US20080138394A1 - Composite For Liver-Specific Delivery and Release of Therapeutic Nucleic Acids or Drugs - Google Patents

Composite For Liver-Specific Delivery and Release of Therapeutic Nucleic Acids or Drugs Download PDF

Info

Publication number
US20080138394A1
US20080138394A1 US11/741,287 US74128707A US2008138394A1 US 20080138394 A1 US20080138394 A1 US 20080138394A1 US 74128707 A US74128707 A US 74128707A US 2008138394 A1 US2008138394 A1 US 2008138394A1
Authority
US
United States
Prior art keywords
composite
mixture
group
dtc
apo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/741,287
Inventor
Meehyein Kim
Soo In Kim
Duckhyang Shin
Mahnhoon Park
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.)
Mogam Biotechnology Research Institute
Original Assignee
Mogam Biotechnology Research Institute
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 Mogam Biotechnology Research Institute filed Critical Mogam Biotechnology Research Institute
Assigned to MOGAM BIOTECHNOLOGY RESEARCH INSTITUTE reassignment MOGAM BIOTECHNOLOGY RESEARCH INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KIM, MEEHYEIN, KIM, SOO IN, PARK, MAHNHOON, SHIN, DUCKHYANG
Publication of US20080138394A1 publication Critical patent/US20080138394A1/en
Priority to US12/791,600 priority Critical patent/US8318199B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a composite for liver-specific delivery of a therapeutic nucleic acid or a drug, a process for preparing the same and a composition comprising the same with a pharmaceutically acceptable carrier.
  • tissue-specific gene and drug delivery system has long been considered important for drug discovery and pharmaceutical advancement because most drugs are systemically delivered and circulated in the body when administered to a patient, which might adversely affect healthy organs or cells.
  • the tissue-specific delivery system allows the accumulation of a high drug concentration at the target tissue which eliminating adverse side effects, leading to efficient treatment of tissue-specific diseases.
  • liver diseases arise from infection by pathogenic viruses, e.g., HBV (hepatitis B virus) and HCV (hepatitis C virus), while non-infectious liver diseases result from exposure to liver-toxic materials, or genetic or environmental disorders.
  • pathogenic viruses e.g., HBV (hepatitis B virus) and HCV (hepatitis C virus)
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • HCC hepatocellular carcinoma
  • a lipoprotein system mainly that of HDL (high density lipoprotein) has advantages over other delivery systems which use viral vectors (Wang X., et al., Gene Ther. (2006), 13: 1097-1103), non-viral complexes (Landen C.
  • the lipoprotein can be preferably recognized and taken up via cell surface receptors specific for liver cells (Firestone R. A., Bioconjug. Chem.
  • HDL high density lipoprotein
  • the present inventors have therefore endeavored to develop an effective system for liver-specific delivery of a therapeutic drug, and have found that a composite comprising an apolipoprotein A-I and a liposome-forming material can specifically deliver and release therapeutic drugs to the liver.
  • a composite comprising an apolipoprotein A-I (Apo A-I) and a liposome-forming material.
  • composition comprising the composite and a pharmaceutically acceptable carrier.
  • FIG. 1A Purified human Apo A-I from adult blood separated by 4-20% sodium dodecyl sulfate-polyacrylamide gel electrophoresis;
  • FIG. 1B In vivo images of a mouse intravenously injected with Apo A-I labeled with an infrared fluorescent dye at several times after the injection;
  • FIG. 2A Whole body images for radioiodine signals captured using a gamma camera in a mouse intravenously injected with the inventive composite (DTC-Apo*/RLuc) which contains a Renilla luciferase expression plasmid, phRL-CMV and 131 I label;
  • FIG. 2B Whole body images captured several times after mice were intravenously injected with the inventive composite (DTC*-Apo/RLuc) and a comparative composite (DTC*/RLuc), which are labeled with rhodamine, respectively;
  • FIG. 3A Relative levels of secreted HBsAg determined by ELISA in mice which were intravenously injected with a mock control (5% dextrose); naked siHBV; comparative composites containing HBV-specific siRNA (DTC/siHBV) and irrelevant control siRNA (DTC/siCont); and the inventive composites containing HBV-specific siRNA (DTC-Apo/siHBV) and irrelevant control siRNA (DTC-Apo/siCont), respectively, at days 2, 4, 6 and 8 after the injection;
  • FIG. 3B Serum HBsAg levels measured by ELISA in in vivo mouse models of HBV replication which were intravenously injected with the inventive composites containing HBV-specific siRNA (DTC-Apo/siHBV) and irrelevant control siRNA (DTC-Apo/siCont), respectively, at doses of 0.5, 1 or 2 mg/kg, at day 4 after the injection;
  • DTC-Apo/siHBV HBV-specific siRNA
  • DTC-Apo/siCont irrelevant control siRNA
  • FIG. 4A In vivo images of luciferase gene expression in mice which were administered with a luciferase expression plasmid, pEGFPLuc, and one day after administration, intravenously injected with the inventive composites containing luciferase-specific siRNA (DTC-Apo/siLuc) or irrelevant control siRNA (DTC-Apo/siCont), respectively; and
  • FIG. 4B Relative luciferase expression levels measured by counting bioluminescent signals emitted from the liver of the mice shown in FIG. 4A .
  • the composite of the present invention may be in the form of nanoparticles having an average particle size ranging from 50 to 400 nm, preferably 100 to 250 nm, and the apolipoprotein A-I (Apo A-I) used in the present invention may be obtained from human blood by cold ethanol precipitation in accordance with a conventional method (e.g., Lerch, P. G., et al., Vox. Sang. (1996), 71: 155-164).
  • a conventional method e.g., Lerch, P. G., et al., Vox. Sang. (1996), 71: 155-164.
  • the liposome-forming material employed in the inventive composite may be a cationic or neutral liposome-forming material, or a mixture thereof, which play a role of avoiding undesirable interactions between the inventive composite and unknown serum components.
  • Representative examples of the cationic liposome-forming material include DOTAP (1,2-dioleoyl-3-trimethylammonium-propane), DC-cholosterol (3 ⁇ -[N-(N′,N′-dimethylaminoethane)-carbamyl]cholesterol), DDAB (dimethyldioctadecylammonium bromide), and a mixture thereof
  • the neutral liposome-forming material may be DOPE (L-alpha-dioleoyl phosphatidylethanolamine), cholesterol, or a mixture thereof.
  • the inventive composite may comprise Apo A-I and the liposome-forming material at a weight ratio ranging from 1:0.1 to 1:1000, preferably 1:1 to 1:100.
  • the composite of the present invention may further comprise a therapeutic nucleic acid and/or drug.
  • the therapeutic nucleic acid may be a DNA such as plasmid and PCR product, an RNA such as siRNA and ribozyme, or a derivative thereof obtained by chemical modification, preferably siRNA specific for HBV or HCV genome.
  • the therapeutic drug may be an active polypeptide, anticancer agent, or antivirus agent, which does not limit the scope of the present invention.
  • the active polypeptide used in the inventive composition may be selected from the group consisting of epidermal growth factor (EGF), erythropoietin (EPO), coagulation factors VIII, IX and VIIa, follicle stimulating hormone (FSH), granulocyte colony-stimulating factor (GCSF), granulocyte-macrophage colony stimulating factor (GM-CSF), insulin, insulin-like growth factor (IGF), interferon- ⁇ , - ⁇ and - ⁇ (IFN- ⁇ , - ⁇ and - ⁇ ), interleukin-1, -2, -11, -12 and -15 (IL-1, -2, -11, -12 and -15), parathyroid hormone (PTH), platelet-derived growth factor (PDGF), human growth hormone (hGH), tissue plasminogen activator (tPA), vascular endothelial growth factor (VEGF), and a mixture thereof, which does not limit the scope of the present invention.
  • EGF epidermal growth factor
  • EPO erythropoi
  • the anticancer agent may be selected from the group consisting of carboplatin, cisplatin, oxaliplatin, heptaplatin, etoposide, semustine, hydroxycarbamide, citarabine, fludarabine, doxorubicin, epirubicin, idarubicin, pirarubicin, fluorouracil (5-FU), fluoxuridine, mitomycin, bleomycin, clofazimine, interferon, streptozocin, gemcitabine, enocitabine, capecitabine, ursodeoxycholic acid, sorafenib, tegafur, holmium and a holmium-chitosan complex
  • the antivirus agent may be selected from the group consisting of atazanavir, ribavirin, zanamivir, acyclovir, entecavir, didanosin, nevirapine, valaciclovir, nelfinavir, ef
  • the composite of the present invention may be prepared by a method comprising (i) dispersing liposome-forming materials in an organic solvent to form a liposome, (ii) dispersing the liposome in a dextrose solution, and sonicating the mixture to obtain a liposome suspension, and (iii) adding a solution containing Apo A-I thereto to allow forming the inventive composite.
  • the method of the present invention may further comprise (iv) adding a therapeutic nucleic acid or a drug to the suspension of the inventive composite obtained in step (iii).
  • compositions for liver-specific delivery of a therapeutic nucleic acid or drug comprising the inventive composite and a pharmaceutically acceptable carrier.
  • inventive composition may further comprise the therapeutic nucleic acid or drug as described above.
  • composition of the present invention may be formulated for oral or parenteral administration according to any one of the procedures well known in the art, so as to take the form of sterilized aqueous solution, hydrophobic solvent, suspension, emulsion, lyophilized formulation or suppository.
  • the hydrophobic solvent or suspension may further comprise a vegetable oil such as propylene glycol, polyethylene glycol and olive oil; an ester such as ethyloleate; or a mixture thereof
  • the suppository may further comprise witepsol, macrogol, Tween 61, cacao butter, laurel oil, glycerol, gelatine, or a mixture thereof.
  • a proposed daily dose of the composition of the present invention for administration to a human is about from 0.1 mg to 1000 mg, more preferably about from 1 mg to 500 mg. It should be understood that the daily dose should be determined in light of various relevant factors including the condition to be treated, the severity of the patient's symptoms, the route of administration, or the physiological form of the anticancer agent; and, therefore, the dosage suggested above does not limit the scope of the invention in anyway.
  • A-I High purity human apolipoprotein A-I (Apo A-I, 28 kDa) was obtained from serum fractions of normal healthy adults not infected with viral pathogens such as HBV, HCV or HIV by cold ethanol precipitation according to the established protocol (Lerch, P. G., et al., Vox. Sang. (1996), 71: 155-164).
  • FIG. 1A After sodium dodecyl sulfate-polycrylamide gel electrophoresis (SDS-PAGE), and the purified Apo A-I was characterized by Coomassie blue staining. The result is shown in FIG. 1A .
  • the identity of the purified Apo A-I was confirmed by western blot analysis using a goat anti-human Apo A-I antibody (Academy Biomedical Company, USA) which has cross-reactivity to mouse Apo A-I, and a secondary antibody, rabbit anti-goat IgG-HRP (KPL, USA).
  • the purified protein (0.6 mg) was labeled with an infrared dye using IRDye 800 CW in vivo imaging agent (LI-COR Biosciences, USA), and purified using a dextran desalting column (Pierce Biotechnology, Inc., USA) to remove unincorporated dye.
  • the labeled Apo A-I (200 ⁇ g) was administered to 6- to 8-week-old female nude mice (Charles River Laboratories, Inc., USA) via tail vein injection.
  • test mice After 7, 40, 90, 150, 240 or 360 min, the test mice anesthetized with 2% isoflurane were placed in a supine position in a light tight chamber, and their whole body images were obtained using IVIS 200 imaging system (Xenogen, USA) and Living Image Software (Xenogen, USA). The resulting images are shown in FIG. 1B .
  • Apo A-I can be specifically delivered to and stably maintained in the liver for at least 6 hours when systemically administered.
  • FIG. 1C reveals that the uptake yield of the administered Apo A-I by liver are maximal at approximately 45% within 150 min after administration.
  • DOTAP 1,2-dioleoyl-3-trimethylammonium-propane
  • cholesterol Sigma, USA
  • DOTAP/cholesterol DTC
  • the organic solvent was removed by evaporation under a stream of N 2 gas and the residue was kept in a vacuum desiccator for 2 hours to ensure the removal of the residual organic solvent.
  • the resulting dried film was hydrated in a 5% dextrose solution and the suspension thus obtained was sonicated using a bath sonicator.
  • HBV X gene-specific siRNA SEQ ID NOs: 1 (sense) and 2 (antisense); Shin, D., Virus Res. (2006), 119: 146-153
  • inventive composite DTC-Apo
  • DTC-Apo/siHBV inventive composite containing HBV X gene-specific siRNA
  • Example 2 The procedure of Example 2 was repeated except for using firefly luciferase-specific siRNA (SEQ ID NOs: 3 (sense) and 4 (antisense); Elbashir, S. M., Nature (2001), 411: 494-498) instead of HBV-specific siRNA, to obtain the inventive composite containing firefly luciferase-specific siRNA, named DTC-Apo/siLuc.
  • SEQ ID NOs: 3 sense
  • 4 antisense
  • Example 2 The procedure of Example 2 was repeated except for using 0.3 mg of a plasmid phRL-CMV encoding Renilla luciferase (Promega, WI) and 3 mg of DTC-Apo instead of 40 ⁇ g of HBV-specific siRNA and 400 ⁇ g of DTC-Apo, to obtain the inventive composite containing a plasmid phRL-CMV, named DTC-Apo/RLuc.
  • Example 2 The procedure of Example 2 was repeated except for using control double stranded RNA (SEQ ID NOs: 5 (sense) and 6 (antisense)) instead of HBV-specific siRNA, to obtain the inventive composite containing control double stranded RNA, named DTC-Apo/siCont.
  • control double stranded RNA SEQ ID NOs: 5 (sense) and 6 (antisense)
  • the inventive composite has an average particle size in the nanoscale range, suitable for systemic administration, and it is positive charged no matter whether it contained a nucleic acid or not, which showed that the inventive composite would not occur undesirable interaction with unknown serum components.
  • DTC-Apo/RLuc obtained in Example 4 was labeled with 0.6 mCi 131 I (The Korea Atomic Energy Research Institute, Daejeon, South Korea) by the chloramines-T method (named DTC-Apo*/RLuc).
  • 200 ⁇ Ci of the purified DTC-Apo*/RLuc was intravenously injected into nude mice (Charles River Laboratories), and the radioactivity from the whole body of each mouse was monitored using a gamma-camera (Medical imaging Electronics, USA) at 40, 120 and 240 min postinjection, respectively. The results are shown in FIG. 2A .
  • FIG. 2A clearly shows that the inventive composite is accumulated in the hepatic tissue at 40 min after administration.
  • the cationic liposomes of the comparative and inventive composites, DTC/RLuc and DTC-Apo/RLuc obtained in Comparative Example 3 and Example 4, respectively, were labeled with a fluorescent dye, rhodamine using lissamine rhodamine B-diacyl phosphatidylethanolamine (Avanti Polar Lipids), to obtain labeled composities, DTC*/RLuc and DTC*-Apo/RLuc, respectively.
  • the labeled composites were each injected into nude mice (Charles River Laboratories), and the whole body was monitored using IVIS 200 imaging system (Xenogen, USA) at 20, 60 and 100 min postinjection.
  • the accumulation level of the inventive composite is enhanced in the liver more prominently than that of the comparative composite.
  • the fluorescent signal noise detected at the ends of the limb may be due to the overlapped emission wavelengths between rhodamine and red blood cells.
  • mice (Charles River Laboratories) were intravenously treated with 200 ⁇ Ci of the unlabeled DTC/RLuc or DTC-Apo/RLuc, or naked phRL-CMV or a 5% dextrose solution, as controls, and sacrificed the following day.
  • Heart, lung, kidney and liver were each harvested from each mouse and homogenized.
  • the bioluminescent intensity of each tissue homogenate was measured using a renilla luciferase assay system (Promega) to determine the luciferase expression level per total protein. The results are shown in FIG. 2C .
  • luminescence signals are particularly prominent in the liver of mice injected with DTC-Apo/RLuc in an amount ranging from 6,700 to 50,300 RLU/mg.
  • luciferase signals were strong in the lung and kidney but only modest in the liver.
  • the inventive composite can liver-specifically deliver a therapeutic gene or drug to hepatic cells the therapeutic gene being expressed therein.
  • DTC-Apo containing HBV-specific siRNA DTC-Apo/siHBV was examined using a mouse model for acute HBV infection as follows.
  • HBV replication competent plasmid pCpGHBV-MBRI
  • pCpGHBV-MBRI HBV replication competent plasmid
  • mice 10 ⁇ g of pCpGHBV-MBRI was hydrodynamically injected into the tail veins of female C57BL/6 mice (Charles River Laboratories) of 8-9 weeks of age weighing approximately 20 g to induce the acute HBV infectious.
  • the HBV-infected model mice were intravenously administered with 2 mg/kg (i. e., 40 ⁇ g of nucleic acid per mouse) of DTC-Apo/siHBV, DTC-Apo/siCont, DTC/siHBV and DTC/siCont obtained in Examples 2 and 5, and Comparative Examples 1 and 4, respectively.
  • 2 mg/kg of naked HBV-specific siRNA or 5% dextrose solution was also administrated to control mice groups.
  • HBV surface antigen (HBsAg) level one of the major viral structural proteins
  • ELISA DiaSorin, USA
  • mice with acute HBV replication were treated with 0.5, 1, or 2 mg/kg doses of DTC-Apo/siHBV, while 2 mg/kg of DTC-Apo/siCont or a 5% dextrose solution was also administrated to the model mice as control groups.
  • the serum viral antigen levels in each mouse was monitored at day 4 post-injection. The results are shown in FIG. 3B .
  • the treatment of the inventive composite containing HBV-specific siRNA reduces the viral antigen expression in mice with acute HBV replication in a does-dependent manner, unlike the control groups.
  • inventive composite can promote the hepatic tissue-specific delivery of a therapeutic nucleic acid or drug and lead to potent therapeutic effects in vivo, only through a intravenous treatment of the inventive composite containing a therapeutic nucleic acid or drug.
  • DTC-Apo/siLuc or DTC-Apo/siCont obtained in Example 3 or 5, or a 5% dextrose solution control was injected at a dose of 1 mg/kg via the tail veins of the mice under ambient pressure, and the following day, the treated mice were anaesthetized with 2% isoflurane, and intraperitoneally injected with 200 ⁇ l of 15 mg/ml D-luciferin (Molecular Imaging Products Company, USA). Ten minutes later, photon signals from the whole body of each mouse was analyzed using an IVIS imaging system (Xenogen). The results are shown in FIGS. 4A and 4B .

Abstract

The inventive composite having a nanoscale particle size can specifically deliver therapeutic nucleic acids or drugs to the liver and selectively release them into hepatic cells to manifest potent therapeutic effects without inducing any enzymatic abnormalities or pathological damage to the normal liver function, when administered together with the therapeutic agents.

Description

    FIELD OF THE INVENTION
  • The present invention relates to a composite for liver-specific delivery of a therapeutic nucleic acid or a drug, a process for preparing the same and a composition comprising the same with a pharmaceutically acceptable carrier.
  • BACKGROUND OF THE INVENTION
  • A tissue-specific gene and drug delivery system has long been considered important for drug discovery and pharmaceutical advancement because most drugs are systemically delivered and circulated in the body when administered to a patient, which might adversely affect healthy organs or cells. The tissue-specific delivery system allows the accumulation of a high drug concentration at the target tissue which eliminating adverse side effects, leading to efficient treatment of tissue-specific diseases.
  • Some liver diseases arise from infection by pathogenic viruses, e.g., HBV (hepatitis B virus) and HCV (hepatitis C virus), while non-infectious liver diseases result from exposure to liver-toxic materials, or genetic or environmental disorders. The progression of early-stage liver diseases caused by biological stimuli ultimately leads to chronic hepatitis, liver cirrhosis or hepatocellular carcinoma (HCC). Among several drug or gene delivery systems currently studied in the treatment of such liver diseases, a lipoprotein system, mainly that of HDL (high density lipoprotein), has advantages over other delivery systems which use viral vectors (Wang X., et al., Gene Ther. (2006), 13: 1097-1103), non-viral complexes (Landen C. N., et al., Cancer Res. (2005), 65: 6910-6918; Morrissey D. V., et al., Nat. Biotechnol. (2005), 23: 1002-1007; Sorensen D. R., et al., J. Mol. Biol. (2003), 327: 761-766; and Urban-Klein B., et al., Gene Ther. (2005), 12: 461-466) and antibodies (Song E., et al., Nat. Biotechnol. (2005), 23: 709-717). For example, the lipoprotein can be preferably recognized and taken up via cell surface receptors specific for liver cells (Firestone R. A., Bioconjug. Chem. (1994), 5: 105-113; de Smidt P. C., et al., Crit. Rev. Ther. Drug Carrier Syst. (1990), 7: 99-120; and Filipowska D., et al., Cancer Chemother Pharmacol. (1992), 29: 396-400), and it is an endogenous product which is not detrimental to human and does not trigger immunological responses in clinical applications (Pussinen P. J., et al., Biochem. Biophys. Acta. (2000), 1485: 129-144).
  • Recently, there has been a report that a recombinant high density lipoprotein (HDL) can be used as a carrier for delivering a lipophilic antitumor drug into human hepatocellular carcinoma cells by taking advantage of the hydrophobic cholesterol ester-loading properties of HDL (Lou B., et al., World J. Gastroenterol. (2005), 11: 954-959). However, it has merely been demonstrated in vitro, but not in vivo, that the cellular uptake of the HDL carrier by a hepatoma cell line, SMMC-7721, is higher in compared with a normal liver cell line, L02, and the limitation in tissue-specific targeting remains to be solved.
  • The present inventors have therefore endeavored to develop an effective system for liver-specific delivery of a therapeutic drug, and have found that a composite comprising an apolipoprotein A-I and a liposome-forming material can specifically deliver and release therapeutic drugs to the liver.
  • SUMMARY OF THE INVENTION
  • Accordingly, it is an object of the present invention to provide a composite capable of specifically delivering and releasing a therapeutic nucleic acid or a drug to the liver when administered via a systemic route.
  • It is another object of the present invention to provide a process for the preparation of said system.
  • It is further object of the present invention to provide a composition for liver-specific delivery of a therapeutic nucleic acid or drug, comprising said composite.
  • In accordance with one aspect of the present invention, there is provided a composite comprising an apolipoprotein A-I (Apo A-I) and a liposome-forming material.
  • In accordance with another aspect of the present invention, there is provided a process for the preparation of the composite.
  • In accordance with further another aspect of the present invention, there is provided a composition comprising the composite and a pharmaceutically acceptable carrier.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The above and other objects and features of the present invention will become apparent from the following description of the invention, when taken in conjunction with the accompanying drawings, which respectively show:
  • FIG. 1A: Purified human Apo A-I from adult blood separated by 4-20% sodium dodecyl sulfate-polyacrylamide gel electrophoresis;
  • FIG. 1B: In vivo images of a mouse intravenously injected with Apo A-I labeled with an infrared fluorescent dye at several times after the injection;
  • FIG. 1C: Percent uptake rate of Apo A-I in the livers of mice injected with Apo A-I at different time points (n=4) after the injection;
  • FIG. 2A: Whole body images for radioiodine signals captured using a gamma camera in a mouse intravenously injected with the inventive composite (DTC-Apo*/RLuc) which contains a Renilla luciferase expression plasmid, phRL-CMV and 131I label;
  • FIG. 2B: Whole body images captured several times after mice were intravenously injected with the inventive composite (DTC*-Apo/RLuc) and a comparative composite (DTC*/RLuc), which are labeled with rhodamine, respectively;
  • FIG. 2C: Luciferase levels measured in tissue homogenates from heart, lung, kidney and liver of mice (n=3) which were intravenously administered with a mock control (5% dextrose), naked DNA, or the inventive or a comparative composite containing a Renilla luciferase expression plasmid, phRL-CMV (DTC/RLuc or DTC-Apo/RLuc);
  • FIG. 3A: Relative levels of secreted HBsAg determined by ELISA in mice which were intravenously injected with a mock control (5% dextrose); naked siHBV; comparative composites containing HBV-specific siRNA (DTC/siHBV) and irrelevant control siRNA (DTC/siCont); and the inventive composites containing HBV-specific siRNA (DTC-Apo/siHBV) and irrelevant control siRNA (DTC-Apo/siCont), respectively, at days 2, 4, 6 and 8 after the injection;
  • FIG. 3B: Serum HBsAg levels measured by ELISA in in vivo mouse models of HBV replication which were intravenously injected with the inventive composites containing HBV-specific siRNA (DTC-Apo/siHBV) and irrelevant control siRNA (DTC-Apo/siCont), respectively, at doses of 0.5, 1 or 2 mg/kg, at day 4 after the injection;
  • FIG. 4A: In vivo images of luciferase gene expression in mice which were administered with a luciferase expression plasmid, pEGFPLuc, and one day after administration, intravenously injected with the inventive composites containing luciferase-specific siRNA (DTC-Apo/siLuc) or irrelevant control siRNA (DTC-Apo/siCont), respectively; and
  • FIG. 4B: Relative luciferase expression levels measured by counting bioluminescent signals emitted from the liver of the mice shown in FIG. 4A.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The composite of the present invention may be in the form of nanoparticles having an average particle size ranging from 50 to 400 nm, preferably 100 to 250 nm, and the apolipoprotein A-I (Apo A-I) used in the present invention may be obtained from human blood by cold ethanol precipitation in accordance with a conventional method (e.g., Lerch, P. G., et al., Vox. Sang. (1996), 71: 155-164).
  • The liposome-forming material employed in the inventive composite may be a cationic or neutral liposome-forming material, or a mixture thereof, which play a role of avoiding undesirable interactions between the inventive composite and unknown serum components. Representative examples of the cationic liposome-forming material include DOTAP (1,2-dioleoyl-3-trimethylammonium-propane), DC-cholosterol (3β-[N-(N′,N′-dimethylaminoethane)-carbamyl]cholesterol), DDAB (dimethyldioctadecylammonium bromide), and a mixture thereof, and the neutral liposome-forming material may be DOPE (L-alpha-dioleoyl phosphatidylethanolamine), cholesterol, or a mixture thereof.
  • The inventive composite may comprise Apo A-I and the liposome-forming material at a weight ratio ranging from 1:0.1 to 1:1000, preferably 1:1 to 1:100.
  • The composite of the present invention may further comprise a therapeutic nucleic acid and/or drug.
  • The therapeutic nucleic acid may be a DNA such as plasmid and PCR product, an RNA such as siRNA and ribozyme, or a derivative thereof obtained by chemical modification, preferably siRNA specific for HBV or HCV genome.
  • The therapeutic drug may be an active polypeptide, anticancer agent, or antivirus agent, which does not limit the scope of the present invention.
  • The active polypeptide used in the inventive composition may be selected from the group consisting of epidermal growth factor (EGF), erythropoietin (EPO), coagulation factors VIII, IX and VIIa, follicle stimulating hormone (FSH), granulocyte colony-stimulating factor (GCSF), granulocyte-macrophage colony stimulating factor (GM-CSF), insulin, insulin-like growth factor (IGF), interferon-α, -β and -γ (IFN-α, -β and -γ), interleukin-1, -2, -11, -12 and -15 (IL-1, -2, -11, -12 and -15), parathyroid hormone (PTH), platelet-derived growth factor (PDGF), human growth hormone (hGH), tissue plasminogen activator (tPA), vascular endothelial growth factor (VEGF), and a mixture thereof, which does not limit the scope of the present invention.
  • Further, the anticancer agent may be selected from the group consisting of carboplatin, cisplatin, oxaliplatin, heptaplatin, etoposide, semustine, hydroxycarbamide, citarabine, fludarabine, doxorubicin, epirubicin, idarubicin, pirarubicin, fluorouracil (5-FU), fluoxuridine, mitomycin, bleomycin, clofazimine, interferon, streptozocin, gemcitabine, enocitabine, capecitabine, ursodeoxycholic acid, sorafenib, tegafur, holmium and a holmium-chitosan complex, and the antivirus agent may be selected from the group consisting of atazanavir, ribavirin, zanamivir, acyclovir, entecavir, didanosin, nevirapine, valaciclovir, nelfinavir, efavirenz, ganciclovir, lamivudine, famciclovir, stavudine, abacavir, indinavir, oseltamivir, inosiplex, and adefovir, which does not limit the scope of the present invention.
  • The composite of the present invention may be prepared by a method comprising (i) dispersing liposome-forming materials in an organic solvent to form a liposome, (ii) dispersing the liposome in a dextrose solution, and sonicating the mixture to obtain a liposome suspension, and (iii) adding a solution containing Apo A-I thereto to allow forming the inventive composite. The method of the present invention may further comprise (iv) adding a therapeutic nucleic acid or a drug to the suspension of the inventive composite obtained in step (iii).
  • In accordance with further aspect of the present invention, there is provided a composition for liver-specific delivery of a therapeutic nucleic acid or drug, comprising the inventive composite and a pharmaceutically acceptable carrier. The inventive composition may further comprise the therapeutic nucleic acid or drug as described above.
  • The composition of the present invention may be formulated for oral or parenteral administration according to any one of the procedures well known in the art, so as to take the form of sterilized aqueous solution, hydrophobic solvent, suspension, emulsion, lyophilized formulation or suppository. In the formulation of the inventive composition, the hydrophobic solvent or suspension may further comprise a vegetable oil such as propylene glycol, polyethylene glycol and olive oil; an ester such as ethyloleate; or a mixture thereof, and the suppository may further comprise witepsol, macrogol, Tween 61, cacao butter, laurel oil, glycerol, gelatine, or a mixture thereof.
  • Further, a proposed daily dose of the composition of the present invention for administration to a human (of approximately 70 kg body weight) is about from 0.1 mg to 1000 mg, more preferably about from 1 mg to 500 mg. It should be understood that the daily dose should be determined in light of various relevant factors including the condition to be treated, the severity of the patient's symptoms, the route of administration, or the physiological form of the anticancer agent; and, therefore, the dosage suggested above does not limit the scope of the invention in anyway.
  • The following Examples are intended to further illustrate the present invention without limiting its scope.
  • TEST EXAMPLE 1 Liver-specificity of Purifed Apo A-I
  • High purity human apolipoprotein A-I (Apo A-I, 28 kDa) was obtained from serum fractions of normal healthy adults not infected with viral pathogens such as HBV, HCV or HIV by cold ethanol precipitation according to the established protocol (Lerch, P. G., et al., Vox. Sang. (1996), 71: 155-164).
  • After sodium dodecyl sulfate-polycrylamide gel electrophoresis (SDS-PAGE), and the purified Apo A-I was characterized by Coomassie blue staining. The result is shown in FIG. 1A. The identity of the purified Apo A-I was confirmed by western blot analysis using a goat anti-human Apo A-I antibody (Academy Biomedical Company, USA) which has cross-reactivity to mouse Apo A-I, and a secondary antibody, rabbit anti-goat IgG-HRP (KPL, USA).
  • For in vivo imaging, the purified protein (0.6 mg) was labeled with an infrared dye using IRDye 800 CW in vivo imaging agent (LI-COR Biosciences, USA), and purified using a dextran desalting column (Pierce Biotechnology, Inc., USA) to remove unincorporated dye. The labeled Apo A-I (200 μg) was administered to 6- to 8-week-old female nude mice (Charles River Laboratories, Inc., USA) via tail vein injection. After 7, 40, 90, 150, 240 or 360 min, the test mice anesthetized with 2% isoflurane were placed in a supine position in a light tight chamber, and their whole body images were obtained using IVIS 200 imaging system (Xenogen, USA) and Living Image Software (Xenogen, USA). The resulting images are shown in FIG. 1B.
  • As shown in FIG. 1B, Apo A-I can be specifically delivered to and stably maintained in the liver for at least 6 hours when systemically administered.
  • Further, the photon intensities in the liver of the test mice were measured using Living Image Software (Xenogen, USA) at each time point after the administration, and the result is shown in FIG. 1C.
  • FIG. 1C reveals that the uptake yield of the administered Apo A-I by liver are maximal at approximately 45% within 150 min after administration.
  • Taken together, these data demonstrate that the purified Apo A-I maintains its native conformation required for cell-surface receptor recognition and catabolic circulation in vivo, suggesting that it might be applicable as a potent candidate carrier for targeting the liver in feasibility studies for therapeutic drug delivery.
  • EXAMPLE 1 Preparation of the Inventive Composite
  • An equimolar mixture of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP; Avanti Polar Lipids, USA) and cholesterol (Sigma, USA) was dispersed in chloroform and mixed to form a cationic liposome of DOTAP/cholesterol (DTC). After the liposome assembly was formed, the organic solvent was removed by evaporation under a stream of N2 gas and the residue was kept in a vacuum desiccator for 2 hours to ensure the removal of the residual organic solvent. The resulting dried film was hydrated in a 5% dextrose solution and the suspension thus obtained was sonicated using a bath sonicator. A solution containing 10% the Apo A-I purified in Test Example 1 was added thereto at a DTC: Apo A-I mix ratio of 10:1 (w/w), and the mixture was kept overnight at 4° C. to obtain the inventive composite (DTC-Apo).
  • EXAMPLE 2 Preparation of the Inventive Composite Containing Nucleic Acid
  • 40 μg of HBV X gene-specific siRNA (SEQ ID NOs: 1 (sense) and 2 (antisense); Shin, D., Virus Res. (2006), 119: 146-153) was mixed with 400 μg of the inventive composite, DTC-Apo, in 200 μl of 5% dextrose solution and the mixture was incubated at room temperature for 30 min, to obtain the inventive composite containing HBV X gene-specific siRNA, named DTC-Apo/siHBV.
  • EXAMPLE 3 Preparation of the Inventive Composite Containing Nucleic Acid
  • The procedure of Example 2 was repeated except for using firefly luciferase-specific siRNA (SEQ ID NOs: 3 (sense) and 4 (antisense); Elbashir, S. M., Nature (2001), 411: 494-498) instead of HBV-specific siRNA, to obtain the inventive composite containing firefly luciferase-specific siRNA, named DTC-Apo/siLuc.
  • EXAMPLE 4 Preparation of the Inventive Composite Containing Nucleic Acid
  • The procedure of Example 2 was repeated except for using 0.3 mg of a plasmid phRL-CMV encoding Renilla luciferase (Promega, WI) and 3 mg of DTC-Apo instead of 40 μg of HBV-specific siRNA and 400 μg of DTC-Apo, to obtain the inventive composite containing a plasmid phRL-CMV, named DTC-Apo/RLuc.
  • EXAMPLE 5 Preparation of the Inventive Composite Containing Nucleic Acid
  • The procedure of Example 2 was repeated except for using control double stranded RNA (SEQ ID NOs: 5 (sense) and 6 (antisense)) instead of HBV-specific siRNA, to obtain the inventive composite containing control double stranded RNA, named DTC-Apo/siCont.
  • COMPARATIVE EXAMPLES 1 TO 4 Preparation of the Comparative Composite Containing Nucleic Acid
  • The procedures of Examples 2 to 5 were repeated except for using DTC instead of DTC-Apo, to obtain comparative composites named DTC/siHBV, DTC/siLuc, DTC/RLuc and DTC/siCont, respectively.
  • TEST EXAMPLE 2 Characterization of the Inventive and Comparative Composite
  • The inventive and comparative composites obtained in Examples 2 to 5 and Comparative Examples 1 to 4 were characterized by measuring their size and charge using a Zetasizer 3000 apparatus (Malvern Instruments, Malvern, Worcestershire, United Kingdom), respectively, to determine the average diameters and zeta potential values thereof. The results are shown in Table 1.
  • TABLE 1
    Formulation Size (nm) ζ pot (mV)
    DTC 176.5 ± 1.4 53.3 ± 4.0
    DTC with DNA 205.5 ± 4.2 42.7 ± 1.8
    DTC with siRNA 196.0 ± 1.8 44.6 ± 2.2
    DTC-Apo 147.9 ± 2.8 49.5 ± 6.3
    DTC-Apo with DNA 179.5 ± 3.4 38.6 ± 4.0
    DTC-Apo with siRNA 177.1 ± 1.4 39.1 ± 2.8
  • As shown in Table 1, the inventive composite has an average particle size in the nanoscale range, suitable for systemic administration, and it is positive charged no matter whether it contained a nucleic acid or not, which showed that the inventive composite would not occur undesirable interaction with unknown serum components.
  • TEST EXAMPLE 3 Liver-specific Gene Delivery In Vivo
  • In order to facilitate the systemic and sensitive detection of the in vivo migration route, Apo A-I of DTC-Apo/RLuc obtained in Example 4 was labeled with 0.6 mCi 131I (The Korea Atomic Energy Research Institute, Daejeon, South Korea) by the chloramines-T method (named DTC-Apo*/RLuc). 200 μCi of the purified DTC-Apo*/RLuc was intravenously injected into nude mice (Charles River Laboratories), and the radioactivity from the whole body of each mouse was monitored using a gamma-camera (Medical imaging Electronics, USA) at 40, 120 and 240 min postinjection, respectively. The results are shown in FIG. 2A.
  • FIG. 2A clearly shows that the inventive composite is accumulated in the hepatic tissue at 40 min after administration.
  • Further, the cationic liposomes of the comparative and inventive composites, DTC/RLuc and DTC-Apo/RLuc obtained in Comparative Example 3 and Example 4, respectively, were labeled with a fluorescent dye, rhodamine using lissamine rhodamine B-diacyl phosphatidylethanolamine (Avanti Polar Lipids), to obtain labeled composities, DTC*/RLuc and DTC*-Apo/RLuc, respectively. The labeled composites were each injected into nude mice (Charles River Laboratories), and the whole body was monitored using IVIS 200 imaging system (Xenogen, USA) at 20, 60 and 100 min postinjection.
  • As shown in FIG. 2B, the accumulation level of the inventive composite is enhanced in the liver more prominently than that of the comparative composite. The fluorescent signal noise detected at the ends of the limb may be due to the overlapped emission wavelengths between rhodamine and red blood cells.
  • Further, in order to examine nucleic acid release by the inventive composite after systemic injection, mice (Charles River Laboratories) were intravenously treated with 200 μCi of the unlabeled DTC/RLuc or DTC-Apo/RLuc, or naked phRL-CMV or a 5% dextrose solution, as controls, and sacrificed the following day. Heart, lung, kidney and liver were each harvested from each mouse and homogenized. The bioluminescent intensity of each tissue homogenate was measured using a renilla luciferase assay system (Promega) to determine the luciferase expression level per total protein. The results are shown in FIG. 2C.
  • As shown in FIG. 2C, consistently with liver-specific accumulation of isotope or rhodamine-labeled DTC-Apo composites (FIGS. 2A and 2B), luminescence signals are particularly prominent in the liver of mice injected with DTC-Apo/RLuc in an amount ranging from 6,700 to 50,300 RLU/mg. In contrast, in mice treated with DTC/RLuc, luciferase signals were strong in the lung and kidney but only modest in the liver.
  • The results suggest that the inventive composite can liver-specifically deliver a therapeutic gene or drug to hepatic cells the therapeutic gene being expressed therein.
  • TEST EXAMPLE 4 Therapeutic Effect of the Inventive Composite with Nucleic Acid
  • To examine the therapeutic activity of the inventive composite, in vivo antiviral effect of DTC-Apo containing HBV-specific siRNA (DTC-Apo/siHBV) was examined using a mouse model for acute HBV infection as follows.
  • First, in order to establish an acute HBV-infected mouse model, HBV replication competent plasmid, pCpGHBV-MBRI, was created by excision of the viral genome from the mother clone pHBV-MBRI (Shin, D., et al., Virus Res. (2006), 119: 146-153) and religated into SpeI and XbaI digested pCpG-mcs (InvivoGen, USA), which is known to be not inducible nonspecific inflammatory responses in mammalian hosts. Then, 10 μg of pCpGHBV-MBRI was hydrodynamically injected into the tail veins of female C57BL/6 mice (Charles River Laboratories) of 8-9 weeks of age weighing approximately 20 g to induce the acute HBV infectious. After 8 hours, the HBV-infected model mice were intravenously administered with 2 mg/kg (i. e., 40 μg of nucleic acid per mouse) of DTC-Apo/siHBV, DTC-Apo/siCont, DTC/siHBV and DTC/siCont obtained in Examples 2 and 5, and Comparative Examples 1 and 4, respectively. 2 mg/kg of naked HBV-specific siRNA or 5% dextrose solution was also administrated to control mice groups. Serum samples were collected from each treated mouse on days 2, 4, 6 and 8 after injection, and the serum HBV surface antigen (HBsAg) level, one of the major viral structural proteins, was determined by ELISA (DiaSorin, USA) to monitor the viral protein level secreted into the blood. The results are shown in FIG. 3A.
  • As shown in FIG. 3A, there is a significant reduction of serum HBsAg in mice administered with a single dose of DTC-Apo/siHBV particles, as shown by the average inhibitions degree of 65.1% (P=0.014), 63.4% (P=0.047), 74.9% (P=0.015) and 72.8% (P=0.034) on days 2, 4, 6 and 8 post-injection, respectively, relative to the matched DTC/siHBV or DTC-Apo/siCont treated mice.
  • Further, in order to examine the dose-dependent activity of DTC-Apo/siHBV, the mice with acute HBV replication were treated with 0.5, 1, or 2 mg/kg doses of DTC-Apo/siHBV, while 2 mg/kg of DTC-Apo/siCont or a 5% dextrose solution was also administrated to the model mice as control groups. The serum viral antigen levels in each mouse was monitored at day 4 post-injection. The results are shown in FIG. 3B.
  • As shown in FIG. 3B, the treatment of the inventive composite containing HBV-specific siRNA reduces the viral antigen expression in mice with acute HBV replication in a does-dependent manner, unlike the control groups.
  • These in vivo data indicate that the inventive composite can promote the hepatic tissue-specific delivery of a therapeutic nucleic acid or drug and lead to potent therapeutic effects in vivo, only through a intravenous treatment of the inventive composite containing a therapeutic nucleic acid or drug.
  • TEST EXAMPLE 5 Therapeutic Effect of the Inventive Composite with Nucleic Acid
  • In order to confirm that the target-specific effect of the inventive composite comprising a therapeutic nucleic acid or drug occurs selectively and mainly in the hepatic tissue, 6-7-week-old female Balb/c mice (Charles River Laboratories) were hydrodynamically injected with 10 μg of pEGFPLuc plasmid (Clontech), which is known to express the firefly luciferase and also to facilitate in vivo image analysis, respectively.
  • After one day, DTC-Apo/siLuc or DTC-Apo/siCont obtained in Example 3 or 5, or a 5% dextrose solution control was injected at a dose of 1 mg/kg via the tail veins of the mice under ambient pressure, and the following day, the treated mice were anaesthetized with 2% isoflurane, and intraperitoneally injected with 200 μl of 15 mg/ml D-luciferin (Molecular Imaging Products Company, USA). Ten minutes later, photon signals from the whole body of each mouse was analyzed using an IVIS imaging system (Xenogen). The results are shown in FIGS. 4A and 4B.
  • As shown in FIGS. 4A and 4B, there is no signal change suggesting luciferase expression inhibition in mice injected with DTC-Apo/siCont, while a dramatic reduction in luciferase activity (about 70%) was observed for mice treated with DTC-Apo/siLuc as early as day 1 after treatment.
  • Taken together, these results show that the selective target of the inventive composite administered systemically is the liver and that the therapeutic nucleic acid or drug delivered by the inventive composite is specifically released into hepatic cells to manifest a potent therapeutic effect, without inducing any enzymatic abnormalities or pathological damage of the normal liver function.
  • While the embodiments of the subject invention have been described and illustrated, it is obvious that various changes and modifications can be made therein without departing from the spirit of the present invention which should be limited only by the scope of the appended claims.

Claims (20)

1. A composite comprising an apolipoprotein A-I and a liposome-forming material.
2. The composite of claim 1, wherein the liposome-forming material is a cationic or neutral liposome-forming material, or a mixture of thereof.
3. The composite of claim 2, wherein the cationic liposome-forming material is selected from the group consisting of DOTAP (1,2-dioleoyl-3-trimethylammonium-propane), DC-cholosterol (3β-[N-(N′,N′-dimethylaminoethane)carbamyl]cholesterol), DDAB (dimethyldioctadecylammonium bromide), and a mixture thereof.
4. The composite of claim 2, wherein the neutral liposome-forming material is selected from the group consisting of DOPE (L-alpha-dioleoyl phosphatidylethanolamine), cholesterol, and a mixture thereof.
5. The composite of claim 1, wherein the weight ratio of the apolipoprotein A-I and the liposome-forming material is in the range of 1:0.1 to 1:1000.
6. The composite of claim 1, which further comprise a therapeutic nucleic acid or a drug.
7. The composite of claim 6, wherein the therapeutic nucleic acid is selected from the group consisting of a DNA, an RNA, and a derivative thereof.
8. The composite of claim 7, wherein the RNA is an siRNA specific for HBV or HCV genome.
9. The composite of claim 6, wherein the therapeutic drug is an active polypeptide, an anticancer agent or an antivirus agent.
10. The composite of claim 9, wherein the active polypeptide is selected from the group consisting of epidermal growth factor (EGF), erythropoietin (EPO), coagulation factors VIII, IX and VIIa, follicle stimulating hormone (FSH), granulocyte colony-stimulating factor (GCSF), granulocyte-macrophage colony stimulating factor (GM-CSF), insulin, insulin-like growth factor (IGF), interferon-α, -β and -γ (IFN-α, -β and -γ), interleukin-1, -2, -11, -12 and -15 (IL-1, -2, -11, -12 and -15), parathyroid hormone (PTH), platelet-derived growth factor (PDGF), human growth hormone (hGH), tissue plasminogen activator (tPA), vascular endothelial growth factor (VEGF), and a mixture thereof.
11. The composite of claim 9, wherein the anticancer agent is selected from the group consisting of carboplatin, cisplatin, oxaliplatin, heptaplatin, etoposide, semustine, hydroxycarbamide, citarabine, fludarabine, doxorubicin, epirubicin, idarubicin, pirarubicin, fluorouracil (5-FU), fluoxuridine, mitomycin, bleomycin, clofazimine, interferon, streptozocin, gemcitabine, enocitabine, capecitabine, ursodeoxycholic acid, sorafenib, tegafur, holmium, a holmium-chitosan complex, and a mixture thereof.
12. The composite of claim 9, wherein the antivirus agent is selected from the group consisting of atazanavir, ribavirin, zanamivir, acyclovir, entecavir, didanosin, nevirapine, valaciclovir, nelfinavir, efavirenz, ganciclovir, lamivudine, famciclovir, stavudine, abacavir, indinavir, oseltamivir, inosiplex, adefovir, and a mixture thereof.
13. A composition comprising the composite of claim 1 and a pharmaceutically acceptable carrier.
14. The composition of claim 13, which further comprise a therapeutic nucleic acid or a drug.
15. The composition of claim 14, wherein the therapeutic nucleic acid is selected from the group consisting of a DNA, an RNA, and a derivative thereof.
16. The composition of claim 15, wherein the RNA is an siRNA specific for HBV or HCV genome.
17. The composition of claim 14, wherein the therapeutic drug is an active polypeptide, an anticancer agent or an antivirus agent.
18. The composition of claim 17, wherein the active polypeptide is selected from the group consisting of epidermal growth factor (EGF), erythropoietin (EPO), coagulation factors VIII, IX and VIIa, follicle stimulating hormone (FSH), granulocyte colony-stimulating factor (GCSF), granulocyte-macrophage colony stimulating factor (GM-CSF), insulin, insulin-like growth factor (IGF), interferon-α, -β and -γ (IFN-α, -β and -γ), interleukin-1, -2, -11, -12 and -15 (IL-1, -2, -11, -12 and -15), parathyroid hormone (PTH), platelet-derived growth factor (PDGF), human growth hormone (hGH), tissue plasminogen activator (tPA), vascular endothelial growth factor (VEGF), and a mixture thereof.
19. The composition of claim 17, wherein the anticancer agent is selected from the group consisting of carboplatin, cisplatin, oxaliplatin, heptaplatin, etoposide, semustine, hydroxycarbamide, citarabine, fludarabine, doxorubicin, epirubicin, idarubicin, pirarubicin, fluorouracil (5-FU), fluoxuridine, mitomycin, bleomycin, clofazimine, interferon, streptozocin, gemcitabine, enocitabine, capecitabine, ursodeoxycholic acid, sorafenib, tegafur, holmium, a holmium-chitosan complex, and a mixture thereof.
20. The composition of claim 17, wherein the antivirus agent is selected from the group consisting of atazanavir, ribavirin, zanamivir, acyclovir, entecavir, didanosin, nevirapine, valaciclovir, nelfinavir, efavirenz, ganciclovir, lamivudine, famciclovir, stavudine, abacavir, indinavir, oseltamivir, inosiplex, adefovir, and a mixture thereof.
US11/741,287 2006-11-09 2007-04-27 Composite For Liver-Specific Delivery and Release of Therapeutic Nucleic Acids or Drugs Abandoned US20080138394A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/791,600 US8318199B2 (en) 2006-11-09 2010-06-01 Liposome for liver-specific delivery and release of therapeutic nucleic acids or drugs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020060110402A KR100817024B1 (en) 2006-11-09 2006-11-09 Composite for specifically transporting a nucleic acid or a drug to liver and pharmaceutical composition comprising the same
KR10-2006-0110402 2006-11-09

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/791,600 Division US8318199B2 (en) 2006-11-09 2010-06-01 Liposome for liver-specific delivery and release of therapeutic nucleic acids or drugs

Publications (1)

Publication Number Publication Date
US20080138394A1 true US20080138394A1 (en) 2008-06-12

Family

ID=39411771

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/741,287 Abandoned US20080138394A1 (en) 2006-11-09 2007-04-27 Composite For Liver-Specific Delivery and Release of Therapeutic Nucleic Acids or Drugs
US12/791,600 Active US8318199B2 (en) 2006-11-09 2010-06-01 Liposome for liver-specific delivery and release of therapeutic nucleic acids or drugs

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/791,600 Active US8318199B2 (en) 2006-11-09 2010-06-01 Liposome for liver-specific delivery and release of therapeutic nucleic acids or drugs

Country Status (2)

Country Link
US (2) US20080138394A1 (en)
KR (1) KR100817024B1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110739A1 (en) * 2007-05-15 2009-04-30 University Of North Texas Health Science Center At Forth Worth Targeted cancer chemotherapy using synthetic nanoparticles
WO2009150284A3 (en) * 2008-06-13 2010-12-09 Proyecto De Biomedicina Cima, S.L. Apo-a conjugates for the administration of biologically active compounds
WO2011038205A2 (en) * 2009-09-25 2011-03-31 Curna, Inc. Treatment of growth hormone (gh) related diseases by inhibition of natural antisense transcript to gh
ES2362062A1 (en) * 2009-12-11 2011-06-28 Research Center Borstel New conjugates and compositions for immunotherapy and anti-tumor treatment. (Machine-translation by Google Translate, not legally binding)
CN103169660A (en) * 2013-04-15 2013-06-26 石正国 Prepared ribavirin lipidosome oral emulsion with high encapsulation efficiency
US8734853B2 (en) 2008-11-17 2014-05-27 University Of North Texas Health Science Center At Fort Worth HDL particles for delivery of nucleic acids
US8771664B2 (en) 2009-12-11 2014-07-08 Fundacion Para La Investigacion Medica Aplicada Compositions comprising apolipoprotein A polypeptide and interleukin 15, and methods of treatment using the same
US20160176946A1 (en) * 2013-06-05 2016-06-23 Csl Limited Process for preparing apolipoprotein a-i (apo a-i)
US9763892B2 (en) 2015-06-01 2017-09-19 Autotelic Llc Immediate release phospholipid-coated therapeutic agent nanoparticles and related methods
US11712407B2 (en) * 2014-12-30 2023-08-01 Celltrion, Inc. Hybrid-type multi-lamellar nanostructure of epidermal growth factor and liposome and method for manufacturing same

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
WO2009129227A1 (en) 2008-04-17 2009-10-22 Pds Biotechnology Corporation Stimulation of an immune response by enantiomers of cationic lipids
KR101131202B1 (en) 2009-02-23 2012-04-05 (주)에이피테크놀로지 Method for preparation of nanostructured complex of hydrophobic drug
JP6348489B2 (en) 2012-06-15 2018-07-04 ピーディーエス バイオテクノロジー コーポレイションPds Biotechnology Corporation Cationic lipid vaccine composition and methods of use
CN103655477B (en) * 2012-09-03 2017-06-23 唐为钢 A kind of Entecavir HDL encapsulating preparation and its production and use
TWI672149B (en) 2012-09-21 2019-09-21 美商Pds生技公司 Improved vaccine compositions and methods of use
WO2017083820A1 (en) 2015-11-13 2017-05-18 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
US11712418B2 (en) 2017-05-26 2023-08-01 Bruin Biosciences, Inc. Chemoembolization agents
CN107998080A (en) * 2017-11-21 2018-05-08 东南大学 A kind of active targeting of coupled antibody carries medicine long circulating liposome and preparation method thereof
KR102280309B1 (en) 2019-06-13 2021-07-22 부산대학교 산학협력단 Increased drug delivery platform using W/O/W triolein emulsion in the liver
WO2020252375A1 (en) * 2019-06-14 2020-12-17 Dnalite Therapeutics, Inc. Compositions and methods for biological delivery vehicles

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050008617A1 (en) * 2002-09-28 2005-01-13 Massachusetts Institute Of Technology Compositions and methods for delivery of short interfering RNA and short hairpin RNA
US20050042632A1 (en) * 2002-02-13 2005-02-24 Sirna Therapeutics, Inc. Antibodies having specificity for nucleic acids
US20050222064A1 (en) * 2002-02-20 2005-10-06 Sirna Therapeutics, Inc. Polycationic compositions for cellular delivery of polynucleotides
US20060008910A1 (en) * 2004-06-07 2006-01-12 Protiva Biotherapeuties, Inc. Lipid encapsulated interfering RNA
US20060019912A1 (en) * 2003-12-19 2006-01-26 Chiron Corporation Cell transfecting formulations of small interfering RNA related compositions and methods of making and use
US7402404B2 (en) * 2001-10-03 2008-07-22 The Children's Hospital Of Philadelphia Assay method for measurement of net cholesterol flux

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6287590B1 (en) 1997-10-02 2001-09-11 Esperion Therapeutics, Inc. Peptide/lipid complex formation by co-lyophilization
JP2008001003A (en) * 2006-06-23 2008-01-10 Konica Minolta Holdings Inc Inkjet image recording method

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7402404B2 (en) * 2001-10-03 2008-07-22 The Children's Hospital Of Philadelphia Assay method for measurement of net cholesterol flux
US20050042632A1 (en) * 2002-02-13 2005-02-24 Sirna Therapeutics, Inc. Antibodies having specificity for nucleic acids
US20050222064A1 (en) * 2002-02-20 2005-10-06 Sirna Therapeutics, Inc. Polycationic compositions for cellular delivery of polynucleotides
US20050008617A1 (en) * 2002-09-28 2005-01-13 Massachusetts Institute Of Technology Compositions and methods for delivery of short interfering RNA and short hairpin RNA
US20060019912A1 (en) * 2003-12-19 2006-01-26 Chiron Corporation Cell transfecting formulations of small interfering RNA related compositions and methods of making and use
US20060008910A1 (en) * 2004-06-07 2006-01-12 Protiva Biotherapeuties, Inc. Lipid encapsulated interfering RNA

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110739A1 (en) * 2007-05-15 2009-04-30 University Of North Texas Health Science Center At Forth Worth Targeted cancer chemotherapy using synthetic nanoparticles
WO2009150284A3 (en) * 2008-06-13 2010-12-09 Proyecto De Biomedicina Cima, S.L. Apo-a conjugates for the administration of biologically active compounds
US8734853B2 (en) 2008-11-17 2014-05-27 University Of North Texas Health Science Center At Fort Worth HDL particles for delivery of nucleic acids
WO2011038205A2 (en) * 2009-09-25 2011-03-31 Curna, Inc. Treatment of growth hormone (gh) related diseases by inhibition of natural antisense transcript to gh
WO2011038205A3 (en) * 2009-09-25 2011-10-27 Opko Curna, Llc Treatment of growth hormone (gh) related diseases by inhibition of natural antisense transcript to gh
ES2362062A1 (en) * 2009-12-11 2011-06-28 Research Center Borstel New conjugates and compositions for immunotherapy and anti-tumor treatment. (Machine-translation by Google Translate, not legally binding)
US8771664B2 (en) 2009-12-11 2014-07-08 Fundacion Para La Investigacion Medica Aplicada Compositions comprising apolipoprotein A polypeptide and interleukin 15, and methods of treatment using the same
CN103169660A (en) * 2013-04-15 2013-06-26 石正国 Prepared ribavirin lipidosome oral emulsion with high encapsulation efficiency
US20160176946A1 (en) * 2013-06-05 2016-06-23 Csl Limited Process for preparing apolipoprotein a-i (apo a-i)
JP2016521694A (en) * 2013-06-05 2016-07-25 シーエスエル、リミテッド Method for producing apolipoprotein AI (Apo AI)
US9890203B2 (en) * 2013-06-05 2018-02-13 Csl Limited Process for preparing apolipoprotein A-I (Apo A-I)
RU2668831C2 (en) * 2013-06-05 2018-10-02 СиЭсЭл ЛИМИТЕД Process for preparing apolipoprotein a-i (apo a-i)
US10421799B2 (en) 2013-06-05 2019-09-24 Csl Limited Process for preparing Apolipoprotein A-I (Apo A-I)
US11279750B2 (en) 2013-06-05 2022-03-22 Csl Limited Process for preparing apolipoprotein A-I (Apo A-I)
US11712407B2 (en) * 2014-12-30 2023-08-01 Celltrion, Inc. Hybrid-type multi-lamellar nanostructure of epidermal growth factor and liposome and method for manufacturing same
US9763892B2 (en) 2015-06-01 2017-09-19 Autotelic Llc Immediate release phospholipid-coated therapeutic agent nanoparticles and related methods

Also Published As

Publication number Publication date
KR100817024B1 (en) 2008-03-26
US20100239657A1 (en) 2010-09-23
US8318199B2 (en) 2012-11-27

Similar Documents

Publication Publication Date Title
US8318199B2 (en) Liposome for liver-specific delivery and release of therapeutic nucleic acids or drugs
Ding et al. A biomimetic nanovector-mediated targeted cholesterol-conjugated siRNA delivery for tumor gene therapy
JP5749155B2 (en) Protease stabilized acylated insulin analogue
Wan et al. Enzyme-responsive liposomes modified adenoviral vectors for enhanced tumor cell transduction and reduced immunogenicity
JP2021522228A (en) Lipid-based formulations for RNA delivery
JP5600299B2 (en) Vesicle formulation
CN103881084B (en) The phospholipid derivant of a kind of branched polyethylene glycol and the lipid membrane structure body of composition thereof
JP2018529738A (en) Methods for therapeutic administration of messenger ribonucleic acid drugs
JP2000506865A (en) Targeted delivery of genes encoding interferons
JP7379325B2 (en) Cationic lipid compositions for tissue-specific delivery
Levačić et al. Dynamic mRNA polyplexes benefit from bioreducible cleavage sites for in vitro and in vivo transfer
Chen et al. A TRAIL-delivered lipoprotein-bioinspired nanovector engineering stem cell-based platform for inhibition of lung metastasis of melanoma
CN105012956A (en) Difunctional tumor targeted liposome drug-delivery system and preparation and application thereof
KR20060012661A (en) Methods and compositions for interferon therapy
Gupta et al. Exploring the therapeutic potential of the bioinspired reconstituted high density lipoprotein nanostructures
Sun et al. Arf6-mediated macropinocytosis-enhanced suicide gene therapy of C16TAB-condensed Tat/pDNA nanoparticles in ovarian cancer
US20190275345A1 (en) Remote control of light-triggered virotherapy
CN115487150B (en) Liver-targeted traceable drug delivery carrier, preparation method and application thereof, and diabetes treatment drug
Jung et al. Low immunogenic bio-nanocapsule based on hepatitis B virus escape mutants
US11801304B2 (en) Formulated and/or co-formulated liposome compositions containing TFGB antagonist prodrugs useful in the treatment of cancer and methods thereof
CN114452407A (en) Gene editing delivery system and preparation method and application thereof
US20040014709A1 (en) Methods and compositions for interferon therapy
Gao et al. PreS/2-21-guided siRNA nanoparticles target to inhibit hepatitis B virus infection and replication
JP7016084B2 (en) Lipid derivative for nucleic acid introduction
Sun et al. A biomimic reconstituted high density lipoprotein nanosystem for enhanced VEGF gene therapy of myocardial ischemia

Legal Events

Date Code Title Description
AS Assignment

Owner name: MOGAM BIOTECHNOLOGY RESEARCH INSTITUTE, KOREA, REP

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, MEEHYEIN;KIM, SOO IN;SHIN, DUCKHYANG;AND OTHERS;REEL/FRAME:019222/0874

Effective date: 20070403

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION