm sections, washed once with PBS, and fixed with 2% Formaldehyde-0.2% Glutaraldehyde for 4 hours at room temperature.
  • the fixed tumor sections were washed 4 times, each for 1 hour, and stained with X-Gal solution plus 0.1% NP-40 (pH 8.5) at 37°C overnight.
  • the stained tumor sections were embedded and sectioned using normal histological procedures and counter-stained with nuclear fast red. Four sections per tumor were examined to evaluate the ⁇ -galactosidase gene expression, as indicated by the blue stained cells.
  • Figures 4A-I show the "post-coated" folate-PEG-liposome-mediated systemic gene transfection in vivo.
  • Figures 4A and 4B 3 days after the i.v. injection of LipA-PEG- F complexed with pSVb, 30-60%. of tumor cells in nude mouse xenograft stained blue, indicating the tumor delivery and expression of reporter gene ⁇ -galactosidase.
  • the normal mouse tissues, including lung and liver, were not stained blue, except a few macrophages ( Figures 4G and 4H), demonstrating that the PEG "post-coated" liposomes can deliver the complexed gene selectively to tumors in vivo.
  • the ligand-PEG "post-coated" liposomes can deliver the complexed genes selectively to the tumor in vivo after systemic administration, and give sustained gene expression in the tumor, with an in vivo transfection efficiency of 30-60%o. Therefore, this system is very useful and promising for in vivo gene delivery and gene therapy.
  • EM can reveal the structure and size distribution of liposomes. EM can also be used for quality control of liposomal preparation. We observed the ligand-cationic liposomes under Transmission
  • Ligand-PEG "post-coated" cationic liposome-pSVb complexes were prepared as described in Examples 1 -3. One drop of the liposome complex was added to the grid. 5 minutes later, excess liquid was removed by attaching a filter paper to the edge of the grid. One drop of 4% Uranium Acetate was then added onto the grid for negative staining. 5 minutes later, excess liquid was also removed by attaching a filter paper to the edge of the grid. The grid was air dried at room temperature for 15 minutes before being put into the sample chamber of the TEM. The TEM JEOL-1200EX was used in the study according to the manufacturer's instruction. Photos were taken at magnitudes of 10-50k, 60 kVolt. The liposome samples on the grid were prepared and stained freshly and observed within one hour.
  • cationic liposome-DNA complexes have a diverse structure and size ranging from 100 nm to 1000 nm.
  • the ligand-liposome-DNA complexes disclosed in the invention have much smaller size and much more even size distribution.
  • Cationic liposome LipA itself has a size of 25-50 nm, average 35 nm (in diameter).
  • an interesting "irregular or acentric onion-like core structure" in the core of the LipA-DNA complex was observed, with evenly distributed sizes of 35-65 nm (50 nm in average).
  • the size of liposomes increased only slightly, becoming 35-80 nm (60 nm in average), indicating a 10-15 nm thick layer of folate-PEG was "post-coated” on the LipA-DNA complex.
  • the liposomes must be first resistant to serum and then pass through the blood vessel (capillary) wall.
  • the ligand-PEG "post-coated” liposomes disclosed in the invention can meet these two requirements.
  • the ligand-PEG-Lip-DNA is very resistant to serum as shown in Example 4.
  • ligand-PEG was "post-coated"
  • the liposomes became stabilized and retained transfecting activity for a prolonged period of time. 100% of their activity was evident for one month and 50-60% transfecting activity still remained after 6 months in storage. Therefore, the ligand-PEG "post-coated" cationic liposomes disclosed in the invention are more stable and have a longer shelf life, which are important considerations for practical use.
  • Example 8 Preparation of Galactose-PEG post-coated polye hylenimine-DNA complex (polyplex)
  • This example discloses a procedure to prepare a galactose-PEG post-coated PEI-DNA complex.
  • Galactose or galactosylated ligands have high affinity to asialoglycoprotein receptor which is present at high density on hepatocytes and hepatomas (13).
  • Galactose-PEG post-coated PEI-DNA complex will be useful for targeted gene delivery to liver for gene therapy of liver diseases.
  • Branched PEI with average molecular weight of 25 kDa (designated P25) was purchased from Aldrich.
  • Linear PEI with average molecular weight of 25 kDa (designated LP25) was purchased from Polysciences, Inc. Both PEIs were reacted with maleimide-containing active ester to introduce maleimide group, similar to that of DOPE-MB in Example 3.
  • the followings are examples of (but not limited to) the maleimide-containing active esters that can be used: 3-
  • MBS Maleimidobenzoic acid N-hydroxysuccinimide ester
  • SMPB Succinimidyl 4-(p- maleimidophenyl)butyrate
  • SMCC Succinimidyl 4-(N- maleimidomethyl)cyclohexane-l -carboxylate
  • the Gal-PEG-PDP was reduced by DTT similar to that of F-PEG-PDP as described in Example 3.
  • Polyplexes were prepared by adding DNA into P25-MB or LP25-MB solution in water or 10-150 mM NaCl, at a N/P ratio (amine nitrogen of PEI vs. phosphate of DNA, molar ratio) 1-50, preferably 5-15, as described (66, 67).
  • the freshly reduced ligand- PEG-SH (F-PEG-SH or Gal-PEG-SH) is added to the polyplex and reacted for 2-6 hours at room temperature or overnight at 4°C.
  • the post-coated polyplex can be used directly or purified by
  • Sepharose CL-4B chromatography to get rid of the unconjugated ligand-PEG. Sepharose CL-4B chromatography confirmed that more than 50%> of ligand-PEG-SH was linked to the polyplex. 50%) dextrose was added to a final concentration of 5% for i.v. administration.
  • Example 9 Ligand-PEG post-coating stabilized polyplex mediated transfection in vitro and in vivo
  • This example demonstrates the in vitro and in vivo transfection activity of F-PEG and Gal-PEG post-coating stabilized polyplexes.
  • the plasmid, pLuc which contains the firefly luciferase gene under CMV promoter was used in the study (35).
  • the polyplex P25-pLuc was prepared by adding P25-MB (10 mM of monomer in water, pH 7.4 by HCl) to the pLuc DNA solution while vortexing and vortexed an additional 1 minute, at a N/P ratio 6-12.
  • the F-PEG and Gal-PEG were post-coated to the polyplex as described in Example 8.
  • the in vitro transfection was performed in 24-well plates, as described in Example 4 and references 27 and 35.
  • the transfection reagent solutions were added to the cells, JSQ-3, in the presence of 10% serum. 24 hours later, the cells were washed and lysed to measure the luciferase activity and protein concentration (35).
  • the results are expressed as 10 3 relative light units (RLU) per ⁇ g protein in the lysate, as shown in Table 2.
  • the results show that the ligand-PEG post-coated polyplexes have very high transfection activity in the presence of serum, significantly higher than non-coated polyplex. Also it is worthy to note that the ligand-PEG post-coated polyplexes were much less cytotoxic than non-coated polyplex P25-pLuc, as indicated by the protein assay and observation under a microscope.
  • C57BK 6 black mice were i.v. injected with the ligand-PEG post- coated polyplexes as well as non-coated polyplexes, at 80-100 ⁇ g DNA/mouse in 0.4-0.5 ml/injection via tail vein. 24 hours later, the organs were excised and homogenized in IX lysis buffer (Promega). The luciferase activities and protein concentrations were measured as described (35), and the results are expressed as 10 3 RLU/mg protein as shown in Table 3.
  • Table 3 shows that polyplexes without PEG coating deliver genes predominantly to the lungs and little to liver, spleen or other organs (data not shown).
  • the ligand-PEG post-coating can stabilize and shield the polyplex in vivo to survive the lung's first filtration effect (like other PEG-coated stealth vectors), and can deliver the carried gene selectively to the target tissue.
  • the complexes prepared by this Gal-PEG post-coating method can deliver DNA selectively to the liver after systemic administration, and would be useful for liver-targeted gene therapy.
  • FIGS. 5A-D show the spectrophotograms of the scans.
  • Figure 5A shows the scan of F-PEG-PDP.
  • Figure 5B shows a new peak at 342 nm arises, representing the cleaved 2-thiopyridone from the PDP group.
  • Figure 5C shows a 240 nm peak of galactose as well as a 280 nm peak of PEG.
  • Figure 5D shows up. The results confirm that both PDP and the ligands were conjugated to PEG and the PDP was functional and can be reduced.
  • Zalipski S Polyethylene glycol-lipid conjugates. In: Stealth Liposomes, Lasic DD and Martin FJ, eds., 1995, CRC Press, Inc., Boca Raton, FL. 93-102.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nanotechnology (AREA)
  • Immunology (AREA)
  • Dispersion Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention relates to the area of systemic, tissue-specific non-viral gene delivery. The present invention provides a novel method to prepare ligand-directed, PEG-stabilized complex as gene delivery systems for targeted gene therapy. Due to the presence of PEG these novel complexes have longer circulating times than conventional ligand-liposome complexes. In addition, due to the presence of the ligand in the complex with PEG, these complexes are tissue targeting. Their small size further makes them very desirable for in vivo use.

Description

TITLE OF THE INVENTION
LIGAND-PEG POST-COATING STABILIZED LIPOPLEX AND POLYPLEX FOR
TARGETED GENE DELIVERY
FIELD OF THE INVENTION
This invention relates to the methods to prepare ligand-PEG post-coating stabilized liposome-DNA complex (Lipoplex) or polymer-DNA complex (Polyplex) for targeted gene delivery in vivo. The complexes prepared by ligand-PEG post-coating after complexing with
DNA have a defined size with the ligand-PEG coating outside and have the capability of targeted gene delivery with systemic administration.
BACKGROUND OF THE INVENTION
Due to the rapid advancement of genetics, gene therapy is becoming a promising strategy for the treatment of cancer and other diseases (1-3). Current gene therapy approaches employ either viral or non- viral vector systems (4, 5). The limitations of a viral approach are related to their lack of targeting and to residual viral elements that can be immunogenic, cytopathic, or recombinogenic (5). Non-viral gene transfer vectors could circumvent some of the problems associated with using viral vectors. Progress has been made toward developing non-viral, pharmaceutical formulations of genes for in vivo human therapy, particularly cationic liposome- mediated gene transfer systems (4-8). Cationic liposomes are composed of positively charged lipid bilayers and can be complexed to negatively charged, naked DNA by simple mixing of lipids and DNA such that the resulting complex has a net positive charge. The liposome-DNA complex (lipoplex) is easily bound and taken up by cells with a relatively high transfection efficiency (8). Features of cationic liposomes that make them versatile and attractive for DNA delivery include: simplicity of preparation; the ability to complex large amounts of DNA; versatility in use with any type and size of DNA or RNA; the ability to transfect many different types of cells, including non-dividing cells; and lack of immunogenicity or biohazardous activity (reviewed in 4, 9). Notably, the introduction of wtp53 by a liposome-DNA complex partially inhibited the growth of mammary (10) or glioblastoma (1 1) tumors in nude mice. More importantly from the perspective of human cancer therapy, cationic liposomes have been proven to be safe and efficient for in vivo gene delivery (8, 12). More than tv/enty clinical trials are now underway using cationic liposomes for gene delivery, and liposomes for delivery of small molecule therapeutics (e.g., chemotherapeutic and antifungal agents) are already on the market
(2).
A. Tumor Targeted Liposome Delivery One disadvantage of cationic liposomes is that they lack tumor specificity and have relatively low transfection efficiencies as compared to viral vectors. However, this can be dramatically increased when the liposomes bear a ligand recognized by a cell surface receptor. Receptor-mediated endocytosis represents a highly efficient internalization pathway in eukaryotic cells (13-15). The presence of a ligand on a liposome facilitates the entry of DNA into cells through initial binding of ligand by its receptor on the cell surface followed by internalization of the bound complex. Once internalized, sufficient DNA escapes the endocytic pathway to be expressed in the cell nucleus. A variety of ligands have been examined for their liposome- targeting ability, including folic acid, a vitamin necessary for DNA synthesis (13, 16). Folate receptor levels are found elevated in various types of cancer including breast cancer (17-22), and most significantly, the folate receptor recycles during the internalization of folate in rapidly dividing cells such as cancer cells (23). Folate-conjugated macromolecules and liposomes have also been shown to be specifically taken up in vitro by receptor-bearing tumor cells (24, 25). Thus the folate receptor is considered to be useful as a prognostic tumor marker for cancer and as a potential target for drug delivery in the therapy of malignant cell growth (16, 22, 24, 26). Recently, we have developed a folate-liposome-DNA system for systemic wtp53 gene therapy of SCCHN and examined its ability to sensitize SCCHN to radiation in vitro and in vivo (27). These experiments demonstrated the tumor selectivity and high in vivo transfection efficiency of this systemically delivered liposome complex, and that the combination of intravenous folate-liposome-p53 and radiation was able to eliminate and prevent the recurrence of established SCCHN xenograft tumors for extended periods of time. Interestingly, this folate- liposome-DNA system can also target to human breast cancer in vitro and in vivo (27).
The limitations of the above ligand-liposome-DNA complex or ligand-lipoplex that we encountered are low stability (must be freshly prepared before use) and high in vivo clearance (low serum half-life). Thus, only a limited amount of i.v. injected lipoplex actually reaches the tumor. These two areas must be improved upon for continued pharmaceutical development of the folate lipoplex. One way to overcome these limitations is to coat the lipoplex with inert polymers to build up a sterically repulsive shield that protects the complex from nonspecific opsonization by plasma proteins, thus interfering with liposome recognition and clearance from circulation by the macrophages of the reticuloendothelial system (RES) (28, 29). B. Sterically Stabilized Liposomes Numerous studies have reported that modifications of unliganded liposomes with polymers such as polyethylene glycol (PEG) prolong the length of time in blood circulation as a result of a reduced rate and extent of uptake by the RES (reviewed in 28). These liposomes have been characterized as "sterically stabilized" liposomes or "Stealth™" liposomes, on the basis of their enhanced in vivo stability and reduced reactivity to plasma proteins (28). Hong K, et al. (30) produced a stable cationic liposome-DNA complex by including a small amount of
PEG-phospholipid conjugate in the formulation. The preparation was reported to be stable for months at 4°C and gave reproducible high transfection activity in mouse lungs after intravenous injection. Recently, Cullis' group (31, 32) described a detergent dialysis procedure to prepare a PEG-lipoplex, which allows encapsulation of plasmid DNA within a lipid envelope, where the resulting particle is stabilized by an existing PEG coating. However, the transfection activity of this PEG-lipoplex relied upon the dissociation of the PEG coating, transforming the complex from a stable particle to a transfection-competent one. Although this procedure works in vitro, the requirement of PEG-coating dissociation severely limits its ability to systemically deliver genes in vivo. It is difficult to control the timing of the PEG-coating dissociation after i.v. injection of the particle, circulation in the blood stream and passage through the blood vessel wall, to reach the target site. As for ligand-targeted liposome-DNA complexes, there is less data in the literature with regards to PEG-stabilization. We prepared cationic folate-PEG-liposomes to complex DNA, using a modification of the methods reported by Dr. Low's group (24, 33). Using these methods, we encountered a problem in that the prepared cationic PEG-liposomes had a PEG-coating layer on their outer surface, which interfered with their subsequent complexing and condensing with DNA, a key step in forming lipoplexes. Therefore, a higher lipid/DNA ratio was required. The result was that the complexing was neither efficient nor complete, and the size of the complexes was too large (300-1000 nm) for the in vivo systemic gene delivery to be feasible. A report by Zalipsky et al. (72) uses a different chemistry and a different methodology than the one described herein to attach a ligand-PEG-lipid conjugate to a liposome. Their methodology employs a lipid anchor (DSPE) to attach a ligand to a liposome vesicle via a PEG linking moiety.
C. Polyethylenimine - the versatile cationic polymer for gene delivery
Polyethylenimine (PEI) is the organic macromolecule with the highest cationic-charge- density potential, and a versatile vector for gene and oligonucleotide transfer in vitro and in vivo, as first reported by Boussif et al. (66). Since then, there has been a flurry of research aimed at this polycation and its role in gene therapy (73). Cell-binding ligands can be introduced to the polycation to 1) target specific cell types and 2) enhance intracellular uptake after binding the target cell (13). Erbacher et al. (67) conjugated the integrin-binding peptide 9-mer RGD via a disulfide bridge and showed physical properties of interest for systemic gene delivery. Ogris et al. (68) conjugated transferrin to PEI 800 kDa and compared the in vitro and in vivo transfection activities of the DNA/transferrin-PEI complex before and after PEGylation (i.e., covalent coupling of PEG). PEGylation of the complexes strongly reduced plasma protein binding and erythrocyte aggregation, their size was stabilized and the surface charge was reduced. The PEGylation prolonged the circulation of the complex in the blood after i.v. injection. Targeted gene delivery into subcutaneously growing tumors after systemic application was achieved using this PEGylation sterically stabilized DNA/transferrin-PEI complex, whereas non-PEGylated complexes gave predominant gene expression in the lungs associated with considerable toxicity (69). This strategy differs from the PEG post-coating method in that the ligand transferrin is already conjugated to the PEI prior to addition of PEG. Although the results are encouraging, the major shortcoming of the strategy is that the ligand transferrin lies inside the PEG shield and thus limits its accessibility to the corresponding receptor on target cells. Another drawback is that the DNA/transferrin-PEI was PEGylated with a succinimidyl derivative of PEG propionic acid (M-PEG-SPA), which reacts randomly with primary amino groups present on both the PEI and the transferrin molecules (69). Therefore, the ligand transferrin itself can be reacted by M-
PEG-SPA interfering with or blocking binding to its receptor, or even completely inactivating the ligand.
The above problems can be avoided by using the "ligand-PEG post-coating" method disclosed in the present invention, while preserving all the advantages of PEGylation. SUMMARY OF THE INVENTION
The present invention discloses a novel strategy for preparing the ligand-PEG-liposome- DNA or ligand-PEG-PEI-DNA complexes. This is referred to as a ligand-PEG post-coating method. With this strategy, ligand-PEG is linked after the DNA has been condensed inside the liposomes (or PEI). Hence the PEG layer will coat only the outside of the DNA-liposome complex (lipoplex) or DNA-PEI complex (polyplex), and will not interfere with the internal structure of the complexes. Therefore, the complex has a condensed DNA-cationic lipid or PEI structure inside and a PEG coating outside, with ligand at the distal end of the coating PEG. We designate this strategy a "ligand-PEG post-coating" method to distinguish it from previous methods (24, 29, 33, 68, 72).
The ligand-PEG post-coating method takes advantage of the protective and long- circulating properties of the reported PEG-liposomes such as "sterically stabilized" liposomes or "Stealth™" liposomes, while keeping the unique characteristics of ligand-cationic liposome- DNA complexes. The ligand-PEG post-coated lipoplexes have low cytotoxicity and the capability of SCCNH tumor-targeted gene delivery after systemic administration. This strategy is a very useful and promising way to design and develop a targeted gene delivery system for systemic gene therapy.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows the scheme for preparation of a folate-liposome-DNA complex by a
"Post-coating Method". Note that before folate-PEG-SH, the solid lines show the SPDP method and the dashed lines show the 2-iminothiolane method.
Figure 2 shows the folate-PEG-liposome-mediated transfection of JSQ-3 cells and the effect of serum on their transfection. The LipA-PEG-F complexed with plasmid pSVb DNA (27) was prepared by the "Post-coating Method". LipA consists of equal molar DOTAP and DOPE plus 5% molar DOPE-MB. LipA-PEG complexed with pSVb was prepared similarly by post- coating PEG without folate. JSQ-3 cells in a 96-well plate (lxlO4 cells/well) were transfected by 1 μg DNA/well of LipA-PEG-F(pSVb) or LipA-PEG(pSVb) with or without 10% fetal bovine serum. 2 days later, the cells were lysed and β-galactosidase expression was measured by colorimetric assay. Means of duplicates were plotted. Figure 3 shows the specificity of folate-PEG-liposome-mediated gene transfection of JSQ-3 cells. LipA-PEGl-F and LipA-PEG2-F were prepared by the "Post-coating Method", SPDP method and 2-iminothiolane method, respectively . JSQ-3 cells in A 96-well plate (lxlO4 cells/well) were transfected with serially diluted LipA-PEGl-F(pSVb) or LipA-PEG2-F(pSVb), starting from 0.32 μg DNA/well. For ligand competition, free folic acid (FA) was added just before transfection to a final concentration of 1 mM. 2 days later, the cells were lysed and β- galactosidase expression was measured by colorimetric assay. Means of duplicates were plotted.
Figures 4A-I show the results of "post-coated" folate-PEG-liposome-mediated systemic gene transfection in vivo. LipA-PEGl-F was prepared by the SPDP method and LipA-PEG2-F by the 2-iminothiolane method.
Figures 5A-D show spectrophotograms of purified F-PEG-PDP and Gal-PEG-PDP, before and after the addition of DTT, using a Beckman DU640 spectrophotometer from 200 nm to 700 nm.
DETAILED DESCRIPTION OF THE INVENTION
The ligand-PEG post-coating method takes advantage of the protective and long- circulating properties of the reported PEG-liposomes such as "sterically stabilized" liposomes or "Stealth™" liposomes, while keeping the unique characteristics of ligand-cationic liposome- DNA complexes. The ligand-PEG post-coated lipoplexes have low cytotoxicity and the capability of SCCNH tumor-targeted gene delivery after systemic administration. This strategy is a very useful and promising way to design and develop a targeted gene delivery system for systemic gene therapy.
It is important to note that the methodology described herein does not employ a lipid anchor, nor is PEG used as a "linking moiety". Moreover, it should be noted that the liposome used by Zalipsky et al. (72) is an empty vesicle, not one encapsulating DNA. Most significantly, the conjugate described by Zalipsky et al. (72) would not work to deliver the DNA-liposome complex produced by our method which has demonstrated small size and demonstrated efficacy. Attachment of the DSPE-lipid anchor to this liposome-DNA complex would effectively destroy its structure and therefore its potential as a deliver}- system for gene therapy. Folate is used as an example of a ligand for lipoplex formation, as illustrated in Figure
1 which depicts a scheme for preparation of folate-PEG liposome-DNA complexes. First, DNA is complexed and condensed with cationic liposomes, containing 1-10%, preferably 5%, molar of DOPE-Maleimidophenyl butyrate (DOPE-MPB), or any other sulfhydryl-reacting molecule- lipid conjugate, in their formulation. The sulfhydryl group is necessary for subsequent conjugation. A dithiopyridine (PDP) group is first introduced at one end of PEG-bis-amine and then folate is introduced at the other end. The folate-PEG-PDP is then reduced by dithiothreitol
(DTT), or any other reducing reagent, to produce free sulfhydryl group, folate-PEG-SH. This complex is then reacted with the maleimide group of MPB, or any other sulfhydryl-reacting molecule, on the DNA-liposomes to link the folate-PEG to the DNA-liposomes (Fig.1 , solid line flowchart). This method is called the SPDP method although other amine-reacting crosslinkers can also be used. An alternative method of the strategy is shown in Fig.l , dashed line, wherein the folate is first linked to one end of PEG-bis-amine by reacting it with folate-NHS. A free sulfhydryl (-SH) group is then introduced directly to the other end by reacting it with 2- iminothiolane. The resulting folate-PEG-SH is then post-coated onto DNA-liposomes. This method is called the 2-iminothiolane method, although other thiol-introducing reagents can also be used. The advantage of the 2-iminothiolane method is that the whole process can be carried out in aqueous phase, which is especially suitable for ligands unstable in organic phase, e.g., peptide or protein ligands such as RGD, Fas-ligands, EGF, FGF, antibodies or their fragments, etc.
Cationic polymers, e.g., polylysine, protamine or polyethylenimine can also be used to replace the cationic liposome. PEI is used as an example, similar to that of liposomes. The maleimide groups of MB, or other sulfhydryl-reacting molecules, are introduced to PEI, in the range of 0.1-25% modification of amines in the PEI molecule. DNA is complexed and condensed first with PEI-MB, at a N/P ratio (amine nitrogen of PEI vs. phosphate of DNA, molar ratio) of 1-50, preferably 5-15, as described (66, 67). After addition of HEPES buffer to a final concentration of 10-20 mM, to adjust pH to 7.4-8.0, the freshly reduced ligand-PEG-SH is added to the polyplex and reacted for 2-6 hours at room temperature, or overnight at 4°C. The post- coated polyplex can be used directly, or purified by Sepharose CL-4B chromatography to remove the unconjugated ligand-PEG. A galactose-PEG-SH post-coated PEI-DNA complex prepared as described above and detailed below in Example 8 showed enhanced stability, increased resistance to serum, and reduced toxicity both in vitro and in vivo. The intravenous injection of the galactose-PEG post-coated polyplex resulted in significantly increased reporter gene expression in mouse liver and much reduced expression in mouse lung, whereas the non- PEGylated polyplex gave predominant gene expression in mouse lung and was associated with considerable toxicity
Other conjugation strategies can also be used to link a ligand at one end of PEG and an active group, e g , a thiol group (-SH), at the other end for the subsequent "post-coating"
Alternatively, an active group (with protection) can be linked first, followed by a ligand at the other end After deprotection (like the reduction step in the SPDP method), the free active group is ready for "post-coatmg" An alternative strategy is to make gand-PEG-succinimidyl (active) ester, then react this with the primary amino groups in lipoplex (from DOPE) or polyplex (from PEI) for post-coating In this case, no prior modification of lipids or PEI to introduce a reactive group (e g , maleimide group of MB) is required The chemical or biochemical procedures involved in the invention are common techniques familiar to those skilled in the art
Since ligand-PEG is linked after the DNA has been condensed inside the liposomes, the PEG layer will coat the outside of the DNA-liposome complex and will not interfere with the internal structure of the DNA-liposome complex Therefore, the complex has a condensed DNA- cationic hpid structure or acentric onion-like core structure (see Example 6 below, detailing an electron microscopy analysis) inside and a PEG coating outside, with folate at the distal end of PEG
The ligand-PEG "post-coating" method takes advantage of the protective and long- circulating properties of the reported PEG-hposomes such as "sterically stabilized" liposomes or "Stealth™" liposomes, while keeping the unique characteristics of hgand-cationic liposome- DNA complexes The ligand-PEG post-coated liposomes have the capability of tumor-targeted gene delivery after systemic administration and are suitable for systemic gene therapy in vivo The invention is not limited to the use of any specific targeting ligand Folate is used as an example of a targeting ligand Other ligands can easily be used in the invention with minor modifications The ligand can be any ligand the receptor for which is differentially expressed on the target cell Examples include other vitamins, EGF, insulin, FGF, Heregulm, RGD peptides or other polypeptides reactive to lntegπn receptors, antibodies or their fragments In a preferred embodiment the ligand is folate Foi ligands unstable in oiganic phase e g , polypeptide oi protein ligands such as EGF, FGF, antibodies or their fragments, etc , the reactions to make the ligand-PEG-SH can be carried out in aqueous phase with minor modifications familiar to those skilled in the art.
The invention is not limited to the use of any specific coating polymer for "post-coating". The coating polymer can be any polymer with inert chemical activity within the polymer chain and active groups at the two ends for conjugation. Zalipsky S et al., US Patent No. 5,395,619
(74) disclosed, other than PEG, a series of polymers capable of forming long-circulating liposomes. In the preferred embodiment the polymer is PEG with primary amine groups at the two ends as active groups.
The invention is not limited to the delivery of any specific type of DNA. Any polynucleotides that can be complexed by cationic liposomes or polymers can be used in the invention. Examples of the polynucleotides include but are not limited to: plasmid DNA, DNA fragments, oligonucleotides, oligodeoxynucleotides, chimeric RNA/DNA oligonucleotides, RNA, ribozymes, etc. Anionic peptides, polymers, synthetic or natural molecules can also be used in the invention so long as they can be complexed by cationic liposomes or polymers. Other examples of molecules which can be delivered via ligand-PEG post coated cationic liposomes include a gene, high molecular weight DNA, plasmid DNA, an antisense oligonucleotide, peptide nucleic acids, a chemical agent such as a chemotherapeutic molecule or any large molecule including, but not limited to DNA, RNA, viral particles, growth factors, cytokines, immunomodulating agents and other proteins, including proteins which when expressed present an antigen which stimulates or suppresses the immune system.
The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art, that the techniques disclosed in the examples which follow represent techniques discovered by the inventors to function well in the practice of the invention, and thus can be considered to constitute preferred modes for practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure. Example 1 Preparation of ligand-PEG-SH for post-coating by SPDP method This example discloses the SPDP reaction procedure for preparation of ligand-PEG-SH for post-coating. This is illustrated in Figure 1 by the solid line flowchart. A. Preparation of PDP-PEG-NH2
30 μmol Polyoxyethylene bis(amine) (NH2-PEG-NH2) (M.W. 3350, Sigma) in 2 ml dry chloroform was stirred at room temperature while a solution of 30 μmol N-succinimido-3-(2- pyridyldithio)propionate (SPDP) (Sigma) in 1 ml dry chloroform was added dropwise over a period of 10-15 minutes. 10 μl TEA was added to the solution and stirred for an additional 15-30 minutes. This solution was purified to remove PDP-PEG-PDP or unreacted ΝH2-PEG-ΝH2 as described (48). The product was confirmed by thin layer chromatography (TLC) (chloroform/methanol/acetic acid 100/30/2) as described by Haselgrubler et al. (48).
B . Preparation of Folate-PEG-PDP
N-Hydroxysuccinimide ester of folic acid (folate-NHS) was prepared by reacting 1 gram folic acid (F, Sigma), in 30 ml dry dimethyl sulfoxide plus 0.5 ml triethylamine (TEA, Sigma), with 0.52 g N-hydroxysuccinimide (NHS, Sigma) in the presence of 0.94 g dicyclohexylcarbodiimide (DCC, Fluka) overnight at room temperature (75). The solution was filtered and purified as described by Lee RJ and Low PS (75). PDP-PEG-NH2 was reacted with 2-5 molar excess of folate-NHS in dry chloroform plus 2 molar of TEA overnight at room temperature. One volume of chloroform was added to the solution then washed with PBS 4-6 times, centrifuging at 1000-2000 rpm for 15 minutes between each wash. The clear yellow chloroform solution was evaporated to dryness to obtain the yellow powder product, folate-PEG- PDP. The product was confirmed by TLC (chloroform/methanol/acetic acid 100/30/2).
C. Reduction of PDP to obtain folate-PEG-SH Folate-PEG-PDP was dissolved in 10 mM HEPES buffer, pH 7.4. Freshly prepared 500 mM dithiothreitol (DTT, Sigma) in water was added to reach 50 mM final concentration. The solution was stirred for 30 minutes at room temperature and then desalted by column chromatography (10DG. BioRad) or by Centricon 3 ultrafiltration to obtain reduced folate- PEG-SH. T he free -SH group is not stable and will oxidize easily. Therefore, the reduction should be done immediately before "post-coating" and folate-PEG-SH should be used within 1 -2 hours after desalting. EDTA may be added to a 1 mM final concentration to help stabilize the folate-PEG-SH. The purified folate-PEG-SH can be frozen at -20°C under inert gas for later use. The free -SH group was confirmed by DTNB (5,5'-dithiobis-(2-nitrobenzoic acid)) titration (76). Folate was measured by absorption at 363 nm. The molar ratio of folate/-SH in the purified product was approximately 0.9-1.1.
Example 2 Preparation of ligand-PEG-SH for post-coating by direct thiolation (2-iminothiolane method) This example discloses the reaction procedure of preparing ligand-PEG-SH for post- coating by direct thiolation. This is illustrated in Figure 1 by the dashed line flowchart. Folate is linked to one end of PEG-bis(amine) first. 30 μmol Polyoxyethylene bis(amine)
(NH2-PEG-NH2) in 2 ml dry chloroform was mixed with 30 μmol N-Hydroxysuccinimide ester of folic acid (folate-NHS) in 1 ml dry chloroform. 10 μl TEA was added to the reaction solution and the mixture stirred for 0.5-2 hours at room temperature. One volume of chloroform was added to the solution, and then washed with PBS 4-6 times, centrifuging at 1000-2000 rpm for 15 minutes between each wash. The clear yellow chloroform solution was evaporated to dryness.
The product was purified to remove folate-PEG-folate or unreacted NH2-PEG-NH2 as described (48). The product was confirmed by thin layer chromatography (TLC) (chloroform/me hanol/ acetic acid 100/30/2) with iodine staining.
Free -SH is introduced to the other end of folate-PEG directly by reacting with 2- iminothiolane. The reaction is performed in aqueous solution. Folate-PEG-NH2 was dissolved in 100 mM HEPES, pH 8.0. 2-5 molar excess of 2-iminothiolane HCl (Sigma) in PBS was added and stirred for 0.5-2 hours at room temperature. The resulting folate-PEG-SH was purified by desalting column chromatography (10DG, BioRad) or by Centricon 3 ultrafiltration to obtain reduced folate-PEG-SH. The free -SH group was confirmed by DTNB titration and folate was measured by absorption at 363 nm. The molar ratio of folate/-SH in the purified product was approximately 0.9-1.1.
Example 3 "Post-coating" of cationic liposome-DNA complex (Lipoplex ) This example discloses the procedure of ligand-PEG-SH post-coating onto DNA- liposomes. A. Preparation of cationic liposomes
DOPE-MPB was prepared by reacting DOPE with succinimidyl-4-(/ maleimidophenyl)butyrate (SMPB, Sigma) in dry chloroform in the presence of TEA (65). The maleimide group of MPB is reactive to the sulfhydryl group and serves as the conjugating molecule on the liposomal surface. Other thiol-reactive groups can also be used here, such as other maleimido-containing molecules, etc. DOPE-MPB is also available from Avanti Polar Lipids, Inc., Alabaster, AL.
Cationic liposomes LipA were prepared as follows: a chloroform solution of 5 μmol dioleoyltrimethylammonium-propane (DOTAP), 5 μmol DOPE (Avanti Polar Lipids, Inc., Alabaster, AL) and 0.1-1 μmol DOPE-MPB were mixed together in a round-bottom flask, and the chloroform was evaporated under reduced pressure. 10 ml pure water, was added to the flask to suspend the lipids, then sonicated for 10 minutes in a bath-type sonicator at 4°C. The final concentration of the liposome was 1-2 nmol/μl total lipids. Other cationic liposomes were also prepared in similar fashion with the compositions shown below:
Table 1 Composition of cationic liposomes for post-coating
LipD and LipE were sonicated at 65 °C instead of 4°C.
Other liposome preparation methods can also be used to prepare the cationic liposomes. For example, the ethanol injection method modified from that described by Campbell (80) was used successfully in the present invention. In brief, all lipids were solubilized in ethanol and mixed, injected into vortexing pure water of 50-60°C with a Hamilton syringe. The solution was vortexed for a further 10-15 minutes. The final concentration was 1 -2 μM total lipids. The ethanol injection method is faster, easier and more robust. Since we have found that the maleimide group is not stable in aqueous solution with pH >7, the liposomes should be prepared in water (pH 5-6.5). The pH can be adjusted to 7.5-8.0 before "post-coating" with 1 M HEPES buffer, pH 7.5-8.0, to facilitate the post-coating reaction. B. "Post-coating" of DNA-liposomes complex Plasmid DNA was complexed with MPB-liposomes in water or 10 mM HEPES buffer, pH 7.0, as described (34) at DNA/lipid ratios 1/6-1/26 (μg/nmol), preferably 1/10-1/20. Before "post-coating", 1 M HEPES buffer, pH 7.5-8.0, was added to the MPB-liposome-DNA complexes to a final concentration of 10-20 mM. Folate-PEG-SH was added at 0.5-5 molar excess to MPB-DOPE, preferably 1 -2 molar excess and stirred several hours to overnight at room temperature in the dark. The resulting folate-PEG-LipA-DNA is used directly with 5% dextrose for in vivo gene transfection with pSVb as a reporter gene. For in vitro experiments, we tested the folate-PEG-Lip-DNA solution for transfection without further purification or after purification by Sepharose CL-4B chromatography to eliminate the unlinked folate-PEG-SH. Sepharose CL-4B chromatography confirmed that more than 50% of ligand-PEG-SH was linked to the lipoplex.
Example 4 In vitro gene transfection by "post-coated" folate-PEG-Lip-pSVb This example describes the in vitro gene transfection efficiencies of "post-coated" folate - PEG-liposomes using a reporter gene.
The medium used here was folate-free medium (RPMI-1640 folate-free, Gibco). 1 x 104 cells were plated in each well of a 96-well plate or 5 x 104 cells/well in a 24-well plate. 24 hours later, the cells were washed once with medium without serum or antibiotics and 100 μl transfection solution containing various amounts of folate-PEG-Lip-pSVb or PEG-Lip-pSVb as well as pSVb alone were added to each well. After 5 hours of transfection at 37°C, an equal amount of medium containing 20% fetal bovine serum were added to each well. 48 hours later, the cells were washed once with PBS and lysed in IX reporter lysis buffer (Promega). The cell lysates were treated with 100 μl 150 μM O-nitrophenyl-β-galactopyranoside in 20 mM Tris (pH 7.5) containing 1 mM MgCL and 450 mM β-mercaptoethanol at 37°C for 0.5 hour. The reaction was stopped by the addition of 150 μl/well of 1 M Na2C03. The absorbance was determined at
405 nm. Purified β-galactosidase (Boehringer) was used as a standard. The results were expressed as milliUnits of β-galactosidase equivalent per mg of total protein. For histochemical studies of ligand-liposome-pCMVb transfection, 60% confluent cells in 24-well plates were transfected for 5 hours as described above. After an additional 2 days in culture, the cells were fixed and stained with X-gal. Transfection efficiency was calculated as the percentage of blue- stained cells.
Figure 2 shows the in vitro folate-PEG-liposome-mediated transfection of JSQ-3 cells and the effect of serum on the transfection efficiency. PEG "post-coated" liposomes show resistance to serum and folate-PEG-LipA resulted in two-fold higher reporter gene expression than LipA- PEG without folate ligand. Figure 3 shows the specificity of folate-PEG-liposome transfection. Free folic acid can block the transfection activity of folate-PEG-liposomes to the level of the LipA-PEG, demonstrating that the transfection of the folate-PEG-liposomes is mediated by the folate through the folate-receptor.
Example 5
In Vivo Gene Transfection in Nude Mouse Tumor Model by i.v. Injection of the "post-coated" folate-PEG-Lip-pSVb This example demonstrates the ability of the "post-coated" folate-PEG-Lip-pSVb to selectively target tumor tissue in vivo after systemic administration. NIH3T3 cells transformed with a human Ha-Ras gene were subcutaneously injected in the flank of 4-6 week old female nude (NCr nu-nu) mice. The tumors were allowed to develop to a size of 100 mnr\ The "post-coated" folate-PEG-Lip-pSVb were prepared as described in Examples 1 -3, wherein LipA-PEG 1 -F indicates the "post-coated" folate-PEG-Lip-pSVb prepared by SPDP method (Examples 1 and 3) and LipA-PEG2-F indicates that prepared by the direct thiolation method (Examples 2 and 3). LipA-PEG 1-F, LipA-PEG2-F or pSVb plasmid alone (in
5% dextrose) was injected intravenously via the tail vein, at 50 μg of plasmid DNA/300 μl/animal. Three days and 10 days after DNA injection, the tumors as well as mouse organs were excised, cut into 1 mm sections, washed once with PBS, and fixed with 2% Formaldehyde-0.2% Glutaraldehyde for 4 hours at room temperature. The fixed tumor sections were washed 4 times, each for 1 hour, and stained with X-Gal solution plus 0.1% NP-40 (pH 8.5) at 37°C overnight.
The stained tumor sections were embedded and sectioned using normal histological procedures and counter-stained with nuclear fast red. Four sections per tumor were examined to evaluate the β-galactosidase gene expression, as indicated by the blue stained cells.
Figures 4A-I show the "post-coated" folate-PEG-liposome-mediated systemic gene transfection in vivo. As shown in Figures 4A and 4B, 3 days after the i.v. injection of LipA-PEG- F complexed with pSVb, 30-60%. of tumor cells in nude mouse xenograft stained blue, indicating the tumor delivery and expression of reporter gene β-galactosidase. The normal mouse tissues, including lung and liver, were not stained blue, except a few macrophages (Figures 4G and 4H), demonstrating that the PEG "post-coated" liposomes can deliver the complexed gene selectively to tumors in vivo. Although the folate-liposome without PEG coating showed 30-40% tumor cell blue-staining (Figure 4C), the gene expression disappeared within 6-10 days (Figure 4F). But the PEG "post-coated" liposomes injected groups still showed some (10-40%) blue-staining in the tumors after 10 days, demonstrating that the ligand-PEG "post-coated" liposomes can last longer after systemic injection and give sustained gene expression in the tumors. Plasmid pSVb only injected group showed no tumor cells staining blue (Figure 41). The ligand-PEG "post-coated" liposomes can deliver the complexed genes selectively to the tumor in vivo after systemic administration, and give sustained gene expression in the tumor, with an in vivo transfection efficiency of 30-60%o. Therefore, this system is very useful and promising for in vivo gene delivery and gene therapy.
Example 6
Electron microscopic analysis of the ligand-PEG "post-coated" cationic liposomes
Liposomes can be observed with Electron Microscopy (EM), either Transmission
Electron Microscopy (TEM) with negative staining or Scanning Electron Microscopy (SEM).
EM can reveal the structure and size distribution of liposomes. EM can also be used for quality control of liposomal preparation. We observed the ligand-cationic liposomes under Transmission
Electron Microscope with negative staining.
The copper grid with Formvar and Carbon coating (Electron Microscopy Sciences, Fort
Washington, PA) was used in the study. Ligand-PEG "post-coated" cationic liposome-pSVb complexes were prepared as described in Examples 1 -3. One drop of the liposome complex was added to the grid. 5 minutes later, excess liquid was removed by attaching a filter paper to the edge of the grid. One drop of 4% Uranium Acetate was then added onto the grid for negative staining. 5 minutes later, excess liquid was also removed by attaching a filter paper to the edge of the grid. The grid was air dried at room temperature for 15 minutes before being put into the sample chamber of the TEM. The TEM JEOL-1200EX was used in the study according to the manufacturer's instruction. Photos were taken at magnitudes of 10-50k, 60 kVolt. The liposome samples on the grid were prepared and stained freshly and observed within one hour.
Many publications indicate that cationic liposome-DNA complexes have a diverse structure and size ranging from 100 nm to 1000 nm. In our study, we observed unexpectedly that the ligand-liposome-DNA complexes disclosed in the invention have much smaller size and much more even size distribution. Cationic liposome LipA itself has a size of 25-50 nm, average 35 nm (in diameter). When DNA was complexed with LipA, an interesting "irregular or acentric onion-like core structure" in the core of the LipA-DNA complex was observed, with evenly distributed sizes of 35-65 nm (50 nm in average). After the ligand-PEG "post-coating" process, the size of liposomes increased only slightly, becoming 35-80 nm (60 nm in average), indicating a 10-15 nm thick layer of folate-PEG was "post-coated" on the LipA-DNA complex. To reach target tumor in vivo, the liposomes must be first resistant to serum and then pass through the blood vessel (capillary) wall. The ligand-PEG "post-coated" liposomes disclosed in the invention can meet these two requirements. The ligand-PEG-Lip-DNA is very resistant to serum as shown in Example 4. Using fluorescence labeling and colloidal gold labeling with silver enhancement, it has been demonstrated that 80-100 nm-size PEG-stabilized liposomes could penetrate into the extravascular, interstitial space among tumor calls and scatter throughout the entire tumor region, in several solid tumor models with i.v. injection of the liposomes (77- 79). Therefore, the ligand-PEG "post-coated" liposome-DNA complexes of 35-80 nm size disclosed in the invention could pass through the capillary wall to reach the target, as confirmed by the in vivo gene transfection data disclosed in Example 5.
Example 7 Stability assay of the ligand-PEG "post-coated" liposomes Stability is a critical issue for liposomal pharmaceuticals. Liposome solutions should be stable for an extended period of time after preparation to allow for shipment and storage, without significant loss of their biological/pharmaceutical activities, to be useful as therapeutic agents.
In light of the future clinical use of the ligand-liposome-therapeutic molecule complex of this invention, in this example we examined the stability of the ligand-liposomes and the ligand- liposome-DNA complexes. The ligand-PEG "post-coated" liposome-pSVb complexes were stored under nitrogen in the dark at 4°C for various periods of time, up to 12 months. On the day of the assay, the stored liposomes, as well as freshly prepared liposomes, were used to transfect JSQ-3 cells using the transfection assay as described in Example 4.
LipA-DNA or folate-LipA-DNA complexes without PEG coating lost their transfecting activity in 1-3 days. When ligand-PEG was "post-coated", the liposomes became stabilized and retained transfecting activity for a prolonged period of time. 100% of their activity was evident for one month and 50-60% transfecting activity still remained after 6 months in storage. Therefore, the ligand-PEG "post-coated" cationic liposomes disclosed in the invention are more stable and have a longer shelf life, which are important considerations for practical use.
Example 8 Preparation of Galactose-PEG post-coated polye hylenimine-DNA complex (polyplex) This example discloses a procedure to prepare a galactose-PEG post-coated PEI-DNA complex. Galactose or galactosylated ligands have high affinity to asialoglycoprotein receptor which is present at high density on hepatocytes and hepatomas (13). Galactose-PEG post-coated PEI-DNA complex will be useful for targeted gene delivery to liver for gene therapy of liver diseases. A. Modification of PEIs
Branched PEI with average molecular weight of 25 kDa (designated P25) was purchased from Aldrich. Linear PEI with average molecular weight of 25 kDa (designated LP25) was purchased from Polysciences, Inc. Both PEIs were reacted with maleimide-containing active ester to introduce maleimide group, similar to that of DOPE-MB in Example 3. The followings are examples of (but not limited to) the maleimide-containing active esters that can be used: 3-
Maleimidobenzoic acid N-hydroxysuccinimide ester (MBS) (Sigma M2786), Succinimidyl 4-(p- maleimidophenyl)butyrate (SMPB) (Sigma M6286) and Succinimidyl 4-(N- maleimidomethyl)cyclohexane-l -carboxylate (SMCC) (Sigma M5525). 0.1-0.2 g P25 in 3 ml dry chloroform (LP25 in 2 ml chloroform and 1 ml DMSO) was added to 50 mg MBS/1 mi DMSO and 100 μl triethylamine (Sigma). The mixture was stirred overnight at room temperature. 10 ml pure water was added, vortexed and centrifuged (5000 rpm, 10 minutes). The modified PEI, P25-Maleimidobenzoyl (MB), was solubilized in the aqueous phase, while LP25-MB was a white or light yellow solid between the two phases. The P25-MB was purified by a 10-DG desalting column (BioRad) equilibrated with water. The solid LP25-MB was taken out, washed twice with water, dried and weighed. The solid LP25-MB was solubilized in water by adding 1 N HCl dropwise until totally clear.
B. Preparation of Galactose-PEG-PDP
This procedure is similar to that of F-PEG-PDP in Example 1. 30 μmol Polyoxyethylene bis(amine) (NH2-PEG-NH2) (M.W. 3350, Sigma) in 2 ml dry chloroform was stirred at room temperature. A solution of 30 μmol N-succinimido-3-(2-pyridyldithio)propionate (SPDP) (Sigma) in 1 ml dry chloroform was added dropwise over a period of 10- 15 minutes. 10 μl TEA was added to the solution and stirred for an additional 15-30 minutes. The obtained PDP-PEG- ΝH2 was then reacted with 1 -3 molar excess of D-galactopyranosylphenyl isothiocyanate (Sigma) in dry chloroform plus 2-6 molar of TEA overnight at room temperature. One volume of chloroform was added to the solution then washed with PBS 4-6 times, centrifuging at 1000- 2000 rpm 15 minutes between each wash. The clear chloroform solution was evaporated to dryness to obtain the product, Galactose-PEG-PDP (Gal-PEG-PDP). The product was confirmed by TLC (chloroform/methanol/ acetic acid 100/30/2).
C. Post-coating polyplex
The Gal-PEG-PDP was reduced by DTT similar to that of F-PEG-PDP as described in Example 3. Polyplexes were prepared by adding DNA into P25-MB or LP25-MB solution in water or 10-150 mM NaCl, at a N/P ratio (amine nitrogen of PEI vs. phosphate of DNA, molar ratio) 1-50, preferably 5-15, as described (66, 67). After addition of 1 M HEPES buffer, pH 7.5- 8.0, to a final concentration of 10-20 mM, to adjust to pFI 7.4-8.0, the freshly reduced ligand- PEG-SH (F-PEG-SH or Gal-PEG-SH) is added to the polyplex and reacted for 2-6 hours at room temperature or overnight at 4°C. The post-coated polyplex can be used directly or purified by
Sepharose CL-4B chromatography to get rid of the unconjugated ligand-PEG. Sepharose CL-4B chromatography confirmed that more than 50%> of ligand-PEG-SH was linked to the polyplex. 50%) dextrose was added to a final concentration of 5% for i.v. administration. Example 9 Ligand-PEG post-coating stabilized polyplex mediated transfection in vitro and in vivo
This example demonstrates the in vitro and in vivo transfection activity of F-PEG and Gal-PEG post-coating stabilized polyplexes.
The plasmid, pLuc, which contains the firefly luciferase gene under CMV promoter was used in the study (35). The polyplex P25-pLuc was prepared by adding P25-MB (10 mM of monomer in water, pH 7.4 by HCl) to the pLuc DNA solution while vortexing and vortexed an additional 1 minute, at a N/P ratio 6-12. The F-PEG and Gal-PEG were post-coated to the polyplex as described in Example 8. The in vitro transfection was performed in 24-well plates, as described in Example 4 and references 27 and 35. The transfection reagent solutions were added to the cells, JSQ-3, in the presence of 10% serum. 24 hours later, the cells were washed and lysed to measure the luciferase activity and protein concentration (35). The results are expressed as 103 relative light units (RLU) per μg protein in the lysate, as shown in Table 2.
Table 2
Ligand-PEG Post-coated Polyplex Mediated
Transfection of JSQ-3 Cells In Vitro in the Presence of Serum
The results show that the ligand-PEG post-coated polyplexes have very high transfection activity in the presence of serum, significantly higher than non-coated polyplex. Also it is worthy to note that the ligand-PEG post-coated polyplexes were much less cytotoxic than non-coated polyplex P25-pLuc, as indicated by the protein assay and observation under a microscope.
For the in vivo study, C57BK 6 black mice were i.v. injected with the ligand-PEG post- coated polyplexes as well as non-coated polyplexes, at 80-100 μg DNA/mouse in 0.4-0.5 ml/injection via tail vein. 24 hours later, the organs were excised and homogenized in IX lysis buffer (Promega). The luciferase activities and protein concentrations were measured as described (35), and the results are expressed as 103 RLU/mg protein as shown in Table 3. The Gal-LP25 and Gal-P25 were galactosylated PEIs prepared according to Zenta et al. (70) and Bandyopadhyay et al. (71). All polyplexes were prepared at the same N/P = 10.
Table 3 Systemic Gene Delivery by Ligand-PEG Post-coated Polyplex in C57BL/6 Mice
Table 3 shows that polyplexes without PEG coating deliver genes predominantly to the lungs and little to liver, spleen or other organs (data not shown). When PEIs were galactosylated, the reporter gene expression in liver was increased a little and that in lung was reduced, but not enough to be used for liver-targeted gene delivery (liver/lung = 0.5 or 0.03). However, when the polyplex was post-coated by Gal-PEG, significant reporter gene expression in the liver was observed, much more than that in the lungs (liver/lung = 624). The results demonstrated that the ligand-PEG post-coating can stabilize and shield the polyplex in vivo to survive the lung's first filtration effect (like other PEG-coated stealth vectors), and can deliver the carried gene selectively to the target tissue. The complexes prepared by this Gal-PEG post-coating method can deliver DNA selectively to the liver after systemic administration, and would be useful for liver-targeted gene therapy.
At DNA doses of 80-100 μg/mouse, we observed in the experiments that PEI-DNA polyplexes were very toxic to mice after i.v. injection, especially P25. All mice injected with P25 polyplex died within 12 hours. Even P25 modified by F-NHS (F-P25) or galactosylated (Gal-
P25) showed similar toxicities (2/2 and 3/4 died within 12 hours, respectively). The only mouse which survived in the Gal-P25 group was severely ill and the anatomy showed an enlarged and pale liver. Histology showed liver lesions similar to that of acute hepatitis. All mice injected with F-PEG post-coated polyplexes survived and showed no sign of any of the above toxicities. The Gal-PEG post-coated polyplexes had reduced toxicity and anatomy showed no sign of liver lesions. The results demonstrated that the ligand-PEG post-coating can reduce the toxicity of polyplexes, which is promising for the clinical development of PEI-based gene delivery systems. Example 10 Characterization of ligand-PEG for post-coating This example shows the results of characterization of F-PEG-PDP and Gal-PEG-PDP for post-coating. A. Ultraviolet Spectrophotogram
The purified F-PEG-PDP and Gal-PEG-PDP were scanned, before and after adding DTT, by a Beckman DU640 spectrophotometer from 200 nm to 700 nm. Figures 5A-D show the spectrophotograms of the scans. Figure 5A shows the scan of F-PEG-PDP. There is a 363 nm peak of folate and a 280 nm peak of PEG. After reduction by DTT (Figure 5B), a new peak at 342 nm arises, representing the cleaved 2-thiopyridone from the PDP group. Figure 5C shows a 240 nm peak of galactose as well as a 280 nm peak of PEG. After reduction by DTT (Figure 5D), the 342 nm 2-thiopyridone peak shows up. The results confirm that both PDP and the ligands were conjugated to PEG and the PDP was functional and can be reduced.
REFERENCES CITED
1. Meng RD and EL-Deiry WS. (1999). Tumor suppressor genes as targets for cancer gene therapy. In: Lattime EC and Gerson SL, ed., Gene Therapy of Cancer, Academic Press, San Diego, CA, 3-20
2. Zhang, W., Fujiwara, T., Grimm, E.A., & Roth, J.A.. Advances in Cancer Gene Therapy. Advances in Pharmacology 1997, 32, 289-333.
3. Chen QR and Mixson JA, ( 1998). Systemic gene therapy with p53 inhibits breast cancer: recent advances and therapeutic implications. Front Biosci 3:D997-D1004
4. Huang L and Viroonchatapan E. (1999). Introduction. In: Non-viral Vectors for Gene Therapy. Huang L. Hung MC and Wagner E. eds., Academic Press, San Diego, CA. 3-22.
5. Ledley, F. D. (1995). Nonviral gene therapy: the promise of genes as pharmaceutical products. Hum Gene Ther, 6(9), 1 129-1 144.
6. Feigner PL, Tsai YJ, Sukhu L, Wheeler CJ, Manthorpe M, Marshall J, Cheng SH. Improved cationic lipid formulations for in vivo gene therapy. Ann N Y Acad Sci 1995; 772:126-139.
7. Lewis JG, Lin KY, Kothavale A, Flanagan WM, Matteucci MD, De Prince RB, Mook RA, Hendren RW, Wagner RW. A serum-resistant cytofectin for cellular delivery of antisense oligodeoxynucleotides and plasmid DNA. Proc Natl Acad Sci U S A 1996; 93:3176-3181.
8. Aoki K, Yoshida T. Sugimura T, Terada M. Liposome-mediated in vivo gene transfer of antisense K-ras construct inhibits pancreatic tumor dissemination in the murine peritoneal cavity. Cancer Research, 1995, 55, 3810-3816.
9. Feigner PL. Progress in gene delivery research and development. In: Non-viral Vectors for Gene Therapy. Huang L, Hung MC and Wagner E. eds., Academic Press, San Diego, CA. 26-38.
10. Xu M, Kumar D, Srinivas S, Detolla LJ, Yu SF, Stass SA, Mixson AJ. Parenteral gene therapy with p53 inhibits human breast tumors in vivo through a bystander mechanisim without evidence of toxicity. Human Gene Therapy 1997; 8:177-185.
1 1 . Hsiao M. Tse V. Carmel J, Tsai Y. Feigner PL, Haas M, Silverberg GD. Intracavitary liposome-mediated p53 gene transfer into glioblastoma with endogenous wild-type p53 in vivo results in tumor suppression and long-term survival. Biochem Biophys Res Commun 1997 Apr 17; 233:359-364.
12. Thierry, A.R., Lunardi-Iskandar, Y., Bryant, J.L., Rabinovich, P., Gallo, R.C., & Mahan, L.C. (1997). Systemic Gene Therapy: Biodistrobution and long-term expression of transgene in mice. Proc Natl Acad Sci, 92, 9742-9746.
13. Wagner E. Ligand-polycation conjugates for receptor-targeted gene transfer. In: Non-viral Vectors for Gene Therapy. Huang L, Hung MC and Wagner E. eds., Academic Press, San Diego, CA. 208-227.
14. Christiano RJ, Curiel DT. Strategies to accomplish gene therapy via the receptor- mediated endocytosis pathways. Cancer Gene Therapy 1996; 3:457-497.
15. Cheng PW. Receptor ligand- facilitated gene transfer: enhancement of liposome-mediated gene transfer and expression by transferrin. Hum Gene Ther 1996; 7:275-282.
16. Lee RJ, Huang L. Folate-targeted, anionic liposome-entrapped polylysine-condensed dna for tumor cell-specific gene transfer. J Biol Chem 1996; 271 : 8481-8487.
17. Campbell IG, Jones TA, Foulkes WD, Trowsdale J. Folate-binding protein is a marker for ovarian cancer. Cancer Res 1991 ; 51 :5329-5338.
18. Weitman SD, Lark RH, Coney LR, Fort DW, Frasca V. Zurawski VR, Kamen BA. Distribution of the folate receptor GP38 in normal and malignant cell lines and tissues. Cancer Res 1992; 52:3396-3401.
19. Rochman H, Selhub J, Karrison T. Folate binding protein and the estrogen receptor in breast cancer. Cancer Detect Prev 1985;8(l-2):71-75
20. Pinard MF, Jolivet J, Ratnam M, Kathmann I, Molthoff C, Westerhof R, Schornagel JH, Jansen G. Functional aspects of membrane folate receptors in human breast cancer cells with transport-related resistance to methotrexate. Cancer Chemother Pharmacol 1996, 38(3):281-288
21. Holm J, Hansen SI, Sondergaard K, Hoier-Madsen M. The high-affinity folate binding protein in normal and malignant mammary gland tissue. Adv Exp Med Biol 1993;338:757-760
22. Holm J, Hansen SI, Hoier-Madsen M, Sondergaard K, Bzorek M. Folate receptor of human mammary adenocarcinoma. APMIS 1994 Jun;102(6):413-419 23. Kamen BA, Wang MT, Steckfuss AJ, Peryea X, Anderson RGW. Delivery of folates to the cytoplasm of MAI 04 cells is mediated by a surface membrane receptor that recycles. J Biol Chem 1988; 263:13602-13609.
24. Lee R.J, Low PS. Folate-mediated tumor cell targeting of liposome-entrapped doxorubicin in vitro. Biochim Biophys Acta, 1995, 1233, 134-144.
25. Gottschalk S, Cristiano RJ, Smith LC, Woo SL. Folate receptor mediated DNA delivery into tumor cells: potosomal disruption results in enhanced gene expression. Gene Ther, 1994, 1 : 185-191.
26. Antony AC.(1996) Folate receptors. Annu Rev Nutr; 16, 501-521.
27. Xu L, Pirollo KF, Rait A, Murray AL and Chang EH. Systemic p53 gene therapy in combination with radiation results in human tumor regression. Tumor Targeting 1999, 4, 92-104.
28. Papahadjopoulos D. Stealth liposomes: from steric stabilization to targeting. In: Stealth Liposomes, Lasic DD and Martin FJ, eds., 1995, CRC Press, Inc., Boca Raton, FL. 1-6.
29. Gabizon A, Horowitz AT, Goren D, Tzemach D, Mandelbaum-Shavit F, Qazen MM, Zalipsky S. Targeting folate receptor with folate linked to extremities of poly(ethylene glycol)-grafted liposomes: in vitro studies. Bioconjug Chem 1999 Mar-Apr;10(2):289-98
30. Hong K, Zheng W, Baker A, Papahadjopoulos D. Stabilization of cationic liposome- plasmid DNA complexes by polyamines and poly(ethylene glycol)-phospholipid conjugates for efficient in vivo gene delivery. FEBS Lett 1997; 400(2):233-237
31. Mok KW, Lam AM and Cullis PR. Stabilized plasmid-lipid particles: factors influencing plasmid entrapment and transfection properties. Biochim Biophys Acta 1999, 1419(2):137-50
32. Wheeler JJ, Palmer L, Ossanlou M, MacLachlan I, Graham RW, Zhang YP, Hope MJ, Scherrer P and Cullis PR. Stabilized plasmid-lipid particles: construction and characterization. Gene Ther 1999, 6(2):271-81
33. Wang S, Lee RJ, Cauchon G, Gorenstein DG, Low PS. Delivery of antisense oligodeoxyribonucleotides against the human epidermal growth factor receptor into cultured KB cells with liposomes conjugated to folate via polyethylene glycol. Proc Natl Acad Sci U S A 1995, 92(8):3318-3322. 34. Xu, L., Pirollo, K.F.. and Chang, E.H., Transferrin-Liposome-Mediated p53 Sensitization of Squamous Cell Carcinoma of the Head and neck to Radiation In Vitro. Human Gene Therapy, 8, 467-475 (1997).
35. Xu, L., Pirollo, K.F., Tang, W-H., Rait, A. and Chang, E.H. Transferrin-liposome- mediated Systemic p53 Gene Therapy in Combination with Radiation Results in Regression of Human Head and Neck Cancer Xenografts. Human Gene Therapy 1999, 10:2941-2952.
36. Tranum-Jensen J. Electron microscopy: assays involving negative staining. Methods Enzymol 1988,165:357-374.
37. Templeton NS, Lasic DD, Frederik PM, Strey HH, Roberts DD, Pavlakis GN. Improved DNA: liposome complexes for increased systemic delivery and gene expression. Nat Biotechnol 1997, 15(7):647-52
38. Lasic DD. Structure and structure-activity relationships of lipid-gased gene delivery systems. In: Non-viral Vectors for Gene Therapy. Huang L, Hung MC and Wagner E. eds., Academic Press, San Diego, CA. 69-89.
39. Sternberg B, Hong K, Zheng W, Papahadjopoulos D. Ultrastructural characterization of cationic liposome-DNA complexes showing enhanced stability in serum and high transfection activity in vivo. Biochim Biophys Acta 1998, 1375(l-2):23-35
40. Dvorak HF, Nagy JA, Feng D, Brown LF, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol 1999;237:97-132
41. Feng D, Nagy JA, Pyne K, Hammel I, Dvorak HF, Dvorak AM. Pathways of macromolecular extravasation across microvascular endothelium in response to VPF/VEGF and other vasoactive mediators. Microcirculation 1999 Mar;6(l):23-44
42. Jin AJ, Huster D, Gawrisch K and Nossal R. Light scattering characterization of extruded lipid vesicles. Eur Biophys J. 1999, 28(3): 187- 199.
43. Korgel BA, van Zanten JH and Monbouquette HG. Vesicle size distributions measured by flow field-flow fractionation coupled with multiangle light scattering. Biophys J 1998, 74(6):3264-72 44. Battersby BJ, Grimm R, Huebner S and Cevc G. Evidence for three-dimensional interlayer correlations in cationic lipid-DNA complexes as observed by cryo-electron microscopy. Biochim Biophys Acta 1998, 1372(2):379-383.
45. Skalko N, Bouwstra J, Spies F, Stuart M, Frederik PM and Gregoriadis G. Morphological observations on liposomes bearing covalently bound protein: studies with freeze-fracture and cryo electron microscopy and small angle X-ray scattering techniques. Biochim Biophys Acta 1998, 1370(1): 151 -160
46. Kuhl TL, Majewski J, Wong JY, Steinberg S, Leckband DE, Israelachvili JN and Smith GS. A neutron reflectivity study of polymer-modified phospholipid monolayers at the solid-solution interface: polyethylene glycol-lipids on silane-modified substrates. Biophys J 1998, 75(5):2352-2362.
47. Kawaura C, Noguchi A, Furuno T and Nakanishi M. Atomic force microscopy for studying gene transfection mediated by cationic liposomes with a cationic cholesterol derivative. FEBS Lett 1998, 421(l):69-72
48. Haselgrubler T, Amerstorfer A, Schindler H and Gruber HJ. Synthesis and applications of a new poly(ethylene glycol) derivative for the crosslinking of amines with thiols. Bioconjug Chem 1995, 6(3):242-248
49. Zalipski S. Polyethylene glycol-lipid conjugates. In: Stealth Liposomes, Lasic DD and Martin FJ, eds., 1995, CRC Press, Inc., Boca Raton, FL. 93-102.
50. Price, J.E., Polyzos, A., Zhang, R.D., & Daniels, L.M. (1990). Tumorigenicity and Metastasis of Human Breast Carcinoma Cell Lines in Nude Mice. Cancer Research, 50, 717-721.
51. Couffinhal T, et al. Histochemical staining following lacZ gene transfer underestimates transfection efficiency. Hum Gen Ther. 8:929-934. (1997).
52. Barron LG, Gagne L and Szoka FC Jr. Lipoplex-mediated gene delivery to the lung occurs within 60 minutes of intravenous administration. Hum Gene Ther 1999, 10(10): 1683-1694.
53. Noguchi A, Furuno T, Kawaura C and Nakanishi M. Membrane fusion plays an important role in gene transfection mediated by cationic liposomes. FEBS Lett 1998, 433(1 -2): 169-173. 54. Song Y.K, Liu F, Chu S and Liu D. Characterization of cationic liposome-mediated gene transfer in vivo by intravenous administration. Hum Gene Ther 1997, 8(13): 1585-1594.
55. Sharma US, Sharma A, Chau RI and Straubinger RM Liposome-mediated therapy of intracranial brain tumors in a rat model. Pharm Res 1997, 14(8):992-998.
56. Niks M and Otto M. Towards an optimized MTT assay. J Immunol Methods 1990, 130(1): 149-151.
57. Ruf H, Georgalis Y and Grell E. Dynamic laser light scattering to determine size distributions of vesicles. Methods Enzymol 1989;172:364-390.
58. Friend DS, Papahadjopoulos D and Debs RJ. Endocytosis and intracellular processing accompanying transfection mediated by cationic liposomes. Biochim Biophys Acta 1996, 1278:41-50.
59. Noguchi A, Furuno T, Kawaura C, Nakanishi M. Membrane fusion plays an important role in gene transfection mediated by cationic liposomes. FEBS Lett 1998 Aug 14;433(1 - 2): 169-73
60. Miller CR, Bondurant B, McLean SD, McGovern KA, O'Brien DF. Liposome-cell interactions in vitro: effect of liposome surface charge on the binding and endocytosis of conventional and sterically stabilized liposomes. Biochemistry 1998 Sep 15;37(37): 12875-83
61. Hui SW, Langner M, Zhao YL, Ross P, Hurley E and Chan K. The role of helper lipids in cationic liposome-mediated gene transfer. Biophys J 1996, 71 :590-599.
62. Dunphy EJ, Redman RA, Herweijer H and Cripe TP. Reciprocal enhancement of gene transfer by combinatorial adenovirus transduction and plasmid DNA transfection in vitro and in vivo. Hum Gene Ther 1999, 10(14):2407-1417.
63. Tseng WC, Haselton FR and Giorgio TD. Transfection by cationic liposomes using simultaneous single cell measurements of plasmid delivery and transgene expression. J Biol Chem 1997, 272(41):25641-25647.
64. Cremaschi D, Porta C, Ghirardelli R, Manzoni C, Caremi I. Endocytosis inhibitors abolish the active transport of polypeptides in the mucosa of the nasal upper concha of the rabbit. Biochim Biophys Acta 1996; 1280:27-33.
65. Heath TD. Covalent attachment of proteins to liposomes. Methods Enzymol 1987;149: 1 1 1. 66. Boussif et al. Proc. Natl. Acad. Sci. USA 1995, 92, 7297-7301
67. Erbacher et al. Gene Ther 1999, 6, 138-145
68. Ogris et al. Gene Ther 1999, 6, 595-605
69. Kircheis et al, J Gene Med 1999, 1, 1 1 1-120
70. Zenta et al. Bioconj Chem 1997, 8:839-844
71. Bandyopadhyay P, et al. J Biol Chem 1999, 274:10163-10172.
72. Zalipsky et al. Biochemical Conjugates 1997, 8(2), 1 11-1 18.
73. Godbey et al. J Controlled Release 1999, 60, 149-16
74. US Patent No. 5,395,619.
75. Lee RJ and Low PS. J Biol Chem 1994; 269:3198-3204
76. Reddy YV, Rao DN, J Biol Chem 1998; 273(37):23866-76
77. Huang, SK, et al. Biochim Biophys Acta 1991 ; 1069: 1 17.
78. Huang, SK, et al. Cancer Res 1992; 52:5135-5143.
79. Stealth Liposomes, Lasic D. and Martin F. ed., 1995; 1 19-125.
80. Campbell MJ Biotechniques 1995; 18(6): 1027-1032.

Claims

WHAT IS CLAIMED IS:
1. A method of preparing a post-coated cationic vehicle for targeted delivery of a therapeutic molecule comprising the steps of:
(a) complexing said therapeutic molecule with said cationic vehicle to form a cationic vehicle-therapeutic molecule complex;
(b) conjugating a polymer with a targeting ligand to form a polymer-ligand complex wherein said polymer is conjugated to said ligand; and
(c) conjugating said preformed cationic vehicle-therapeutic molecule complex with said polymer-ligand complex wherein said polymer-ligand complex will coat said preformed cationic vehicle-therapeutic molecule complex thereby forming said post-coated cationic vehicle for targeted delivery of said therapeutic molecule.
2. The method of claim 1 wherein said cationic vehicle is a cationic liposome, a cationic polymer, polylysine, protamine or polyethyleneimine.
3. The method of claim 1 wherein said cationic vehicle comprises i) dioleoyltrimethylammonium-propane (DOTAP), dioleoylphosphatidylethanolamine (DOPE) and DOPE-maleimidophenyl butyrate (DOPE-MPB); ii) dimethyldioctadecylammonium bromide (DDAB), DOPE and DOPE-MPB; iii) DOTAP, cholesterol and DOPE-MPB; or iv) DDAB, cholesterol and DOPE-MPB.
4. The method of claim 3 wherein said cationic vehicle further comprises cholesterol.
5. The method of claim 2 wherein said cationic vehicle comprises 1-10 mole percent of a reactive linking group.
6. The method of claim 5 wherein said reactive linking group is a maleimido group.
7. The method of claim 6 wherein said maleimido group is maleimidophenyl butyrate.
8. The method of claim 1 wherein said polymer-ligand complex comprises a reactive group.
9. The method of claim 8 wherein said reactive group is a sulfhydryl.
10. The method of claim 1 wherein said ligand is a sugar, vitamin, protein, antibody or growth factor.
11. The method of claim 10 wherein said ligand is folate, galactose, peptide, polypeptide or antibody fragment.
12. The method of claim 1 wherein said ligand is folate.
13. The method of claim 1 wherein said polymer is polyethyleneglycol, polyvinylpyrrolidone, p o l yv i ny l m ethy l ether , p o l yhydro x ypropy l meth acryl ate , polyhydroxylpropylmethacrylamide, polyhydroxyethyl acrylate, polymethacrylamide, polydimethylacrylamide, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyloxazoline, polyhydroxypropyloxazoline, or polyaspartamide.
14. The method of claim 1 wherein said polymer is polyethyleneglycol.
15. The method of claim 1 wherein said cationic vehicle is polyethyleneimine and wherein 0.1 -25% of amines in said polyethyleneimine are bound to a sulfhydryl reacting molecule.
16. The method of claim 15 wherein DNA and polyethyleneimine are at a ratio of N to P of 1-50.
17. The method of claim 1 wherein said post-coated cationic vehicle has a diameter smaller than 80 nm.
18. The method of claim 1 wherein said post-coated cationic vehicle has a diameter in the range 35-80 nm.
19. The method of claim 1 wherein said post-coated cationic vehicle has an average diameter of approximately 60 nm.
20. The method of claim 1 wherein said ligand is covalently bound directly to said polymer.
21. The method of claim 1 wherein said therapeutic molecule is a gene, a high molecular weight DNA, plasmid DNA, an antisense oligonucleotide, a peptide nucleic acid, a chemical agent, DNA, RNA, ribozyme, CpG sequences, a viral particle, a growth factor, cytokine, an immunomodulating agent, or a protein which stimulates or suppresses the immune system.
22. A method of preparing a ligand-polymer complex capable of covalently binding with a reactive group on a cationic vehicle, comprising the steps of: a) selecting a polymer with two ends which has a functional moiety at each of the two ends, wherein the functional moieties can be the same or different; b) adding a chemical entity comprising a moiety capable of being converted to a reactive crosslinker a first end of said polymer of step (a); c) adding said ligand to a second end of said polymer of step (a); and d) converting said moiety to reactive moiety; wherein said reactive moiety can form a covalent bond with a reactive group.
23. The method of claim 22 wherein said functional moiety is selected from the group consisting of amine, sulfhydryl, carboxyl and hydrazide.
24. The method of claim 22 wherein said chemical entity comprises a sulfur capable of being converted to a sulfhydryl and d) converting said moiety to a moiety with a free sulfhydryl; wherein said free sulfhydryl can form a covalent bond with a reactive group on said cationic vehicle.
25. The method of claim 22 wherein said chemical moiety is dithiopyridine.
26. A method of preparing a ligand-polymer complex capable of covalently binding with a reactive group on a cationic vehicle, comprising the steps of: a) selecting a polymer with two ends which has a functional moiety at each of the two ends, wherein the functional moieties can be the same or different; b) covalently bonding a ligand to a first end of said polymer; c) adding a chemical entity comprising a reactive crosslinker to a second end of said polymer of step (b); wherein said reactive crosslinker can form a covalent bond with a reactive group of a cationic vehicle.
27. The method of claim 26 wherein said ligand is folate.
28. The method of claim 26 wherein said chemical entity is 2-iminothiolane.
29. A cationic vehicle prepared by a method of any one of claims 1-21.
30. A polymer-ligand complex prepared by a method of any one of claims 22-28.
EP00904440A 1999-01-21 2000-01-21 Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery Withdrawn EP1144012A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US11679299P 1999-01-21 1999-01-21
US116792P 1999-01-21
PCT/US2000/001346 WO2000043043A1 (en) 1999-01-21 2000-01-21 Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery

Publications (1)

Publication Number Publication Date
EP1144012A1 true EP1144012A1 (en) 2001-10-17

Family

ID=22369253

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00904440A Withdrawn EP1144012A1 (en) 1999-01-21 2000-01-21 Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery

Country Status (5)

Country Link
EP (1) EP1144012A1 (en)
JP (1) JP2002535290A (en)
AU (1) AU2620000A (en)
CA (1) CA2358497A1 (en)
WO (1) WO2000043043A1 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2370690A1 (en) * 1999-04-20 2000-10-26 The University Of British Columbia Cationic peg-lipids and methods of use
US6852334B1 (en) 1999-04-20 2005-02-08 The University Of British Columbia Cationic peg-lipids and methods of use
AU5454801A (en) * 2000-04-20 2001-11-07 Univ British Columbia Methods of enhancing splp-mediated transfection using endosomal membrane destabilizers
US7189705B2 (en) 2000-04-20 2007-03-13 The University Of British Columbia Methods of enhancing SPLP-mediated transfection using endosomal membrane destabilizers
GB0129121D0 (en) * 2001-12-05 2002-01-23 Ic Vec Ltd Compound
US20030229013A1 (en) 2001-12-07 2003-12-11 Shih-Kwang Wu Solid phase method for synthesis peptide-spacer-lipid conjugates, conjugates synthesized thereby and targeted liposomes containing the same
WO2005039558A1 (en) * 2003-10-24 2005-05-06 Transgene S.A. Targeted delivery of therapeutically active compounds
TW200819137A (en) * 2006-10-31 2008-05-01 Iner Aec Executive Yuan Method for preparation of lipid-spacer radical- reactions of functional group-peptide
US20130011333A1 (en) * 2010-01-18 2013-01-10 Board Of Regents, The University Of Texas System Methods and Compositions for Nanoparticle-Mediated Cancer Cell-Targeted Delivery
US20120237565A1 (en) * 2011-03-14 2012-09-20 Intezyne Technologies, Incorporated Pegylated polyplexes containing two or more different polymers for polynucleotide delivery
CN104922068B (en) * 2015-05-22 2017-10-13 江苏凯基生物技术股份有限公司 A kind of Decoy nucleic acid cationics liposome vectors and preparation method thereof
CN106267224B (en) * 2015-06-01 2019-05-28 湖南师范大学 The preparation method of galactose derivative cationic-liposome nano particle
WO2017220099A1 (en) * 2016-06-24 2017-12-28 Statens Serum Institut Adjuvants with modified drainage properties
CN113332442A (en) * 2021-06-10 2021-09-03 愈美明德(成都)生物医药科技有限公司 Targeted delivery molecule, particle, preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0043043A1 *

Also Published As

Publication number Publication date
CA2358497A1 (en) 2000-07-27
JP2002535290A (en) 2002-10-22
AU2620000A (en) 2000-08-07
WO2000043043B1 (en) 2000-09-21
WO2000043043A1 (en) 2000-07-27

Similar Documents

Publication Publication Date Title
Durymanov et al. Non-viral delivery of nucleic acids: insight into mechanisms of overcoming intracellular barriers
Pichon et al. Histidine-rich peptides and polymers for nucleic acids delivery
Ogris et al. Tumor-targeted gene therapy: strategies for the preparation of ligand–polyethylene glycol–polyethylenimine/DNA complexes
Brownlie et al. PEI-based vesicle-polymer hybrid gene delivery system with improved biocompatibility
US6586524B2 (en) Cellular targeting poly(ethylene glycol)-grafted polymeric gene carrier
AU681735C (en) Self-assembling polynucleotide delivery system comprising dendrimer polycations
Simões et al. Cationic liposomes for gene delivery
Wolschek et al. Specific systemic nonviral gene delivery to human hepatocellular carcinoma xenografts in SCID mice
Zauner et al. Polylysine-based transfection systems utilizing receptor-mediated delivery
US5990089A (en) Self-assembling polynucleotide delivery system comprising dendrimer polycations
Erbacher et al. Transfection and physical properties of various saccharide, poly (ethylene glycol), and antibody‐derivatized polyethylenimines (PEI)
Reddy et al. Optimization of folate-conjugated liposomal vectors for folate receptor-mediated gene therapy
Yang et al. Design, preparation and application of nucleic acid delivery carriers
US20030203865A1 (en) Lipid-comprising drug delivery complexes and methods for their production
CA2220950A1 (en) Delivery vehicles comprising stable lipid/nucleic acid complexes
WO1997000965A2 (en) A lipidic vector for nucleic acid delivery
JP2007521247A (en) Biocompatibility-novel cationic lipopolymers as gene delivery agents
EP1144012A1 (en) Ligand-peg post-coating stabilized lipoplex and polyplex for targeted gene delivery
US20050163832A1 (en) Intracellular delivery of therapeutic agents
Wang et al. Double click-functionalized siRNA polyplexes for gene silencing in epidermal growth factor receptor-positive tumor cells
US20090232762A1 (en) Compositions for delivery of therapeutic agents
WO2008070141A2 (en) Compositions for delivery of therapeutic agents
US20010044147A1 (en) Delivery vehicles comprising stable lipid/nucleic acid complexes
US20080031883A1 (en) Condition-dependent, multiple target delivery system
Khare et al. Bioconjugates: harnessing potential for effective therapeutics

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20010809

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20040311