WO2022250430A1 - Procédé de préparation d'arnm pour exprimer une protéine, et utilisation de l'arnm ainsi préparé - Google Patents

Procédé de préparation d'arnm pour exprimer une protéine, et utilisation de l'arnm ainsi préparé Download PDF

Info

Publication number
WO2022250430A1
WO2022250430A1 PCT/KR2022/007377 KR2022007377W WO2022250430A1 WO 2022250430 A1 WO2022250430 A1 WO 2022250430A1 KR 2022007377 W KR2022007377 W KR 2022007377W WO 2022250430 A1 WO2022250430 A1 WO 2022250430A1
Authority
WO
WIPO (PCT)
Prior art keywords
mrna
car
cells
nucleotide sequence
poly
Prior art date
Application number
PCT/KR2022/007377
Other languages
English (en)
Korean (ko)
Inventor
김태돈
강희영
Original Assignee
한국생명공학연구원
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 한국생명공학연구원 filed Critical 한국생명공학연구원
Publication of WO2022250430A1 publication Critical patent/WO2022250430A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464404Epidermal growth factor receptors [EGFR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46448Cancer antigens from embryonic or fetal origin
    • A61K39/464482Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • 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
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/50Vector systems having a special element relevant for transcription regulating RNA stability, not being an intron, e.g. poly A signal
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the present invention relates to a technology for improving mRNA stability and protein translation in a host cell with respect to mRNA for transformation for protein expression.
  • mRNA is an intermediate link between the translation of DNA encoding protein information and the protein production process by cytoplasmic ribosomes.
  • Optimal in vitro transcription mRNA preparations for therapeutic purposes are produced from linear DNA templates using T7, T3 or Sp6 phage RNA polymerase.
  • the RNA thus prepared must have an open reading frame (ORF) encoding the protein of interest, an untranslated region, a 5'-Cap and a poly (A) tail sequence.
  • ORF open reading frame
  • the engineered mRNA is similar to the fully mature natural mRNA molecule and is identical to that naturally present in the cytoplasm of eukaryotic cells, thereby avoiding the activation of the host cell's innate immune system.
  • poly(A) tail plays an important regulatory role in mRNA translation and stability
  • RNA transcripts are prepared from DNA templates and then poly(A) polymerase is used. Tailing, which extends the adenosine sequence at the 3' end of the mRNA, is essential.
  • the length of the poly (A) tail affects mRNA stability and protein expression efficiency, and the optimal poly (A) tail length may vary depending on the type of cell and the protein to be expressed.
  • TIL Tumor Infiltrating Lymphocytes
  • CAR Chimeric Antigen Receptor
  • TCR T-Cell Receptor
  • a transgenic CAR-NK cell Chimeric Antigen Receptor - Natural killer cell
  • DNA has a problem in that safety is not guaranteed because the CAR gene is integrated into the genome or mutated.
  • attempts have been made to use transiently transformed CAR-NK cells using mRNA, but in the case of transient transformation, mRNA instability, low transfection rate, and target protein expression rate act as obstacles, resulting in poor performance. No studies were shown. Accordingly, it is necessary to develop an mRNA construct capable of stably expressing CAR without being degraded in the cytoplasm of natural killer cells.
  • One object of the present invention is to provide an mRNA construct having a long half-life and a high expression rate of a target protein in the cytoplasm of a host cell and a method for preparing the same.
  • Another object of the present invention is to provide a transient transformant transformed with the mRNA construct and the transformant for use in the treatment of cancer.
  • one aspect of the present invention is a nucleotide sequence encoding a target protein or peptide; and a poly(A) sequence having a length of 20 nt to 500 nt linked downstream of the mRNA encoding the target protein or peptide.
  • Another aspect of the present invention provides a gene construct comprising a DNA sequence corresponding to or complementary to the nucleotide sequence of the mRNA construct, and a vector comprising the gene construct.
  • another aspect of the present invention provides a method for preparing mRNA with improved stability, comprising performing in vitro transcription using the vector as a template.
  • poly(A) tailing may not be performed in vitro.
  • another aspect of the present invention provides a transformant in which the mRNA construct is introduced into a host cell, and a cell therapy composition comprising the same as an active ingredient.
  • the present invention provides a cancer treatment method comprising administering the transformant to a subject.
  • the present invention provides the use of the transformant for the preparation of anticancer drugs.
  • the mRNA construct of the present invention is stable and can express a target protein that fully functions after being injected into a cell for a long time, it is expected to contribute to the active use of transient transformation technology.
  • transiently transfected CAR-NK cells prepared by selecting a Chimeric Antigen Receptor (CAR) as a target protein and applying it to the mRNA construct of the present invention have excellent cytotoxicity against cancer cells while stably expressing the CAR for a long time.
  • CAR Chimeric Antigen Receptor
  • FIG. 1 shows the structure of the CAR mRNA of the present invention and the domain and motif configuration of the CAR.
  • Figure 2 shows the results of optimizing transfection, showing the results of establishing transformation conditions for NK92 cells using the Nepa21 system.
  • Figure 3 shows the results of transfection optimization, showing the results of establishing transfection conditions for primary NK cells using the Nepa21 system.
  • Figure 4 shows the results of the confirmation of CAR expression over time in Transient CAR NK92 cells and Stable CAR NK92 cells.
  • Figure 5 shows the results of confirming the cancer cell killing ability over time for Transient CAR NK92 cells and Stable CAR NK92 cells.
  • Figure 6 shows the domains and motifs constituting the vector construction or CAR for the confirmation experiment on the expression rate and cancer cell killing ability of the chimeric antigen receptor including DAP10,
  • FIG. 7 shows the confirmation of the CAR expression rate according to the presence or absence of DAP10 during CAR mRNA transfection using FACS, in relation to FIG. 6 above.
  • Figure 11a is a schematic diagram of mRNA constructs designed differently with 5'- ⁇ -globin UTR, 3'-BGH, EMCV-IRES structure, forward/reverse direction or 5'/3' position of EMCV-IRES
  • Figure 11b is a schematic view of the mRNA construct manufacturing process.
  • FIG. 12 is a diagram comparing the reduction ratios of mRNA constructs 1 to 7 injected into cells over time.
  • FIG. 13 is a diagram confirming comparison of CAR protein expression levels in transient NK92 cells injected with mRNA constructs 1 to 7.
  • FIG. 14 is a view comparing and confirming the toxicity of transient NK92 cells injected with mRNA constructs 1 to 7 in lung cancer cell lines and breast cancer cell lines.
  • 15A is a view confirming, through electrophoresis, a transcript obtained from a vector containing a DNA sequence corresponding to or complementary to the nucleotide sequence of mRNA in which the order of BGH and IRES is different at the 3' end.
  • 15B is a view confirming the efficiency of target protein expression and the performance of the target protein in cells of the mRNA construct in which the order of BGH and IRES is different at the 3' end.
  • 16a and 16b are views confirming the efficiency of expression of the target protein and the performance of the target protein in cells of the mRNA construct according to the presence or absence of the poly(A) sequence at the 3' end.
  • One aspect of the present invention provides an mRNA construct that is structurally stable and capable of actively inducing the expression of a target protein having intact functions by being injected into a cell for a long time.
  • the mRNA construct of the present invention includes a nucleotide sequence encoding a target protein or peptide and a poly(A) nucleotide sequence.
  • the target protein or peptide is a target protein or peptide to be produced in a recombinant transformant, and refers to any protein or peptide that can be expressed using the transformant's own transcription or translation system.
  • the target protein or peptide includes hormones, hormone analogues, enzymes, enzyme inhibitors, receptors and receptor fragments, antigens and fragments or analogs of antigens, antibodies and antibody fragments, monoclonal antibodies, structural proteins, toxin proteins, etc., It may be a protein in which some or some or all of them are fused.
  • a chimeric antigen receptor (CAR) was used as the target protein or peptide.
  • the nucleotide sequence encoding the target protein or peptide may be an RNA sequence, and in particular, an mRNA sequence.
  • the poly(A) nucleotide sequence refers to a nucleotide sequence in which a plurality of adenosine monophosphates are linked through phosphodiester bonds.
  • the poly(A) sequence may have a length of 20 nt to 500 nt, for example, 30 nt to 450 nt, 50 nt to 400 nt, 100 nt to 350 nt, or 150 nt to 310 nt.
  • the poly(A) nucleotide sequence may be arranged downstream of the nucleotide sequence encoding the target protein or peptide.
  • the downstream means the next part of the 3'-end of the nucleotide sequence encoding the target protein or peptide. Therefore, the poly(A) nucleotide sequence may be placed after the 3'-end of the nucleotide sequence encoding the target protein or peptide.
  • the poly(A) nucleotide sequence may be connected immediately after the 3'-end of the nucleotide sequence encoding the target protein or peptide, and the 3'-end of the nucleotide sequence encoding the target protein or peptide and the An arbitrary base sequence may be interposed between the poly(A) base sequences.
  • the mRNA construct of the present invention may further include a 5'- ⁇ -globin untranslated region (UTR) region upstream of the nucleotide sequence encoding the target protein or peptide.
  • the upstream refers to the front part of the 5'-end of the nucleotide sequence encoding the target protein or peptide. Therefore, the 5'- ⁇ -globin UTR region may be placed in front of the 5'-end of the nucleotide sequence encoding the target protein or peptide.
  • the 5'-end of the nucleotide sequence encoding the target protein or peptide may be connected immediately after the 5'- ⁇ -globin UTR region, and the 5'- ⁇ -globin UTR region and the target protein or peptide Any nucleotide sequence may be interposed between the 5'-end of the nucleotide sequence encoding .
  • the 5'- ⁇ -globin UTR region is an untranslated portion present in the front part of the 5'-end of the nucleotide sequence of the gene encoding ⁇ -globin, that is, upstream of the gene encoding ⁇ -globin.
  • the 5'- ⁇ -globin UTR region may include or consist of the nucleotide sequence of SEQ ID NO: 1.
  • the mRNA construct of the present invention may further include BGH between the nucleotide sequence encoding the target protein or peptide and the poly(A) nucleotide sequence. Therefore, the BGH may be placed between the 3'-end of the nucleotide sequence encoding the target protein or peptide and the 5'-end of the poly(A) nucleotide sequence, and similarly, the target protein or peptide encoding the target protein or peptide Any base sequence may be interposed between the 3'-end of the base sequence and the BGH, and between the BGH and the 3'-end of the poly(A) base sequence.
  • the BGH refers to a polyadenylation signal of bovine growth hormone (bGH).
  • the BGH may include or consist of the nucleotide sequence of SEQ ID NO: 2.
  • the mRNA construct of the present invention may further include an IRES between the nucleotide sequence encoding the target protein or peptide and the poly(A) nucleotide sequence. Therefore, the IRES may be placed between the 3'-end of the nucleotide sequence encoding the target protein or peptide and the 5'-end of the poly(A) nucleotide sequence, and similarly, the target protein or peptide encoding the target protein or peptide Any base sequence may be interposed between the 3'-end of the base sequence and the IRES, and between the IRES and the 3'-end of the poly(A) base sequence.
  • the IRES is i) between the nucleotide sequence encoding the target protein or peptide and the BGH and ii) between the BGH and the poly(A) sequence.
  • the IRES is an internal ribosome entry site or a ribosome binding site, and forms a loop structure on mRNA to initiate translation of mRNA.
  • the IRES may be an IRES region of encephalomyocarditis virus (EMCV), and may specifically include or consist of the nucleotide sequence of SEQ ID NO: 3.
  • EMCV encephalomyocarditis virus
  • the mRNA construct of the present invention constructed as described above may be intended to be transformed into cells.
  • the transformation means introducing a foreign nucleotide sequence into a cell, and the cell into which the foreign gene is introduced by transformation is called a transformant.
  • the mRNA construct of the present invention may be transformed into cells as mRNA itself, and in this case, the mRNA itself is not in a form capable of independently replicating in cells, and is degraded in cells over time. It is extinguished.
  • the mRNA construct of the present invention may be for temporary transformation, or may be for maintaining the expression of the target protein or peptide by being stably maintained for a long time in the cytoplasm of the transformed cell.
  • an mRNA construct was designed in which poly(A) sequences of 40 nt, 150 nt and 310 nt lengths were linked downstream of the mRNA encoding the target protein, the chimeric antigen receptor. It was confirmed that the expression rate of the protein from the mRNA construct and the activity (cytotoxicity to cancer cells) by the protein were remarkably improved and sustained in the transient CAR-NK cells prepared by direct transformation of cells and primary NK cells.
  • Another aspect of the present invention provides a gene construct comprising a DNA sequence corresponding to or complementary to the nucleotide sequence of the mRNA construct of the present invention, and a vector comprising the gene construct.
  • the DNA nucleotide sequence corresponding to the nucleotide sequence of the mRNA construct means that only uracil (U) is changed to thymine (T) in the nucleotide sequence of the mRNA construct, and all other nucleotide sequences are the same DNA sequence. do.
  • the DNA nucleotide sequence complementary to the mRNA construct means a DNA sequence having an antisense nucleotide sequence to a DNA nucleotide sequence corresponding to the nucleotide sequence of the mRNA construct.
  • a gene construct comprising a DNA sequence corresponding to or complementary to the nucleotide sequence of the mRNA construct of the present invention may be used in a form included in a vector for storage or utilization.
  • the vector itself can be transformed into cells.
  • the gene construct is stably or permanently transformed, and the target protein or peptide can be expressed by transcribing and translating the target protein or peptide from the vector using the transcription and translation system of the transformant.
  • mRNA may be prepared using the vector as a template by in vitro transcription and transformed into cells.
  • the mRNA may be transiently transformed, and the target protein or peptide may be translated using the transformant's translation system to express the target protein or peptide.
  • the poly(A) nucleotide sequence is included downstream of the nucleotide sequence encoding the target protein or peptide, even when introduced into the cell, it exists very stably in the cytoplasm of the cell It is possible to express the target protein or peptide for a long time.
  • another aspect of the present invention provides a method for producing mRNA with improved stability, comprising performing in vitro transcription using the vector as a template.
  • the in vitro transcription refers to a process of synthesizing RNA in vitro using RNA polymerase, its cofactor, NTP substrate, and template DNA, and is well known to those skilled in the art to which the present invention pertains. Therefore, those skilled in the art to which the present invention pertains can perform the above process in an appropriate manner.
  • a nucleotide sequence corresponding to or complementary to the poly(A) nucleotide sequence is included in the gene construct included in the vector, only in vitro transcription as described above is performed. and poly(A) tailing may not be performed separately or additionally in vitro after in vitro transcription.
  • another aspect of the present invention provides a transformant in which an mRNA construct in which the target protein or peptide is a chimeric antigen receptor (CAR) in the mRNA construct of the present invention is introduced into a cell.
  • CAR chimeric antigen receptor
  • the cells may be immune cells, and the immune cells may be used without limitation as long as they are cells capable of inducing immunity to induce a desired therapeutic effect, and peripheral blood, umbilical cord blood, bone marrow, tumor-infiltrating lymphocytes, lymph node tissue or thymus tissue may be used. It can be obtained from, and can be obtained by differentiating from placental cells, embryonic stem cells, induced pluripotent stem cells or hematopoietic stem cells. In addition, the immune cells can be obtained from human, monkey, chimpanzee, dog, cat, mouse, rat and transgenic species thereof as well as established cell lines.
  • the method for obtaining immune cells can use any means known in the art, and can be obtained from autologous, allogeneic or heterologous sources.
  • autologous means any cell derived from the same individual that is subsequently reintroduced into the individual
  • allogeneic means any cell derived from another animal of the same species as the individual into which the cell is being introduced
  • xenogeneic means a cell derived from another species of animal.
  • the immune cells are natural killer cells (NK cells), T cells, natural killer T cells (NKT cells), cytokine induced killer cells (CIK), macrophages , It may be any one selected from dendritic cells, etc., but is not limited thereto. Therefore, immune cells expressing the chimeric antigen receptor according to the present invention on the cell surface include CAR-NK cells (Chimeric Antigen Receptor Natural Killer Cell), CAR-T cells (Chimeric Antigen Receptor T Cell), and CAR-NKT cells (Chimeric Antigen Receptor Cell). Receptor Natural killer T Cell), CAR-macrophage (Chimeric Antigen Receptor Macrophage), and the like.
  • the immune cells provided in the present invention can control the onset (on) / stop (off) of a conventional chimeric antigen receptor response to target cells, subsequent cell therapy, the need to increase or decrease the activity of therapeutic cells It includes a safety switch that can be very beneficial in situations. For example, when immune cells expressing a chimeric antigen receptor are provided to a patient, in some circumstances there may be side effects, such as off-target toxicity. Or, for example, the therapeutic cells may act to reduce the number of tumor cells or tumor size, and may no longer be needed. In this situation, the treatment cells can be controlled so that they are no longer activated.
  • the CAR-NK cells into which the chimeric antigen receptor was introduced into natural killer cells have problems due to the persistent toxicity of cancer immunotherapy when using conventional T cell-based CAR-T therapeutics, and autologous Not only can the risk of immune disease, the problem of graft-versus-host disease (GVHD) for xenogeneic cell transplantation, and the problem of off-target toxicity be solved through the on/off switch, but also various cancer cells It has the advantage of being able to be used as a general-purpose therapeutic agent by allowing it to be targeted.
  • GVHD graft-versus-host disease
  • the transformant of the present invention can be used as an active ingredient in a pharmaceutical composition for preventing or treating cancer, and another aspect of the present invention is for preventing or treating cancer comprising the transformant as an active ingredient.
  • a pharmaceutical composition is provided.
  • cancer is used synonymously with “tumor” and refers to or refers to a physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
  • Cancer or carcinoma that can be treated with the pharmaceutical composition of the present invention is not particularly limited, and includes both solid cancer and hematological cancer.
  • it may be a blood cancer such as the leukemia or lymphoma, specifically thymic carcinoma, non-Hodgkin lymphoma, diffuse large cell lymphoma, small lymphocytic cell lymphoma ( small lymphocytic lymphoma, T-cell neoplasma, peripheral T-cell lymphoma, mantle cell lymphoma, T-cell acute lymphocytic leukemia lymphobalstic lymphoma), chronic lymphoblastic leukemia, and the like.
  • the leukemia or lymphoma specifically thymic carcinoma
  • non-Hodgkin lymphoma diffuse large cell lymphoma
  • small lymphocytic cell lymphoma small lymphocytic lymphoma, T-cell neoplasma, peripheral T-cell lymphoma, mantle cell lymphoma, T
  • the pharmaceutical composition of the present invention may include 1 to 10 times, 2 to 10 times, or 5 to 8 times the number of immune cells compared to the number of tumor cells of the subject to be treated, but is limited thereto. it is not going to be
  • Anticancer agents used in chemotherapy induce apoptosis of proliferating cells, and radiation to enhance the therapeutic effect of anticancer agents increases such apoptosis.
  • cells whose apoptosis is induced by anticancer agents and radiation are not limited to cancer cells, and may also affect immune cell therapy agents administered to individuals for immunotherapy.
  • Chemotherapy includes, but is not limited to, CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone), EPOCH (etoposide, vincristine, doxorubicin, cyclophosphamide, prednisone) or any other multi-drug regimen.
  • cell therapy product refers to cells and tissues manufactured through isolation, culture, and special manipulation from a subject, and is a drug (US FDA regulation) used for the purpose of treatment, diagnosis, and prevention, and is used to restore the function of cells or tissues. It refers to drugs used for the purpose of treatment, diagnosis, and prevention through a series of actions such as proliferation and selection of living autologous, allogeneic, or heterogeneous cells in vitro or changing the biological characteristics of cells in other ways.
  • the pharmaceutical composition of the present invention may additionally include a pharmaceutical or pharmaceutically acceptable carrier.
  • a pharmaceutical or pharmaceutically acceptable carrier is that it does not inhibit the activity of the active ingredient and does not have toxicity more than is adaptable to the subject of application (prescription), and the 'carrier' facilitates the addition of the compound into cells or tissues. defined as a compound that
  • compositions of the present invention may be formulated in various forms for administration to a subject, and a representative formulation for parenteral administration is an injectable formulation, preferably an isotonic aqueous solution or suspension.
  • Formulations for injection may be prepared according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. For example, it may be formulated for injection by dissolving each component in saline or a buffer solution.
  • dosage forms for oral administration include, for example, ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, and wafers.
  • the tablet may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and optionally starch, agar, alginic acid or Disintegrants such as sodium salts, absorbents, colorants, flavors and/or sweeteners may further be included.
  • the formulation may be prepared by conventional mixing, granulating or coating methods.
  • composition of the present invention may further include adjuvants such as preservatives, hydrating agents, emulsification accelerators, salts or buffers for osmotic pressure control, and other therapeutically useful substances, and may be formulated according to conventional methods. .
  • the pharmaceutical composition according to the present invention can be administered through various routes including oral, transdermal, subcutaneous, intravenous or intramuscular, and the dosage of the active ingredient depends on the route of administration, age, sex, weight and severity of the patient. It can be appropriately selected according to several factors.
  • the composition of the present invention can be administered in parallel with a known compound capable of enhancing the desired effect.
  • the route of administration of the pharmaceutical composition according to the present invention may be administered to humans and animals orally or parenterally, such as intravenously, subcutaneously, intranasally or intraperitoneally.
  • the total effective amount of the transformant according to the present invention can be administered to the patient in a single dose, and a split treatment method in which multiple doses are administered for a long period of time ( fractionated treatment protocol).
  • the pharmaceutical composition of the present invention may vary the content of the active ingredient depending on the severity of the disease, but is typically administered repeatedly several times a day at an effective dose of 100 ⁇ g to 3,000 mg per administration based on adults. .
  • the concentration of the transformant can be determined by considering various factors such as the patient's age, weight, health condition, sex, disease severity, diet and excretion rate, as well as the drug administration route and number of treatments.
  • composition of the present invention may further include a known anticancer agent in addition to the above-described transformant as an active ingredient, and may be used in combination with other known therapies for the treatment of these diseases.
  • another aspect of the present invention provides a method for preventing or treating cancer, comprising administering the transformant of the present invention to a subject in need of treatment.
  • the transformant of the present invention may be in the form of the above-described pharmaceutical composition, and a person skilled in the art to which the present invention belongs can determine an appropriate administration route and dosage of the pharmaceutical composition.
  • Another aspect of the present invention provides the use of the transformant for the preparation of a drug for preventing or treating cancer.
  • a chimeric antigen receptor (CAR) configured as shown in FIG. 1 was designed as a target protein to be applied to the mRNA construct of the present invention and temporarily expressed in cells.
  • the chimeric antigen receptor includes an ectodomain that recognizes a target, a spacer (Myc-Hinge) that provides connection and flexibility of the ectodomain, a transmembrane domain that penetrates cell membranes, and It is constructed by sequentially connecting endodomains that induce cell signaling.
  • the ScFv of an anti-CEACAM6 antibody recognizing CEACAM6 as an antigen with the ectodomain the ScFv of an anti-cotinine antibody recognizing cotinine as an antigen, or an anti-EGFR affibody recognizing EGFR as an antigen
  • CD28 and DAP10 as co-activators along with CD3 ⁇ responsible for cytotoxic signaling as an endodomain
  • a chimeric antigen receptor having an anti-CEACAM6 scFv Anti-CEACAM6-CAR
  • a chimeric antigen receptor (anti-cotinine-CAR) having an anti-cotinine scFv and a chimeric antigen receptor (ZEGFR-CAR) having an anti-EGFR Affibody were respectively designed.
  • the nucleotide sequence of the gene construct encoding it was inserted into the pBluescript SK(-) vector (Addgene) and the lenti-virus vector (
  • GFP mRNA was injected into NK92 cells in an amount of 2 ⁇ g per 1 ⁇ 10 6 cells while varying the voltage from 110 V to 200 V using the Nepa21 system. After transfection, expression of GFP in NK92 cells was examined.
  • the survival rate of NK92 cells gradually decreased as the voltage increased, but it was confirmed that GFP was expressed in more than 90% of the surviving NK92 cells at a voltage of 150V or less, and in 110V conditions It was confirmed to exhibit the best cell viability and GFP expression rate.
  • the expression rate was increased, and GFP expression was observed until 3 days after transfection. Based on this, 20 ⁇ g of mRNA per 1X10 7 of NK92 cells was transfected at a voltage of 110 V to scale-up, and it was confirmed that the expression rate and persistence of expression were excellently maintained even in this case.
  • GFP mRNA was transfected in an amount of 5 ⁇ g per cell 1X10 6 while changing the voltage in the same manner as in the NK92 cells, and then the expression of GFP in primary NK cells was examined.
  • the mRNAs of the anti-CEACAM6-CAR and anti-cotinine-CAR obtained as above were transfected into NK92 cells with a voltage of 110 V using the Nepa 21 system used in Example 2 to obtain transient CAR-NK cells.
  • the CAR expression pattern expression rate and expression persistence
  • FIG. 4 In the case of anti-CEACAM6-CAR, a high expression rate of 90% or more was maintained until 48 hours after mRNA injection, and the expression continued for 4 days, and in the case of anti-cotinine-CAR, an expression rate of 70% or more was shown until 16 hours after mRNA injection.
  • the cytotoxicity (or cell killing ability, cytolytic activity) of the transient CAR-NK cells prepared as described above was confirmed by Calcein AM assay. Specifically, after reacting by treating AU565 cells with Calcein-AM (life technologies; C1430) at a concentration of 5ug/ml (37°C, 5% CO2, 1 hour in the dark), AU565 cells stained with Calcein The transient CAR-NK cells were treated at a ratio of 10:1, 5:1, and 1:1 (natural killer cells:cancer cells), respectively, in 200ul of RPMI1640 (10% FBS) for 5.5 hours, 13 hours, 25 hours, After reacting for 50 hours and 82 hours (37 ° C, 5% CO 2 ), 100 ul of the supernatant was taken to determine the amount of calcein present in the supernatant, and the cytotoxicity according to each condition was calculated in the following manner. At this time, the anti-cotinine-CAR-expressing trans
  • Cytotoxicity (%) (Calcein release value-spontaneous value according to conditions) / (maximum value-spontaneous value) x 100
  • each lenti-viral vector into which the nucleotide sequences of the gene constructs encoding the two target proteins, anti-CEACAM6-CAR and anti-cotinine-CAR, designed in Example 1 were inserted were converted into viral packaging vectors (viral packaging vectors).
  • viral packaging vectors viral packaging vectors.
  • vector; PMDLg/RRE, RSV/REV, VSVG were transfected into HEK293T cells, and anti-CEACAM6-CAR and anti-cotinine-CAR-expressing lentiviruses were obtained therefrom.
  • the amount of the Myc epitope of CD5-CAR was confirmed by flow cytometry to determine the infection unit was calculated. Calculate the number of natural killer cells and the amount of lentivirus so that the multiplicity of infection (MOI) is 30, and spinoculate lentiviruses expressing anti-CEACAM6-CAR and anti-cotinine-CAR in NK92 cells, respectively (360g, 90min, RT) to construct stable CAR-NK cells.
  • MOI multiplicity of infection
  • the CAR expression pattern (expression rate and expression persistence) and cytotoxicity over time were confirmed in the same manner as in the transient CAR-NK cells.
  • the ZEGFR- Based on CAR, ⁇ Ecto-TM-10z, a modified ZEGFR-CAR without an ectodomain, and three types of modified ZEGFR-CARs were designed, and the nucleotide sequences of gene constructs encoding them were inserted into a lenti-viral vector (Clontech, 632155).
  • the mRNAs of 5 CARs were obtained.
  • the mRNAs were transfected into NK92 cells with a voltage of 110V using the Nepa 21 system used in Example 2 to prepare transient CAR-NK cells.
  • CAR expression and cytotoxicity of the transient CAR-NK cells were measured in the same manner as in Example 3 for the 5 types of transient CAR-NK cells prepared as described above.
  • a modified ZEGFR-CAR (ZEGFR-MH-TM-10z-B) in which the ITAM motif of CD28 was removed was designed, respectively, and obtained by the method described above.
  • the mRNA of the modified ZEGFR-CAR was transformed into NK92 cells as described above to prepare transient CAR-NK cells, and CAR expression and cytotoxicity of the transient CAR-NK cells were measured as described above. measured.
  • Example 1 in the case of the modified ZEGFR-CAR (ZEGFR-MH-TM-10z-B) in which the ITAM motif of CD28 is removed, the ZEGFR-CAR designed in Example 1 (in FIG. 8) Example containing the ITAM motif of CD28 as well as the cytotoxicity of transient CAR-NK cells against the breast cancer cell line AU565, as well as the lowered CAR expression rate compared to 'ZEGFR-MH-TM-28-10z-B') It was confirmed that it fell short of the ZEGFR-CAR designed in 1.
  • the mRNA of the modified ZEGFR-CARs was transformed into NK92 cells as described above to prepare transient CAR-NK cells, and CAR expression and cytotoxicity of the transient CAR-NK cells were measured as described above. .
  • FIG. 11a is a schematic diagram of the constructs arranged next to the T7 promoter in the template vector for synthesizing the 7 types of mRNA constructs listed in Table 1 below, and Table 2 below is introduced into the 5'-UTR and 3'-UTR of ZEGFR-CAR. It shows the DNA sequence of one component.
  • the base sequences of the gene constructs encoding the 7 types of mRNA constructs designed as described above were inserted into the pBluescript SK(-) vector (Addgene), and in vitro transcription and mRNA constructs 1 to 7 were prepared by poly(A) tailing.
  • 5'-Cap and 3'-Poly (A) tails are bound to the 7 types of mRNAs obtained as described above.
  • Example [5-1] The 7 types of mRNAs obtained in Example [5-1] were transfected into NK92 cells with a voltage of 110 V using the Nepa 21 system used in Example 2 to prepare transient CAR-NK cells. .
  • the amount of mRNA remaining in the cytoplasm after being injected into NK92 cells over time was confirmed by RT-PCR.
  • Example [5-2] Furthermore, for the 7 types of transient CAR-NK cells prepared in Example [5-2], the expression level of the CAR protein expressed at 8 hours after the mRNA was injected into NK92 cells was measured. As criterion (1), the translational stability of the mRNA construct over time was evaluated.
  • transient CAR-NK cells prepared in Example [5-2] were injected into NK92 cells, and 18 hours later, the same method as in Example 3 was performed.
  • the transient CAR-NK cells were treated with breast cancer cell line AU565 and lung cancer cell line A549, respectively, and cytotoxicity to cancer cells was evaluated.
  • the transient CAR-NK cells had a level and pattern corresponding to the protein expression rate confirmed in Example 5-3.
  • the cytotoxicity of was confirmed, and the secretion of IFN ⁇ and TNF ⁇ , which are a series of reactions occurring during the cell lysis process, was also confirmed to appear at the level and pattern corresponding to the protein expression rate confirmed in Example 5-3.
  • Example 1 In order to confirm the effect of the IRES and BGH sequences located in the 3'-UTR of the mRNA structure on the structural stability, translational stability and cytotoxicity of mRNA, the anti-cotinine-CAR designed in Example 1 was targeted. 5'-UTR and 3'-UTR were designed as shown in Table 3 below, and mRNA was obtained in the same manner as in Example [5-1], followed by electrophoresis. The mRNA obtained as described above has a 5'-Cap and a 3'-Poly(A) tail bound thereto, similarly to the mRNA obtained in Example [5-1].
  • the IRES is included during mRNA synthesis during in vitro transcription. Two bands of the transcript and the IRES-deleted transcript were identified, whereas in the case of the 'UTR-CAR-IRES-BGH' construct in which the IRES is located in front of the BGH, a single band of the transcript containing the IRES was identified. only confirmed
  • the mRNA obtained as described above was transformed into NK92 cells in the same manner as in Examples [5-2] to [5-4], and structural stability, translational stability, and cytotoxicity of the mRNA construct were evaluated.
  • structures No. 2, 3, and 4 in Table 4 are designed so that poly(A) tails exist in the structures themselves, so poly(A) tailing does not proceed separately after in vitro transfer, and structure No. 5 in Table 4 is Although the poly(A) tail does not exist in the structure itself, the poly(A) tail is attached by additional poly(A) tailing after in vitro transcription.
  • the 5 types of mRNs designed and obtained as described above were transformed into NK92 cells and primary NK cells under optimal conditions in the same manner as in Example 2 above to construct transient CAR-NK cells.
  • the protein expression level of CAR and its cytotoxicity to cancer cells were confirmed.
  • construct No. 2 in the case of constructs No. 2 to 4 having a poly(A) tail in the construct itself, in NK92 cells, construct No. 2 is more effective in protein expression or breast cancer cell line AU65 than construct No. 1 without the poly(A) tail. Although the cytotoxicity was high, it was confirmed that the protein expression or cytotoxicity to the breast cancer cell line AU565 was lower than that of constructs 3 and 4, respectively. In addition, constructs 3 and 4 of Nos. 3 and 4 were confirmed to stably express the protein and exhibit cytotoxicity at a level or aspect corresponding to the expression rate (FIG. 16a).
  • construct 5 showed the highest level of protein expression, but the cytotoxicity against breast cancer cell line AU565 was found to be highest in construct 3 containing 150 bp of adenosine (A) (FIG. 16b ).

Abstract

La présente invention concerne une structure d'ARNm comprenant une séquence d'acide nucléique, codant pour une protéine ou un peptide cible, et une séquence d'acide nucléique poly A, ayant une longueur de 20 nt à 500 nt et connectée en aval de la séquence d'acide nucléique codant pour une protéine ou un peptide cible. On s'attend à ce que la structure de l'ARNm soit structurellement stable dans le cytoplasme après avoir été introduite dans les cellules et permette à une protéine cible remplissant pleinement ses fonctions d'être exprimée pendant une longue période, et contribue ainsi à une utilisation robuste d'une technologie de transformation transitoire.
PCT/KR2022/007377 2021-05-25 2022-05-24 Procédé de préparation d'arnm pour exprimer une protéine, et utilisation de l'arnm ainsi préparé WO2022250430A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20210066744 2021-05-25
KR10-2021-0066744 2021-05-25

Publications (1)

Publication Number Publication Date
WO2022250430A1 true WO2022250430A1 (fr) 2022-12-01

Family

ID=84229356

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/007377 WO2022250430A1 (fr) 2021-05-25 2022-05-24 Procédé de préparation d'arnm pour exprimer une protéine, et utilisation de l'arnm ainsi préparé

Country Status (2)

Country Link
KR (1) KR20220159909A (fr)
WO (1) WO2022250430A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807707A (en) * 1995-02-10 1998-09-15 Mcmaster University High efficiency translation of mRNA molecules
US20070298417A1 (en) * 2001-03-09 2007-12-27 John Daly Constructs for gene expression analysis
WO2016197121A1 (fr) * 2015-06-05 2016-12-08 Dana-Farber Cancer Institute, Inc. Compositions et procédés pour thérapie génique transitoire à stabilité améliorée
CN109097396A (zh) * 2018-09-10 2018-12-28 上海细胞治疗工程技术研究中心集团有限公司 一种制备靶向间皮素的car-t细胞的方法
KR20210065065A (ko) * 2019-11-26 2021-06-03 한국생명공학연구원 단백질 발현용 mRNA 구조체 및 이의 용도

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807707A (en) * 1995-02-10 1998-09-15 Mcmaster University High efficiency translation of mRNA molecules
US20070298417A1 (en) * 2001-03-09 2007-12-27 John Daly Constructs for gene expression analysis
WO2016197121A1 (fr) * 2015-06-05 2016-12-08 Dana-Farber Cancer Institute, Inc. Compositions et procédés pour thérapie génique transitoire à stabilité améliorée
CN109097396A (zh) * 2018-09-10 2018-12-28 上海细胞治疗工程技术研究中心集团有限公司 一种制备靶向间皮素的car-t细胞的方法
KR20210065065A (ko) * 2019-11-26 2021-06-03 한국생명공학연구원 단백질 발현용 mRNA 구조체 및 이의 용도

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HU, Y. ET AL.: "Chimeric antigen receptor (CAR)-transduced natural killer cells in tumor immunotherapy", ACTA PHARMACOLOGICA SINICA, vol. 39, 2018, pages 167 - 176, XP055625054, DOI: 10.1038/aps.2017.125 *

Also Published As

Publication number Publication date
KR20220159909A (ko) 2022-12-05

Similar Documents

Publication Publication Date Title
EP4166655A1 (fr) Cellule immunitaire modifiée et son utilisation
US20180327470A1 (en) Chimeric antigen receptor-modified immune effector cell carrying pd-l1 blocking agent
AU601675B2 (en) Purification, production and use of tumor necrosis factors
WO2020038490A1 (fr) Agent thérapeutique comprenant un acide nucléique et des cellules immunitaires modifiées pour exprimer un car, et application de celui-ci
WO2019080537A1 (fr) Agent thérapeutique comprenant un virus oncolytique et des cellules tueuses naturelles-récepteur antigénique chimérique, utilisation, kit, et méthode de traitement d'une tumeur et/ou d'un cancer
WO2017135800A1 (fr) Cellule souche mésenchymateuse exprimant trail et cd, et son utilisation
SK74793A3 (en) Pharmaceutical agent for treatment of tumors and its using
WO2018106068A1 (fr) Composition anticancéreuse comprenant un adénovirus recombiné exprimant un facteur de dégradation pour matrice extracellulaire
US20220290176A1 (en) mRNA CONSTRUCT FOR PROTEIN EXPRESSION AND USE OF SAME
WO2013027988A2 (fr) Adénovirus recombiné comprenant un ribozyme présentant une activité de trans-épissage et un gène destiné à traiter le cancer, et son utilisation
BR112020011898A2 (pt) receptores daric nkg2d
EP3831945A1 (fr) Hétérodimère protéique et son utilisation
CN117295515A (zh) 用于癌症免疫治疗的嵌合抗原受体修饰的粒细胞-巨噬细胞祖细胞
WO2022250430A1 (fr) Procédé de préparation d'arnm pour exprimer une protéine, et utilisation de l'arnm ainsi préparé
WO2013180473A1 (fr) Vecteur d'expression bicistronique pour l'expression d'un anticorps et procédé de production d'un anticorps l'utilisant
WO2024053933A1 (fr) Construction comprenant une séquence utr améliorant la stabilité intracellulaire et la biosynthèse d'arnm et son utilisation
IL292004A (en) Composition for reprogramming cells into dendritic plasma cells or type I interferon-producing cells, methods and uses thereof
US20230279352A1 (en) Methods for generating primary immune cells
CN111499766B (zh) 针对慢性淋巴细胞白血病的免疫效应细胞、其制备方法和应用
WO2021232050A1 (fr) Immunothérapie basée sur des cellules tueuses naturelles pour le traitement ou la prévention d'une infection par sars-cov-2
KR100869861B1 (ko) 고양이 cd80, 고양이 cd86, 고양이 cd28, 및 고양이ctla4 핵산과 폴리펩티드
CN109554349B (zh) Pd-1基因表达沉默的工程化免疫细胞
TWI830771B (zh) 包含核酸及car修飾的免疫細胞的治療劑及其應用
WO2024071544A1 (fr) Nouveau récepteur fc recombinant et cellules le comprenant
WO2023101277A1 (fr) Cellules immunitaires à efficacité améliorée

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: 22811623

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE