WO2008064540A1 - Compositions comprenant le facteur hgfk1 anti-angiogène véhiculé par un virus adéno-associé et utilisations correspondantes - Google Patents

Compositions comprenant le facteur hgfk1 anti-angiogène véhiculé par un virus adéno-associé et utilisations correspondantes Download PDF

Info

Publication number
WO2008064540A1
WO2008064540A1 PCT/CN2007/001343 CN2007001343W WO2008064540A1 WO 2008064540 A1 WO2008064540 A1 WO 2008064540A1 CN 2007001343 W CN2007001343 W CN 2007001343W WO 2008064540 A1 WO2008064540 A1 WO 2008064540A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
hgfk1
gene
aav
group
Prior art date
Application number
PCT/CN2007/001343
Other languages
English (en)
Chinese (zh)
Inventor
Marie C. M. Lin
Zan Shen
Hsiang Fu Kung
Original Assignee
Lin Marie C M
Zan Shen
Hsiang Fu Kung
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 Lin Marie C M, Zan Shen, Hsiang Fu Kung filed Critical Lin Marie C M
Publication of WO2008064540A1 publication Critical patent/WO2008064540A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1833Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • composition containing adeno-associated virus-mediated anti-angiogenic factor HGFK1 and application thereof
  • the present invention relates to a construct or composition for preventing or treating cancer, in particular to an adeno-associated virus vector or an adenovirus vector-mediated expression of the anti-angiogenic factor HGFK1 construct AAV-HGFK1 or Adv-HGFK1, and a construct containing the same A composition of AAV-HGFK1 with an adenoviral vector or a construct of an adenoviral vector expressing an anti-oncogene, tumor suppressor gene or cytokine gene.
  • sustained angiogenesis is one of the key factors influencing this process, as neovascularization provides the nutrient shield, oxygen, and various growth factors necessary for tumor cells to proliferate.
  • HGFK1 is the Kringle 1 domain of HGF (HGFK1) of human hepatocyte growth factor and contains amino acid sequences 127 to 214 of HGF. HGFK1 is a recently discovered anti-angiogenic factor. In vitro experiments have shown that the anti-angiogenic effect of recombinant HGFK1 protein is significantly stronger than that of angiostatin (Xin L. et al., Biochem Biophys Res Commun, 2000; 277: 186-190). ).
  • Adenovirus is a DNA double-stranded, non-enveloped virus with approximately 36 kb of genomic DNA encoding 14 proteins.
  • the adenoviral vector is also an ideal viral vector.
  • the virus strain used at the beginning has strong immunogenicity and may cause a strong allergic reaction. Encouragingly, Adv has recently been improved and its immunogenicity has been greatly reduced.
  • adenoviral vectors In general, the main advantages of current adenoviral vectors are: (1) stable sexual shields, high expression, relatively safe for humans; (2) a wide range of infected host cells that can infect both mitotic and quiescent cells; 3) Adenovirus infection cells do not need to integrate into the host cell genome, there is no risk of activating oncogenes or insertion mutations; ( 4 ) are defective viruses that cannot replicate autonomously; ( 5 ) are easy to prepare, recombinant viruses can be injected intravenously , spray, intratracheal instillation or preparation into capsules orally and enter the body by intestinal absorption, etc., to obtain a high-priced viral vector.
  • AAV adeno associated virus
  • P53 is a protein product of tumor suppressor gene expression, which inhibits cell division, induces apoptosis, up-regulates multiple tumor suppressor genes and down-regulates multiple oncogenes, and inhibits vascular endothelial growth factor (VEGF) gene and The role of drug resistance (MDR) gene expression (Sherr CJ, Cell, 2004; 1 16: 235-46.).
  • VEGF vascular endothelial growth factor
  • MDR drug resistance
  • hTERTC27 is an anti-cancer gene discovered by our research team. It encodes a 27 kDa amino acid sequence at the C-terminus of human telomerase, which can be specifically expressed in tumor cells and has tumor cell-specific cytotoxicity. It can cause tumor cell growth inhibition and apoptosis, but does not inhibit and apoptosis in normal cells. In vivo test-face confirmed that hTERTC27 can inhibit tumor formation and growth (Huang J. et al" Biochem Biophys Res Commun, 2003 Feb 14; 301: 627-32. 1 Huang J. et al., Cancer Res, 2002 Jun 1 ; 62: 3226-32.) The results of the hTERTC27 study have been filed for US patents (US Patent Application No. 10/449,565 filed 30 May 2003).
  • the cytokine IL-2 can significantly enhance the immune function of T cells, sputum cells, macrophages, and sputum cells, and also induce killer LAK cells and activate tumor infiltrating lymphocytes (TIL).
  • TIL tumor infiltrating lymphocytes
  • IL-2 synergizes with many other cytokines to enhance immune function, so IL-2 has significant immune enhancement and anti-tumor effects (Waldmann TA, Annu Rev Med, 2006; 57: 65-81).
  • the present inventors have selected a tumor suppressor, an anticancer or a cytokine which can be used in combination with HGFK1 through creative labor, and have utilized different expression vectors to enable them to fully exert synergistic effects in preventing or treating cancer. Summary of the invention
  • the present invention provides a method capable of inhibiting tumor blood vessel growth and inhibiting cancer cell growth and metastasis.
  • Recombinant constructs In particular, the inventors selected a tumor suppressor gene such as p53, such as an anti-cancer gene of hTERT C27 or a cytokine gene such as IL-2, and expressed by creative labor.
  • the construct of the anti-angiogenic factor constitutes a composition for synergistic prevention or treatment of cancer.
  • the constructs and compositions of the present invention are not easily degraded, have a long action time, and are stable in effect, thereby being capable of effectively preventing and/or treating cancer growth, infiltration, metastasis, and recurrence.
  • a construct for preventing or treating cancer comprising a nucleic acid sequence of an anti-angiogenic factor HGFK1 and an adeno-associated virus expression vector or an adenovirus expression vector, and referred to as AAV-HGFK1 or Adv-HGFK1, is provided.
  • the nucleic acid sequence of HGFK1 is SEQ ID NO: 1 or a homologous sequence thereof.
  • composition for preventing or treating cancer which comprises an effective dose:
  • a first construct comprising an anti-angiogenic factor gene and an adeno-associated virus expression vector; an adenovirus expression vector or a second construct comprising a tumor suppressor gene, an anti-oncogene, and a cytokine or Combination and adenoviral expression vectors.
  • a construct or composition of the invention for the manufacture of a medicament for the prophylaxis or treatment of cancer, wherein said medicament comprises a prophylactically or therapeutically effective amount of said construct or composition.
  • a specific antibody to the HGFK1 protein is provided.
  • This antibody can be used for immunohistochemical staining and Western Blot detection of HGFK1 protein.
  • the antibodies can also be used in the preparation of related kits.
  • kits for detecting a HGFK1 protein or gene comprising a specific antibody of the HGFK1 protein of the present invention, or a probe or primer for detecting the HGFK1 gene.
  • a further aspect of the invention provides a method of preventing or treating cancer in a subject comprising administering to the individual a prophylactically or therapeutically effective amount of a construct or composition of the invention, the individual in need of administration.
  • the administering comprises administering by intravenous or intratumoral injection.
  • the individual is a mammal, preferably a human.
  • a method of inhibiting tumor or cancer cell growth comprising contacting an effective amount of a construct or composition of the invention with a tumor or cancer cell.
  • the method of inhibiting tumor or cancer cell growth of the present invention can be carried out in vivo or in vitro.
  • Contact in the method comprises transfecting or transforming a vector or composition of the invention into a tumor or cancer Cell.
  • the anti-angiogenic factor gene used in the present invention may be selected from the group consisting of angiostatin, an endostatin, and a human HGFK1 gene.
  • the anti-angiogenic factor gene comprises a human HGFK1 gene encoding the amino acid sequence of positions 127 to 214 of HGF, and the nucleic acid sequence is SEQ ID NO: 1.
  • the anti-angiogenic factor HGFK1 gene preferably encodes the amino acid sequence of SEQ ID NO: 5 or a homologous sequence thereof.
  • the amino acid sequence of the anti-angiogenic factor HGFK1 in addition to SEQ ID NO: 5, further comprises six histidine sequences, i.e., the amino acid sequence of SEQ ID NO: 6.
  • the tumor suppressor gene used in the present invention is preferably p53; the anticancer gene is preferably TERTC27; and the cytokine is preferably a gene encoding IL-2.
  • the composition may contain any pharmaceutically acceptable carrier.
  • the cancer is a solid tumor, preferably hepatocellular carcinoma, colorectal cancer, and glial cell carcinoma, preferably primary and metastatic liver cancer.
  • the construct or composition of the present invention can be used for the prevention or treatment of primary cancer, metastatic cancer. It is preferred to treat metastatic cancer.
  • the HGFK1-expressing construct constructed using an adenovirus or an adeno-associated virus vector can stably and stably express the anti-angiogenic gene HGFK1 protein in vivo as compared with the expression of a conventional vector in the prior art.
  • the anti-angiogenic construct of the composition of the present invention can exert a synergistic effect with an adenovirus expression vector or a construct containing an adenoviral vector inhibiting a tumor cell growth gene, and a construct containing anti-angiogenesis is used alone.
  • the body can significantly enhance the effect of inhibiting tumors.
  • Figure 1A shows the construct AAV-HGFK 1 of an adeno-associated virus vector containing the HGFK1 gene.
  • Figure IB shows a schematic representation of the construct pAd-X of an adenoviral vector (Adv) comprising the p53, hTERTC27, HGF 1 or IL-2 gene, wherein X is any of the p53, hTERTC27, HGFK1 or IL-2 genes. .
  • Figure 2 is a bar graph showing the transfection efficiency of the adenoviral vector Adv significantly enhancing the construct of the adeno-associated virus AAV vector.
  • Adenovirus Adv significantly enhanced the transfection efficiency of adeno-associated virus AAV recombinant vector by more than 10 times.
  • the abbreviation Adv used in the figure is Adeno virus; LD-AAV-EGFP is low dose AAV-EGFP (MOI is lxlO 3 ); HD-AAV-EGFP is high dose AAV-EGFP (MOI is lx lO 4 ); EGFP is an enhanced green fluorescent protein.
  • Figure 3 shows the results of gene therapy for AAV-HGFK1 that significantly inhibited tumor growth, metastasis, tumor cell apoptosis, and significantly prolonged survival in experimental animals.
  • PBS is PBS buffer
  • EGFP is enhanced green fluorescent protein.
  • Figure 3A shows the tumor volume of each group of hepatocellular carcinoma animal models at different time points after treatment.
  • Figure 3B shows the pathological anatomical features of ascites, intrahepatic metastasis, lung metastasis, abdominal metastasis, and primary tumor volume at death in each group of hepatocellular carcinoma model animals.
  • Figure 3C shows the apoptosis of tumor cells in each group of hepatocellular carcinoma models by TUNEL staining at 21 days after treatment. The arrows indicate apoptotic cells.
  • Figure 3D shows the survival of each group of hepatocellular carcinoma models. ** indicates a value ⁇ 0.01 compared with the control group.
  • Figure 4 shows the survival curve of gene therapy in a nude mouse model of subcutaneously implanted human malignant glioma. Divided into seven groups: Adv-hTERTC27 Adv-HGFK1 or AAV-HGFK1 alone treatment group; AAV-HGFKl+Adv-hTERT C27 composition or AAV-HGFKl+Adv-IL-2 composition treatment group; control group was PBS or AAV-EGFP injection group.
  • the graph shows the change in the percentage of animal survival at different times (days) after treatment.
  • Figure 5 shows the therapeutic end of a gene therapy composition for an animal model of liver metastases from colorectal cancer. fruit.
  • Figure 5A shows the percentage of survival of the AAV-HGFK1 group, the AAV-EGFP group, the AAV-EGFP+Adv-p53 group, and the AAV-HGFK1+Adv-p53 group at different times;
  • Figure 5B shows: AAV-HGFK1 group Survival time, mean survival time, and number of animals with liver metastasis and primary tumorigenesis, AAV-EGFP group, AAV-EGFP+Adv-p53 group, and AAV-HGFK1+Adv-p53 group. ** indicates that the survival time of the AAV-HGFKl+Adv-p53 treatment group was significantly longer than that of the AAV-EGFP control group. Value ⁇ 0.01.
  • the abbreviation EGFP in the figure is an enhanced green fluorescent protein.
  • Figure 6 shows the survival curve treatment results of the gene therapy composition of Buffalo rat primary hepatocellular carcinoma.
  • the graph shows changes in the percentage of animal survival at different times after treatment (days:).
  • PBS is PBS buffer
  • HD-AAV-HGFK1 is high dose AAV-HGFKL
  • Figure 7 shows the results of the toxicity test.
  • Abbreviations in the figure ALT is alanine aminotransferase; AST is aspartate aminotransferase; TB is total bilirubin; LDH is lactate dehydrogenase; CK is creatine kinase; PBS is PBS buffer.
  • the invention thus uses the term "human hepatocyte growth factor Kringle 1 domain (HGFK1)" to include alternative variants of its conserved amino acids that do not alter its ability to inhibit cell growth and angiogenesis.
  • HGFK1 human hepatocyte growth factor Kringle 1 domain
  • stringent hybridization conditions refers to hybridization and washing conditions of a gene having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% of the core with the gene. When the nucleotide sequence is complementary, it can still hybridize to the gene.
  • Such hybridization conditions are well known in the art, for example, see Current Protocols in Molecular Biology, John Wiley & Sons, NY (1989), 6.3.1-6.3.6; Molecular Cloning, Cold Spring Harbor Laboratory, NY (1982), pp. 387. -389.
  • homologous sequence refers to a nucleotide sequence identity of at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% of the gene. And does not substantially affect the function of the encoded protein. Gene same The source sequence may also be a fragment of the gene as long as the function of the encoded protein is substantially unchanged.
  • homologous sequence of the amino acid sequence as used herein means having at least
  • preventing or treating cancer means that the construct or composition used can prevent the metastasis or recurrence of cancer, alleviate, alleviate, control, ameliorate or cure the primary cancer or metastatic cancer or its associated symptoms; It can prolong the life span or survival time of cancer patients and reduce mortality.
  • control group refers to a administration group of a construct consisting of green fluorescent protein (EGFP) and an adeno-associated virus vector (AAV) and/or a PBS buffer group.
  • EGFP green fluorescent protein
  • AAV adeno-associated virus vector
  • a first construct comprising an anti-angiogenic factor gene and an adeno-associated virus vector
  • HGFK1 Xin L. et al., Biochem Biophys Res Commun, 2000; 277: 186-190 gene
  • HGFK1 Xin L. et al., Biochem Biophys Res Commun, 2000; 277: 186-190 gene
  • a preferred nucleic acid sequence of the invention is the HGFK1 gene of SEQ ID NO: 1 and homologous sequences thereof.
  • the SEQ ID NO: 1 nucleic acid sequence encodes the amino acid sequence of positions 127 to 214 of HGF (SEQ ID NO: 5).
  • the nucleic acid sequence of HGFK1 further comprises a nucleic acid sequence which hybridizes to SEQ ID NO: 1 under stringent hybridization conditions or a homologous sequence thereof.
  • the adeno associated virus (AAV) used in the present invention includes all types of AAV, such as AAV1, AAV2, AAV5, AAV8, AAVl, preferably AAV2 (Choi VW et al. Curr. Gene Titer. 2005; 5 : 299-310.).
  • the AAV selected by the present invention can efficiently transmit the HGFK1 gene and improve the expression efficiency.
  • the construct expressing the HGFK1 gene of the present invention constructed from the AAV, particularly AAV2 can synergize with the construct constructed by the adenovirus expression vector in the composition of the present invention, and significantly enhance the treatment or The effect of preventing cancer.
  • an anti-angiogenic factor gene sequence can be obtained and recombined with an adeno-associated virus vector to constitute a construct of the present invention.
  • Phase For more information see Sambrook et al (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbour Laboratory, NY; Marston, F (1987) DNA Cloning Techniques: A Practical Approach Vol III IRL Press, Oxford UK; DNA Cloning : FM Ausubel et al, Current Protocols in - Molecular Biology, John Wiley & Sons, Inc. (1994).
  • a construct comprising the HGFK1 gene and an adenoviral vector was constructed according to conventional methods well known in the art and methods of the first construct.
  • Adenoviral vector comprising a packaging system used in the construction, preferably pAd Adeno-X TM packaging systems, packaging systems AdEasyTM, AdMax TM packaging system, Ad5 / F35Max TM packaging system, pAd / CMV / V5-DEST TM packaging system / CMV /V5-DESTTM packaging system.
  • the adenovirus expression vector selected by the present invention particularly the pAd/CMV/V5-DESTTM packaging system, can efficiently transmit HGFK1 factor and has the effect of preventing and treating cancer when used alone or in combination. For example, see the embodiments of the present invention.
  • a nucleic acid sequence comprising a tumor suppressor gene, an anti-oncogene or a cytokine and a second construct of an adenoviral vector
  • a foreign gene that can be used to construct a second construct including a tumor suppressor gene such as p53, such as an anti-oncogene of hTERT C27 or a nucleic acid sequence of a cytokine gene such as IL-2.
  • the genes of these factors can be combined with adenoviral vectors to form a second construct.
  • these second constructs containing a tumor suppressor gene, an anti-oncogene or a cytokine and an adenoviral vector are combined with a first construct comprising an anti-angiogenic factor gene and an adeno-associated virus vector, resulting in an unexpected The effect of preventing or treating cancer.
  • the foreign gene for use in the second construct of the present invention is preferably a nucleic acid sequence of p53, hTERT C27 or IL-2.
  • the genes encoding p53, hTERT C27 and IL-2 have the nucleic acid sequences of SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively.
  • Adenoviral vector comprising a packaging system used in the construction, Adeno-X TM packaging systems, packaging systems AdEasyTM, AdMax TM packaging system, Ad5 / F35Max TM packaging system, pAd / CMV / V5-DEST TM packaging system, preferably pAd / CMV / V5-DEST TM packaging system.
  • a construct containing a tumor suppressor gene, an anti-oncogene gene or a cytokine gene and an adenoviral vector can be constructed by the method of the above 2.
  • constructs comprising a foreign gene and an adenoviral vector are well known in the art, for example, see the construction of a construct of the HGFK1 gene and an adenoviral vector described above.
  • composition for preventing and treating cancer of the present invention comprises a prophylactically or therapeutically effective amount of a first construct comprising an anti-angiogenic factor gene and an adeno-associated virus vector; an adenoviral vector, or a tumor suppressor gene, an anti-oncogene or a second construct of a cytokine and an adenoviral vector; and optionally, a pharmaceutically acceptable carrier.
  • compositions for administration to a cancer patient for the construct or composition of the invention.
  • Any suitable for example parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraorbital, ocular, intraventricular, intraorbital, intraspinal, intracranial, intraperitoneal, nasal Internal, aerosol or oral administration methods.
  • the dosage form can be a liquid solution or suspension; a tablet or capsule; a powder, a nasal drop or an aerosol.
  • compositions suitable for parenteral administration include sterile aqueous or non-aqueous formulations of the compositions of the invention which are preferably isotonic with the blood of the recipient.
  • Formulations are prepared according to known methods using suitable dispersing or wetting agents and suspending agents as pharmaceutical carriers.
  • the sterile injectable preparation may also be a sterile injection solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, non-volatile oils can be used conventionally as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Biocompatible, biodegradable lactide polymers, lactide/glycolide polymers or polyoxyethylene-polyoxypropylene copolymers can also be employed to control the release of the compounds.
  • Other potential systems for regulating parenteral delivery of the constructs or compositions of the present invention include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable perfusion systems, and liposomes.
  • Formulations suitable for inhalation may include a pharmaceutical carrier such as lactose, or may be an aqueous solution containing, for example, polyoxyethylene-9-laurate, glycocholate, and deoxycholate, or may be administered as a nasal drop. Oily solution or as a gel.
  • an “effective amount” based on a construct or composition of the invention includes a therapeutically effective dose or a prophylactically effective dose.
  • therapeutically effective dose is meant an effective dose at the dose, and within the necessary time period, the desired therapeutic outcome is achieved, such as inhibition of cancer cell growth, reduction of lesions, reduction in relapse rate, prolonged survival, and the like.
  • the therapeutically effective dose of the construct or composition will also vary depending on factors such as the patient's disease state, age, sex, and weight, as well as the ability of the construct or composition to elicit the desired response in the patient.
  • the dosage regimen can be adjusted to provide an optimal therapeutic response.
  • the therapeutically effective dose can also be an amount that the therapeutically effective effect exceeds any toxic or detrimental effects of the compound.
  • prophylactically effective dose is meant an effective dose at the dose, and within the necessary time period, the desired prophylactic result is achieved, for example to prevent metastasis and recurrence of cancer.
  • construct or composition for treating or prophylactically effective dosage range may be effective dosage is any integer of 6 lx lO 3 to lx lO, e.g., a first prophylactic or therapeutic body constructed from a MOI (multiplicity of infection) of the present invention.
  • a preferred effective dose of the composition of the present invention is AAV-HGFK1: 4 ⁇ 10 ⁇ vg/kg to l l0 13 vg/kg; Adv-X: lx l0 1() vp/kg to lx lO 11 vp/kg.
  • the dosage may vary depending on the severity of the cancer to be prevented or treated.
  • the specific dosage can be configured according to the individual's needs and adjusted according to the judgment of the professional who is administering or supervising the administration.
  • the dosage ranges set forth herein are merely exemplary and are not intended to limit the dosage range selected by the medical personnel.
  • the amount of construct in the composition can vary depending on factors such as the disease state, age, sex, and weight of the individual.
  • the dosage form can be adjusted to provide optimal therapeutic results. 5.
  • the invention further relates to a method of preventing or treating cancer in a subject, the method comprising administering to the individual an effective amount of a construct or composition of the invention.
  • the individual has cancer, is suspected of having cancer, or has a susceptibility (risk) of cancer, and thus the individual needs to be prevented or treated for cancer.
  • the judgment of the above individual state is well known to those skilled in the art or clinical practitioners and physicians.
  • the individual is a mammal, preferably a human.
  • the cancer may be a primary cancer or a metastatic cancer, preferably a solid tumor, more preferably a hepatocellular carcinoma, a colon cancer, and a sphincter cell carcinoma.
  • the route of administration to the individual includes intravenous, intraperitoneal, intralesional or intratumoral, sustained release of the implant, intramuscular, intrathecal and oral administration. It is preferably administered intravenously or intratumorally.
  • the vein is preferably administered intravenously from a vein near the tumor, for example, for liver cancer, the portal vein is selected for administration.
  • the invention further relates to a method of inhibiting tumor or cancer cell growth, the method comprising contacting an effective amount of a construct or composition of the invention with said tumor or cancer cell.
  • the method of inhibiting tumor or cancer cell growth may comprise transfecting or transforming the tumor or cancer cell in vivo or in vitro using the construct or composition of the present invention, thereby inhibiting growth of the cell.
  • the cancer or tumor cell is selected from the group consisting of liver cancer, colorectal cancer, and glial cancer cells.
  • the construct or composition of the invention may be administered to an individual or to a tumor cell in a single dose or in multiple doses.
  • a single dose can be, for example, the above effective dose.
  • the multiple dose may be from about 1/2 to about 1/100, preferably from about 1/2 to about 1/10, more preferably from about 1/2 to about 1/4 of the above effective amount.
  • One skilled in the art can determine or adjust the dosage to be administered based on the specific circumstances of the patient or tumor cell growth and the toxicity of the drug. It is also permissible to administer doses above or below the effective dose.
  • the inventors designed AAV-HGFK1 and Adv eg Adv-p53, Adv-hTERTC27, Adv-IL-2
  • a composition of Adv itself eg Adv-p53, Adv-hTERTC27, Adv-IL-2
  • the therapeutic effect of the composition is significantly better than the therapeutic effect of any of AAV-HGFK1, Adv-p53, Adv-hTERTC27 or Adv-IL-2 alone.
  • AAV-HGFK1 In orthotopic hepatocellular carcinoma in rats, prevention and treatment with AAV-HGFK1 is carried out by an injection route including portal vein and intratumoral injection. The results showed that AAV-HGFK1 can cause apoptosis and necrosis of tumor cells, significantly prolong the survival time of animal models of hepatocellular carcinoma (average 30 days from the control group to 49 days in the treatment group), and completely inhibit the metastasis of cancer (Fig. 3).
  • the therapeutic effect of AAV-HGFK1 and AAV-EGFP+Adv-p53 was significantly better than that of the control treatment group; while the average survival time of the AAV-HGFK1+Adv-p53 composition treatment group was significantly longer than AAV-HGFK1.
  • the group and the AAV-EGFP+Adv-p53 group were directly smaller than 0.001; the treatment of the AAV-HGFKl+ Adv-p53 composition even reached a complete cure, resulting in long-term tumor-free survival of the experimental animals (Fig. 5).
  • a route of administration including a para-tumor injection and a tail vein injection.
  • Adv-hTERTC27, Adv-HGFK1 and AAV-HGFK1 alone extended the mean survival time from 28.3 days to 42.8, 47.5 and 60.6 days, respectively; and AAV-HGFKl+Adv-hTERTC27 composition and AAV-HGFK1+ Adv-IL-2
  • the efficacy of the composition was more pronounced: up to 80 days after treatment, none of the treatment groups administered with the composition of AAV-HGFK1 + Adv-hTERTC27 died, and only one treatment group of the composition administered with AAV-HGFK1 + Adv-IL-2 Death ( Figure 4).
  • the present invention may encompass other cancers.
  • the basis is as follows: 1. Several primary and metastatic cancer models are selected as the more common types of cancer, which are representative; 2. All cancers invade and metastasize depend on angiogenesis, and The invention can provide long-term, high-level prevention and treatment of anti-angiogenesis; 3.
  • the tumor suppressor gene selected in the invention has broad-spectrum anti-cancer properties; 4.
  • the composition of the invention can prevent cancer recurrence and metastasis .
  • constructs or compositions of the invention can be used to prevent or treat the growth, infiltration, metastasis, and recurrence of solid tumors. 7. Toxicity study
  • the present invention provides an evaluation of the toxicity of a composition comprising a recombinant expression construct of AAV-HGFK1 and a composition of an Adv-p53 and/or Adv-hTERTC27 expression construct.
  • a composition comprising a recombinant expression construct of AAV-HGFK1 and a composition of an Adv-p53 and/or Adv-hTERTC27 expression construct.
  • We administered high doses of the composition to experimental animals and then tested for indicators of liver function in serum: alanine aminotransferase, aspartate aminotransferase, and total bilirubin; indicators that reflect myocardial damage: lactate dehydrogenase, Creatine kinase and Tn-T did not detect signs of liver function and myocardial damage.
  • no significant damage was found in the ⁇ & ⁇ staining of myocardial tissue.
  • the constructs of the invention can be purified to the extent necessary to immunize an animal, such as a rabbit.
  • an animal such as a rabbit.
  • one or three constructs can be prepared against angiogenic factors, and each construct is injected into at least two animals.
  • the antiserum can be produced by a series of injections, preferably including at least three booster injections.
  • Primary immunization can be performed with Freund's complete adjuvant, followed by booster immunization with Freund's incomplete adjuvant.
  • Antibody titers can be monitored using Western Blot and immunoprecipitation analysis using the purified protein.
  • the immune serum can be affinity purified using a C Br-dextran coupled protein.
  • Anti-serum specificity can be determined using an unrelated protein plate.
  • a polypeptide corresponding to a relatively unique immunogenic region of an anti-angiogenic factor of the invention can be produced and introduced by C-terminal lysine and keyhole velocin (KLH) Coupling.
  • KLH keyhole velocin
  • Antisera against each of these polypeptides can be affinity purified by polypeptide conjugated to BSA, using polypeptide conjugates in ELISA and Western Blot and assayed for specificity by Western Blot and immunoprecipitation.
  • monoclonal antibodies that specifically bind to the anti-angiogenic factors of the invention can be prepared according to standard hybridoma methods. Once produced, specific recognition of monoclonal antibodies can be determined by Western blotting or immunoprecipitation. Antibodies which bind to the factors described herein are believed to be useful; such antibodies can be used, for example, as immunoassays.
  • antibodies can be prepared using immunogenic polypeptide fragments.
  • immunogenic polypeptide fragments For example, at least 75%, at least 50%, at least 25%, or at least 5% of the amino acid sequence of SEQ ID NO: 5 of the present invention is used as an immunogenic polypeptide fragment from the end of several bases to the amino terminal to produce an immunogenic polypeptide fragment to produce antibody.
  • the amino acid sequence RSK KGT VSI TKS GIKC (SEQ ID NO: 7) is preferred as the immunogen of HGFK1 in the present invention.
  • the present invention also relates to a kit for detecting an anti-angiogenic factor HGFK1 protein or a gene thereof, the kit comprising the anti-angiogenic factor HGFK1 protein of the present invention, or according to an anti-angiogenic factor HGFK1 gene sequence, preferably A primer or probe designed according to SEQ ID NO: 1 or a homologous sequence thereof.
  • the kit is for detection analysis of ELISA, Western blot, co-immunoprecipitation, and the like. In another embodiment, the kit is for detection analysis of Northern Blot, Southern Blot, PCR, and the like. Thus, in one embodiment of the invention, the kit further includes instructions for directing the assay and other commonly used reagents for the assay, such as labeling reagents.
  • the present invention also relates to the use of the antibody against the angiogenic factor HGFK1 protein for the preparation of the kit.
  • the human HGFK1 cDNA (SEQ ID NO: 1 nucleic acid sequence) was inserted into the AAV packaging vector of pAM/CAG/EGR-1-pL-WPRE-BGH-polyA which had been previously digested with the same endonuclease to constitute AAV.
  • -HGFK1 recombinant vector The three-vector helper virus-deficient packaging system (AAV, H22 and pFD6) was used for packaging, that is, the recombinant vector AAV-HGFK1 and the helper plasmids H22 and pFD6 were co-transfected into HEK-293 cells by calcium carbonate method, and transfected for 60-72 hours. Thereafter, the cells were harvested, and the isolated recombinant virus particles were purified by HiTrap Heparin affinity chromatography, and the final titer was determined by real-time PCR.
  • the present invention provides a method for preparing and packaging a construct of a recombinant defective adenovirus expression vector encoding human p53, hTERTC27 IL-2 or HGFK1 gene, comprising the steps of: human p53 gene, hTERTC27 gene, IL-2 gene or HGFK1 gene are respectively connected to the shuttle plasmid pENTR TM 2B, and then the recombinant shuttle plasmid pENTR-p53, pENTR-hTERTC27, pENTR-IL-2 or pENTR-HGFKl and pAd / CMV / V5-DEST expression vectors obtained by homologous recombination pAd- P53, pAd-hTERTC27, pAd-IL-2 or pAD-HGFK1, the resulting expression vector was transfected into 293 A cells with LipofectamineTM 2000 Reagent (Invitrogen), lysed, concentrated and purified by cesium
  • the purified recombinant adenovirus carrying human p53, hTERTC27, IL-2 or HGFK1 gene was transfected into the rabbit carotid artery and used to carry the LacZ reporter gene.
  • Recombinant adenovirus (Adv-LacZ) was used as a control. After 3 days, the expression of mRNA and protein of human p53, hTERTC27 or IL-2 gene was detected by RT-PCR and ELISA.
  • Rat hepatocyte cancer cell line McA-RH-7777 cells were surgically injected Buffalo rats under the left hepatic subcapsular (lxlO 6 cells / only, one-time injection). After 7 days, the rat abdominal cavity was opened by surgery, and it was found that there was a 3x3nmi 2 tumor tissue on the left lobe surface of the liver, which confirmed the establishment of the model.
  • a primary hepatocellular carcinoma model was established as described in Example 3.
  • the treatment was started immediately after the tumor formation was confirmed. They were randomly divided into three groups, six in each group: 1. PBS buffer group; 2. AAV-EGFP group (1.2 x 10 12 vg) /only); 3. AAV-HGFK1 group (1.2x10 12 vg/only). The PBS group and the AAV-EGFP group were used as a control group, and AAV-HGFK1 was used as a treatment group.
  • the injection route included two types of portal vein and intratumoral injection, in which the injected dose in the tumor was 0.2 ⁇ 10 12 vg/only, and the dose in the portal vein was lx l0 12 vg/only, which was confirmed to be a one-time injection immediately after the tumor model was established. .
  • Figure 3A shows the tumor volume of each group of hepatocellular carcinoma animal models at different time points after treatment. There was no significant difference in tumor volume between the treatment group (AAV-HGFK1 injection) and the control group (PBS or AAV-EGFP injection) on day 7 and day 14 (P>0.05), and the tumor volume of the treatment group was significantly higher on the 21st day after treatment. Less than the control group (P ⁇ 0.01).
  • Figure 3B shows the pathological anatomical features of each group of hepatocellular carcinoma model animals at death, in which ascites, intrahepatic metastasis, lung metastasis, abdominal metastasis, and primary tumor volume were observed.
  • ascites 6 animals in each of the two control groups had ascites (100%), and treatment Only 2 of the group had ascites (33%); intrahepatic metastasis: 6 animals in both control groups had intrahepatic metastasis (100%), but no intrahepatic metastasis was observed in all animals in the treatment group; lung metastasis: PBS control There were 5 lung metastases in the group (83.3%) and 6 lung metastases in the AAV-EGFP control group (100%), but no lung metastasis in all animals in the treatment group; abdominal metastasis: 6 animals in both control groups There was abdominal metastasis (100%), and no abdominal metastases were seen in all animals in the treatment group.
  • Primary tumor volume The treatment group was significantly smaller than the control group (P ⁇
  • Fig. 3C shows the apoptosis of tumor cells in each group of hepatocellular carcinoma models by TUNEL staining at 21 days after treatment.
  • the arrow shows apoptotic cells.
  • the tumor cell apoptosis index of the treatment group was significantly lower than that of the two control groups (P ⁇ 0.01).
  • Figure 3D shows the survival of each group of hepatocellular carcinoma models.
  • the mean survival time (49 days) in the AAV-HGFK1 treatment group was significantly longer than the mean survival time of both controls (both 30 days).
  • a human malignant glioma nude mouse model was established as described in Example 3.
  • AAV-EGFP group (l xloHyg/only);
  • AAV-HGFK1 group (UxloUyg/only);
  • the tumor is administered immediately after the tumor is formed, and the administration route is a tumor injection and a tail vein injection. All are one-time injections.
  • the survival time of the three treatment groups of AAV-HGFKl was significantly prolonged, corpse ⁇ 0.01); while the composition of AAV-HGF 1+ Adv-hTERTC27 and AAV-HGFK1 + Adv-IL-2 were more effective (** indicates The composition of the treatment group of AAV-HGFKl + Adv-hTERTC27 and AAV-HGFKl+Adv-IL-2 was significantly longer than the treatment group of Adv-hTERTC27, Adv-HGFK1, AAV-HGFK1 alone, and the corpse was prolonged.
  • liver metastasis of colorectal cancer in mice After the liver metastasis of colorectal cancer in mice, they were randomly divided into four groups, six in each group:
  • AAV-EGFP group (Ux loUyg / only);
  • AAV-HGFKl group (1.5xlO u vg / only);
  • AAV-HGFK1+Ad-P53 (1.5x l0 u vg/only +2.5 x 10 9 vp/only).
  • Figure 5A shows that in the animal model of liver metastasis of colorectal cancer, the AAV-HGFK1 group and the AAV-EGFP+Adv-p53 group were significantly better than the AAV-EGFP treatment group; and the AAV-HGFKl+Adv-p53 composition treatment The mean survival time of the group was significantly longer than that of the AAV-HGFK1 group and the AAV-EGFP+Adv-p53 group, which was less than 0.001.
  • Figure 5B shows that the mean survival time of the AAV-HGFKl+Adv-p53 treatment group was over 61 days, while the average survival time of the other groups was 21 days in the AAV-EGFP group and 30 days in the AAV-HGFK1 group, AAV-EGFP+Adv. -p53 group for 30 days.
  • one of the six animals in the AAV-HGFKl+Adv-p53 treatment group achieved long-term disease-free survival; The tumors were all eliminated and some of the primary tumors were eliminated.
  • AAV-HGFK1 (3 ⁇ 10 ⁇ vg/only);
  • AAV-HGFKl + Adv- p53 composition (AAV-HGFK1 of 3x 10 "vg / only; Adv-p53 is 2.5x l0 9 vp / only).
  • the route of administration includes portal vein and intratumoral injection, both of which are single injections.
  • the test results are shown in Figure 6.
  • composition of AAV-HGFKl+Adv-p53 was able to extend the mean survival time from 30 days to 61 days, even more than 10 times higher doses of AAV-HGFK1 ( 3 ⁇ 10 12 vg/) compared with the control group. Only) 53 days to treat.
  • the mean survival time of the Adv-p5 3 treatment group alone was 37 days.
  • the mean survival time of the AAV-HGFK1 (3x10" vg/only) treatment group was 39 days.
  • the present invention provides an evaluation test for detecting the toxicity of the AAV-HGFK1 recombinant expression construct and the composition composed of Adv-p53 and Adv-hTERTC27 to the body. High doses of the composition were administered to the experimental animals.
  • PBS buffer injection group as a control
  • AAV-HGFK1 4.8x l0 12 vg/only injection group
  • AAV-HGFKl+Adv-hTERTC27 (4.8x l0 12 vg/only +1 ⁇ 10 10 ⁇ /only) injection group.
  • the administration methods are portal vein injection and intrahepatic injection, all of which are one-time injections, and the intrahepatic injection dose is one-third of the dose of the portal vein.
  • the test results are shown in Figure 7.
  • liver function damage On the 7th and 60th day after administration, the indicators reflecting liver function in the serum were detected: alanine aminotransferase, aspartate aminotransferase and total bilirubin, and no signs of liver function damage were found (Fig. ⁇ .
  • Synthetic peptide RSYKGT VSI TKS GIKC, whose sequence is identical to the amino acid sequence at the end of HGFK1 ⁇
  • Lymph node injection method 1 subcutaneous (or intradermal) injection of live BCG 50 mg (about 0.30 ml per side) in the two hind paws of the rabbit. After 7-10 days, the rabbit iliac crest and diaphragmatic lymph nodes are swollen; the synthetic polypeptide emulsion with complete adjuvant is injected into the swollen bilateral lymph nodes.
  • 0.50ml (containing synthetic peptide 5mg/ml, penicillin 1 000U/ml, streptomycin 1 OOO ig/ml); 3 if necessary, after 14 days, repeat step 2; 4 after 7 days, in both lymph nodes Injection of 0.50 ml of the synthetic polypeptide emulsion with complete adjuvant (containing synthetic peptide 5 mg/ml, penicillin 1 000 U/ml, streptomycin 1 OOO ⁇ g/ml); 5 5-7 days later, ear vein blood collection . Serum titer was determined.
  • the serum was diluted and added to the peripheral wells.

Abstract

L'invention concerne des constructions AVV-HGFK1 et Adv-HGFK1 exprimant le facteur-HGFK1 anti-angiogène médié par le virus et par l'adénovirus adéno-associé. L'invention concerne également des compositions comprenant AVV-HGFK1 et la construction médiée par l'adénovirus exprimant l'antioncogène, le gène supresseur de tumeur ou la cytokine. Ces constructions ou ces compositions peuvent être utilisées pour traiter, contrôler ou empêcher des carcinomes primaires et des carcinomes métastatiques. L'invention concerne les utilisations de ces constructions ou des ces compositions pour préparer les médicaments de lutte contre les cancers et de leur traitement.
PCT/CN2007/001343 2006-11-27 2007-04-23 Compositions comprenant le facteur hgfk1 anti-angiogène véhiculé par un virus adéno-associé et utilisations correspondantes WO2008064540A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNA2006101403471A CN1958075A (zh) 2006-11-27 2006-11-27 含腺伴随病毒介导的抗血管生成因子的组合物及其应用
CN200610140347.1 2006-11-27

Publications (1)

Publication Number Publication Date
WO2008064540A1 true WO2008064540A1 (fr) 2008-06-05

Family

ID=38070028

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2007/001343 WO2008064540A1 (fr) 2006-11-27 2007-04-23 Compositions comprenant le facteur hgfk1 anti-angiogène véhiculé par un virus adéno-associé et utilisations correspondantes

Country Status (2)

Country Link
CN (1) CN1958075A (fr)
WO (1) WO2008064540A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1254378A (zh) * 1997-04-28 2000-05-24 罗纳-布朗克罗莱尔股份有限公司 腺病毒介导的肿瘤内投送血管生成拮抗剂用于肿瘤的治疗
US20030148950A1 (en) * 2001-10-09 2003-08-07 Li Xin Kringle domain 1 of human hepatocyte growth factor and uses therefor
US20030225027A1 (en) * 2002-05-31 2003-12-04 Huang Jun Jian Telomerase reverse transcriptase fragments and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1254378A (zh) * 1997-04-28 2000-05-24 罗纳-布朗克罗莱尔股份有限公司 腺病毒介导的肿瘤内投送血管生成拮抗剂用于肿瘤的治疗
US20030148950A1 (en) * 2001-10-09 2003-08-07 Li Xin Kringle domain 1 of human hepatocyte growth factor and uses therefor
US20030225027A1 (en) * 2002-05-31 2003-12-04 Huang Jun Jian Telomerase reverse transcriptase fragments and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HUANG J.J. ET AL.: "A human TERT C-terminal polypeptide sensitizes HeLa cells to H2O2-induced senescence without affecting telomerase enzymatic activity", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 301, no. 3, 14 February 2003 (2003-02-14), pages 627 - 632 *
JUN JIAN HUANG ET AL.: "Ectopic Expression of a COOH-terminal Fragment of the Human Telomerase Reverse Transcriptase Leads to Telomere Dysfunction and Reduction of Growth and Tumorigenicity in HeLa Cells", CANCER RESEARCH, vol. 63, 1 June 2002 (2002-06-01), pages 3226 - 3232 *
XIN L. ET AL.: "Kringle 1 of human hepatocyte growth factor inhibits bovine aortic endothelial cell proliferation stimulated by basic fibroblast growth factor and causes cell apoptosis", BIOCHEM. BIOPHYS. RE. COMMUN., vol. 277, no. 1, 14 October 2000 (2000-10-14), pages 186 - 190 *

Also Published As

Publication number Publication date
CN1958075A (zh) 2007-05-09

Similar Documents

Publication Publication Date Title
JP6740340B2 (ja) 免疫優性アデノウイルスエピトープに突然変異を有する腫瘍溶解性アデノウイルスおよび癌処置におけるそれらの使用
KR102351555B1 (ko) Hpv 및 관련 질환용 면역 증강 치료 백신
US9744224B2 (en) Methods for treating cancer by administration of nucleic acids encoding FAP and cancer antigens
KR102409147B1 (ko) 종양분해성 백시니아 바이러스
KR102020018B1 (ko) 종양 선택적 e1a 및 e1b 돌연변이
US20080213220A1 (en) Cancer-targeted viral vectors
US20150250837A1 (en) Oncolytic virus encoding pd-1 binding agents and uses of the same
US7772367B2 (en) C-terminal p53 palindromic peptide that induces apoptosis of cells with aberrant p53 and uses thereof
Zolochevska et al. Pigment epithelial-derived factor and melanoma differentiation associated gene-7 cytokine gene therapies delivered by adipose-derived stromal/mesenchymal stem cells are effective in reducing prostate cancer cell growth
Ma et al. Oral adeno‐associated virus‐sTRAIL gene therapy suppresses human hepatocellular carcinoma growth in mice
US10617729B2 (en) Multitargeting onocolytic adenovirus, methods of use, and methods of making
JP2020503390A (ja) 治療用タンパク質の標的細胞特異的な産生のため、および標的細胞に関連する疾患、状態、または障害の治療のための、膜融合性脂質ナノ粒子、ならびにその作製および使用のための方法
Deng et al. Oncolytic and immunologic cancer therapy with GM-CSF-armed vaccinia virus of Tian Tan strain Guang9
KR20200026894A (ko) 개인 맞춤형 백신
KR20180015113A (ko) 암을 치료하기 위한 치료 조성물 및 사용 방법
JP2003504052A (ja) 複製−コンピテント抗癌ベクター
US20140147917A1 (en) Reic-expressing adenovirus vector
KR102167934B1 (ko) 재조합 아데노바이러스 및 이를 포함하는 줄기세포
JP2002515442A (ja) 癌を処置するためのアデノウィルス−化学治療剤のコンビネーション
Sutter et al. Gene therapy for gastric cancer: is it promising?
WO2022077591A1 (fr) Utilisation d'adénovirus recombinant dans la préparation de médicaments pour la prévention contre les virus
WO2020091066A1 (fr) Polythérapie pour le traitement du cancer thoracique faisant appel à ad-reic/dkk-3 et à un inhibiteur de point de contrôle
WO2008074189A1 (fr) Adénovirus recombinant comprenant un gène khp50 recombinant et son procédé de préparation et ses utilisations
WO2006125381A1 (fr) Virus du gene de ciblage tumoral zd55-il-24, son procede de construction et son application
WO2008064540A1 (fr) Compositions comprenant le facteur hgfk1 anti-angiogène véhiculé par un virus adéno-associé et utilisations correspondantes

Legal Events

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

Ref document number: 07720916

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07720916

Country of ref document: EP

Kind code of ref document: A1