WO2008065726A1 - Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale - Google Patents

Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale Download PDF

Info

Publication number
WO2008065726A1
WO2008065726A1 PCT/JP2006/324316 JP2006324316W WO2008065726A1 WO 2008065726 A1 WO2008065726 A1 WO 2008065726A1 JP 2006324316 W JP2006324316 W JP 2006324316W WO 2008065726 A1 WO2008065726 A1 WO 2008065726A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
antitumor
gene
tumor
cells
Prior art date
Application number
PCT/JP2006/324316
Other languages
English (en)
Japanese (ja)
Inventor
Toshiyoshi Fujiwara
Noriaki Tanaka
Yasuo Urata
Original Assignee
Oncolys Biopharma Inc.
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 Oncolys Biopharma Inc. filed Critical Oncolys Biopharma Inc.
Priority to PCT/JP2006/324316 priority Critical patent/WO2008065726A1/fr
Publication of WO2008065726A1 publication Critical patent/WO2008065726A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/255Esters, e.g. nitroglycerine, selenocyanates of sulfoxy acids or sulfur analogues thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to an anti-tumor drug resistance overcome agent comprising a recombinant virus in which a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence and an E1B gene in this order is incorporated.
  • Cancer cells acquire resistance to anticancer drugs through various molecular mechanisms, such as transporter activation and receptor kinase changes (Bush JA, Li G. Cancer chemoresistance: the relationship between p53 and multidrugtransporters. Int J Cancer 98:, 323 ⁇ 330, 2002.) Although research on anticancer drug resistance has been conducted, there is no solution yet. Cell death due to anticancer drugs is apoptosis, and resistance to anticancer drugs is apoptotic resistance (Schmitt CA, Lowe SW. Apoptosis and chemoresistance in transgenic cancer models.
  • telomerisin adenovirus
  • Cell death by telomerisin is a different mechanism of action from apoptosis.
  • telomerisin and the microtubule agent docetaxel each drug exhibited an independent antitumor effect, and as a result, a combination effect independent of each other was observed in vivo. Disclosure of the invention
  • An object of the present invention is to provide an anti-tumor drug resistance overcome agent comprising a recombinant virus in which a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence, and an E1B gene in this order is incorporated.
  • telomerisin showed an extremely excellent cell killing effect against antitumor agent-resistant tumor cells, and the present invention was completed. That is, the present invention is as follows.
  • An anti-tumor agent resistance-resolving agent comprising a recombinant virus incorporating a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence, and an E1B gene in this order.
  • the promoter for human telomerase is, for example, hTERT.
  • the virus is preferably adenovirus, for example.
  • Antitumor agents are, for example, platinum preparations, microtubule inhibitory activators, alkylating activators, antimetabolite activators, anticancer antibiotics and topoisomerase inhibitory activators as well as their pharmacologically acceptable It is possible to use at least one selected from the group consisting of salts.
  • antitumor agents include docetaxel, .paclitaxel, cisplatin, psulfan, methotrexate, fluorouracil, tegafur, bleomycin, adriamycin, mitomycin C, vinorelbine and irinotecan and their pharmacologically acceptable conditions. And at least one selected from the group consisting of salts.
  • the tumor is, for example, lung cancer, colon cancer, stomach cancer, breast cancer, esophageal cancer, head and neck cancer, liver cancer, pancreas cancer, gallbladder or bile duct cancer, prostate cancer, bladder cancer, cervical cancer, thyroid cancer, And at least one selected from the group consisting of ovarian cancer, leukemia, lymphoma, sarcoma, and mesenchymal tumor.
  • a pharmaceutical composition for combination therapy of a tumor that has acquired antitumor drug resistance comprising a substance having a use.
  • the substance having an antitumor action is, for example, 'docetaxel, pinolerubin, irinotecan and histone deacetylase inhibitory activity inhibitor and their pharmacological (group consisting of this acceptable salt). At least one selected from '.
  • a tumor that has acquired resistance to an antitumor agent comprising administering to a mammal a recombinant virus incorporating a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence, and an E1B gene in this order.
  • a method to suppress cell proliferation comprising administering to a mammal a recombinant virus incorporating a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence, and an E1B gene in this order.
  • Recombinant virus in which a polynucleotide containing a human telomerase promoter, _E1A gene, IRES sequence and E1B gene in this order is incorporated, and a substance having antitumor activity or pharmacologically acceptable
  • a method of inhibiting the growth of tumor cells that have acquired resistance to an antitumor agent comprising administering to a mammal in combination with a salt.
  • an example of a promoter for human telomerase is hTERT.
  • the virus is preferably an adenovirus.
  • Antitumor agents include, for example, platinum preparations, microtubule inhibitory activators, alkylating activators, antimetabolite activators, anticancer antibiotics and tobodies.Somerase inhibitor activators and their pharmacologically acceptable And at least one selected from the group consisting of possible salts.
  • antineoplastic agents include docetaxel, paclitaxel, cisplatin, psnorephan, methotrexate, funoleolauracinole, tegafunore, preomesin, adriamycin, mitomycin C, vinorelbine and irinotecan and their pharmacologically acceptable conditions. And at least one selected from the group consisting of possible salts.
  • Tumors include, for example, lung cancer, colon cancer, stomach cancer, breast cancer, esophageal cancer, head and neck cancer, liver cancer, knee cancer, gallbladder or bile duct cancer, prostate cancer, bladder cancer, cervical cancer, thyroid And at least one selected from the group consisting of adenocarcinoma, ovarian cancer, leukemia, lymphoma, sarcoma, and mesenchymal tumor.
  • the substance having an antitumor action is, for example, a group consisting of docetaxenole, pinorelbine, irinotecan and histone deacetylase inhibitory activity inhibitors and pharmacologically acceptable salts thereof. At least one selected from. Brief Description of Drawings
  • FIG. 1 shows the structure of OBP-301 (telomerisin) and OBP-401.
  • Figure 2 shows the characterization of drug resistance sublines. a) CDDP, b) Parkitaxel, —
  • FIG. 3 is a diagram in which anticancer agents were administered to various cancer cell resistant strains and the antitumor effect was observed with a microscope.
  • Fig. 4 is a diagram showing the results of an experiment in which cell death was performed.
  • Fig. 5 shows the analysis of MDR1 protein expression by Western blot.
  • FIG. 6 shows the analysis of CAE expression using flow cytometry.
  • FIG. 7 shows the results of an XTT assay performed after infecting a cancer cell resistant strain with OBP-301. 'a) Series A431,' b) Series DU145
  • FIG. 8 shows the results of an experiment in which a cancer cell resistant strain was infected with OBP-301 and then subjected to XTT assembly over time.
  • Fig. 9 shows the results of CBB staining after infection of a cancer cell resistant strain with OBP-301.
  • FIG. 10 is a view observed with a microscope after infecting a cancer cell resistant strain with OBP-301 and OBP-401.
  • FIG. 11 is a view observed with a microscope after infecting a cancer cell resistant strain with OBP-301 and OBP-401. Quantitative real time PCR was used to angularly analyze the intracellular growth of OBP-301.
  • Figure 12 shows the results of an experiment analyzing the intracellular growth of OBP-301 using quantitative real-time PCR. a) A431 series, b) DU145 series Best mode for carrying out the invention
  • the present invention relates to an anti-tumor drug resistance overcome agent comprising a recombinant virus in which a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence and an E1B gene in this order is incorporated.
  • a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence and an E1B gene in this order is incorporated.
  • the present invention relates to an anti-tumor agent resistance overcomer that exerts a cell-killing effect on tumor cells that have acquired anti-tumor agent resistance.
  • Anti-tumor drug resistance refers to the fact that the effect of a tumor cell gradually acquires resistance to these anti-tumor drugs when the treatment with the same anti-tumor drug is repeated.
  • Various molecular mechanisms such as transporter activation and receptor kinase changes are involved in the mechanism by which tumor cells acquire antitumor drug resistance.
  • resistance to antitumor agents can be said to be apoptosis resistance.
  • the recombinant virus contained in the antitumor agent resistance overcomer of the present invention starts to proliferate when the telomerase promoter is expressed.
  • the recombinant virus since the expression of telomerase is extremely high in tumor cells as compared with normal cells, the recombinant virus has a feature that it does not grow in normal cells but grows only in tumor cells.
  • the mechanism of cell death that this recombinant virus exerts on tumor cells is different from the mechanism of apoptosis by conventional anti-tumor agents, so that tumor cells are treated with anti-tumor agents. Even if it is controlled, it can proliferate and replicate in the cell without any influence. As a result, as described above, even when resistance to an antitumor agent is acquired in a tumor cell, the tumor cell can be specifically killed. Thus, as a result of repeated use of antitumor agents, tumor cells that have acquired antitumor agent resistance and no longer have a cytocidal effect are administered with the recombinant virus included in the present invention. It can be said that “resistance to anti-tumor agents can be overcome”. Therefore, the recombinant virus included in the present invention is useful as an anti-tumor drug resistance overcomer.
  • the recombinant virus used in the present invention refers to a virus in which a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence, and an E.1B gene in this order is incorporated into a genome.
  • the virus to be used is not particularly limited, but an adenovirus is preferable from the viewpoint of safety. Among adenoviruses, type 5 adenovirus is particularly preferable from the viewpoint of ease of use.
  • the E1A gene, the IRES sequence and the E1B gene are driven by the human telomerase promoter. Since the expression of telomerase is extremely high in tumor cells as compared with normal cysts, the telomerase promoter is expressed in tumor cells containing telomerase, whereby the recombinant virus included in the present invention grows. As a result, cell damage due to virus growth occurs in the tumor cells, and the recombinant virus used in the present invention can specifically kill the tumor cells.
  • telomerase promoter determines the transcription start site of telomerase and directly regulates its frequency. Telomerase is an enzyme that maintains telomere length by antagonizing shortening during replication of eukaryotic chromosomes.
  • the type of such telomerase promoter is not particularly limited, but for example, a promoter of human telomerase reverse transcriptase QiTERT) is preferable.
  • hTERT is a 1.4 kbp region upstream of its 5 'end, and many transcription factor binding sequences have been confirmed, and this region is thought to be the hTERT promoter. Among them, the 181 bp sequence upstream of the translation start site is downstream. It is an important core region for gene expression.
  • this core area As long as it contains a region, it can be used without limitation, but it is preferable to use an upstream sequence of about 378 bp completely including this core region as the hTERT promoter. It has been confirmed that the gene expression efficiency of this sequence of about 378 bp is equivalent to that of the 181 bp core region alone.
  • the base sequence of such hTERT is not shown in SEQ ID NO: 1.
  • hTERT hybridizes under stringent conditions with DNA consisting of a complementary base sequence to DNA consisting of the base sequence shown in SEQ ID NO: 1 in addition to the base sequence shown in SEQ ID NO: 1, and has hTERT activity Nucleotide base sequences are also included.
  • T stringent conditions for the above hybridization include lxSSC to 2xSSC, 0.1 to 0.5% SDS, and 42 to 68 ° C, and more specifically. For example, after pre-hybridization at ⁇ 68 ° C for 30 minutes or more, washing in 2xSSC, 0.1% SDS at room temperature for 5 to 15 minutes is performed 4 to 6 times. .
  • the ⁇ 1 ⁇ gene, the IRES sequence and the E1B gene are included in this order because the IRES sequence inserted between the E1A gene and the E1B gene is used. This is because the proliferation ability is increased.
  • the £ 1 gene and 18 genes are included in the E1 gene.
  • the E1 gene is one of the early gene (early: E) and late gene (late: L) related to DNA replication of the virus. It codes for a protein involved in the control of transcription of the viral genome.
  • the E1A protein encoded by the E1A gene activates transcription of genes (E1B, E2, E4, etc.) necessary for the production of infectious viruses.
  • the E1B protein encoded by the E1B gene promotes viral replication by helping the late gene (L gene) mRNA accumulate in the cytoplasm of the infected host cell and inhibit host cell protein synthesis. To do.
  • the base sequences of E1A gene and E1B gene are shown in SEQ ID NO: 2 and SEQ ID NO: 3, respectively.
  • E1A and E1B are stringent with DNA consisting of a base sequence complementary to the DNA consisting of the base sequences shown in SEQ ID NO: 2 and SEQ ID NO: 3 in addition to the base sequences shown in SEQ ID NO: 2 and SEQ ID NO: 3, respectively. And a base sequence encoding a protein that hybridizes under various conditions and has E1A and E1B activities, respectively.
  • stringent conditions in the hybridization include, for example, lxSSC to 2xSSC, 0.1 to 0.5% SDS, and 42 to 68. More specifically, after prehybridization at 60 to 68 ° C for 30 minutes or more, wash in 2x SSC, 0.1% SDS at room temperature for 5 to 15 minutes. The conditions for performing 6 times are mentioned.
  • IRESdnternal Ribosome Entry Site is a protein synthesis initiation signal specific to the Picornaviridae family, and since it has a sequence complementary to the 3 'end of 18S ribosomal RNA, it is thought to serve as a ribosome binding site. It has been. It is known that mRNA derived from the Picornaviridae virus is translated via this sequence. The translation efficiency from the IRES sequence is high, and protein synthesis is performed even in the middle of mRNA independent of the cap structure. Therefore, in this virus, both the E 1 A gene and the E 1B gene downstream of the IRES sequence are independently translated by the promoter of human telomerase.
  • the expression of the telomerase promoter is controlled independently by the E1A gene and the E1B gene. Therefore, compared to the case where either the E1A gene or the E1B gene is controlled by the telomerase promoter, virus growth Can be more strictly limited to cells having telomerase activity.
  • the IRES sequence is shown in SEQ ID NO: 4.
  • IRES is highly stringent under stringent conditions with DNA consisting of a base sequence complementary to the DNA consisting of the base sequence shown in SEQ ID NO: 4, with the help of the base sequence shown in SEQ ID NO: 4.
  • a base sequence encoding a protein having IRES activity is also included.
  • stringent conditions in the above hybridization include lxSSC to 2xSSC, 0.1 to 0.5% 'SDS, and 42 to 68 ° C, and more specifically, 30 to 60 to 68 ° C. After prehybridization for at least 5 minutes, the conditions include washing in 2xSSC and 0.1% SDS at room temperature for 5 to 15 minutes, 4 to 6 times.
  • the promoter of human telomerase is located upstream of the E1 gene. This is because proliferation can be promoted in cells having telomerase activity.
  • the gene contained in the recombinant virus of the present invention can be obtained by ordinary genetic engineering techniques. The case where hTERT is used as a human telomerase is described below.
  • E1A-S By performing RT-PCR and / or DNA-PCR from cells expressing the E1 gene, such as 293 cells, using primers such as E1A-S, El A-AS, E1B-S, and E1B-AS After amplifying the E1A gene and E1B gene and confirming the sequence using a known method such as TA cloning, if necessary, the E 1 A and E 1B DNA fragments can be excised with a known restriction enzyme.
  • E1A-IRES-E1B can be inserted into a known vector (eg, pIRES), and then the hTERT promoter sequence excised with a restriction enzyme such as Mul or Bgllll can be inserted upstream of> E1A.
  • a known vector eg, pIRES
  • a restriction enzyme such as Mul or Bgllll
  • telomelysin a virus incorporating the cassette consisting of hTERT-ElA-IRES- ⁇ used in the present invention (FIG. 1a) is referred to as “telomerisin” or “Telomelysin”.
  • the following method can be used.
  • cells such as human colon cancer cell SW620, human lung cancer cell A549, H1299, etc. are plated on a culture plate containing an appropriate culture medium and cultured at 37 ° C in the presence of carbon dioxide.
  • As the culture solution DMEM, MEM, RPMI-1640 or the like generally used for animal cell culture is adopted, and serum, antibiotics, vitamins and the like can be added as necessary.
  • the cultured cells are infected by inoculating a certain amount of the virus, for example, 0.:! To 10 MOI, preferably 0.1 to 1 MOI (multiplicity of infection).
  • MOI is the ratio between the amount of virus (infectious unit) and the number of cells when a certain amount of cultured cells are infected with a certain amount of virus particles, and is used as an index for infecting cells with viruses. To confirm virus growth, collect virus-infected cells, extract DNA, and perform quantitative analysis by performing real-time PCR using a primer that targets the appropriate gene of the virus. be able to. 3. Antitumor agents
  • Antitumor agents refer to drugs that have the effect of inhibiting the growth and proliferation of tumor cells (cancer cells) that become tumor masses as a result of repeated cell division due to disorder of the body's regulatory mechanism and excessive cell proliferation. Also included are drugs that inhibit growth by inhibiting nucleic acid synthesis in cancer cells or inhibiting metabolism. Specifically, the following alkylation activators, antimetabolite activators, antibiotics, microtubule inhibitor activators, platinum preparations, topoisomerase inhibitor activators, etc. Absent. These antitumor agents may form pharmacologically acceptable salts.
  • Alkylation activator This preparation has the effect of introducing an alkyl group into the nucleic acid protein of cancer cells to cause cell damage.
  • alkyl group For example, carbo, kon, psulfan (mustard drug), dimustine ( Nitrosourea)).
  • Antimetabolite active agent This preparation has the action of antagonizing enzymes in the metabolic process and inhibiting cell synthesis, such as methotrexate (folic acid), mercaptopurine, (purine), Examples include cytarabine (pyrimidine), fluorouracil, tegafur, and carmofur.
  • Antibiotic examples include anti-cancer activity, actinomycin D, bleomycin, adriamycin, mitomycin C and the like.
  • Microtubule inhibitory active agent This preparation acts on microtubules and exhibits an antitumor effect. For example, docetaxel, paclitaxel (taxane), vinorelbine, pinklistin, vinblastine (alkaloid) Can be mentioned.
  • Platinum preparation This preparation has an action of inhibiting DNA synthesis by constituting intra-DNA chain or inter-chain bond or DNA protein bond, and examples thereof include cisplatin, alpoplatin, nedaplatin and the like. .
  • Topoisomerase inhibitor Irinotecan (topoisomerase I inhibitor), podophyllotoxin derivative (tobosomerase II inhibitor), which inhibits topoisomerase. What is topoisomerase?
  • the antitumor agent in the present invention is preferably a microtubule inhibitory activity '"herbal agent and a platinum preparation, and more preferably docetaxel and cisbratin, but is not limited thereto. It is not a thing.
  • Docetaxel is classified as a taxane among the above microtubule inhibitors and is mainly applied to the treatment of breast cancer and non-small cell lung cancer.
  • Docetaxel is obtained as a semi-synthetic product from Sewyichii needle extract, which promotes polymerization with tubulin and suppresses microtubule depolymerization as well as microtubule formation and stops cell mitosis. It has a function.
  • Cisbratin is mainly applied to the treatment of testicular tumor, bladder cancer, renal pelvic and ureteral tumor, prostate cancer, ovarian cancer, 'head and neck cancer, lung cancer,' esophageal cancer, cervical cancer.
  • the types of tumor (cancer) cells to which the antitumor agent resistance-resolving agent of the present invention acts are not limited, and all types of tumor cells can be used.
  • leukemia, lymphoma, sarcoma, mesenchymal tumor This is effective.
  • Most of the tumor cells derived from chick tissue show an increase in telomerase activity, and the antitumor agent resistance overcomer of the present invention generally acts on tumor cells whose proliferation has been activated by such telomerase activity. sell.
  • inorganic acid salts for example, hydrochloride, hydrobromide, sulfate, nitrate, perchlorate, phosphate
  • organic acid salts for example, vinegar Acid salt, trifluoroacetate, maleate, oxalate, tartrate, malonate, succinate, fumarate, sulfonate, malate, etc.
  • organic sulfonate eg, methanesulfonate Salts, trifluoromethanesulfonate, ethanesulfonate, benzenesulfonate, toluenesulfonate, etc.
  • amino acid salts eg aspartate, glutamate, etc.
  • quaternary amine salts alkali metal salts (eg, Sodium salt, strong salt, etc.), alkaline earth metal salt (eg, magnesium salt, calcium salt, etc.).
  • the antitumor agent resistance overcomer of the present invention is characterized in that it comprises a recombinant virus in which a polynucleotide containing a human telomerase promoter, an E 1A gene, an IRES sequence, and an E1B gene in this order is incorporated.
  • telomerisin the mechanism of action of cell death by telomerisin on tumor cells is different from the mechanism of action of apoptosis caused by conventional anticancer drugs. This is achieved by the nature of telomerisin, such that even when treated, the cells can proliferate and replicate in the cells without any influence.
  • the anti-tumor agent resistance overcomer of the present invention can be applied to the affected area as it is, or any known method, for example, injection such as vein, muscle, intraperitoneal or subcutaneous, or nasal cavity, oral cavity or It can also be introduced into a living body (target cells or organs) by inhalation from the lung, oral administration, intravascular administration using a catheter or the like.
  • it can be used as it is after handling or squeezing by a method such as freezing, or it can be used as it is, or a known pharmaceutically acceptable carrier such as an excipient, a bulking agent, a binder, a lubricant, etc. (Including buffering agents, tonicity agents, chelating agents, coloring agents, preservatives, fragrances, flavoring agents, sweetening agents, etc.) and the like.
  • the anti-tumor agent resistance overcomer of the present invention includes tablets, capsules, powders, granules, pills, liquids, syrups and other oral administration agents, injections, external preparations, suppositories, eye drops, etc. Depending on the dosage form, it can be administered orally or parenterally. Preferably, local injection into muscles, abdominal cavity, etc., injection into veins, etc. are exemplified. The dose is appropriately selected according to the type of active ingredient, administration route, administration subject, patient age, body weight, sex, symptom and other conditions.
  • the daily dose of the virus included in the present invention is 10 6 to about lOnPFl plaque forming units), preferably about 10 9 to lOuPFU, can be administered once a day, or can be divided into several times. Further, when using the virus of the present invention, it is possible to suppress the immunity of the living body by using a known immunosuppressive agent or the like so that the virus can be easily infected.
  • the virus of the present invention can be used as a pharmaceutical composition in combination with at least one substance having antitumor activity.
  • the substance having an antitumor action of the present invention refers to the growth and proliferation of tumor cells (cancer cells) that become tumor masses as a result of cell proliferation that is disrupted due to disorder of the body's regulatory mechanism and cell overgrowth.
  • tumor cells cancer cells
  • Substances that have the action of suppressing cancer including substances that suppress the nucleic acid synthesis of cancer cells or inhibit metabolism by inhibiting metabolism. Specific examples include the following.
  • Antibiotics actinomycin D, bleomycin, adriamycin, mitomycin C, etc.
  • Substances having microtubule inhibitory activity docetaxel, paclitaxel, pinolenolevin, pinklistin, vinblastine, etc.
  • Substances having topoisomerase inhibitory activity irinotecan, podophyllotoxin derivatives, etc.
  • the substances having antitumor activity include pharmaceutically acceptable salts. Examples of such salts include the salts described above.
  • the dose of the substance having an antitumor activity or a pharmaceutically acceptable salt thereof included in the present invention also depends on the type of active ingredient, administration route, administration subject, patient age, body weight, sex, symptom and other conditions. It is selected appropriately. For example, in the case of docetaxel, usually, once a day in adults, 60mg / m 2 of body surface area over a period of over one hour more than 1 intravenous drip infusion at 3-4 week intervals. The dose may be adjusted according to the symptoms. However, the highest dose once a 70mg / m 2.
  • irinotecan for example, irinotecan hydrochloride is usually administered to an adult once a day, 150 mg / m 2 intravenously 3-4 times at 2-week intervals, and the drug is withdrawn for at least 3 weeks. This can be repeated as one course.
  • the combined use of the recombinant virus contained in the antitumor agent regimen overcomer of the present invention and the substance having an antitumor action can suppress the growth of tumor cells.
  • the mode of combination is not limited to the mode in which the recombinant virus and the substance having an antitumor effect are mixed and administered at the same time, and the mode in which either one is administered first and the other is administered later is also possible. included.
  • both a recombinant virus and an antitumor substance are contained in one treatment schedule, they are included in the “combination” in the present invention. Specifically, the following treatment methods are mentioned, for example.
  • a substance having antitumor activity can be administered after telomerisin is administered.
  • Telomerisin may be administered intratumorally and at the same time a substance having antitumor activity may be administered systemically. In animal experiments, a clear concomitant effect is recognized by simultaneous administration as described above.
  • telomerisin it is also possible to administer telomerisin after administering the substance having antitumor activity. Administration of substances with anti-tumor activity may be short-term or long-term.
  • the antitumor agent resistance overcomer of the present invention is considered to have a very low possibility of causing side effects for the following reasons, and can be said to be a very safe preparation.
  • the virus of the present invention has a proliferative ability, it can be used at a lower concentration than a non-proliferative virus used in normal gene therapy.
  • an XTT assay can be performed.
  • XTT (2,3 bis [2-Methoxy 4-nitro-5-sulfpphenyl] -2H-tetrazolium-5-carboxyanilide inner salt) assay measures the activity of living cells by dehydrogenase of mitochondria in living cells This is a suitable method for monitoring cytotoxicity in vitro. The following is a detailed explanation of the XTT Atsay.
  • the XTT solution shows a yellow color when it does not contain phenol red or when dissolved in a balanced salt solution.
  • the mitochondrial dehydrogenase of the viable cells cleaves the tetrazolium ring of XTT and becomes soluble in aqueous solution. Colored formazan crystals are produced.
  • an electron conjugate substance such as phenazine methosulfate (PMS) is often added to the reaction solution as a reduction accelerator. Then, the cytotoxicity of the desired substance can be measured by measuring the resulting orange solution by the colorimetric method by utilizing the change in the amount of formazan produced by increasing or decreasing the number of cells.
  • the recombinant virus of the present invention is infected at an appropriate concentration (MOI: multiplicity of infection) to various cancer cells cultured in vitro. After culturing the cells under appropriate conditions, various concentrations of antitumor agents are administered to the virus-infected cell culture. After virus infection, after culturing for an appropriate period, the above XTT assembly can be performed to analyze the antitumor effect.
  • MOI multiplicity of infection
  • Adenoviruses infect and enter cells via the Coxsackie virus adenovirus receptor (CAR), but the efficiency of virus infection is reduced in cancer cells that are negative for CAE. Thus, the more CAR expression in cancer cells, the easier adenovirus enters the cancer cells.
  • Whether or not CAR is expressed in tumor cells may be determined by analyzing CAE expression using flow cytometry after culturing various tumor cells under appropriate conditions.
  • the anticancer drug FR901228 (Fujisawa Pharmaceutical Co., Ltd. (currently Astellas Pharma Inc.)) can enhance the expression of CAR in cancer cells.
  • FR901228 when used in combination with the recombinant adenovirus of the present invention, it is preferable in that it also has the action of improving the infection efficiency of the combined recombinant adenovirus in tumor cells.
  • Flow cytometry is a method of measuring the size of individual cells, DNA content, etc. in a cell population by measuring the fluorescence emitted from individual particles by flowing a cell suspension at high speed. Flow cytometry not only analyzes the differences in the relative size, shape, and internal structure of each cell, but also measures fluorescence intensity and type of fluorescence by fluorescent labeling to identify cells. And the abundance ratio of various cells constituting the cell group can be analyzed in a short time.
  • the nucleus Since the cell membrane cannot maintain its state due to cell death, the nucleus is easily stained with PI (propidium iodide) described later. Therefore, the amount of DNA and the cell cycle can be determined from the result of nuclear staining with PI.
  • the effect of the recombinant virus in the anti-tumor drug resistance overcoming agent of the present invention on the target cell cycle is determined by PI using the flow cytometry described above to stain the nucleus. Can be analyzed. Tumor cells are considered to be highly malignant when the proportions of S phase and G2 / M phase in the cell cycle are high. Therefore, it can be said that an antitumor effect is observed if a result that suppresses the ratio of G2 / M phase is obtained.
  • the proliferation of the recombinant virus in the target cell of the antitumor agent resistance overcomer of the present invention can be measured, for example, using quantitative real-time PCR as follows. In other words, after culturing the recombinant virus with an anti-tumor agent for an appropriate period, collect the virus-infected cells, extract the DNA, and use real-time PCR with primers that target the appropriate gene of the virus. By performing the above, it is possible to quantitatively analyze an appropriate gene possessed by the virus.
  • a gene encoding a fluorescent substance is incorporated into the recombinant virus of the present invention
  • cells in which the virus has been propagated are irradiated with excitation light to give a predetermined fluorescence (for example, in the case of GFP). Emits green fluorescence), allowing virus growth in cells to be visualized.
  • a predetermined fluorescence for example, in the case of GFP
  • GFP fluorescence expression is seen in the cell.
  • It is also possible to observe virus-infected cells over time by observing the expression of GFP fluorescence over time using a CCD camera.
  • Cells can also be labeled and detected in real time in the body.
  • the present invention will be further described by way of examples. However, the present invention is not limited to these examples.
  • RT-PCR was performed under the following conditions using RNA extracted from human fetal kidney cells 293 cells using a known method and the following specific primers (E1A-S, El A-AS). The bp E1A gene was amplified.
  • E1A-S 5'-aca ccg gga ctg aaa atg ag-3 '
  • DNA-PCR was performed from the DNA extracted from 293 cells using the following primers (E 1B-S, E 1B-AS) at 4 ° C for 5 min. To amplify the l, 822 bp E 1B gene .
  • the PCR solution composition and reaction conditions were the same as for the E1A gene.
  • E1B-S 5'-ctg acc tea tgg agg ctt gg-3 '(SEQ ID NO: 7)
  • E1B-AS 5 'gcc cac aca ttt cag tac ctc-3' (SEQ ID NO: 8) TA Cloning of each PCR product (TA Cloning Kit Dual Promoter;
  • DNA fragments of 911 bp (ElA) and 1836 bp (ElB) were cut out with the restriction enzyme EcoRI.
  • E1A was inserted into the Mul cleavage site of pIRES Vector I (CLONTECH), and E1B was inserted into the Sail site in the forward direction (E1A-IRES-E1B).
  • a 455 bp hTERT promoter sequence excised with restriction enzymes Mul and Bglll was inserted into the Xhol site upstream of E 1A of E 1A IRES-E 1B in the forward direction (phTERT-E 1A- IRES-E 1B)
  • CMV cytomegalovirus
  • a 4,38 lbp sequence was excised from pSh-hAIB using restriction enzymes I-Ceul and Pl-Scel and inserted into Adeno'X Viral DNA of Adeno-X Expression System (CLONTECH) ⁇ (AdenoX hAIB) AdenoX-hAIB After linearization by treatment with the restriction enzyme Pacl, 293 cells were transfected to produce an infectious recombinant adenovirus (OBP-301; Telomelysin; Telomelysin).
  • Figure la shows a schematic diagram of the duplicate cassette incorporated into telomerisin.
  • pEGFP-l (CLONTECH) was cleaved with Agel / Nhel, smoothed with a talenau fragment, and self-ligated (pEGFP-N2).
  • This pEGFP-N2 was cleaved with Nsil / Aflll, blunted with T4 DNA polymerase, and Bglll site was prepared using Bglll linker. This Bglll fragment was inserted into the BamHI site of pHMl l (pHMl l-EGFP-N2)
  • a 4381 bp sequence was excised from the prepared recombinant gene using restriction enzymes I-Ceul and PI-SceI and inserted into Adeno—X Viral DNA of Adeno—X Expression System (CLONTECH) (AdenoX—hAIB) AdenoX—hAIB After linearization by Pacl treatment, 293 cells were transfected to produce infectious recombinant adenovirus (hereinafter referred to as “OBP-401”) (Fig. Lb).
  • OBP-401 infectious recombinant adenovirus
  • CD431 (cisplatin) is administered to A431 / CDDP2 cells, human prostate cancer cells DU145 cells, and clone 2, 3, and 4 cells that have acquired resistance are administered paclitaxel, and how much antitumor resistance each cell has Confirmed whether to win.
  • cells are seeded at a density of 1,000 cells / well in a 96-well plate, and CDDP 0, 0.1, 1, 3, 10, 30, 50, ⁇ or paclitaxel 0, 0.1, 1, 2, 5, 10, 50, 100 ⁇ was added.
  • a kit Roche Diagnostics
  • the culture solution is removed from the well, and the reaction solution containing the sputum reagent is prepared and added and cultured. About 4 hours later, the absorbance was measured with a microtiter plate (ELISA) reader, the number of viable cells was calculated, and the antitumor effect was confirmed.
  • ELISA microtiter plate
  • the relative resistance of the parent strain was about 3-4 times higher in the A431 system and about 6-7 times higher in the DU145 system (Fig. 2).
  • A431 and CDDP resistance A431 strain the DU145 and paclitaxel-resistant DU145 strain was CDDP 10 mu Micromax respectively, paclitaxel ⁇ infected. The specimen was observed with a microscope on the third day after infection. As a result, a marked antitumor effect was observed in both parental strains, but resistance was confirmed in resistant strains (Fig. 3).
  • A431 / P is the parent strain
  • A431 / CDDP1 and A431 / CDDP2 are resistant strains.
  • Dul45 / P is the parent strain and clones 2-4 are resistant strains. '
  • A431 and CDDP resistant strains, DU145 and paclitaxel resistant strains were infected with CDDP and paclitaxel, respectively, and CBB staining was performed 7 days after infection. As a result, both the parent strains showed a marked antitumor effect, but the resistant strains were also resistant to high concentrations of anticancer drugs (Fig. 4).
  • MDR1 multidrug resistance gene
  • CAR Coxsaki virus-adenovirus receptor
  • A431 and CDDP resistant strains were infected with OBP-301 and OBP-401 at 10 MOI for 2 hours. 5 days after infection Morphological changes and fluorescence expression were observed with an fc fluorescence inverted microscope. As a result, in the bright field, although it was slightly resistant to A431 / P, a marked cell killing effect was observed in the resistant strain. In addition, when the fluorescence of OBP-401 was observed, strong fluorescence was observed in all cell lines, indicating that intracellular proliferation was sufficient (FIG. 10). '
  • DU145 and paclitaxel resistant strains were infected with OBP-301 and OBP-401 at 0.1 MOI for 2 hours, and observed with a microscope on the fifth day after infection.
  • a marked cell killing effect was observed in all parental and resistant strains.
  • fluorescence strong fluorescence was observed in all cell lines, indicating that intracellular proliferation was sufficient (Fig. 11).
  • ABP and 301 were infected with OBP-301 at 0.1 MOI or 1 MOI for 2 hours using PCR. Cells were collected at 2, 24, 48, and 72 hours after infection, DNA was extracted, and viral growth was quantitatively analyzed by real-time PCR.
  • an antitumor agent resistance overcome agent comprising a recombinant virus in which a polynucleotide comprising a human telomerase promoter, an E1A gene, an IRES sequence and an E1B gene in this order is incorporated.
  • the anti-tumor drug resistance overcomer of the present invention is useful in that it can be applied as a new therapeutic strategy for anti-tumor drug resistant tumors. Sequence listing free text

Landscapes

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

Abstract

La présente invention concerne un agent destiné à rompre la tolérance anti-tumorale. Ledit agent destiné à rompre la tolérance anti-tumorale est caractérisé par le fait qu'il contient un virus recombinant porteur d'un polynucléotide qui présente, dans l'ordre suivant, un promoteur de télomérase humain, un gène E1A, une séquence IRES et un gène E1B ayant été intégrés dans ledit polynucléotide.
PCT/JP2006/324316 2006-11-29 2006-11-29 Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale WO2008065726A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/JP2006/324316 WO2008065726A1 (fr) 2006-11-29 2006-11-29 Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/JP2006/324316 WO2008065726A1 (fr) 2006-11-29 2006-11-29 Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale

Publications (1)

Publication Number Publication Date
WO2008065726A1 true WO2008065726A1 (fr) 2008-06-05

Family

ID=39467530

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/324316 WO2008065726A1 (fr) 2006-11-29 2006-11-29 Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale

Country Status (1)

Country Link
WO (1) WO2008065726A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004005511A1 (fr) * 2002-07-08 2004-01-15 Kansai Technology Licensing Organization Co., Ltd. Croissance selective d'un virus entrainant une lyse tumorale dans des cellules tumorales
WO2006085689A1 (fr) * 2005-02-10 2006-08-17 Oncolys Biopharma, Inc. Agent anticancereux a associer a telomelysin
JP2007015997A (ja) * 2005-07-11 2007-01-25 Oncolys Biopharma Inc テロメライシン含有抗腫瘍剤耐性克服剤

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004005511A1 (fr) * 2002-07-08 2004-01-15 Kansai Technology Licensing Organization Co., Ltd. Croissance selective d'un virus entrainant une lyse tumorale dans des cellules tumorales
WO2006085689A1 (fr) * 2005-02-10 2006-08-17 Oncolys Biopharma, Inc. Agent anticancereux a associer a telomelysin
JP2007015997A (ja) * 2005-07-11 2007-01-25 Oncolys Biopharma Inc テロメライシン含有抗腫瘍剤耐性克服剤

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ABEI M. ET AL.: "Shokakigan no Idenshi Kagaku Ryoho no Tenbo: Gan Sentakuteki Zoshokugata Adenovirus o Mochiita Idenshi Chiryo ni yoru Gan Saibo no Koganzai Taisei no Kokufuku", JAPANESE JOURNAL OF GASTROENTEROLOGY, vol. 102, 2005, pages A486 + ABSTR. NO. SHO PD5-11, XP003022673 *
FUJIWARA T. ET AL.: "Konganzai Taisei Saibo ni Okeru Telomerase Tokuiteki Seigen Zoshokugata Adenovirus Telomelysin (OBP-301) no Koshuyo Koka no Kento", DAI 64 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 2005, pages 434 + ABSTR. NO. W-674, XP003022672 *

Similar Documents

Publication Publication Date Title
EP1795604B1 (fr) Virus recombinant contenant un gene de telomelysin-gfp
Sobhakumari et al. NOX4 mediates cytoprotective autophagy induced by the EGFR inhibitor erlotinib in head and neck cancer cells
CN108603196A (zh) Rna向导的对人类jc病毒和其他多瘤病毒的根除
EP2987858B1 (fr) Virus coxsackie génétiquement modifié
JP2013500339A (ja) 癌を治療するための送達ツールとしての脂肪由来間質細胞(asc)
JP2019533697A (ja) 5−ハロウラシル修飾マイクロrna及びがんの処置におけるその使用
AU2015279834A1 (en) Use of peptides that block metadherin-SND1 interaction as treatment for cancer
EP1553178B1 (fr) Virus oncolytique se propageant de manière sélective dans les cellules tumorales
CA3236236A1 (fr) Compositions et systemes pour l'edition cellulaire programmable par l'arn et leurs procedes de fabrication et d'utilisation
WO2013066485A2 (fr) Compositions et méthodes de traitement d'un cancer métastatique
Alipour et al. DNAi-peptide nanohybrid smart particles target BCL-2 oncogene and induce apoptosis in breast cancer cells
KR101835759B1 (ko) Phf 20 를 이용한 암세포에 대한 정보 제공 방법
KR102132198B1 (ko) Tert 프로모터 돌연변이에 특이적인 crispr 시스템 및 그의 이용
JP2007015997A (ja) テロメライシン含有抗腫瘍剤耐性克服剤
Qi et al. Twenty years of Gendicine® rAd-p53 cancer gene therapy: The first-in-class human cancer gene therapy in the era of personalized oncology
WO2008065726A1 (fr) Agent contenant du télomelysin destiné à rompre la tolérance anti-tumorale
WO2006085689A1 (fr) Agent anticancereux a associer a telomelysin
CN116670172A (zh) 表达免疫检查点抑制剂的癌症特异性反式剪接核酶及其用途
KR102133205B1 (ko) PNA-pHLIP 접합체를 유효성분으로 포함하는 암 예방 또는 치료용 약학적 조성물
KR102471898B1 (ko) 면역관문 억제제를 발현하는 암 특이적 트랜스-스플라이싱 리보자임 및 이의 용도
WO2015020215A1 (fr) Vecteur lentiviral thtd, agent de vieillissement, inhibiteur du cancer et composition médicinale contenant ledit thtd, protéine intégrée dans un particule creuse de type virus et procédé de production de particule creuse de type virus
US20230390320A1 (en) Cancer-specific trans-splicing ribozyme expressing immune checkpoint inhibitor, and use thereor
TW202128986A (zh) 表現出突變人類細胞色素p450 2b6蛋白的基因改造人類幹細胞及其在癌症治療上的應用
Zhang Antisense Therapy in Mouse Models of Histone H3. 3k27m Diffuse Midline Glioma Inhibits Tumor Growth, Promotes Neural and Glial Differentiation
JP2024504367A (ja) がんを治療及び改善するための方法

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: 06834071

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: 06834071

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP