WO2021235697A1 - Cd22-specific antibody and use thereof - Google Patents

Cd22-specific antibody and use thereof Download PDF

Info

Publication number
WO2021235697A1
WO2021235697A1 PCT/KR2021/004566 KR2021004566W WO2021235697A1 WO 2021235697 A1 WO2021235697 A1 WO 2021235697A1 KR 2021004566 W KR2021004566 W KR 2021004566W WO 2021235697 A1 WO2021235697 A1 WO 2021235697A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
antibody
cells
region represented
Prior art date
Application number
PCT/KR2021/004566
Other languages
French (fr)
Korean (ko)
Inventor
김기태
김승구
이병은
Original Assignee
주식회사 이노베이션바이오
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 주식회사 이노베이션바이오 filed Critical 주식회사 이노베이션바이오
Publication of WO2021235697A1 publication Critical patent/WO2021235697A1/en

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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464413CD22, BL-CAM, siglec-2 or sialic acid binding Ig-related lectin 2
    • 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
    • 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
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/563Immunoassay; Biospecific binding assay; Materials therefor involving antibody fragments
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • the present invention relates to an antibody specific for CD22 and uses thereof, and more particularly, to an antibody that specifically binds to CD22, a chimeric antigen receptor comprising the antibody, a CAR-T cell expressing the chimeric antigen receptor, and It relates to a pharmaceutical composition for preventing or treating diseases mediated by cells expressing CD22, including them.
  • CD22 is a 135 kDa membrane glycoprotein belonging to a family of sialic acid-binding proteins called sialoadhesins. CD22 is detected in the cytoplasm at the beginning of B cell development, appears on the cell surface simultaneously with IgD, and is found in most mature B cells. When B cells are activated, their expression is increased. It has been reported that CD22 is lost at the end of differentiation and is generally absent in plasma cells.
  • CD22 ⁇ immunoglobulin-like domains in the extracellular region.
  • the CD22 ⁇ mutant lacks Ig-like domain 4 and may have a truncated cytoplasmic domain. Antibodies that block the binding of CD22 to monocytes, neutrophils, lymphocytes and erythrocytes have been observed to bind within the first or second Ig-like domain.
  • CD22 In the cytoplasmic domain of CD22, tyrosine is phosphorylated upon binding to the B-cell antigen receptor and binds to Lyk, Syk and phosphatidylinositol 3-kinase.
  • the function of CD22 is to down-modulate the B cell activation threshold. It can also mediate cell binding through binding to cells containing appropriate sialoglycoconjugates.
  • CD22 includes NHL, acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (B-CLL) and especially acute non-lymphocytic leukemia (ANLL), It is expressed in most B-cell leukemias and lymphomas.
  • B-ALL acute lymphoblastic leukemia
  • B-CLL chronic lymphocytic leukemia
  • ANLL acute non-lymphocytic leukemia
  • an antibody that binds to CD22 was screened to establish two novel antibodies (3F11 and 7B5), and the two types of antibodies selected in the present invention are CD22 antigen It was confirmed that it specifically binds to In addition, using the CD22-specific antibody of the present invention, a chimeric antigen receptor targeting CD22 and CAR-T cells were prepared. ) was confirmed to effectively kill.
  • Another object of the present invention is to provide a polynucleotide encoding the antibody, a vector expressing the antibody, and a recombinant cell transformed with the vector.
  • Another object of the present invention is a chimeric antigen receptor comprising the antibody, a polynucleotide encoding the chimeric antigen receptor targeting CD22, a vector comprising the same, and an immune effector expressing the polynucleotide or the chimeric antigen receptor comprising the vector to provide cells.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating a disease mediated by CD22-expressing cells, including immune effector cells expressing the antibody or chimeric antigen receptor targeting CD22.
  • Another object of the present invention is to provide a composition for diagnosing or monitoring a disease mediated by a cell expressing CD22 comprising the antibody.
  • the present invention relates to (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3, and the amino acid of SEQ ID NO: 4 a light chain variable region comprising a CDR1 region represented by , a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid sequence represented by SEQ ID NO: 6; or
  • a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14
  • An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15 and a CDR3 region represented by the amino acid of SEQ ID NO: 16 is provided.
  • the antibody may be a monoclonal antibody, preferably a single-chain variable fragment (scFv).
  • scFv single-chain variable fragment
  • the antibody (a) has a heavy chain variable region represented by the amino acid of SEQ ID NO: 7 and a light chain variable region represented by the amino acid of SEQ ID NO: 8,
  • the antibody (b) may be composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 17 and a light chain variable region represented by the amino acid of SEQ ID NO: 18.
  • the present invention provides a polynucleotide encoding the antibody that specifically binds to CD22.
  • the present invention provides a vector comprising a polynucleotide encoding the antibody that specifically binds to CD22.
  • the present invention provides a recombinant cell that produces an antibody or fragment thereof that specifically binds to CD22 transformed with the vector.
  • the present invention provides a CD22-binding domain; transmembrane domain; costimulatory domain; and a chimeric antigen receptor (CAR) comprising an intracellular signal transduction domain,
  • the CD22-binding domain may be selected from an antibody or fragment thereof capable of specifically binding to CD22 of the present invention.
  • the transmembrane domain may be derived from a protein selected from the group consisting of CD8 ⁇ , CD4, CD28, CD137, CD80, CD86, CD152 and PD1.
  • the costimulatory domain may be derived from a protein selected from the group consisting of CD28, 4-1BB, OX-40 and ICOS, and the signaling domain may be derived from CD3 ⁇ .
  • it may further include a hinge region located between the C-terminus of the CD22-binding domain and the N-terminus of the transmembrane domain, wherein the hinge region is CD8 ⁇ may be of origin.
  • the present invention provides a polynucleotide encoding the chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the present invention also provides a vector comprising a polynucleotide encoding a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the vector may be a plasmid, a retroviral vector or a lentiviral vector.
  • the present invention provides an immune effector cell comprising the polynucleotide encoding the chimeric antigen receptor (CAR) or a polynucleotide encoding the chimeric antigen receptor (CAR), and expressing the chimeric antigen receptor (CAR). do.
  • the immune effector cells may be T cells.
  • the present invention provides an immune effector cell expressing a chimeric antigen receptor targeting CD22; Or an antibody or fragment thereof that specifically binds to CD22; provides a pharmaceutical composition for preventing or treating a disease mediated by cells expressing CD22, including.
  • the disease mediated by cells expressing CD22 is lymphoma, non-Hogkins lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed delayed NHL, refractory sexual NHL, refractory delayed NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL) ), Burkitt's lymphoma, and mantle cell lymphoma.
  • NHL non-Hogkins lymphoma
  • aggressive NHL relapsed aggressive NHL
  • relapsed delayed NHL refractory Sexual NHL
  • CLL chronic lymphocytic leukemia
  • small lymphocytic lymphoma small lymphocytic lymphoma
  • leukemia hairy cell leukemia
  • HCL hairy cell leukemia
  • ALL acute lymphocytic leukemia
  • Burkitt's lymphoma and mantle cell lympho
  • the composition may include a therapeutic agent for a disease mediated by cells expressing CD22.
  • the present invention provides a composition for diagnosing or monitoring a disease mediated by cells expressing CD22, including an antibody that specifically binds to CD22.
  • two novel antibodies (3F11 and 7B5) were established by screening antibodies that more specifically bind to CD22, and it was confirmed that the novel antibodies specifically bind to the CD22 antigen.
  • the CAR-T cells of the present invention effectively kill CD22-expressing cells, the CD22-specific antibody, the CD22-targeting chimeric antigen receptor, and CAR-T cells of the present invention are related to CD22 expression. It can be usefully used as a composition for preventing or treating diseases.
  • 1 is data confirming the binding ability of 3F11 and 7B5 antibodies selected in the present invention to CD22 by FACS.
  • FIG. 2 is a schematic diagram showing a lentiviral vector expressing a chimeric antigen receptor (CD22-CAR) targeting CD22 and a chimeric antigen receptor expressed in T cells.
  • CD22-CAR chimeric antigen receptor
  • FIG. 3 is a schematic diagram showing a method for preparing CD22-CAR-T cells using a lentivirus expressing CD22-CAR.
  • the present invention provides an antibody or fragment thereof that specifically binds to CD22,
  • a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and the amino acid of SEQ ID NO: 4 an antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 6; or
  • a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 It relates to an antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 16.
  • the antibody may be a monoclonal antibody.
  • the term "monoclonal antibody” is also called a monoclonal antibody or monoclonal antibody, and is an antibody produced by a single antibody-forming cell, and has a uniform primary structure (amino acid sequence). It recognizes only one antigenic determinant and is generally produced by culturing a hybridoma cell in which cancer cells and antibody-producing cells are fused. can also be produced.
  • the antibody may be used by humanizing the remaining parts except for the CDR parts, if necessary.
  • CDR complementarity determining region
  • humanized antibody is an antibody that possesses an amino acid sequence corresponding to that of an antibody produced by a human and/or is made using one of the techniques for making human antibodies as disclosed herein. This definition of a humanized antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • antibody can be used not only in a complete form having two full-length light chains and two full-length heavy chains, but also as a fragment of an antibody molecule.
  • a fragment of an antibody molecule refers to a fragment having at least a peptide tag (epitope) binding function, and includes scFv, Fab, F(ab'), F(ab') 2 , a single domain, and the like.
  • Fab has a structure having variable regions of light and heavy chains, a constant region of a light chain and a first constant region (CH1) of a heavy chain, and has one antigen-binding site.
  • Fab' differs from Fab in that it has a hinge region comprising one or more cysteine residues at the C terminus of the heavy chain CH1 domain.
  • the F(ab')2 antibody is generated by forming a disulfide bond between the cysteine residues of the hinge region of Fab', and Fv is the smallest antibody fragment having only a heavy chain variable region and a light chain variable region.
  • dsFv double chain Fv
  • scFv single chain Fv
  • the heavy chain variable region and the light chain variable region are covalently linked via a peptide linker.
  • antibody fragments can be obtained using proteolytic enzymes (for example, by restriction digestion of the entire antibody with papain to obtain Fab, and by digestion with pepsin to obtain F(ab')2 fragments), preferably It can be produced through genetic recombination technology.
  • the monoclonal antibody that specifically binds to CD22 of the present invention can be prepared by using all or part of the CD22 protein as an immunogen (or antigen). More specifically, first, as an immunogen, CD22, a fusion protein containing CD22 protein, or a carrier containing CD22 protein, if necessary, together with an adjuvant (eg, Freund adjuvant) as an adjuvant (eg, Freund adjuvant) except for humans Immunization is achieved by subcutaneous, intramuscular, intravenous, intraperitoneal or one or more injections in mammals.
  • an adjuvant eg, Freund adjuvant
  • the mammals other than humans are preferably mice, rats, hamsters, malmots, chickens, rabbits, cats, dogs, pigs, goats, sheep, donkeys, horses or cattle (transgenic mice that produce human antibodies) (including transgenic animals engineered to produce antibodies from other animals such as From the first immunization, immunization is performed 1 to 4 times every 1 to 21 days, and antibody-producing cells can be obtained from the immune-sensitized mammal about 1 to 10 days after the final immunization. The number of times and time intervals for immunization can be appropriately changed depending on the characteristics of the immunogen to be used, and the like.
  • Preparation of a hybridoma secreting a monoclonal antibody can be carried out according to the method of Keira and Mirstein et al. (Nature, 1975, Vol. 256, p. 495-497) and a method similar thereto.
  • Hybridomas can be prepared by cell fusing of myeloma cells.
  • the mammal may be a mouse, a rat, a guinea pig, a hamster, a chicken, a rabbit or a human, preferably a mouse, a rat, a chicken or a human.
  • a fusion promoter such as polyethylene glycol or Sendai virus, or a method by electric pulse is used, for example, an antibody-producing cell and a mammalian-derived cell capable of immortal growth in a fusion medium containing a fusion promoter. is suspended at a ratio of about 1:1 to 1:10, and in this state, incubated at about 30 to 40° C. for about 1 to 5 minutes.
  • the fusion medium for example, MEM medium, RPMI1640 medium and Iscove's Modified Dulbecco's Medium may be used, and it is preferable to exclude sera such as bovine serum.
  • the fusion cells obtained as described above are transferred to a selection medium such as HAT medium, and cultured at about 30 to 40° C. for about 3 days to 3 weeks. It kills cells other than hybridomas. Then, after culturing the hybridomas on a microtiter plate, etc., the immunogen used for the above-described immune response of animals other than humans and the part with increased reactivity with the culture supernatant were subjected to radioactive substance-marked immunotherapy (RIA). antibody) or ELISA (Enzyme-Linked Immunosorbent Assay). And the clone producing the monoclonal antibody found above shows a specific binding ability to the immunogen.
  • a selection medium such as HAT medium
  • the monoclonal antibody of the present invention can be obtained by culturing such a hybridoma in vitro or in vivo.
  • a conventional method for culturing cells derived from mammals is used, and for collecting monoclonal antibodies from a culture or the like, a conventional method in this field for purifying antibodies in general is used. Examples of each method include salting out, dialysis, filtration, concentration, centrifugation, fractional precipitation, gel filtration chromatography, ion exchange chromatography, affinity chromatography, high-performance liquid chromatography, and gel electrophoresis. and isoelectric point electrophoresis, and these are applied in combination as needed.
  • the purified monoclonal antibody is then concentrated and dried to obtain a liquid or solid state depending on the intended use.
  • the monoclonal antibody of the present invention comprises DNAs encoding heavy chain and light chain variable regions, respectively, with known DNA encoding the constant regions of heavy chain and light chain (e.g., Japan 2007-252372). No. publication) and each ligated gene is synthesized by PCR or chemical synthesis, and the transformant is obtained by transplanting a known expression vector (pcDNA 3.1 (sold by Invitrogen), etc.) that enables expression of the gene. It can be obtained by preparing an antibody and expressing it in a host such as CHO cells or Escherichia coli, and purifying the antibody from this culture solution using a protein A or G (Protein A or G) column or the like.
  • pcDNA 3.1 sold by Invitrogen
  • a hybridoma producing an anti-CD22 antibody is prepared and screened, and an antibody (scFv) that specifically binds to CD22 (scFv) 2 Species were selected and designated as 3F11 and 7B5, respectively.
  • the 3F11 antibody comprises a CDR1 region represented by the amino acid of SEQ ID NO: 1 (GFTFSNYW), a CDR2 region represented by the amino acid of SEQ ID NO: 2 (IRLKSNNYAT) and a CDR3 region represented by the amino acid of SEQ ID NO: 3 (TSIYYYGRDYAMDY)
  • a light chain comprising a heavy chain variable region and a CDR1 region represented by the amino acid of SEQ ID NO: 4 (SSVSSSY), a CDR2 region represented by the amino acid of SEQ ID NO: 5 (STS) and a CDR3 region represented by the amino acid of SEQ ID NO: 6 (HQYHRSPYT) It was confirmed that it consists of a variable region.
  • the 3F11 antibody is composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 7 and a light chain variable region represented by the amino acid of SEQ ID NO: 8, wherein the heavy chain variable region is the nucleotide sequence of SEQ ID NO: 9, and the light chain variable region is It was confirmed that it is encoded by the nucleotide sequence of SEQ ID NO: 10.
  • the 7B5 antibody comprises a CDR1 region represented by the amino acid of SEQ ID NO: 11 (GFSLTRYD), a CDR2 region represented by the amino acid of SEQ ID NO: 12 (MWTGGGT) and a CDR3 region represented by the amino acid of SEQ ID NO: 13 (VRSYYGSAMDY)
  • a light chain comprising a heavy chain variable region and a CDR1 region represented by the amino acid of SEQ ID NO: 14 (DHINNW), a CDR2 region represented by the amino acid of SEQ ID NO: 15 (GAT) and a CDR3 region represented by the amino acid of SEQ ID NO: 16 (QQHWSTPFT) It was confirmed that it consists of a variable region.
  • the 7B5 antibody is composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 17 and a light chain variable region represented by the amino acid of SEQ ID NO: 18, the heavy chain variable region is the nucleotide sequence of SEQ ID NO: 19, and the light chain variable region is It was confirmed that it is encoded by the nucleotide sequence of SEQ ID NO: 20.
  • the CD22-specific antibody of the present invention is preferably scFv (single chain variable fragment), and can be produced through genetic recombination technology so that the heavy chain variable region and the light chain variable region can be linked with a linker.
  • the linker may preferably be represented by the amino acid sequence of SEQ ID NO: 21 or the nucleotide sequence of SEQ ID NO: 22, but is not limited thereto.
  • the 3F11 antibody When linked by a light chain variable region-linker-heavy chain variable region, the 3F11 antibody may be represented by the amino acid sequence of SEQ ID NO: 23 or the nucleotide sequence of SEQ ID NO: 24, and the 7B5 antibody may be represented by the amino acid sequence of SEQ ID NO: 25 or the nucleotide sequence of SEQ ID NO: 26 have.
  • the present invention relates to a polynucleotide encoding the antibody that specifically binds to CD22.
  • polynucleotide generally refers to a nucleic acid molecule, deoxyribonucleotide or ribonucleotide, or an analog thereof, separated by any length.
  • the polynucleotides of the present invention are administered by (1) in-vitro amplification, such as polymerase chain reaction (PCR) amplification; (2) cloning and recombination; (3) purification such as digestion and gel electrophoretic separation; (4) It may be prepared through synthesis such as chemical synthesis, and preferably, the isolated polynucleotide is prepared by recombinant DNA technology.
  • the nucleic acid for encoding the antibody or antigen-binding fragment thereof can be prepared by various methods known in the art, including, but not limited to, restriction fragment operation of synthetic oligonucleotides or application of SOE PCR. can be manufactured.
  • the present invention relates to a vector comprising a polynucleotide encoding an antibody that specifically binds to CD22, and a recombinant cell transformed with the vector.
  • vector refers to a gene preparation including essential regulatory elements such as a promoter so that a target gene can be expressed in an appropriate host cell.
  • the vector may be selected from one or more of a plasmid, a retroviral vector and a lentiviral vector. Upon transformation into an appropriate host, the vector may replicate and function independently of the host genome, or in some cases may be integrated into the genome itself.
  • the vector may contain expression control elements that allow the coding region to be accurately expressed in a suitable host.
  • regulatory elements include, for example, promoters, ribosome-binding sites, enhancers and other regulatory elements for regulating gene transcription or mRNA translation. can do.
  • the specific structure of the expression control sequence may vary depending on the function of the species or cell type, but generally 5' ratios participating in transcription initiation and translation initiation, respectively, such as TATA box, capped sequence, CAAT sequence, etc. - contains a transcribed sequence, and a 5' or 3' non-translated sequence.
  • a 5' non-transcriptional expression control sequence may include a promoter region that may include a promoter sequence for transcription and control of a functionally linked nucleic acid.
  • promoter means a minimal sequence sufficient to direct transcription.
  • promoter constructs sufficient to allow expression of a regulatable promoter-dependent gene induced by cell type-specific or external signals or agents may be included, and these constructs may be located in the 5' or 3' portion of the gene. . Both conservative and inducible promoters are included.
  • Promoter sequences may be derived from prokaryotes, eukaryotes or viruses.
  • the term "transformant” refers to a cell transformed by introducing a vector having a polynucleotide encoding one or more target proteins into a host cell, and introducing the expression vector into the host cell to prepare a transformant
  • a method for this the calcium phosphate method or the calcium chloride/rubidium chloride method described in the literature (Sambrook, J., et al., Molecular Cloning, A Laboratory Manual (2nd edition), Cold Spring Harbor Laboratory, 1. 74, 1989) , an electroporation method, an electroinjection method, a chemical treatment method such as PEG, a method using a gene gun, or the like.
  • the transformant expressing the vector is cultured in a nutrient medium, it is possible to manufacture and isolate antibody proteins in large quantities.
  • Medium and culture conditions can be appropriately selected and used depending on the host cell. In culture, conditions such as temperature, medium pH, and culture time should be appropriately adjusted to be suitable for cell growth and mass production of protein.
  • the vector according to the present invention can be transformed into a host cell, preferably a mammalian cell, for the production of an antibody.
  • a host cell preferably a mammalian cell
  • suitable host cell lines capable of expressing fully glycosylated proteins have been developed in the art and include COS-1 (eg ATCC CRL 1650), COS-7 (eg ATCC CRL-1651), HEK293 , BHK21 (eg ATCC CRL-10), CHO (eg ATCC CRL 1610) and BSC-1 (eg ATCC CRL-26) cell lines, Cos-7 cells, CHO cells, hep G2 cells , P3X63Ag8653, SP2/0-Agl4, 293 cells, HeLa cells, etc., which are readily available from, for example, the American Type Culture Collection (ATCC, USA).
  • Preferred host cells include cells of lymphocyte origin, such as melanoma and lymphoma cells.
  • the present invention from another point of view,
  • a chimeric antigen receptor comprising an intracellular signal transduction domain
  • the CD22-binding domain comprises (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and SEQ ID NO: An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region represented by the amino acid of 4, a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid of SEQ ID NO: 6. ; or
  • a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14
  • An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 16.
  • chimeric antigen receptor generally refers to a fusion protein containing an antigen and an extracellular domain having the ability to bind one or more intracellular domains.
  • a CAR is a key part of a chimeric antigen receptor T cell (CAR-T) and may include an antigen binding domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain.
  • a CAR can be combined with a T cell receptor-activating intracellular domain based on the antigen (eg, CD22) specificity of the antibody.
  • Genetically modified CAR-expressing T cells can specifically identify and eliminate target antigen-expressing malignant cells.
  • CD22-binding domain generally refers to a domain capable of specifically binding to a CD22 protein.
  • the CD22-binding domain may contain an anti-CD22 antibody or fragment thereof capable of specifically binding to a human CD22 polypeptide or fragment thereof expressed in a B cell.
  • binding domain refers to "extracellular domain”, “extracellular binding domain”, “antigen-specific binding domain” and “Extracellular antigen-specific bidding domain” can be used interchangeably and refers to a CAR domain or fragment having the ability to specifically bind a target antigen (eg, CD22). refers to
  • the anti-CD22 antibody or fragment thereof is the above-described anti-CD22 antibody, a monoclonal antibody, preferably a single chain variable fragment (scFv). Specifically, it can be prepared using 3F11 and 7B5 antibodies specific for CD22 of the present invention.
  • scFv single chain variable fragment
  • a signal peptide may be further included at the N-terminus of the CD22-binding domain, and the "signal peptide” generally refers to a peptide chain for guiding protein transduction.
  • the signal peptide may be a short peptide having a length of 5 to 30 amino acids, and the amino acid sequence of SEQ ID NO: 34 is preferably used in the present invention.
  • the present invention may further comprise a hinge region located between the C terminus of the CD22-binding domain and the N terminus of the transmembrane domain, wherein the hinge region is derived from CD8 ⁇ , preferably SEQ ID NO: It can be represented by the amino acid sequence of 35.
  • the "hinge region” generally refers to the linking region between the antigen-binding region and the immune cell Fc receptor (FcR)-binding region.
  • transmembrane domain generally refers to a domain of a CAR that passes through a cell membrane and is connected to an intracellular signaling domain to play a signaling role.
  • the transmembrane domain may be derived from a protein selected from the group consisting of CD8 ⁇ , CD4, CD28, CD137, CD80, CD86, CD152 and PD1, and may preferably be represented by the amino acid sequence of SEQ ID NO: 36.
  • costimulatory domain generally refers to an intracellular domain capable of providing immune-stimulatory molecules, which are cell surface molecules necessary for an effective response of lymphocytes to antigens.
  • the costimulatory domain described above may comprise a costimulatory domain of CD28, and may comprise a costimulatory domain of the TNF receptor family such as the costimulatory domain of OX40 and 4-1BB, preferably SEQ ID NO: It may be 4-1BB represented by the amino acid sequence of 37.
  • intracellular signal transduction domain generally refers to a domain located inside a cell and capable of transmitting a signal.
  • the intracellular signaling domain is the intracellular signaling domain of the chimeric antigen receptor.
  • the intracellular signaling domain may be selected from CD3 ⁇ intracellular domain, CD28 intracellular domain, CD28 intracellular domain, 4-1BB intracellular domain and OX40 intracellular domain, preferably the amino acid of SEQ ID NO: 38 It may be CD3 ⁇ represented by the sequence.
  • the present invention relates to a polynucleotide encoding the chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the polynucleotide encoding the chimeric antigen receptor (CAR) comprises a polynucleotide encoding a CD22-binding domain; a polynucleotide encoding a transmembrane domain; polynucleotides coating the co-stimulatory domain; and a polynucleotide encoding an intracellular signaling domain.
  • the polynucleotide encoding the CD22-binding domain may be a polynucleotide encoding 3F11 and 7B5 antibodies specific for CD22 of the present invention, in the form of an scFv in which the light chain variable region and the heavy chain variable region are linked by a linker, and the specific nucleotide sequence is As described above.
  • the polynucleotide encoding the chimeric antigen receptor (CAR) of the present invention comprises: a signal peptide represented by the nucleotide sequence of SEQ ID NO: 28;
  • transmembrane domain represented by the nucleotide sequence of SEQ ID NO: 30;
  • 4-1BB (costimulatory domain) represented by the nucleotide sequence of SEQ ID NO: 31;
  • CD3 ⁇ intracellular signaling domain
  • SEQ ID NO: 32 The nucleotide sequence of SEQ ID NO: 32.
  • a polynucleotide encoding a hinge region may be further included, preferably a CD8 hinge region represented by the nucleotide sequence of SEQ ID NO: 29 can be
  • the present invention relates to a vector comprising a polynucleotide encoding the chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the vector is a recombinant viral vector, preferably a lentiviral vector, comprising an operably linked EF1 ⁇ promoter; a polynucleotide encoding a signal peptide; a polynucleotide encoding a CD22-binding domain; a polynucleotide encoding a transmembrane domain; It includes a polynucleotide encoding an intracellular signaling domain, and may further include a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) to increase protein expression ( FIG. 2 ).
  • WPRE woodchuck hepatitis virus post-transcriptional regulatory element
  • the EF1 ⁇ promoter may be represented by the nucleotide sequence of SEQ ID NO: 27, and if necessary, 90% or more, 93% or more, 95% or more, 96% or more, 97% or more, 98% or more of the nucleotide sequence of SEQ ID NO: 27 , or sequences that are at least 99% identical.
  • the promoter is operably linked to drive expression of the CD22-binding domain, anti-CD22 antibody (scFv).
  • Biological methods for introducing polynucleotides into host cells include the use of DNA and RNA vectors.
  • Viral vectors, and in particular retroviral vectors have become the most widely used methods for inserting genes into mammalian, eg, human cells.
  • Other viral vectors may be derived from lentiviruses, poxviruses, herpes simplex viruses, adenoviruses and adeno-associated viruses, and the like.
  • Chemical means for introducing polynucleotides into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads, and lipid-including oil-in-water emulsions, micelles, mixed micelles, and liposomes. including the underlying system.
  • An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (eg, an artificial membrane vesicle).
  • Other methods are available for state-of-the-art targeted delivery of nucleic acids, for example, delivery of polynucleotides using targeted nanoparticles or other suitable sub-micron sized delivery systems.
  • an exemplary delivery vehicle is a liposome.
  • lipid preparations is contemplated for the introduction of nucleic acids into host cells (in vitro, ex vivo or in vivo).
  • the nucleic acid may be associated with a lipid.
  • Nucleic acids associated with lipids may be encapsulated within the aqueous interior of the liposome, interspersed within the lipid bilayer of the liposome, attached to the liposome via a linking molecule associated with both the liposome and oligonucleotide, captured within the liposome, complexed with the liposome, or , dispersed in a lipid containing solution, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with micelles, or otherwise associated with a lipid.
  • the lipid, lipid/DNA or lipid/expression vector association composition is not limited to any particular structure in solution.
  • the present invention includes a vector comprising a polynucleotide encoding the chimeric antigen receptor (CAR) or a polynucleotide encoding a chimeric antigen receptor (CAR), wherein the chimeric antigen receptor (CAR) It relates to immune effector cells expressing
  • the immune effector cells may be mammalian-derived cells, preferably T cells or natural killer (NK) cells.
  • immune effector cells expressing the chimeric antigen receptor can be prepared by introducing the CAR vector of the present invention into immune effector cells, for example, T cells or NK cells.
  • the CAR vector may be introduced into cells by methods known in the art such as electroporation, lipofectamine 2000, Invitrogen, and the like.
  • immune effector cells can be transfected with a lentiviral vector to integrate the viral genome carrying the CAR molecule into the host genome to ensure long-term and stable expression of the target gene.
  • a transposon can be used to introduce a CAR transport plasmid and a transferase transport plasmid into a target cell.
  • a CAR molecule can be added to the genome by a gene editing method (eg, CRISPR/Cas9).
  • a lentiviral vector into which a polynucleotide coating CD22-CAR was inserted was prepared, and the prepared vector was transformed into T cells to prepare CD22-CAR-T cells. did.
  • the prepared CD22-CAR-T cells express the chimeric antigen receptor targeting CD22 of the present invention.
  • Immune effector cells for making immune effector cells expressing a chimeric antigen receptor (CAR) can be obtained from a subject, wherein the "subject” is a living organism (eg, a mammal) in which an immune response can be elicited. includes Examples of subjects include humans, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from numerous sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, umbilical cord blood, thymus tissue, tissue from the site of infection, ascites, pleural effusion, splenic tissue, and tumors.
  • CAR chimeric antigen receptor
  • T cells can be obtained from blood units collected from a subject using any of a number of techniques known to those of ordinary skill in the art, for example, FicollTM separation.
  • Cells from blood are obtained by apheresis, and apheresis products typically contain T cells, monocytes, granulocytes, lymphocytes including B cells, other nucleated leukocytes, red blood cells, and platelets.
  • T cells are isolated from peripheral blood lymphocytes by lysing red blood cells and depleting monocytes, for example, by centrifugation through a PERCOLLTM gradient or by countercurrent centrifugation.
  • activated T cells are isolated from peripheral blood mononuclear cells (PBMCs), and then CD22-CAR lentivirus is transduced into T cells. to prepare CD22-CAR-T cells.
  • PBMCs peripheral blood mononuclear cells
  • CD22-CAR lentivirus is transduced into T cells.
  • FIG. 4 it was confirmed that the CD22-CAR-T cells expressing CD4 or CD8 increased as the CD22-peptide increased. This means that the CD22-CAR-T cells prepared in the present invention effectively bind to CD22.
  • the level of IFN ⁇ expression by CD22-CAR-T cells in the presence of target cells was checked.
  • T cells were not activated in K562 cells that do not express CD22, whereas T cells were activated in the presence of U2932 cells expressing CD22, thereby increasing IFN ⁇ expression. .
  • the chimeric antigen receptor and CAR-T cell targeting CD22 of the present invention can be usefully used as a composition for preventing or treating diseases related to B cells or CD22 expression.
  • composition for preventing or treating diseases mediated by CD22 expression Composition for preventing or treating diseases mediated by CD22 expression
  • the present invention provides a pharmaceutical for the prevention or treatment of diseases mediated by CD22-expressing cells, including immune effector cells expressing an antibody that specifically binds to CD22 or a chimeric antigen receptor targeting CD22. to the composition.
  • the disease mediated by cells expressing CD22 is lymphoma, non-Hogkins lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed delayed NHL, refractory NHL, refractory Delayed NHL, chronic lymphocytic leukemia (CLL), small lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL), Burkitt's lymphoma and mantle cell lymphoma.
  • NHL non-Hogkins lymphoma
  • aggressive NHL relapsed aggressive NHL
  • refractory NHL refractory Delayed NHL
  • CLL chronic lymphocytic leukemia
  • small lymphoma small lymphoma
  • leukemia hairy cell leukemia
  • HCL hairy cell leukemia
  • ALL acute lymphocytic leukemia
  • Burkitt's lymphoma mantle cell lymphoma.
  • the composition may include a therapeutic agent for a disease mediated by a cell expressing CD22, and the therapeutic agent is present in a state covalently bound to the heavy and/or light chain of an antibody that specifically binds to CD22.
  • the CD22-specific antibody or CD22-CAR-T cell of the present invention may be co-administered.
  • the therapeutic agent includes a small molecule drug, a peptide drug, a toxin (eg, a cytotoxin), and the like.
  • the low molecular weight drug may be a compound that exhibits a pharmaceutical activity of interest and generally has a molecular weight of about 800 Da or less or 2000 Da or less.
  • An inorganic small molecule refers to a molecule containing no carbon atoms, whereas an organic small molecule refers to a compound containing at least one carbon atom.
  • the peptidic drug refers to amino acids containing polymeric compounds, including naturally occurring and non-naturally occurring peptides, oligopeptides, cyclic peptides, polypeptides and proteins, as well as peptide mimics.
  • the peptide drug may be obtained by chemical synthesis or produced from a genetically encoded source (eg, a recombinant source).
  • the molecular weight of the peptide drug may range from 200 Da to 10 kDa or more.
  • the toxin is preferably a cytotoxin
  • the cytotoxin includes, but is not limited to, ricin, abrin, diphtheria toxin, Pseudomonas exotoxin (eg, PE35, PE37, PE38, PE40, etc.), saporin, gelonin, USA. antiviral protein (PAP), botulinum toxin, bryodin, momordin and buganin.
  • PAP antiviral protein
  • the therapeutic agent may be an anticancer agent.
  • Anticancer agents reduce the proliferation of cancer cells and include non-peptidyl (ie, non-proteinaceous) compounds, including cytotoxic agents and cytostatic agents.
  • Non-limiting examples of anticancer agents include alkylating agents, nitrosourea, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids and steroid hormones. Peptide compounds may also be used.
  • an antibody that specifically binds to CD22 or an immune effector cell expressing a chimeric antigen receptor targeting CD22 is the only active ingredient in the composition for treatment or diagnosis, or, for example, an anti-T cell, anti It can be used in combination with other active ingredients, including other antibody components such as -IFN ⁇ or anti-LPS antibodies, or non-antibody components such as xanthine.
  • the pharmaceutical composition preferably comprises a therapeutically effective amount of an antibody of the invention.
  • therapeutically effective amount means an amount of a therapeutic agent required to treat, ameliorate, or prevent a target disease or condition, or the amount of a therapeutic agent necessary to exhibit a detectable therapeutic or prophylactic effect.
  • a therapeutically effective dosage can be initially determined by cell culture assays or animal models, usually rodents, rabbits, dogs, pigs, or primates. Animal models can also be used to determine appropriate concentration ranges and routes of administration. Such information can be used to determine useful dosages and routes for dosing in humans.
  • an effective dosage is 0.01-50 mg/kg, preferably 0.1-20 mg/kg, more preferably about 15 mg/kg.
  • composition may be administered to the patient individually or in combination with other agents, agents, or hormones.
  • the dosage at which the antibody of the present invention is administered depends on the nature of the condition to be treated, the grade of malignant lymphoma or leukemia, and whether the antibody is used to prevent disease or to treat an existing condition.
  • the frequency of administration depends on the half-life of the antibody molecule and the duration of the drug's effect. If the antibody molecule has a short half-life (eg, 2 to 10 hours), it may be necessary to provide one or more doses per day. Alternatively, if the antibody molecule has a long half-life (eg, 2 to 15 days), it may be necessary to provide a dose once a day, once a week, or once every 1 or 2 months.
  • the pharmaceutical composition may contain a pharmaceutically acceptable carrier for administration of the antibody.
  • the carrier itself must not cause the production of antibodies that are harmful to the individual receiving the composition, and must be non-toxic.
  • Suitable carriers may be slowly metabolized macromolecules, such as proteins, polypeptides, liposomes, polysaccharides, polylactic acid, polyglycolic acid, amino acid polymers, amino acid copolymers and inactive viral particles.
  • salts are, for example, mineral acid salts such as hydrochloride, hydrobromide, phosphate and sulfate, or acetic acid, propionic acid. Salts of organic acids such as malonic acid and benzoic acid may be used.
  • Pharmaceutically acceptable carriers in therapeutic compositions may additionally include liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances such as wetting agents, emulsifying agents or pH buffering agents may be present in such compositions.
  • the carrier may be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions for ingestion of the pharmaceutical composition by a patient.
  • Preferred forms for administration include those suitable for parenteral administration, eg, by injection or infusion (eg, bolus injection or continuous infusion).
  • parenteral administration eg, by injection or infusion (eg, bolus injection or continuous infusion).
  • the product may take the form of suspensions, solutions or emulsions in oil or water-soluble excipients, which may contain prescription agents such as suspending, preservative, stabilizing and/or dispersing agents.
  • the antibody molecule may be in anhydrous form and reconstituted with an appropriate sterile solution prior to use.
  • compositions of the present invention can be administered directly to a patient.
  • the patients to be treated may be animals.
  • the composition is preferably adapted for administration to human patients.
  • the pharmaceutical composition of the present invention is not limited, but oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intraventricular, transdermal, transcutaneous (see, e.g., WO 98/20734), subcutaneous, Administration may be by any route, including intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal routes.
  • a hypospray may be used to administer the pharmaceutical composition of the present invention.
  • therapeutic compositions may be prepared as injectables as liquid solutions or suspensions.
  • solid forms suitable for solution or suspension in liquid excipients prior to injection may be prepared.
  • Direct delivery of the composition may generally be achieved by injection, subcutaneous injection, intraperitoneal injection, intravenous injection, intramuscular injection, or may be delivered to the interstitial space of a tissue.
  • the composition may be administered to the wound site. Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • the active ingredient in the composition may be an antibody molecule. As such, it may be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is administered by a route using the gastrointestinal tract, the composition will need to contain an agent that protects the antibody from degradation but releases the antibody once absorbed from the gastrointestinal tract.
  • the present invention relates to a composition for diagnosing or monitoring a disease mediated by a CD22-expressing cell comprising an antibody that specifically binds to CD22.
  • the antibody that specifically binds to CD22 may be directly or indirectly labeled.
  • Indirect labels include secondary antibodies comprising a detectable label, wherein the secondary antibody binds to an antibody that specifically binds to CD22.
  • Other indirect labels include biotin, wherein an antibody that specifically binds to biotinylated CD22 can be detected using avidin or streptavidin comprising a detectable label.
  • Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Suitable labels include, but are not limited to, magnetic beads, fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, green fluorescent protein, red fluorescent protein, yellow fluorescent protein, etc.), radioactive labels (e.g., For example, 3 H, 125 I, 35 S, 14 C or 32 P), enzymes (eg, mustard radish peroxidase, alkaline phosphatase, luciferase and enzyme-linked immunosorbent assay (ELISA)) commonly used) and colorimetric labels such as colloidal gold or colored glass or plastic (eg, polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein isothiocyanate, Texas red, rhodamine, green fluorescent protein, red fluorescent protein, yellow fluorescent protein, etc.
  • the antibody may be labeled with a fluorescent protein, and may contain a contrast agent or a radioisotope.
  • the antibody specifically binding to CD22 of the present invention is used in a diagnostic kit
  • the antibody is immobilized on a support
  • the support may be a microplate, microarray, chip, glass, bead or particle, or a membrane.
  • Example 1 Preparation and selection of antibodies that specifically bind to CD22
  • CD22 peptide-specific antibody To select the CD22 peptide-specific antibody, a hybridoma producing an antibody binding to CD22 was prepared and the antibody was selected.
  • splenocytes were extracted by immunization with CD22 protein (ACRObiosystems Inc., cat# CD2-H52H8, USA), and hybridoma cells were prepared by cell fusion with mouse myeloma cells.
  • HAT medium Human myeloma cells used for cell fusion cannot survive in HAT medium because they do not have HGPRT (HypoxanthineGuanidine-Phosphoribosyl-Transferase), but hybridomas can survive in HAT medium by fusion with splenocytes. Since only hybridomas can be grown using this, it is usually grown in HAT medium until hybridomas are established.
  • HGPRT HypoxanthineGuanidine-Phosphoribosyl-Transferase
  • the limiting dilution method was used to select hybridomas producing an antibody binding to CD22 from among the proliferated hybridomas. First, it was made to be less than one cell per 96 well, and then, it was confirmed by ELISA whether the antibody obtained from the clones proliferated from one cell binds to CD22, and clones that bind to CD22 were selected. The above process was repeated three times to select hybridomas producing an antibody binding to CD22. In this way, two types of antibodies that bind to CD22 were obtained.
  • the two antibodies were named 3F11 and 7B5, respectively, and their base and amino acid sequences were analyzed. Sequence information on the heavy chain variable region and the light chain variable region of each antibody according to the sequencing results is shown in Tables 1 and 2 below, and the underlined portions in Tables 1 and 2 below are complementarity determining regions; CDR).
  • CD22-His tag CD22 extracellular domain; ACRObiosystems Inc.
  • 3F11 and 7B5 antibodies were treated in each well, and then reacted at room temperature for 2 hours, and then washed 3 times with 1 X PBST.
  • Secondary antibody (anti-HRP, 1:10,000) was treated and reacted at room temperature for 30 minutes, washed 3 times with 1 X PBST, and then treated with TMB for color development and reacted at room temperature for 5 minutes. Finally, the reaction was terminated by treatment with a stop solution of 1N H 2 SO 4 , and then the absorbance was measured at 450 nm.
  • CD22-expressing non-cell lymphoma U2932 (B-cell lymphoma U2932 cell) was reacted with 1 ⁇ 10 6 cells and 1 ⁇ g of 3F11 and 7B5 antibodies for 30 minutes, and then the surface was stained with a secondary antibody, followed by flow cytometry. Measured with an analyzer.
  • PE-conjugated anti-CD22 antibody PE-conjugated anti-CD22 antibody; Biolegend Inc., cat# 302506, USA
  • a PE-conjugated anti-mouse IgG antibody was used as a positive control
  • PE-conjugated goat anti-mouse IgG PE-conjugated goat anti-mouse IgG; Biolegend Inc., cat# 405307, USA
  • Example 4 Construction of a chimeric antigen receptor (CAR) expression vector targeting CD22
  • a lentiviral vector CD22-CAR lentivirus
  • CAR chimeric antigen receptor
  • EF1 ⁇ promoter SEQ ID NO: 27
  • a polynucleotide encoding a signal peptide SEQ ID NO: 28
  • a polynucleotide encoding a CD22-binding domain 3F11 represented by SEQ ID NO: 24 or 7B5 represented by SEQ ID NO: 26
  • a polynucleotide encoding the CD8 hinge region SEQ ID NO: 29
  • a polynucleotide encoding a transmembrane domain SEQ ID NO: 730
  • a polynucleotide encoding 4-1BB (costimulatory domain) SEQ ID NO: 31
  • a polynucleotide encoding CD3 ⁇ intracellular signaling domain SEQ ID NO: 32
  • CAR DNA composed of a polynucleotide encoding WPRE SEQ ID NO: 33
  • Lentiviral vectors were co-transfected into Lenti-X 293T cells with three vectors, pMDLg/pRRE (Addgene, cat# #12251), pMD2.G (Addgene, cat##12259), and pRSV-Rev (Addgene, cat##12253). After co-transfection, CD22-CAR lentivirus was produced. For co-infection, three vectors and Lenti-X 293T cells were cultured for 6 hours using Lipofectamine 3000 transfection kit (Invitrogen, cat# L3000-015) and Opti-MEM+GlutaMAX (gibco, cat# 51985-034) medium. .
  • the CD22-CAR lentiviral vector prepared in Example 4 was transformed into T cells to prepare CD22-CAR-T cells.
  • T cell activation beads (T cell activation bead; Miltenyl Biotec, cat# 130-091-441) was used to activate T cells.
  • CD22-CAR-T cells were prepared by transducing the activated T cells with the CD22-CAR lentivirus prepared in Example 4 into T cells, and the transduction efficiency was increased using Lenti-boost-p.
  • CD22 peptide binding capacity of CD22-CAR-T cells was confirmed by flow cytometry.
  • the CD22-CAR-T cells prepared above were reacted with FITC-CD22 protein and anti-CD3, anti-CD4, and anti-CD8 antibodies, and then fluorescence intensity was measured using a FACS machine.
  • CD3 expressing cells were used as T cells, and the level of FITC expression in T cells was confirmed.
  • CD22-CAR-T cells expressing CD4 or CD8 binding to CD22 increased as the CD22 peptide increased. This means that the CD22-CAR-T cells prepared in the present invention effectively bind to CD22.
  • the level of IFN ⁇ expression by the CD22-CAR-T cells in the presence of target cells was checked.
  • K562 cells that do not express CD22 (ATCC, cat#CCL-243) and U2932 cells that express CD22 (DSMZ, cat#ACC-633) were used. :1, 1:1. After reacting for a certain period of time at a ratio of 0.5:1, 0:1, surface & intra antibody staining was performed for FACS measurement (CD22 protein, INF-r, CD107a, CD3, CD4, CD8 staining). The level of IFN ⁇ expression of CD22-CAR-T reacted with target cells was confirmed on the basis of 0:1 (CAR-T only).
  • T cells were not activated in K562 cells that do not express CD22, whereas T cells were activated in the presence of U2932 cells expressing CD22, thereby increasing IFN ⁇ expression. .
  • K562 cells which do not express CD22
  • U2932 cells which express CD22
  • the ratios of CD22-CAR-T cells were 1:4, 1:2, 1:1, 1:0.5 and 1:0.25.
  • luminescence CytoTox-Glo Cytotoxicity Assay, Promega, cat# G9291 was measured. The degree of cell death was calculated using Equation 1 below as the measured value.
  • Target Spontaneous Luminescence value derived from the medium of target cells only
  • Target Maximum Luminescence value derived from 100% lysis of target cells (using Lysis Reagent)
  • the CD22-CAR-T cells of the present invention specifically killed U2932 cells expressing CD22.
  • the antibody selected in the present invention specifically recognizes CD22-expressing cells, and using the established antibody, the chimeric antigen receptor (CAR) and CAR-T cells targeting CD22 effectively bind to CD22 In addition, it was confirmed that the activation of CAR-T cells bound to CD22 was made.
  • CAR chimeric antigen receptor
  • the CAR-T cells of the present invention effectively kill CD22-expressing cells
  • the CD22-specific antibody, the chimeric antigen receptor targeting CD22, and CAR-T cells of the present invention are related to CD22 expression. It can be usefully used as a composition for preventing or treating diseases.

Abstract

The present invention relates to a CD22-speciifc antibody and a use thereof and, more specifically, to an antibody binding specifically to CD22, a chimeric antigen receptor including the antibody, a CAR-T cell expressing the chimeric antigen receptor, and a pharmaceutical composition comprising same for preventing or treating CD22-expressing cell-mediated diseases. The antibody selected in the present invention was identified to specifically recognize CD22-expressing cells, and it was found that not only do the chimeric antigen receptor (CAR) and CAR-T cells, which employ the established antibody to target CD22, bind effectively to CD22, but also the CD22-bound CAR-T cells are activated. Furthermore, because the CAR-T cells of the present invention were identified to effectively kill CD22-expressing cells, the CD22-speific antibody, the chimeric antigen receptor targeting CD22, and the CAR-T cells according to the present invention can be advantageously utilized in a composition for prevention or treatment of CD22 expression-related diseases.

Description

CD22에 특이적인 항체 및 이의 용도Antibodies specific for CD22 and uses thereof
본 발명은 CD22에 특이적인 항체 및 이의 용도에 관한 것으로, 보다 상세하게는 CD22에 특이적으로 결합하는 항체, 상기 항체를 포함하는 키메라 항원 수용체, 상기 키메라 항원 수용체를 발현하는 CAR-T 세포, 및 이들을 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환 예방 또는 치료용 약학적 조성물에 관한 것이다.The present invention relates to an antibody specific for CD22 and uses thereof, and more particularly, to an antibody that specifically binds to CD22, a chimeric antigen receptor comprising the antibody, a CAR-T cell expressing the chimeric antigen receptor, and It relates to a pharmaceutical composition for preventing or treating diseases mediated by cells expressing CD22, including them.
CD22는 시알로어드헤신(sialoadhesins)이라 불리는 시알산(sialic acid) 결합단백질 족에 속하는 135kDa의 막당단백질이다. CD22는 B세포 발생 초기에 세포질에서 검출되고, IgD와 동시에 세포 표면에 나타나며, 대부분의 성숙한 B세포에서 발견된다. B세포가 활성화되면 발현이 증가한다. CD22는 분화말기에 상실되며, 일반적으로 형질세포(plasma cell)에는 존재하지 않는 것으로 보고되었다.CD22 is a 135 kDa membrane glycoprotein belonging to a family of sialic acid-binding proteins called sialoadhesins. CD22 is detected in the cytoplasm at the beginning of B cell development, appears on the cell surface simultaneously with IgD, and is found in most mature B cells. When B cells are activated, their expression is increased. It has been reported that CD22 is lost at the end of differentiation and is generally absent in plasma cells.
인간의 CD22에는 2종의 동형(isoform)이 존재한다. 지배적인 형태(CD22β)는 세포외부 영역에 7개의 면역글로불린-유사(Ig-like) 도메인을 포함한다. CD22α 변이체는 Ig-유사 도메인 4가 없고, 세포질 도메인이 잘려진 형태를 가질 수 있다. 단핵구, 호중구(neutrophil), 림프구 및 적혈구에 대한 CD22의 결합을 차단하는 항체는, 첫번째 또는 두번째 Ig-유사 도메인내에 결합하는 것이 관찰되었다.There are two isoforms of human CD22. The dominant form (CD22β) contains seven immunoglobulin-like (Ig-like) domains in the extracellular region. The CD22α mutant lacks Ig-like domain 4 and may have a truncated cytoplasmic domain. Antibodies that block the binding of CD22 to monocytes, neutrophils, lymphocytes and erythrocytes have been observed to bind within the first or second Ig-like domain.
CD22의 세포질 도메인은, B세포 항원 수용체의 결합에 따라 티로신이 인산화되며, Lyk, Syk 및 포스파티딜이노시톨 3-키나제와 결합한다. CD22의 기능은 B세포 활성화 역치(threshold)를 하위조절(down-modulation)하는 것이다. 이것은 또한 적당한 시알로당접합체(sialoglycoconjugates)를 포함하는 세포와의 결합을 통해 세포결합을 매개할 수 있다.In the cytoplasmic domain of CD22, tyrosine is phosphorylated upon binding to the B-cell antigen receptor and binds to Lyk, Syk and phosphatidylinositol 3-kinase. The function of CD22 is to down-modulate the B cell activation threshold. It can also mediate cell binding through binding to cells containing appropriate sialoglycoconjugates.
CD22는 NHL, 급성 림프모구백혈병(acute lymphoblastic leukaemia: B-ALL), 만성 림프모구백혈병(chronic lymphocytic leukaemia: B-CLL) 및 특히 급성 비림프구 백혈병(acute non-lymphocytic leukaemia: ANLL)을 포함하는, 대부분의 B세포 백혈병 및 림프종에서 발현된다.CD22 includes NHL, acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (B-CLL) and especially acute non-lymphocytic leukemia (ANLL), It is expressed in most B-cell leukemias and lymphomas.
이러한 CD22 발현과 관련된 질환 등의 치료 또는 진단을 위해 CD22에 특이적인 항체의 개발이 이루어지고 있다. 국제공개특허 WO1998-041641호에는 V H44및 V L100위치에 시스테인 잔기를 갖는 재조합 항-CD22 항체에 대해 개시되어 있으며, 국제공개특허 WO 1998-042378호에는 B세포 악성종양의 치료를 위한 항-CD22 항체에 대해 개시되어 있다.For the treatment or diagnosis of diseases related to CD22 expression, the development of an antibody specific for CD22 is being made. International Patent Publication No. WO1998-041641 discloses a recombinant anti-CD22 antibody having cysteine residues at positions V H 44 and V L 100, and International Patent Publication No. WO 1998-042378 discloses an antibiotic for the treatment of B-cell malignancies. -CD22 antibody is disclosed.
본 발명에서는 CD22에 보다 특이적으로 결합하는 항체를 개발하기 위해 CD22에 결합하는 항체를 스크리닝 하여 신규한 항체 2종(3F11 및 7B5)을 확립하였으며, 본 발명에서 선별한 2종의 항체는 CD22 항원과 특이적으로 결합하는 것을 확인하였다. 또한, 본 발명의 CD22 특이적인 항체를 이용하여 CD22를 표적으로 하는 키메라 항원 수용체 및 CAR-T 세포를 제조하였으며, CD22-CAR-T 세포는 CD22와 결합할 뿐만 아니라, CD22 발현 세포(B 세포 림프종)를 효과적으로 사멸시키는 것을 확인하였다.In the present invention, in order to develop an antibody that more specifically binds to CD22, an antibody that binds to CD22 was screened to establish two novel antibodies (3F11 and 7B5), and the two types of antibodies selected in the present invention are CD22 antigen It was confirmed that it specifically binds to In addition, using the CD22-specific antibody of the present invention, a chimeric antigen receptor targeting CD22 and CAR-T cells were prepared. ) was confirmed to effectively kill.
따라서, 본 발명의 목적은 CD22에 특이적으로 결합하는 항체를 제공하는 데 있다.Accordingly, it is an object of the present invention to provide an antibody that specifically binds to CD22.
본 발명의 다른 목적은 상기 항체를 코딩하는 폴리뉴클레오타이드, 상기 항체를 발현하는 벡터, 상기 벡터로 형질전환된 재조합 세포를 제공하는 데 있다.Another object of the present invention is to provide a polynucleotide encoding the antibody, a vector expressing the antibody, and a recombinant cell transformed with the vector.
본 발명의 또 다른 목적은 상기 항체를 포함하는 키메라 항원 수용체, CD22를 표적하는 키메라 항원 수용체를 코딩하는 폴리뉴클레오타이드, 이를 포함하는 벡터, 및 상기 폴리뉴클레오타이드 또는 벡터를 포함하는 키메라 항원 수용체 발현하는 면역 이펙터 세포를 제공하는 데 있다. Another object of the present invention is a chimeric antigen receptor comprising the antibody, a polynucleotide encoding the chimeric antigen receptor targeting CD22, a vector comprising the same, and an immune effector expressing the polynucleotide or the chimeric antigen receptor comprising the vector to provide cells.
본 발명의 또 다른 목적은 상기 항체 또는 CD22를 표적하는 키메라 항원 수용체를 발현하는 면역 이펙터 세포를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 예방 또는 치료용 약학적 조성물을 제공하는 데 있다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating a disease mediated by CD22-expressing cells, including immune effector cells expressing the antibody or chimeric antigen receptor targeting CD22.
본 발명의 또 다른 목적은 상기 항체를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 진단 또는 모니터링 조성물을 제공하는 데 있다.Another object of the present invention is to provide a composition for diagnosing or monitoring a disease mediated by a cell expressing CD22 comprising the antibody.
상술한 목적을 달성하기 위해, In order to achieve the above object,
본 발명은 (a) 서열번호 1의 아미노산으로 표시되는 CDR1 영역, 서열번호 2의 아미노산으로 표시되는 CDR2 영역 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역, 서열번호 5의 아미노산으로 표시되는 CDR2 영역 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위; 또는 The present invention relates to (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3, and the amino acid of SEQ ID NO: 4 a light chain variable region comprising a CDR1 region represented by , a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid sequence represented by SEQ ID NO: 6; or
(b) 서열번호 11의 아미노산으로 표시되는 CDR1 영역, 서열번호 12의 아미노산으로 표시되는 CDR2 영역 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역, 서열번호 15의 아미노산으로 표시되는 CDR2 영역 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위로구성된 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 제공한다.(b) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15 and a CDR3 region represented by the amino acid of SEQ ID NO: 16 is provided.
본 발명의 바람직한 일실시예에 있어서, 상기 항체는 단클론 항체, 바람직하게는 scFv(Single-chain variable fragment)일 수 있다.In a preferred embodiment of the present invention, the antibody may be a monoclonal antibody, preferably a single-chain variable fragment (scFv).
본 발명의 바람직한 다른 일실시예에 있어서, 상기 (a) 항체는 서열번호 7의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 8의 아미노산으로 표시되는 경쇄 가변부위로,In another preferred embodiment of the present invention, the antibody (a) has a heavy chain variable region represented by the amino acid of SEQ ID NO: 7 and a light chain variable region represented by the amino acid of SEQ ID NO: 8,
상기 (b) 항체는 서열번호 17의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 18의 아미노산으로 표시되는 경쇄 가변부위로 구성될 수 있다. The antibody (b) may be composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 17 and a light chain variable region represented by the amino acid of SEQ ID NO: 18.
다른 목적을 달성하기 위해, 본 발명은 상기 CD22에 특이적으로 결합하는 항체를 코딩하는 폴리뉴클레오타이드를 제공한다.To achieve another object, the present invention provides a polynucleotide encoding the antibody that specifically binds to CD22.
또한, 본 발명은 상기 CD22에 특이적으로 결합하는 항체를 코딩하는 폴리뉴클레오타이드 포함하는 벡터를 제공한다. In addition, the present invention provides a vector comprising a polynucleotide encoding the antibody that specifically binds to CD22.
또한, 본 발명은 상기 벡터로 형질전환된 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 생산하는 재조합 세포를 제공한다. In addition, the present invention provides a recombinant cell that produces an antibody or fragment thereof that specifically binds to CD22 transformed with the vector.
또 다른 목적을 달성하기 위해, 본 발명은 CD22-결합 도메인; 막관통 도메인(transmembrane domain); 공동자극 도메인(costimulatory domain); 및 세포 내 신호전달 도메인(intracellular signal transduction domain)을 포함하는 키메릭 항원 수용체(chimeric antigen receptor: CAR)로,To achieve another object, the present invention provides a CD22-binding domain; transmembrane domain; costimulatory domain; and a chimeric antigen receptor (CAR) comprising an intracellular signal transduction domain,
상기 CD22-결합 도메인은 본 발명의 CD22와 특이적으로 결합할 수 있는 항체 또는 이의 단편 중에 선택될 수 있다.The CD22-binding domain may be selected from an antibody or fragment thereof capable of specifically binding to CD22 of the present invention.
본 발명의 바람직한 일실시예에 있어서, 상기 막관통 도메인은 CD8α, CD4, CD28, CD137, CD80, CD86, CD152 및 PD1로 구성된 군에서 선택되는 단백질로부터 유래될 수 있다. In a preferred embodiment of the present invention, the transmembrane domain may be derived from a protein selected from the group consisting of CD8α, CD4, CD28, CD137, CD80, CD86, CD152 and PD1.
본 발명의 다른 바람직한 일실시예에 있어서, 상기 공동자극 도메인은 CD28, 4-1BB, OX-40 및 ICOS로 구성된 군에서 선택되는 단백질 유래일 수 있고, 상기 신호전달 도메인은 CD3ζ 유래일 수 있다.In another preferred embodiment of the present invention, the costimulatory domain may be derived from a protein selected from the group consisting of CD28, 4-1BB, OX-40 and ICOS, and the signaling domain may be derived from CD3ζ.
본 발명의 또 다른 바람직한 일실시예에 있어서, 상기 CD22-결합 도메인의 C 말단 및 막경유 도메인의 N 말단 사이에 위치된 힌지 부위(hinge region)를 추가로 포함할 수 있으며, 상기 힌지 부위는 CD8α 유래일 수 있다. In another preferred embodiment of the present invention, it may further include a hinge region located between the C-terminus of the CD22-binding domain and the N-terminus of the transmembrane domain, wherein the hinge region is CD8α may be of origin.
다른 목적을 달성하기 위해, 본 발명은 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드를 제공한다.To achieve another object, the present invention provides a polynucleotide encoding the chimeric antigen receptor (CAR).
또한, 본 발명은 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 포함하는 벡터를 제공한다. The present invention also provides a vector comprising a polynucleotide encoding a chimeric antigen receptor (CAR).
본 발명의 바람직한 일실시예에 있어서, 상기 벡터는 플라스미드(plasmid), 레트로바이러스(retroviral) 벡터 또는 렌티바이러스(lentiviral) 벡터일 수 있다. In a preferred embodiment of the present invention, the vector may be a plasmid, a retroviral vector or a lentiviral vector.
또한, 본 발명은 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 또는 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 포함하고, 상기 키메릭 항원 수용체(CAR)를 발현하는 면역 이펙터 세포를 제공한다. In addition, the present invention provides an immune effector cell comprising the polynucleotide encoding the chimeric antigen receptor (CAR) or a polynucleotide encoding the chimeric antigen receptor (CAR), and expressing the chimeric antigen receptor (CAR). do.
본 발명의 바람직한 일실시예 있어서, 상기 면역 이펙터 세포는 T 세포일 수 있다.In a preferred embodiment of the present invention, the immune effector cells may be T cells.
또 다른 목적을 달성하기 위해, 본 발명은 CD22를 표적하는 키메라 항원 수용체를 발현하는 면역 이펙터 세포; 또는 CD22에 특이적으로 결합하는 항체 또는 이의 단편;을 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 예방 또는 치료용 약학적 조성물을 제공한다. To achieve another object, the present invention provides an immune effector cell expressing a chimeric antigen receptor targeting CD22; Or an antibody or fragment thereof that specifically binds to CD22; provides a pharmaceutical composition for preventing or treating a disease mediated by cells expressing CD22, including.
본 발명의 바람직한 일실시예에 있어서, CD22를 발현하는 세포에 의해 매개되는 질환은 림프종, 비호치킨 림프종(non-Hogkins lymphoma: NHL), 공격적 NHL, 재발성 공격적 NHL, 재발성 지연성 NHL, 불응성 NHL, 불응성 지연성 NHL, 만성 림프성 백혈병(chronic lymphocytic leukemia: CLL), 소형 림프성 림프종, 백혈병, 모발성 세포 백혈병(hairy cell leukemia: HCL), 급성 림프성 백혈병(acute lymphocytic leukemia: ALL), 버킷트 림프종 및 외투 세포 림프종로 구성된 군에서 선택될 수 있다. In a preferred embodiment of the present invention, the disease mediated by cells expressing CD22 is lymphoma, non-Hogkins lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed delayed NHL, refractory Sexual NHL, refractory delayed NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL) ), Burkitt's lymphoma, and mantle cell lymphoma.
본 발명의 바람직한 또 다른 일실시예에서, 상기 조성물은 CD22를 발현하는 세포에 의해 매개되는 질환의 치료제를 포함할 수 있다. In another preferred embodiment of the present invention, the composition may include a therapeutic agent for a disease mediated by cells expressing CD22.
또한, 본 발명은 CD22에 특이적으로 결합하는 항체를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 진단 또는 모니터링용 조성물을 제공한다.In addition, the present invention provides a composition for diagnosing or monitoring a disease mediated by cells expressing CD22, including an antibody that specifically binds to CD22.
본 발명에서는 CD22에 보다 특이적으로 결합하는 항체를 스크리닝하여 신규한 항체 2종(3F11 및 7B5)을 확립하였고, 상기 신규한 항체들이 CD22 항원과 특이적으로 결합하는 것을 확인하였다.In the present invention, two novel antibodies (3F11 and 7B5) were established by screening antibodies that more specifically bind to CD22, and it was confirmed that the novel antibodies specifically bind to the CD22 antigen.
또한, 상기 확립된 항체를 이용하여 제조된 CD22를 표적으로 하는 키메라 항원 수용체(CAR) 및 CAR-T 세포는 CD22와 효과적으로 결합할 뿐만 아니라, CD22와 결합한 CAR-T 세포의 활성화가 이루어진 것을 확인하였다. In addition, it was confirmed that the chimeric antigen receptor (CAR) and CAR-T cells targeting CD22 prepared using the established antibody not only effectively bound to CD22, but also activated the CAR-T cells bound to CD22. .
나아가, 본 발명의 CAR-T 세포는 CD22를 발현하는 세포를 효과적으로 사멸시키는 것을 확인하였으므로, 본 발명의 CD22에 특이적인 항체, CD22를 표적으로 하는 키메라 항원 수용체 및 CAR-T 세포는 CD22 발현과 관련된 질환 예방 또는 치료용 조성물로 유용하게 활용할 수 있다. Furthermore, since it was confirmed that the CAR-T cells of the present invention effectively kill CD22-expressing cells, the CD22-specific antibody, the CD22-targeting chimeric antigen receptor, and CAR-T cells of the present invention are related to CD22 expression. It can be usefully used as a composition for preventing or treating diseases.
도 1은 본 발명에서 선별한 3F11 및 7B5 항체의 CD22에 대한 결합력을 FACS로 확인한 데이터이다.1 is data confirming the binding ability of 3F11 and 7B5 antibodies selected in the present invention to CD22 by FACS.
도 2는 CD22를 표적으로 하는 키메라 항원 수용체(CD22-CAR)를 발현하는 렌티바이러스 벡터 및 T 세포에서 발현된 키메라 항원 수용체를 나타낸 모식도이다. 2 is a schematic diagram showing a lentiviral vector expressing a chimeric antigen receptor (CD22-CAR) targeting CD22 and a chimeric antigen receptor expressed in T cells.
도 3은 CD22-CAR를 발현하는 렌티바이러스를 이용한 CD22-CAR-T 세포 제조방법을 나타낸 모식도이다. 3 is a schematic diagram showing a method for preparing CD22-CAR-T cells using a lentivirus expressing CD22-CAR.
도 4는 3F11 및 7B5 항체 각각을 이용하여 제조한 CD22-CAR-T 세포의 CD22 결합능을 확인한 데이터이다.4 is data confirming the CD22 binding ability of CD22-CAR-T cells prepared using each of 3F11 and 7B5 antibodies.
도 5는 3F11 및 7B5 항체 각각을 이용하여 제조한 CD22-CAR-T 세포의 활성화를 확인하기 위해, 표적 세포의 존재 하에 CD22-CAR-T 세포에 의한 IFNγ 발현 정도를 확인한 데이터이다.5 is data confirming the level of IFNγ expression by CD22-CAR-T cells in the presence of target cells in order to confirm the activation of CD22-CAR-T cells prepared using 3F11 and 7B5 antibodies, respectively.
도 6은 3F11 및 7B5 항체 각각을 이용하여 제조한 CD22-CAR-T 세포에 의한 표적 세포의 사멸효과를 확인한 데이터이다.6 is data confirming the killing effect of target cells by CD22-CAR-T cells prepared using each of 3F11 and 7B5 antibodies.
이하, 본 발명을 상세하게 설명한다. Hereinafter, the present invention will be described in detail.
CD22에 특이적으로 결합하는 항체Antibodies that specifically bind to CD22
본 발명은 일관점에서, CD22에 특이적으로 결합하는 항체 또는 이의 단편으로서,In one aspect, the present invention provides an antibody or fragment thereof that specifically binds to CD22,
(a) 서열번호 1의 아미노산으로 표시되는 CDR1 영역, 서열번호 2의 아미노산으로 표시되는 CDR2 영역 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역, 서열번호 5의 아미노산으로 표시되는 CDR2 영역 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편; 또는(a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and the amino acid of SEQ ID NO: 4 an antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 6; or
(b) 서열번호 11의 아미노산으로 표시되는 CDR1 영역, 서열번호 12의 아미노산으로 표시되는 CDR2 영역 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역, 서열번호 15의 아미노산으로 표시되는 CDR2 영역 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편에 관한 것이다.(b) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 It relates to an antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 16.
본 발명에서, 상기 항체는 단클론 항체(monoclonal antibody)일 수 있다. 본 발명에서, 용어 "단클론 항체(monoclonal antibody)"는 모노클로날 항체 또는 단일클론항체라고도 불리며, 단일 항체 형성세포가 생성하는 항체로, 1차 구조(아미노산 배열)가 균일한 특징이 있다. 오직 하나의 항원 결정기만을 인식하며, 일반적으로 암세포와 항체생산세포를 융합한 하이브리도마(hybridoma cell)을 배양하여 생산되지만, 확보된 항체 유전자 서열을 이용하여 다른 재조합 단백질 발현 숙주세포를 이용하여 생산할 수도 있다. 또한, 상기 항체는 필요에 따라 CDR 부분을 제외한 나머지 부분을 인간화시켜 사용할 수도 있다.In the present invention, the antibody may be a monoclonal antibody. In the present invention, the term "monoclonal antibody" is also called a monoclonal antibody or monoclonal antibody, and is an antibody produced by a single antibody-forming cell, and has a uniform primary structure (amino acid sequence). It recognizes only one antigenic determinant and is generally produced by culturing a hybridoma cell in which cancer cells and antibody-producing cells are fused. can also be produced. In addition, the antibody may be used by humanizing the remaining parts except for the CDR parts, if necessary.
본 발명에서, 용어 "CDR", 즉 "상보성 결정 영역"은 중쇄 및 경쇄 폴리펩타이드 모두의 가변 영역 내에서 발견되는 비근접(non-contiguous) 항원 결합 부위를 의미하는 것이다.As used herein, the term "CDR", ie, "complementarity determining region", refers to a non-contiguous antigen binding site found within the variable region of both heavy and light chain polypeptides.
본 발명에서, 용어 "인간화 항체"는 인간에 의해 생산된 항체의 것에 상응하는 아미노산 서열을 소유하고 및/또는 본원에서 개시된 바와 같은 인간 항체를 만들기 위한 기술 중에서 한 가지를 이용하여 만들어진 항체이다. 인간화 항체의 이러한 정의는 비인간 항원 결합 잔기를 포함하는 인간화 항체를 특정적으로 배제한다.As used herein, the term "humanized antibody" is an antibody that possesses an amino acid sequence corresponding to that of an antibody produced by a human and/or is made using one of the techniques for making human antibodies as disclosed herein. This definition of a humanized antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
본 발명에서, 용어 "항체"는 2개의 전체 길이의 경쇄 및 2개의 전체 길이의 중쇄를 가지는 완전한 형태뿐만 아니라 항체 분자의 단편도 사용될 수 있다. 항체 분자의 단편이란 적어도 펩타이드 태그(에피토프) 결합 기능을 보유하고 있는 단편을 뜻하며 scFv, Fab, F(ab'), F(ab') 2,단일 도메인(single domain) 등을 포함한다. In the present invention, the term “antibody” can be used not only in a complete form having two full-length light chains and two full-length heavy chains, but also as a fragment of an antibody molecule. A fragment of an antibody molecule refers to a fragment having at least a peptide tag (epitope) binding function, and includes scFv, Fab, F(ab'), F(ab') 2 , a single domain, and the like.
항체 단편 중 Fab는 경쇄 및 중쇄의 가변영역과 경쇄의 불변 영역 및 중쇄의 첫 번째 불변 영역(CH1)을 가지는 구조로 1개의 항원 결합 부위를 가진다. Fab'는 중쇄 CH1 도메인의 C 말단에 하나 이상의 시스테인 잔기를 포함하는 힌지 부위(hinge region)를 가진다는 점에서 Fab와 차이가 있다. F(ab')2 항체는 Fab'의 힌지 부위의 시스테인 잔기가 디설파이드 결합을 이루면서 생성되며, Fv는 중쇄 가변부위 및 경쇄 가변부위만을 가지고 있는 최소의 항체조각이다. 이중쇄 Fv(dsFv)는 디설파이드 결합으로 중쇄 가변부위와 경쇄 가변부위가 연결되어 있고 단쇄 Fv(scFv)는 일반적으로 펩타이드 링커를 통하여 중쇄의 가변 영역과 경쇄의 가변 영역이 공유 결합으로 연결되어 있다. 이러한 항체 단편은 단백질 가수 분해 효소를 이용해서 얻을 수 있고(예를 들어, 전체 항체를 파파인으로 제한 절단하면 Fab를 얻을 수 있고 펩신으로 절단하면 F(ab')2 단편을 얻을 수 있다), 바람직하게는 유전자 재조합 기술을 통하여 제작할 수 있다. Among the antibody fragments, Fab has a structure having variable regions of light and heavy chains, a constant region of a light chain and a first constant region (CH1) of a heavy chain, and has one antigen-binding site. Fab' differs from Fab in that it has a hinge region comprising one or more cysteine residues at the C terminus of the heavy chain CH1 domain. The F(ab')2 antibody is generated by forming a disulfide bond between the cysteine residues of the hinge region of Fab', and Fv is the smallest antibody fragment having only a heavy chain variable region and a light chain variable region. In a double chain Fv (dsFv), the heavy chain variable region and the light chain variable region are linked by a disulfide bond, and in a single chain Fv (scFv), the heavy chain variable region and the light chain variable region are covalently linked via a peptide linker. Such antibody fragments can be obtained using proteolytic enzymes (for example, by restriction digestion of the entire antibody with papain to obtain Fab, and by digestion with pepsin to obtain F(ab')2 fragments), preferably It can be produced through genetic recombination technology.
본 발명의 CD22에 특이적으로 결합하는 단클론 항체는 CD22 단백질 전체 또는 일부 펩타이드를 면역원(또는 항원)으로 이용하여 제조할 수 있다. 보다 상세하게는, 우선 면역원으로서 CD22, CD22 단백질을 포함하는 융합 단백질 또는 CD22 단백질을 포함하는 캐리어(carrier)를 필요에 따라서 면역증강제인 아주반트(adjuvant)(예, Freund adjuvant)와 함께 인간을 제외한 포유동물의 피하, 근육, 정맥, 발볼록살 또는 복강 내에 1회 내지 그 이상 주사하는 것으로써 면역감작(immunization)을 시킨다. 상기 인간을 제외한 포유동물은 바람직하게는, 마우스, 래트, 햄스터, 몰모트, 닭, 토끼, 고양이, 개, 돼지, 염소, 양, 당나귀, 말 또는 소(인간 항체를 생산하는 형질 전환(transgenic) 마우스와 같은 다른 동물 유래의 항체를 생산하도록 조작된 형질 전환(transgenic) 동물을 포함한다.)이며, 보다 바람직하게는, 마우스, 래트, 햄스터, 몰모트, 닭 또는 토끼이다. 첫 번째 면역으로부터 약 1~21일 마다 1~4회 면역을 실시하여, 최종 면역으로부터 약 1~10일 후에 면역 감작 된 포유동물로부터 항체 생산하는 세포를 수득할 수 있다. 면역을 시키는 회수 및 시간적 간격은 사용하는 면역원의 특징 등에 의하여 적당히 변경할 수 있다.The monoclonal antibody that specifically binds to CD22 of the present invention can be prepared by using all or part of the CD22 protein as an immunogen (or antigen). More specifically, first, as an immunogen, CD22, a fusion protein containing CD22 protein, or a carrier containing CD22 protein, if necessary, together with an adjuvant (eg, Freund adjuvant) as an adjuvant (eg, Freund adjuvant) except for humans Immunization is achieved by subcutaneous, intramuscular, intravenous, intraperitoneal or one or more injections in mammals. The mammals other than humans are preferably mice, rats, hamsters, malmots, chickens, rabbits, cats, dogs, pigs, goats, sheep, donkeys, horses or cattle (transgenic mice that produce human antibodies) (including transgenic animals engineered to produce antibodies from other animals such as From the first immunization, immunization is performed 1 to 4 times every 1 to 21 days, and antibody-producing cells can be obtained from the immune-sensitized mammal about 1 to 10 days after the final immunization. The number of times and time intervals for immunization can be appropriately changed depending on the characteristics of the immunogen to be used, and the like.
단클론 항체를 분비하는 하이브리도마(hybridoma)의 제조는 케이라 및 미르슈타인 등의 방법(Nature, 1975, Vol. 256, p. 495-497) 및 이에 준하는 방법에 따라 실시할 수 있다. 상기와 같이 면역 감작된 인간을 제외한 동물로부터 채취한 비장, 림프절, 골수 또는 편도로 이루어지는 군으로부터 선택되는 어느 하나, 바람직하게는 비장에 포함되는 항체 생산하는 세포와 자가 항체 생산 능력이 없는 포유동물 유래의 골수종 세포(myeloma cells)를 세포 융합시키는 것에 의해 하이브리도마(hybridoma)를 제조할 수 있다. 상기 포유동물은 마우스, 래트, 몰모트, 햄스터, 닭, 토끼 또는 인간일 수 있고, 바람직하게는 마우스, 래트, 닭 또는 인간일 수 있다.Preparation of a hybridoma secreting a monoclonal antibody can be carried out according to the method of Keira and Mirstein et al. (Nature, 1975, Vol. 256, p. 495-497) and a method similar thereto. Any one selected from the group consisting of spleen, lymph node, bone marrow or tonsils collected from animals other than humans who have been immunosensitized as described above, preferably derived from the antibody-producing cells contained in the spleen, and from mammals without autoantibody-producing ability Hybridomas can be prepared by cell fusing of myeloma cells. The mammal may be a mouse, a rat, a guinea pig, a hamster, a chicken, a rabbit or a human, preferably a mouse, a rat, a chicken or a human.
세포 융합은, 예를 들면, 폴리에틸렌글리콜이나 센다이 바이러스를 비롯한 융합 촉진제나 전기 펄스에 의한 방법이 이용되고, 예를 들면, 융합 촉진제를 함유하는 융합 배지에 항체 생산 세포와 무한 증식 가능한 포유류 유래의 세포를 약 1:1 내지 1:10의 비율로 부유시켜, 이 상태로, 약 30 내지 40℃로 약 1 내지 5분간 배양한다. 융합 배지에는, 예를 들면, MEM 배지, RPMI1640 배지 및 이스코브 변형 둘베코 배지(Iscove's Modified Dulbecco's Medium)를 비롯한 통상의 일반적인 것을 이용하면 좋고, 소 혈청 등의 혈청류는 제외해 두는 것이 바람직하다.For cell fusion, for example, a fusion promoter such as polyethylene glycol or Sendai virus, or a method by electric pulse is used, for example, an antibody-producing cell and a mammalian-derived cell capable of immortal growth in a fusion medium containing a fusion promoter. is suspended at a ratio of about 1:1 to 1:10, and in this state, incubated at about 30 to 40° C. for about 1 to 5 minutes. As the fusion medium, for example, MEM medium, RPMI1640 medium and Iscove's Modified Dulbecco's Medium may be used, and it is preferable to exclude sera such as bovine serum.
상기 단클론 항체를 생산하는 하이브리도마 클론을 스크리닝하는 방법은 우선, 상기한 바와 같이 획득한 융합 세포를 HAT 배지 등의 선택용 배지에 옮기고, 약 30 내지 40℃로 약 3일 내지 3주일 배양해서 하이브리도마 이외의 세포를 사멸시킨다. 이어서, 마이크로타이터 플레이트(microtiter plate) 등에서 하이브리도마를 배양한 후, 위에서 기술한 인간을 제외한 동물의 면역반응에 사용한 면역원과 배양 상청액과의 반응성이 증가된 부분을 RIA(radioactive substance-marked immuno antibody) 또는 ELISA(Enzyme-Linked Immunosorbent Assay)같은 면역분석방법을 통하여 찾는 방법을 통해 수행할 수 있다. 그리고 상기에서 찾은 단클론 항체를 생산하는 클론은 상기 면역원에 대하여 특이적인 결합력을 보여준다.In the method of screening hybridoma clones producing the monoclonal antibody, first, the fusion cells obtained as described above are transferred to a selection medium such as HAT medium, and cultured at about 30 to 40° C. for about 3 days to 3 weeks. It kills cells other than hybridomas. Then, after culturing the hybridomas on a microtiter plate, etc., the immunogen used for the above-described immune response of animals other than humans and the part with increased reactivity with the culture supernatant were subjected to radioactive substance-marked immunotherapy (RIA). antibody) or ELISA (Enzyme-Linked Immunosorbent Assay). And the clone producing the monoclonal antibody found above shows a specific binding ability to the immunogen.
본 발명의 단클론 항체는, 이와 같은 하이브리도마를 생체 내외에서 배양함으로써 얻을 수 있다. 배양에는, 포유동물 유래의 세포를 배양하기 위한 통상의 방법이 이용되며, 배양물 등으로부터 단클론 항체를 채취하기 위해서는, 항체 일반을 정제하기 위한 이 분야에서의 통상의 방법이 이용된다. 각각의 방법으로서는, 예를 들면, 염석(鹽析), 투석, 여과, 농축, 원심분리, 분별 침전, 겔 여과 크로마토그래피, 이온 교환 크로마토그래피, 어피니티 크로마토그래피, 고속액체 크로마토그래피, 겔 전기영동 및 등전점 전기영동 등을 들 수 있고, 이들은 필요에 따라서 조합해서 적용된다. 정제한 단클론 항체는, 그 후, 농축, 건조하여, 용도에 따라서 액상 또는 고상으로 한다.The monoclonal antibody of the present invention can be obtained by culturing such a hybridoma in vitro or in vivo. For culture, a conventional method for culturing cells derived from mammals is used, and for collecting monoclonal antibodies from a culture or the like, a conventional method in this field for purifying antibodies in general is used. Examples of each method include salting out, dialysis, filtration, concentration, centrifugation, fractional precipitation, gel filtration chromatography, ion exchange chromatography, affinity chromatography, high-performance liquid chromatography, and gel electrophoresis. and isoelectric point electrophoresis, and these are applied in combination as needed. The purified monoclonal antibody is then concentrated and dried to obtain a liquid or solid state depending on the intended use.
또한, 본 발명의 단클론 항체는, 중쇄(重鎖) 및 경쇄(輕鎖) 가변영역을 코딩하는 DNA를 각각, 중쇄 및 경쇄의 정상영역을 코딩하는 기지의 DNA(예를 들면, 일본 2007-252372호 공보 참조)와 각각 연결한 유전자를, PCR법, 또는, 화학 합성에 의해 합성하고, 그 유전자의 발현을 가능하게 하는 공지의 발현 벡터(pcDNA 3.1(Invitrogen 사 판매) 등에 이식하여 형질 전환체를 제조하고, CHO 세포나 대장균 등의 숙주 중에서 발현시킴으로써 항체를 생산하고, 이러한 배양액으로부터, 프로테인 A 또는 G(Protein A 또는 G) 컬럼 등을 이용해서 항체를 정제함으로써 얻을 수 있다.In addition, the monoclonal antibody of the present invention comprises DNAs encoding heavy chain and light chain variable regions, respectively, with known DNA encoding the constant regions of heavy chain and light chain (e.g., Japan 2007-252372). No. publication) and each ligated gene is synthesized by PCR or chemical synthesis, and the transformant is obtained by transplanting a known expression vector (pcDNA 3.1 (sold by Invitrogen), etc.) that enables expression of the gene. It can be obtained by preparing an antibody and expressing it in a host such as CHO cells or Escherichia coli, and purifying the antibody from this culture solution using a protein A or G (Protein A or G) column or the like.
본 발명의 구체적인 일실시예에서는, CD22에 특이적으로 결합하는 항체를 제조하기 위해, 항-CD22 항체를 생산하는 하이브리도마를 제조 및 스크리닝 하여, CD22에 특이적으로 결합하는 항체(scFv) 2종을 선별하였으며, 이를 각각 3F11 및 7B5로 명명하였다. In a specific embodiment of the present invention, in order to prepare an antibody that specifically binds to CD22, a hybridoma producing an anti-CD22 antibody is prepared and screened, and an antibody (scFv) that specifically binds to CD22 (scFv) 2 Species were selected and designated as 3F11 and 7B5, respectively.
(a) 상기 3F11 항체는 서열번호 1의 아미노산으로 표시되는 CDR1 영역(GFTFSNYW), 서열번호 2의 아미노산으로 표시되는 CDR2 영역(IRLKSNNYAT) 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역(TSIYYYGRDYAMDY)을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역(SSVSSSY), 서열번호 5의 아미노산으로 표시되는 CDR2 영역(STS) 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역(HQYHRSPYT)을 포함하는 경쇄 가변 부위로 구성되는 것을 확인하였다. (a) the 3F11 antibody comprises a CDR1 region represented by the amino acid of SEQ ID NO: 1 (GFTFSNYW), a CDR2 region represented by the amino acid of SEQ ID NO: 2 (IRLKSNNYAT) and a CDR3 region represented by the amino acid of SEQ ID NO: 3 (TSIYYYGRDYAMDY) A light chain comprising a heavy chain variable region and a CDR1 region represented by the amino acid of SEQ ID NO: 4 (SSVSSSY), a CDR2 region represented by the amino acid of SEQ ID NO: 5 (STS) and a CDR3 region represented by the amino acid of SEQ ID NO: 6 (HQYHRSPYT) It was confirmed that it consists of a variable region.
구체적으로, 3F11 항체는 서열번호 7의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 8의 아미노산으로 표시되는 경쇄 가변부위로 구성되며, 상기 중쇄 가변 부위는 서열번호 9의 염기서열로, 경쇄 가변 부위는 서열번호 10의 염기서열로 코딩되는 것을 확인하였다. Specifically, the 3F11 antibody is composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 7 and a light chain variable region represented by the amino acid of SEQ ID NO: 8, wherein the heavy chain variable region is the nucleotide sequence of SEQ ID NO: 9, and the light chain variable region is It was confirmed that it is encoded by the nucleotide sequence of SEQ ID NO: 10.
(b) 상기 7B5 항체는 서열번호 11의 아미노산으로 표시되는 CDR1 영역(GFSLTRYD), 서열번호 12의 아미노산으로 표시되는 CDR2 영역(MWTGGGT) 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역(VRSYYGSAMDY)을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역(DHINNW), 서열번호 15의 아미노산으로 표시되는 CDR2 영역(GAT) 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역(QQHWSTPFT)을 포함하는 경쇄 가변 부위로 구성되는 것을 확인하였다. (b) the 7B5 antibody comprises a CDR1 region represented by the amino acid of SEQ ID NO: 11 (GFSLTRYD), a CDR2 region represented by the amino acid of SEQ ID NO: 12 (MWTGGGT) and a CDR3 region represented by the amino acid of SEQ ID NO: 13 (VRSYYGSAMDY) A light chain comprising a heavy chain variable region and a CDR1 region represented by the amino acid of SEQ ID NO: 14 (DHINNW), a CDR2 region represented by the amino acid of SEQ ID NO: 15 (GAT) and a CDR3 region represented by the amino acid of SEQ ID NO: 16 (QQHWSTPFT) It was confirmed that it consists of a variable region.
구체적으로, 7B5 항체는 서열번호 17의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 18의 아미노산으로 표시되는 경쇄 가변부위로 구성되며, 상기 중쇄 가변 부위는 서열번호 19의 염기서열로, 경쇄 가변 부위는 서열번호 20의 염기서열로 코딩되는 것을 확인하였다. Specifically, the 7B5 antibody is composed of a heavy chain variable region represented by the amino acid of SEQ ID NO: 17 and a light chain variable region represented by the amino acid of SEQ ID NO: 18, the heavy chain variable region is the nucleotide sequence of SEQ ID NO: 19, and the light chain variable region is It was confirmed that it is encoded by the nucleotide sequence of SEQ ID NO: 20.
본 발명의 CD22에 특이적인 항체는 바람직하게 scFv(single chain variable fragment)로, 중쇄 가변 부위 및 경쇄 가변 부위가 링커로 연결될 수 있도록 유전자 재조합 기술을 통하여 제작할 수 있다. 상기 링커는 바람직하게 바람직하게 서열번호 21의 아미노산 서열 또는 서열번호 22의 염기서열로 표시될 수 있으나, 이에 한정되지는 않는다.The CD22-specific antibody of the present invention is preferably scFv (single chain variable fragment), and can be produced through genetic recombination technology so that the heavy chain variable region and the light chain variable region can be linked with a linker. The linker may preferably be represented by the amino acid sequence of SEQ ID NO: 21 or the nucleotide sequence of SEQ ID NO: 22, but is not limited thereto.
경쇄 가변부위-링커-중쇄 가변부위로 연결된 경우 3F11 항체는 서열번호 23의 아미노산 서열 또는 서열번호 24의 염기서열로, 7B5 항체는 서열번호 25의 아미노산 서열 또는 서열번호 26의 염기서열로 표시될 수 있다.When linked by a light chain variable region-linker-heavy chain variable region, the 3F11 antibody may be represented by the amino acid sequence of SEQ ID NO: 23 or the nucleotide sequence of SEQ ID NO: 24, and the 7B5 antibody may be represented by the amino acid sequence of SEQ ID NO: 25 or the nucleotide sequence of SEQ ID NO: 26 have.
본 발명은 다른 관점에서, 상기 CD22에 특이적으로 결합하는 항체를 코딩하는 폴리뉴클레오타이드에 관한 것이다.In another aspect, the present invention relates to a polynucleotide encoding the antibody that specifically binds to CD22.
본 발명에서, 용어 "폴리뉴클레오타이드"는 일반적으로 임의의 길이로 분리된 핵산 분자(nucleic acid molecule), 데옥시리보뉴클레오티드 또는 리보뉴클레오티드, 또는 그의 유사체를 지칭한다. 일부 구현예에서, 본 발명의 폴리뉴클레오타이드는 (1) 중합효소 연쇄반응(PCR) 증폭과 같은 in-vitro 증폭; (2) 클로닝 및 재조합; (3) 절단(digestion) 및 겔 전기영동 분리와 같은 정제; (4) 화학 합성과 같은 합성을 통해 제조될 수 있으며, 바람직하게 분리된 폴리뉴클레오타이드는 재조합 DNA 기술에 의해 제조된다. 본 발명에서, 항체 또는 이의 항원 결합 단편을 코딩하기 위한 핵산은 합성 올리고뉴클레오티드의 제한 단편 조작(restriction fragment operation) 또는 SOE PCR의 적용을 포함하지만 이에 제한하지 않고, 당업계에 공지된 다양한 방법에 의해 제조될 수 있다.As used herein, the term "polynucleotide" generally refers to a nucleic acid molecule, deoxyribonucleotide or ribonucleotide, or an analog thereof, separated by any length. In some embodiments, the polynucleotides of the present invention are administered by (1) in-vitro amplification, such as polymerase chain reaction (PCR) amplification; (2) cloning and recombination; (3) purification such as digestion and gel electrophoretic separation; (4) It may be prepared through synthesis such as chemical synthesis, and preferably, the isolated polynucleotide is prepared by recombinant DNA technology. In the present invention, the nucleic acid for encoding the antibody or antigen-binding fragment thereof can be prepared by various methods known in the art, including, but not limited to, restriction fragment operation of synthetic oligonucleotides or application of SOE PCR. can be manufactured.
본 발명은 또 다른 관점에서, 상기 CD22에 특이적으로 결합하는 항체를 코딩하는 폴리뉴클레오타이드 포함하는 벡터, 및 상기 벡터로 형질전환된 재조합 세포에 관한 것이다.In another aspect, the present invention relates to a vector comprising a polynucleotide encoding an antibody that specifically binds to CD22, and a recombinant cell transformed with the vector.
본 발명에서, 용어 "벡터(expression vector)"는 적당한 숙주세포 내에서 목적 유전자가 발현할 수 있도록 프로모터 등의 필수적인 조절 요소를 포함하는 유전자 제조물이다. 벡터는 플라스미드, 레트로바이러스(retroviral) 벡터 및 렌티바이러스(lentiviral) 벡터 중 하나 이상으로부터 선택될 수 있다. 적당한 숙주로 형질전환되면, 벡터는 숙주 게놈과 무관하게 복제하고 기능할 수 있거나, 또는 일부 경우에 게놈 그 자체에 통합될 수 있다. In the present invention, the term "vector (expression vector)" refers to a gene preparation including essential regulatory elements such as a promoter so that a target gene can be expressed in an appropriate host cell. The vector may be selected from one or more of a plasmid, a retroviral vector and a lentiviral vector. Upon transformation into an appropriate host, the vector may replicate and function independently of the host genome, or in some cases may be integrated into the genome itself.
또한, 벡터는 코딩 영역이 적합한 숙주에서 정확하게 발현될 수 있게 하는 발현 제어 요소를 포함할 수 있다. 이러한 조절 요소는 당업자에게 잘 알려져 있으며, 예를 들어 프로모터, 리보솜 결합 부위(ribosome-binding site), 인핸서(enhancer) 및 유전자 전사(transcription) 또는 mRNA 번역(translation)을 조절하기 위한 다른 조절 요소를 포함할 수 있다. 발현 조절 서열의 특정 구조는 종 또는 세포 유형의 기능에 따라 달라질 수 있으나, 일반적으로 TATA 박스(box), 캡핑된(capped) 서열, CAAT 서열 등과 같은 전사 개시 및 번역 개시에 각각 참여하는 5' 비-전사 서열, 및 5' 또는 3' 비-번역 서열을 함유한다. 예를 들어, 5' 비-전사 발현 조절 서열은 기능적으로 연결된 핵산을 전사 및 조절하기 위한 프로모터 서열을 포함할 수 있는 프로모터 영역을 포함할 수 있다. In addition, the vector may contain expression control elements that allow the coding region to be accurately expressed in a suitable host. Such regulatory elements are well known to those skilled in the art and include, for example, promoters, ribosome-binding sites, enhancers and other regulatory elements for regulating gene transcription or mRNA translation. can do. The specific structure of the expression control sequence may vary depending on the function of the species or cell type, but generally 5' ratios participating in transcription initiation and translation initiation, respectively, such as TATA box, capped sequence, CAAT sequence, etc. - contains a transcribed sequence, and a 5' or 3' non-translated sequence. For example, a 5' non-transcriptional expression control sequence may include a promoter region that may include a promoter sequence for transcription and control of a functionally linked nucleic acid.
본 발명에서, 용어 "프로모터"는 전사를 지시하기에 충분한 최소 서열을 의미한다. 또한, 세포 유형 특이적 또는 외부의 신호 또는 제제에 의해 유도되는 조절 가능한 프로모터 의존적 유전자를 발현하도록 하는 데 충분한 프로모터 구성이 포함될 수 있으며, 이러한 구성들은 유전자의 5' 또는 3' 부분에 위치할 수 있다. 보존적 프로모터 및 유도적 프로모터 둘 다 포함된다. 프로모터 서열은 원핵생물, 진핵생물 또는 바이러스로부터 유래될 수 있다.As used herein, the term "promoter" means a minimal sequence sufficient to direct transcription. In addition, promoter constructs sufficient to allow expression of a regulatable promoter-dependent gene induced by cell type-specific or external signals or agents may be included, and these constructs may be located in the 5' or 3' portion of the gene. . Both conservative and inducible promoters are included. Promoter sequences may be derived from prokaryotes, eukaryotes or viruses.
본 발명에서, 용어 "형질전환체"는 하나 이상의 목적 단백질을 암호화하는 폴리뉴클레오타이드를 갖는 벡터가 숙주세포에 도입되어 형질전환된 세포를 의미하고, 발현 벡터를 숙주세포에 도입하여 형질전환체를 제조하기 위한 방법으로는 문헌(Sambrook, J., et al., Molecular Cloning, A Laboratory Manual(2판), Cold Spring Harbor Laboratory, 1. 74, 1989)에 기재된 인산칼슘법 또는 염화캄슘/염화루비듐법, 일렉트로포레이션법(electroporation), 전기주입법(electroinjection), PEG 등의 화학적 처리방법, 유전자 총(gene gun) 등을 이용하는 방법 등이 있다. In the present invention, the term "transformant" refers to a cell transformed by introducing a vector having a polynucleotide encoding one or more target proteins into a host cell, and introducing the expression vector into the host cell to prepare a transformant As a method for this, the calcium phosphate method or the calcium chloride/rubidium chloride method described in the literature (Sambrook, J., et al., Molecular Cloning, A Laboratory Manual (2nd edition), Cold Spring Harbor Laboratory, 1. 74, 1989) , an electroporation method, an electroinjection method, a chemical treatment method such as PEG, a method using a gene gun, or the like.
상기 벡터가 발현되는 형질전환체를 영양배지에서 배양하면 항체 단백질을 대량으로 제조, 분리 가능하다. 배지와 배양조건은 숙주 세포에 따라 관용되는 것을 적절히 선택하여 이용할 수 있다. 배양시 세포의 생육과 단백질의 대량 생산에 적합하도록 온도, 배지의 pH 및 배양시간 등의 조건들을 적절하게 조절하여야 한다. When the transformant expressing the vector is cultured in a nutrient medium, it is possible to manufacture and isolate antibody proteins in large quantities. Medium and culture conditions can be appropriately selected and used depending on the host cell. In culture, conditions such as temperature, medium pH, and culture time should be appropriately adjusted to be suitable for cell growth and mass production of protein.
본 발명에 따른 벡터는 항체의 생산을 위해 숙주세포, 바람직하게는 포유동물 세포에 형질전환 시킬 수 있다. 완벽한 글리코실화된 단백질을 발현할 수 있는 적합한 숙주 세포주의 수는 당해 분야에서 개발되어 왔으며 COS-1(예를 들면, ATCC CRL 1650), COS-7(예를 들면, ATCC CRL-1651), HEK293, BHK21(예를 들면, ATCC CRL-10), CHO(예를 들면, ATCC CRL 1610) 및 BSC-1(예를 들면, ATCC CRL-26) 세포주, Cos-7 세포, CHO 세포, hep G2 세포, P3X63Ag8653, SP2/0-Agl4, 293 세포, HeLa 세포 등을 포함하며, 이들 세포는 예를 들면, ATCC(American Type Culture Collection, 미국)으로부터 용이하게 이용가능하다. 바람직한 숙주 세포는 흑색종 및 림프종 세포와 같은 림프구 기원의 세포를 포함한다.The vector according to the present invention can be transformed into a host cell, preferably a mammalian cell, for the production of an antibody. A number of suitable host cell lines capable of expressing fully glycosylated proteins have been developed in the art and include COS-1 (eg ATCC CRL 1650), COS-7 (eg ATCC CRL-1651), HEK293 , BHK21 (eg ATCC CRL-10), CHO (eg ATCC CRL 1610) and BSC-1 (eg ATCC CRL-26) cell lines, Cos-7 cells, CHO cells, hep G2 cells , P3X63Ag8653, SP2/0-Agl4, 293 cells, HeLa cells, etc., which are readily available from, for example, the American Type Culture Collection (ATCC, USA). Preferred host cells include cells of lymphocyte origin, such as melanoma and lymphoma cells.
CD22를 표적으로 하는 키메라항원 수용체(Chimeric antigen receptor)Chimeric antigen receptor targeting CD22
본 발명은 다른 관점에서, The present invention from another point of view,
CD22-결합 도메인; CD22-binding domain;
막관통 도메인(transmembrane domain); transmembrane domain;
공동자극 도메인(costimulatory domain); 및 costimulatory domain; and
세포 내 신호전달 도메인(intracellular signal transduction domain)을 포함하는 키메릭 항원 수용체(chimeric antigen receptor: CAR)로,A chimeric antigen receptor (CAR) comprising an intracellular signal transduction domain,
상기 CD22-결합 도메인은 (a) 서열번호 1의 아미노산으로 표시되는 CDR1 영역, 서열번호 2의 아미노산으로 표시되는 CDR2 영역 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역, 서열번호 5의 아미노산으로 표시되는 CDR2 영역 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편; 또는The CD22-binding domain comprises (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and SEQ ID NO: An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region represented by the amino acid of 4, a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid of SEQ ID NO: 6. ; or
(b) 서열번호 11의 아미노산으로 표시되는 CDR1 영역, 서열번호 12의 아미노산으로 표시되는 CDR2 영역 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역, 서열번호 15의 아미노산으로 표시되는 CDR2 영역 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편이다.(b) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid sequence of SEQ ID NO: 16.
본 발명에서, 용어 "키메릭 항원 수용체 (CAR)"는 일반적으로 항원 및 하나 이상의 세포 내 도메인과 결합하는 능력을 갖는 세포 외 도메인을 함유하는 융합 단백질을 지칭한다. CAR는 키메릭 항원 수용체 T 세포(CAR-T)의 핵심 부분이며, 항원 결합 도메인, 막 관통 도메인, 공동 자극 도메인 및 세포 내 신호전달 도메인을 포함할 수 있다. CAR는 항체의 항원(예를 들어, CD22) 특이성에 기초하여 T 세포 수용체-활성화 세포 내 도메인과 조합될 수 있다. 유전자가 변형된 CAR-발현 T 세포는 표적 항원-발현 악성 세포를 특이적으로 식별하고 제거할 수 있다. As used herein, the term “chimeric antigen receptor (CAR)” generally refers to a fusion protein containing an antigen and an extracellular domain having the ability to bind one or more intracellular domains. A CAR is a key part of a chimeric antigen receptor T cell (CAR-T) and may include an antigen binding domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain. A CAR can be combined with a T cell receptor-activating intracellular domain based on the antigen (eg, CD22) specificity of the antibody. Genetically modified CAR-expressing T cells can specifically identify and eliminate target antigen-expressing malignant cells.
본 발명에서, 용어 "CD22-결합 도메인(CD22-binding domain)"은 일반적으로 CD22 단백질에 특이적으로 결합할 수 있는 도메인을 지칭한다. 예를 들어, CD22-결합 도메인은 B 세포에서 발현된 인간 CD22 폴리펩타이드 또는 이의 단편에 특이적으로 결합할 수 있는 항-CD22 항체 또는 이의 단편을 함유할 수 있다. In the present invention, the term "CD22-binding domain" generally refers to a domain capable of specifically binding to a CD22 protein. For example, the CD22-binding domain may contain an anti-CD22 antibody or fragment thereof capable of specifically binding to a human CD22 polypeptide or fragment thereof expressed in a B cell.
본 발명에서, 용어 "결합 도메인(binding domain)"은 "세포 외 도메인(extracellular domain)", "세포 외 결합 도메인(extracellular binding domain)", "항원-특이적 결합 도메인(antigenspecific binding domain)" 및 "세포 외 항원-특이적 결합 도메인(extracellular antigen-specific biding domain)"은 상호 교환적으로 사용될 수 있으며, 표적 항원(예를 들어, CD22)에 특이적으로 결합하는 능력을 갖는 CAR 도메인 또는 단편을 지칭한다. In the present invention, the term "binding domain" refers to "extracellular domain", "extracellular binding domain", "antigen-specific binding domain" and "Extracellular antigen-specific bidding domain" can be used interchangeably and refers to a CAR domain or fragment having the ability to specifically bind a target antigen (eg, CD22). refers to
본 발명에 있어서, 상기 항-CD22 항체 또는 이의 단편은 상술한 항-CD22 항체로, 단클론 항체(monoclonal antibody), 바람직하게는 scFv(single chain variable fragment)이다. 구체적으로, 본 발명의 CD22에 특이적인 3F11 및 7B5 항체를 이용하여 제조할 수 있다.In the present invention, the anti-CD22 antibody or fragment thereof is the above-described anti-CD22 antibody, a monoclonal antibody, preferably a single chain variable fragment (scFv). Specifically, it can be prepared using 3F11 and 7B5 antibodies specific for CD22 of the present invention.
본 발명에 있어서, CD22-결합 도메인의 N 말단에 신호 펩타이드(signal peptide)를 추가로 포함할 수 있으며, 상기 "신호 펩타이드(signal peptide)"는 일반적으로 단백질 전달을 안내하기 위한 펩타이드 사슬을 지칭한다. 신호 펩타이드는 5 내지 30 개의 아미노산 길이를 갖는 짧은 펩타이드일 수 있으며, 본 발명에서는 바람직하게 서열번호 34의 아미노산 서열을 이용하였다.In the present invention, a signal peptide may be further included at the N-terminus of the CD22-binding domain, and the "signal peptide" generally refers to a peptide chain for guiding protein transduction. . The signal peptide may be a short peptide having a length of 5 to 30 amino acids, and the amino acid sequence of SEQ ID NO: 34 is preferably used in the present invention.
본 발명에 있어서, CD22-결합 도메인의 C 말단 및 막관통 도메인의 N 말단 사이에 위치된 힌지 부위(hinge region)를 추가로 포함할 수 있으며, 상기 힌지 부위는 CD8α 유래로, 바람직하게는 서열번호 35의 아미노산 서열로 표시될 수 있다. 상기 "힌지 부위(hinge region)"는 일반적으로 항원-결합 영역과 면역 세포 Fc 수용체 (FcR)-결합영역 사이의 연결 영역을 지칭한다.In the present invention, it may further comprise a hinge region located between the C terminus of the CD22-binding domain and the N terminus of the transmembrane domain, wherein the hinge region is derived from CD8α, preferably SEQ ID NO: It can be represented by the amino acid sequence of 35. The "hinge region" generally refers to the linking region between the antigen-binding region and the immune cell Fc receptor (FcR)-binding region.
본 발명에 있어서, "막관통 도메인(transmembrane domain)"은 일반적으로 세포막을 통과하고 세포 내 신호전달 도메인에 연결되어 신호전달의 역할을 하는 CAR의 도메인을 지칭한다. 상기 막관통 도메인은 CD8α, CD4, CD28, CD137, CD80, CD86, CD152 및 PD1로 구성된 군에서 선택되는 단백질로부터 유래될 수 있으며, 바람직하게는 서열번호 36의 아미노산 서열로 표시될 수 있다. In the present invention, "transmembrane domain" generally refers to a domain of a CAR that passes through a cell membrane and is connected to an intracellular signaling domain to play a signaling role. The transmembrane domain may be derived from a protein selected from the group consisting of CD8α, CD4, CD28, CD137, CD80, CD86, CD152 and PD1, and may preferably be represented by the amino acid sequence of SEQ ID NO: 36.
본 발명에 있어서, "공동 자극 도메인(costimulatory domain)"은 일반적으로 림프구의 항원에 대한 효과적인 반응에 필요한 세포 표면 분자인 면역 자극 분자를 제공할 수 있는 세포 내 도메인을 지칭한다. 상기 기재된 공동자극 도메인(costimulatory domain)은 CD28의 공동 자극 도메인을 포함할 수 있고, OX40 및 4-1BB의 공동 자극 도메인과 같은 TNF 수용체 패밀리의 공동 자극 도메인을 포함할 수 있으며, 바람직하게는 서열번호 37의 아미노산 서열로 표시되는 4-1BB일 수 있다.In the present invention, "costimulatory domain" generally refers to an intracellular domain capable of providing immune-stimulatory molecules, which are cell surface molecules necessary for an effective response of lymphocytes to antigens. The costimulatory domain described above may comprise a costimulatory domain of CD28, and may comprise a costimulatory domain of the TNF receptor family such as the costimulatory domain of OX40 and 4-1BB, preferably SEQ ID NO: It may be 4-1BB represented by the amino acid sequence of 37.
본 발명에 있어서, "세포 내 신호전달 도메인(intracellular signal transduction domain)"은 일반적으로 세포 내부에 위치하고 신호를 전달할 수 있는 도메인을 지칭한다. 본 발명에서, 세포 내 신호전달 도메인은 키메라 항원 수용체의 세포 내 신호전달 도메인이다. 예를 들어, 세포 내 신호전달 도메인은 CD3ζ 세포 내 도메인, CD28 세포 내 도메인, CD28 세포 내 도메인, 4-1BB 세포 내 도메인 및 OX40 세포 내 도메인으로부터 선택될 수 있으며, 바람직하게는 서열번호 38의 아미노산 서열로 표시되는 CD3ζ일 수 있다.In the present invention, "intracellular signal transduction domain" generally refers to a domain located inside a cell and capable of transmitting a signal. In the present invention, the intracellular signaling domain is the intracellular signaling domain of the chimeric antigen receptor. For example, the intracellular signaling domain may be selected from CD3ζ intracellular domain, CD28 intracellular domain, CD28 intracellular domain, 4-1BB intracellular domain and OX40 intracellular domain, preferably the amino acid of SEQ ID NO: 38 It may be CD3ζ represented by the sequence.
키메릭 항원 수용체 코딩 폴리뉴클레오타이드 및 키메릭 항원 수용체 발현 벡터Chimeric Antigen Receptor Encoding Polynucleotides and Chimeric Antigen Receptor Expression Vectors
본 발명은 또 다른 관점에서, 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드에 관한 것이다.In another aspect, the present invention relates to a polynucleotide encoding the chimeric antigen receptor (CAR).
본 발명에 있어서, 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드는 CD22-결합 도메인을 코딩하는 폴리뉴클레오타이드; 막관통 도메인을 코딩하는 폴리뉴클레오타이드; 공동 자극 도메인을 코팅하는 폴리뉴클레오타이드; 및 세포 내 신호전달 도메인을 코딩하는 폴리뉴클레오타이드를 포함할 수 있다. In the present invention, the polynucleotide encoding the chimeric antigen receptor (CAR) comprises a polynucleotide encoding a CD22-binding domain; a polynucleotide encoding a transmembrane domain; polynucleotides coating the co-stimulatory domain; and a polynucleotide encoding an intracellular signaling domain.
상기 CD22-결합 도메인을 코딩하는 폴리뉴클레오타이드는 본 발명의 CD22에 특이적인 3F11 및 7B5 항체를 코딩하는 폴리뉴클레오타이드일 수 있으며, 경쇄가변부위 및 중쇄가변부위가 링커로 연결된 scFv 형태로, 구체적인 염기서열은 상술한 바와 같다. The polynucleotide encoding the CD22-binding domain may be a polynucleotide encoding 3F11 and 7B5 antibodies specific for CD22 of the present invention, in the form of an scFv in which the light chain variable region and the heavy chain variable region are linked by a linker, and the specific nucleotide sequence is As described above.
바람직하게, 본 발명의 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드는, 서열번호 28의 염기서열로 표시되는 신호 펩타이드; Preferably, the polynucleotide encoding the chimeric antigen receptor (CAR) of the present invention comprises: a signal peptide represented by the nucleotide sequence of SEQ ID NO: 28;
서열번호 24의 염기서열로 표시되는 3F11 항체 또는 서열번호 26의 염기서열로 표시되는 7B5 항체;3F11 antibody represented by the nucleotide sequence of SEQ ID NO: 24 or 7B5 antibody represented by the nucleotide sequence of SEQ ID NO: 26;
서열번호 30의 염기서열로 표시되는 막관통 도메인; a transmembrane domain represented by the nucleotide sequence of SEQ ID NO: 30;
서열번호 31의 염기서열로 표시되는 4-1BB(공동자극 도메인); 및 4-1BB (costimulatory domain) represented by the nucleotide sequence of SEQ ID NO: 31; and
서열번호 32의 염기서열로 표시되는 CD3ζ(세포 내 신호전달 도메인)로 구성될 수 있다.It may be composed of CD3ζ (intracellular signaling domain) represented by the nucleotide sequence of SEQ ID NO: 32.
또한, CD22-결합 도메인을 코딩하는 폴리뉴클레오타이드 및 막관통 도메인 사이에, 힌지 부위(hinge region)를 코딩하는 폴리뉴클레오타이드가 추가로 포함될 수 있으며, 바람직하게 서열번호 29의 염기서열로 표시되는 CD8 힌지 부위일 수 있다.In addition, between the polynucleotide encoding the CD22-binding domain and the transmembrane domain, a polynucleotide encoding a hinge region may be further included, preferably a CD8 hinge region represented by the nucleotide sequence of SEQ ID NO: 29 can be
본 발명은 또 다른 관점에서, 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 포함하는 벡터에 관한 것이다. In another aspect, the present invention relates to a vector comprising a polynucleotide encoding the chimeric antigen receptor (CAR).
본 발명의 구체적인 구현예에서, 상기 벡터는 재조합 바이러스 벡터로, 바람직하게는 렌티바이러스 벡터이며, 작동가능하게 연결된 EF1α 프로모터; 시그널 펩타이드를 코딩하는 폴리뉴클레오타이드; CD22-결합 도메인을 코딩하는 폴리뉴클레오타이드; 막관통 도메인을 코딩하는 폴리뉴클레오타이드; 세포 내 신호전달 도메인을 코딩하는 폴리뉴클레오타이드를 포함하며, 단백질 발현을 증가시키기 위해 WPRE(woodchuck hepatitis virus post-transcriptional regulatory element)를 추가로 포함할 수 있다 (도 2). In a specific embodiment of the present invention, the vector is a recombinant viral vector, preferably a lentiviral vector, comprising an operably linked EF1α promoter; a polynucleotide encoding a signal peptide; a polynucleotide encoding a CD22-binding domain; a polynucleotide encoding a transmembrane domain; It includes a polynucleotide encoding an intracellular signaling domain, and may further include a woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) to increase protein expression ( FIG. 2 ).
상기 EF1α 프로모터는 서열번호 27의 염기서열로 표시될 수 있으며, 필요에 따라 상기 서열번호 27의 염기서열과 90% 이상, 93% 이상, 95% 이상, 96% 이상, 97% 이상, 98% 이상, 또는 99% 이상 동일한 서열을 포함할 수 있다.The EF1α promoter may be represented by the nucleotide sequence of SEQ ID NO: 27, and if necessary, 90% or more, 93% or more, 95% or more, 96% or more, 97% or more, 98% or more of the nucleotide sequence of SEQ ID NO: 27 , or sequences that are at least 99% identical.
또한, 상기 프로모터는 CD22-결합 도메인인 항-CD22 항체(scFv)의 발현을 유도하도록 작동 가능하게 연결되어 있다.In addition, the promoter is operably linked to drive expression of the CD22-binding domain, anti-CD22 antibody (scFv).
숙주 세포 내로 폴리뉴클레오티드를 도입하기 위한 생물학적 방법은 DNA 및 RNA 벡터의 사용을 포함한다. 바이러스 벡터, 및 특히 레트로바이러스 벡터는 유전자를 포유동물, 예를 들어 인간 세포 내로 삽입하기 위해 가장 널리 사용되는 방법이 되었다. 다른 바이러스 벡터는 렌티바이러스, 폭스바이러스, 단순 포진 바이러스, 아데노바이러스 및 아데노-연관 바이러스 등으로부터 유래될 수 있다. Biological methods for introducing polynucleotides into host cells include the use of DNA and RNA vectors. Viral vectors, and in particular retroviral vectors, have become the most widely used methods for inserting genes into mammalian, eg, human cells. Other viral vectors may be derived from lentiviruses, poxviruses, herpes simplex viruses, adenoviruses and adeno-associated viruses, and the like.
숙주 세포 내로 폴리뉴클레오티드를 도입하기 위한 화학적 수단은 콜로이드성 분산액 시스템, 예를 들면, 거대분자 복합체, 나노캡슐, 마이크로구체, 비드, 및 수중유 에멀젼, 미셀, 혼합된 미셀, 및 리포솜을 비롯한 지질-기반 시스템을 포함한다. 시험관 내 및 생체 내에서 전달 비히클로서 사용하기 위한 예시적인 콜로이드성 시스템은 리포솜(예를 들면, 인공 막 소포)이다. 핵산의 최신 기술의 표적화된 전달, 예를 들면, 표적화된 나노입자 또는 다른 적합한 마이크로미터-미만 크기의 전달 시스템을 사용한 폴리뉴클레오티드의 전달을 위한 다른 방법이 이용 가능하다.Chemical means for introducing polynucleotides into host cells include colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads, and lipid-including oil-in-water emulsions, micelles, mixed micelles, and liposomes. including the underlying system. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (eg, an artificial membrane vesicle). Other methods are available for state-of-the-art targeted delivery of nucleic acids, for example, delivery of polynucleotides using targeted nanoparticles or other suitable sub-micron sized delivery systems.
비-바이러스 전달 시스템이 이용되는 경우에, 예시적인 전달 비히클은 리포솜이다. 지질 제제의 사용은 숙주세포 내로의 핵산의 도입(시험관 내, 생체 외 또는 생체 내)을 위해 고려된다. 또 다른 측면에서, 핵산은 지질과 회합될 수 있다. 지질과 회합된 핵산은 리포솜의 수성 내부에 캡슐화되거나, 리포솜의 지질 이중층 내에 점재되거나, 리포솜 및 올리고뉴클레오티드 둘 다와 회합된 연결 분자를 통해 리포솜에 부착되거나, 리포솜 내에 포획되거나, 리포솜과 복합체화되거나, 지질 함유 용액 중에 분산되거나, 지질과 혼합되거나, 지질과 조합되거나, 지질 내에 현탁액으로서 함유되거나, 미셀과 함께 함유 또는 복합체화되거나, 또는 지질과 달리 회합될 수 있다. 지질, 지질/DNA 또는 지질/발현 벡터 회합 조성물은 용액 중의 임의의 특정한 구조로 제한되지 않는다.When a non-viral delivery system is used, an exemplary delivery vehicle is a liposome. The use of lipid preparations is contemplated for the introduction of nucleic acids into host cells (in vitro, ex vivo or in vivo). In another aspect, the nucleic acid may be associated with a lipid. Nucleic acids associated with lipids may be encapsulated within the aqueous interior of the liposome, interspersed within the lipid bilayer of the liposome, attached to the liposome via a linking molecule associated with both the liposome and oligonucleotide, captured within the liposome, complexed with the liposome, or , dispersed in a lipid containing solution, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with micelles, or otherwise associated with a lipid. The lipid, lipid/DNA or lipid/expression vector association composition is not limited to any particular structure in solution.
키메릭 항원 수용체(CAR) 발현 면역 이펙터 세포Chimeric Antigen Receptor (CAR) Expressing Immune Effector Cells
본 발명은 또 다른 관점에서, 상기 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 또는 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드를 포함하는 벡터를 포함하고, 상기 키메릭 항원 수용체(CAR)를 발현하는 면역 이펙터 세포에 관한 것이다.In another aspect, the present invention includes a vector comprising a polynucleotide encoding the chimeric antigen receptor (CAR) or a polynucleotide encoding a chimeric antigen receptor (CAR), wherein the chimeric antigen receptor (CAR) It relates to immune effector cells expressing
본 발명에 있어서, 상기 면역 이펙터 세포는 포유동물 유래 세포 일 수 있으며, 바람직하게는 T 세포 또는 자연 살해(NK) 세포일 수 있다.In the present invention, the immune effector cells may be mammalian-derived cells, preferably T cells or natural killer (NK) cells.
본 발명에 있어서, 상기 키메릭 항원 수용체(CAR)를 발현하는 면역 이펙터 세포는 본 발명의 CAR 벡터를 면역 이펙터 세포, 예를 들어 T 세포 또는 NK 세포 내로 도입시켜 제조할 수 있다. In the present invention, immune effector cells expressing the chimeric antigen receptor (CAR) can be prepared by introducing the CAR vector of the present invention into immune effector cells, for example, T cells or NK cells.
구체적으로, CAR 벡터는 전기천공법, 리포펙타민(lipofectamine 2000, Invitrogen) 등과 같은 당업계에 공지된 방법에 의해 세포 내로 도입될 수 있다. 예를 들어, 면역 이펙터 세포는 렌티바이러스 벡터에 의해 형질 감염되어 CAR 분자를 운반하는 바이러스 게놈을 숙주 게놈에 통합시켜 표적 유전자의 장기적이고 안정적인 발현을 보장할 수 있다. 다른 예를 들어, 전이인자(transposon)는 CAR 운반 플라스미드(transposon) 및 전이효소 운반 플라스미드를 표적 세포 내로 도입하는데 이용될 수 있다. 다른 예를 들어, CAR 분자는 유전자 편집방법(예를 들면, CRISPR/Cas9)에 의해 게놈에 첨가될 수 있다.Specifically, the CAR vector may be introduced into cells by methods known in the art such as electroporation, lipofectamine 2000, Invitrogen, and the like. For example, immune effector cells can be transfected with a lentiviral vector to integrate the viral genome carrying the CAR molecule into the host genome to ensure long-term and stable expression of the target gene. For another example, a transposon can be used to introduce a CAR transport plasmid and a transferase transport plasmid into a target cell. For another example, a CAR molecule can be added to the genome by a gene editing method (eg, CRISPR/Cas9).
본 발명의 구체적인 일실시예에서는 도 2에 나타난 바와 같이 CD22-CAR를 코팅하는 폴리뉴클레오타이드가 삽입된 렌티바이러스 벡터를 제조하였으며, 제조된 벡터를 T 세포에 형질전환 시켜 CD22-CAR-T 세포를 제조하였다. 제조된 CD22-CAR-T 세포에서는 본 발명의 CD22를 표적으로 하는 키메라 항원 수용체를 발현하게 된다. In a specific embodiment of the present invention, as shown in FIG. 2 , a lentiviral vector into which a polynucleotide coating CD22-CAR was inserted was prepared, and the prepared vector was transformed into T cells to prepare CD22-CAR-T cells. did. The prepared CD22-CAR-T cells express the chimeric antigen receptor targeting CD22 of the present invention.
키메릭 항원 수용체(CAR)를 발현하는 면역 이펙터 세포 제조를 위한 면역 이펙터 세포는 대상체로 부터 수득할 수 있으며, 상기 "대상체"는 면역 반응이 도출될 수 있는 살아있는 유기체 (예를 들어, 포유동물)를 포함한다. 대상체의 예는 인간, 개, 고양이, 마우스, 래트, 및 그의 트랜스제닉 종을 포함한다. T 세포는 말초 혈액 단핵 세포, 골수, 림프절 조직, 제대혈, 흉선 조직, 감염 부위로부터의 조직, 복수, 흉막 삼출, 비장 조직, 및 종양을 비롯한 수많은 공급원으로부터 수득될 수 있다.Immune effector cells for making immune effector cells expressing a chimeric antigen receptor (CAR) can be obtained from a subject, wherein the "subject" is a living organism (eg, a mammal) in which an immune response can be elicited. includes Examples of subjects include humans, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from numerous sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, umbilical cord blood, thymus tissue, tissue from the site of infection, ascites, pleural effusion, splenic tissue, and tumors.
상기 T 세포는 통상의 기술자에게 공지된 임의의 많은 기술, 예를 들면, 피콜(Ficoll)™ 분리를 사용하여 대상체로부터 수집된 혈액 단위로부터 수득될 수 있다. 혈액으로부터 세포는 분리반출술에 의해 수득되며, 분리반출술 생성물은 전형적으로 T 세포, 단핵구, 과립구, B 세포를 비롯한 림프구, 다른 유핵 백혈구, 적혈구, 및 혈소판을 함유한다. Such T cells can be obtained from blood units collected from a subject using any of a number of techniques known to those of ordinary skill in the art, for example, Ficoll™ separation. Cells from blood are obtained by apheresis, and apheresis products typically contain T cells, monocytes, granulocytes, lymphocytes including B cells, other nucleated leukocytes, red blood cells, and platelets.
분리반출술에 의해 수집된 세포는 혈장 분획을 제거하고 세포를 후속 프로세싱 단계를 위해 적절한 완충제 또는 배지에 두기 위해 세척될 수 있다. T 세포는 적혈구를 용해시키고, 예를 들어 퍼콜(PERCOLL)™ 구배를 통한 원심분리에 의해 또는 역류 원심 분리에 의해 단핵구를 고갈시킴으로써 말초 혈액 림프구로부터 단리된다.Cells collected by apheresis can be washed to remove the plasma fraction and place the cells in an appropriate buffer or medium for subsequent processing steps. T cells are isolated from peripheral blood lymphocytes by lysing red blood cells and depleting monocytes, for example, by centrifugation through a PERCOLL™ gradient or by countercurrent centrifugation.
본 발명의 구체적인 일구현예에서, 도 3에 나타난 바와 같이, 말초 혈액 단핵세포(peripheral blood mononuclear cell, PBMC)로 부터 활성화된 T 세포를 분리한 다음, CD22-CAR 렌티바이러스를 T 세포에 형질도입시켜 CD22-CAR-T 세포를 제조하였다. 제조된 CD22-CAR-T 세포의 CD22-펩타이드 결합능을 확인한 결과, 도 4에 나타난 바와 같이, CD22-펩타이드 증가에 따라, CD4 또는 CD8를 발현하는 CD22-CAR-T 세포가 증가하는 것을 확인하였다. 이는 본 발명에서 제조한 CD22-CAR-T 세포가 효과적으로 CD22와 결합하는 것을 의미한다.In a specific embodiment of the present invention, as shown in FIG. 3 , activated T cells are isolated from peripheral blood mononuclear cells (PBMCs), and then CD22-CAR lentivirus is transduced into T cells. to prepare CD22-CAR-T cells. As a result of confirming the CD22-peptide binding ability of the prepared CD22-CAR-T cells, as shown in FIG. 4 , it was confirmed that the CD22-CAR-T cells expressing CD4 or CD8 increased as the CD22-peptide increased. This means that the CD22-CAR-T cells prepared in the present invention effectively bind to CD22.
본 발명의 구체적인 다른 일구현예에서, CD22-CAR-T 세포의 활성화를 확인하기 위해, 표적 세포의 존재 하에 CD22-CAR-T 세포에 의한 IFNγ 발현 정도를 확인하였다. 그 결과 도 5a 및 도 5b에 나타난 바와 같이, CD22를 발현하지 않는 K562 세포에서는 T 세포가 활성화되지 않은 반면, CD22를 발현하는 U2932 세포의 존재 하에 T 세포가 활성화되어 IFNγ 발현이 증가하는 것을 확인하였다.In another specific embodiment of the present invention, in order to confirm activation of CD22-CAR-T cells, the level of IFNγ expression by CD22-CAR-T cells in the presence of target cells was checked. As a result, as shown in FIGS. 5a and 5b, T cells were not activated in K562 cells that do not express CD22, whereas T cells were activated in the presence of U2932 cells expressing CD22, thereby increasing IFNγ expression. .
본 발명의 구체적인 또 다른 일구현예에서, CD22-CAR-T 세포에 의한 표적 세포의 사멸효과를 확인한 결과, 도 6에 나타난 바와 같이, CD22-CAR-T 세포는 CD22를 발현하는 U2932 세포에 대한 사멸 효과를 보이는 것을 확인하였다. In another specific embodiment of the present invention, as a result of confirming the killing effect of target cells by CD22-CAR-T cells, as shown in FIG. It was confirmed that the killing effect was shown.
즉, 본 발명의 CD22를 표적으로 하는 키메라 항원 수용체 및 CAR-T 세포는 B 세포 또는 CD22 발현과 관련된 질환 예방 또는 치료용 조성물로 유용하게 활용할 수 있다. That is, the chimeric antigen receptor and CAR-T cell targeting CD22 of the present invention can be usefully used as a composition for preventing or treating diseases related to B cells or CD22 expression.
CD22 발현에 의해 매개되는 질환 예방 또는 치료용 조성물Composition for preventing or treating diseases mediated by CD22 expression
본 발명은 또 다른 관점에서, CD22에 특이적으로 결합하는 항체 또는 CD22를 표적하는 키메라 항원 수용체를 발현하는 면역 이펙터 세포를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 예방 또는 치료용 약학적 조성물에 관한 것이다. In another aspect, the present invention provides a pharmaceutical for the prevention or treatment of diseases mediated by CD22-expressing cells, including immune effector cells expressing an antibody that specifically binds to CD22 or a chimeric antigen receptor targeting CD22. to the composition.
본 발명에 있어서, CD22를 발현하는 세포에 의해 매개되는 질환은 림프종, 비호치킨 림프종(non-Hogkins lymphoma: NHL), 공격적 NHL, 재발성 공격적 NHL, 재발성 지연성 NHL, 불응성 NHL, 불응성 지연성 NHL, 만성 림프성 백혈병(chronic lymphocytic leukemia: CLL), 소형 림프성 림프종, 백혈병, 모발성 세포 백혈병(hairy cell leukemia: HCL), 급성 림프성 백혈병(acute lymphocytic leukemia: ALL), 버킷트 림프종 및 외투 세포 림프종로 구성된 군에서 선택될 수 있다. In the present invention, the disease mediated by cells expressing CD22 is lymphoma, non-Hogkins lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed delayed NHL, refractory NHL, refractory Delayed NHL, chronic lymphocytic leukemia (CLL), small lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL), Burkitt's lymphoma and mantle cell lymphoma.
본 발명에 있어서, 상기 조성물은 CD22를 발현하는 세포에 의해 매개되는 질환의 치료제를 포함할 수 있으며, 상기 치료제는 CD22에 특이적으로 결합하는 항체의 중쇄 및/또는 경쇄에 공유결합된 상태로 존재하거나, 본 발명의 CD22에 특이적인 항체 또는 CD22-CAR-T 세포와 병용투여할 수 있다.In the present invention, the composition may include a therapeutic agent for a disease mediated by a cell expressing CD22, and the therapeutic agent is present in a state covalently bound to the heavy and/or light chain of an antibody that specifically binds to CD22. Alternatively, the CD22-specific antibody or CD22-CAR-T cell of the present invention may be co-administered.
상기 치료제는 저분자 약물, 펩타이드성 약물, 독소(예를 들어, 세포독소) 등을 포함한다.The therapeutic agent includes a small molecule drug, a peptide drug, a toxin (eg, a cytotoxin), and the like.
상기 저분자 약물는 관심대상의 약제학적 활성을 나타내고, 일반적으로 분자량이 약 800 Da 이하 또는 2000 Da 이하의 화합물일 수 있다. 무기 저분자는 탄소원자를 하나도 포함하지 않은 분자를 가리키고, 반면 유기저분자는 최소한 하나의 탄소원자를 함유하는 화합물을 가리킨다.The low molecular weight drug may be a compound that exhibits a pharmaceutical activity of interest and generally has a molecular weight of about 800 Da or less or 2000 Da or less. An inorganic small molecule refers to a molecule containing no carbon atoms, whereas an organic small molecule refers to a compound containing at least one carbon atom.
상기 펩타이드성 약물은 중합체성 화합물을 함유하는 아미노산을 가리키고, 이것은 자연발생 및 비-자연발생 펩타이드, 올리고펩타이드, 환형 펩타이드, 폴리펩타이드 및 단백질, 뿐만 아니라 펩타이드 모방물을 포함한다. 상기 펩타이드 약물은 화학적 합성에 의해 획득되거나 또는 유전적으로 인코딩된 공급원(예를 들어, 재조합공급원)에서 생성될 수 있다. 펩타이드 약물의 분자량의 범위는 200 Da 내지 10 kDa 또는그 이상일 수 있다.The peptidic drug refers to amino acids containing polymeric compounds, including naturally occurring and non-naturally occurring peptides, oligopeptides, cyclic peptides, polypeptides and proteins, as well as peptide mimics. The peptide drug may be obtained by chemical synthesis or produced from a genetically encoded source (eg, a recombinant source). The molecular weight of the peptide drug may range from 200 Da to 10 kDa or more.
상기 독소는 바람직하게 세포독소이며, 세포독소에는 비제한적으로, 리신, 아브린, 디프테리아 독소, 슈도모나스 균체외독소(예를 들어, PE35, PE37, PE38, PE40 등), 사포린, 겔로닌, 미국자리공 항바이러스 단백질(PAP), 보툴리눔 독소, 브리오딘, 모모르딘 및 부가닌이 포함된다.The toxin is preferably a cytotoxin, and the cytotoxin includes, but is not limited to, ricin, abrin, diphtheria toxin, Pseudomonas exotoxin (eg, PE35, PE37, PE38, PE40, etc.), saporin, gelonin, USA. antiviral protein (PAP), botulinum toxin, bryodin, momordin and buganin.
또한, 상기 치료제는 항암제일 수 있다. 항암제는 암 세포의 증식을 감소시키고, 세포독성 약제 및 세포 증식 억제제를 아우르는 비-펩타이드성(즉, 비-단백질계) 화합물을 포함한다. 항암제의 비제한적인 예는 알킬화제, 니트로소요소, 항대사물질, 항종양 항생물질, 식물 (빈카) 알칼로이드 및 스테로이드 호르몬을 포함한다. 펩타이드성 화합물 또한 사용될 수 있다.In addition, the therapeutic agent may be an anticancer agent. Anticancer agents reduce the proliferation of cancer cells and include non-peptidyl (ie, non-proteinaceous) compounds, including cytotoxic agents and cytostatic agents. Non-limiting examples of anticancer agents include alkylating agents, nitrosourea, antimetabolites, antitumor antibiotics, plant (vinca) alkaloids and steroid hormones. Peptide compounds may also be used.
상기 약학적 조성물에서 CD22에 특이적으로 결합하는 항체 또는 CD22를 표적하는 키메라 항원 수용체를 발현하는 면역 이펙터 세포는 치료 또는 진단용 조성물 내에서 유일한 활성성분이거나, 또는 예를 들면, 항-T 세포, 항-IFNγ 또는 항-LPS 항체와 같은 다른 항체성분들, 또는 크산틴과 같은 비항체 성분들을 포함하는 다른 활성성분들과 함께 사용 가능하다.In the pharmaceutical composition, an antibody that specifically binds to CD22 or an immune effector cell expressing a chimeric antigen receptor targeting CD22 is the only active ingredient in the composition for treatment or diagnosis, or, for example, an anti-T cell, anti It can be used in combination with other active ingredients, including other antibody components such as -IFNγ or anti-LPS antibodies, or non-antibody components such as xanthine.
약제 조성물은 치료적 유효량의 본 발명의 항체를 포함하는 것이 바람직하다. 여기에서 사용된 용어 "치료적 유효량"은 목표 질환 또는 상태를 치료, 개선 또는 예방하는 데 필요한 치료제의 양을 의미하고, 또는 감지할 수 있는 정도의 치료 또는 예방효과를 나타내는 데 필요한 치료제의 양을 뜻한다. 어떤 항체에 대하여, 치료적 유효 투여량은 세포배양 분석법 또는 보통 설치류, 토끼, 개, 돼지 또는 영장류와 같은 동물 모델로 최초로 결정될 수 있다. 동물 모델은 또한 적절한 농도범위와 투여루트를 결정하는 데 사용될 수 있다. 이러한 정보는 인간의 투약을 위해 유용한 투여량 및 루트를 결정하는 데 사용될 수 있다.The pharmaceutical composition preferably comprises a therapeutically effective amount of an antibody of the invention. As used herein, the term "therapeutically effective amount" means an amount of a therapeutic agent required to treat, ameliorate, or prevent a target disease or condition, or the amount of a therapeutic agent necessary to exhibit a detectable therapeutic or prophylactic effect. means For any antibody, a therapeutically effective dosage can be initially determined by cell culture assays or animal models, usually rodents, rabbits, dogs, pigs, or primates. Animal models can also be used to determine appropriate concentration ranges and routes of administration. Such information can be used to determine useful dosages and routes for dosing in humans.
인간환자를 위한 정밀한 유효량은 질환상태의 심각도, 환자의 일반적 건강 상태, 환자의 나이, 체중 및 성별, 식이요법, 투여시간, 투여빈도, 약제조성, 반응감도 및 치료에 대한 내성/반응에 따라 달라질 수 있다. 상기 양은 통상적인 실험에 의해 결정될 수 있고, 임상의사의 판단의 범위 내에 있다. 일반적으로, 유효 투여량은 0.01~50mg/kg, 바람직하게는 0.1~20mg/kg, 더욱 바람직하게는 약 15mg/kg이다.The precise effective amount for human patients will vary depending on the severity of the disease state, the patient's general health status, the patient's age, weight and sex, diet, administration time, administration frequency, drug composition, response sensitivity, and tolerance/response to treatment. can The amount can be determined by routine experimentation and is within the judgment of the clinician. In general, an effective dosage is 0.01-50 mg/kg, preferably 0.1-20 mg/kg, more preferably about 15 mg/kg.
조성물은 환자에게 개별적으로 투여되거나, 또는 다른 제제, 약제 또는 호르몬과 조합하여 투여될 수 있다.The composition may be administered to the patient individually or in combination with other agents, agents, or hormones.
본 발명의 항체가 투여되는 투여량은 치료될 상태의 성질, 악성 림프종 또는 백혈병의 등급, 및 항체가 질환 예방 차원에서 사용되는지 또는 현존하는 상태를 치료하기 위해 사용되는지에 따라 달라진다.The dosage at which the antibody of the present invention is administered depends on the nature of the condition to be treated, the grade of malignant lymphoma or leukemia, and whether the antibody is used to prevent disease or to treat an existing condition.
투여빈도는 항체분자의 반감기, 약 효과의 지속성에 따라 달라진다. 만약 항체분자가 짧은 반감기(예, 2~10시간)를 가지면, 하루당 1회 또는 그 이상의 투여량을 제공할 필요가 있다. 또는, 항체분자가 긴 반감기(예, 2~15일)를 가지면, 하루에 한번, 일주일에 한차례, 또는 매 1개월 또는 2개월당 한차례의 투여량을 제공할 필요가 있다.The frequency of administration depends on the half-life of the antibody molecule and the duration of the drug's effect. If the antibody molecule has a short half-life (eg, 2 to 10 hours), it may be necessary to provide one or more doses per day. Alternatively, if the antibody molecule has a long half-life (eg, 2 to 15 days), it may be necessary to provide a dose once a day, once a week, or once every 1 or 2 months.
또한, 약제 조성물은 항체의 투여를 위하여 약제학적으로 허용가능한 담체를 함유할 수 있다. 담체는 그 자신이 조성물을 투여받는 개체에 유해한 항체의 생성을 유발해서는 안되고, 독성이 없어야만 한다. 적당한 담체로는 단백질, 폴리펩타이드, 리포오좀, 다당류, 폴리락틱산, 폴리글리콜산, 아미노산 중합체, 아미노산 공중합체 및 비활성 바이러스 입자들과 같은, 서서히 물질대사되는 거대분자일 수 있다.In addition, the pharmaceutical composition may contain a pharmaceutically acceptable carrier for administration of the antibody. The carrier itself must not cause the production of antibodies that are harmful to the individual receiving the composition, and must be non-toxic. Suitable carriers may be slowly metabolized macromolecules, such as proteins, polypeptides, liposomes, polysaccharides, polylactic acid, polyglycolic acid, amino acid polymers, amino acid copolymers and inactive viral particles.
약제학적으로 허용가능한 염들은, 예를 들면, 염화수소산염, 브롬화수소산염, 인산염 및 황산염과 같은 미네랄산염들, 또는 아세트산, 프로피온산. 말론산 및 벤조산 같은 유기산의 염들이 사용될 수 있다.Pharmaceutically acceptable salts are, for example, mineral acid salts such as hydrochloride, hydrobromide, phosphate and sulfate, or acetic acid, propionic acid. Salts of organic acids such as malonic acid and benzoic acid may be used.
치료 조성물 내의 약제학적으로 허용가능한 담체는 부가적으로, 물, 식염수, 글리세롤 및 에탄올과 같은 액체들을 포함할 수 있다. 부가적으로, 습윤제, 유화제 또는 pH 완충물질과 같은 보조 물질들이 이런한 조성물 내에 존재할 수 있다. 상기 담체는 환자에 의한 약제 조성물 섭취를 위해, 정제, 환약, 당의정, 캡슐, 액체, 겔, 시럽, 슬러리 및 현탁제로서 제제화될 수 있다.Pharmaceutically acceptable carriers in therapeutic compositions may additionally include liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances such as wetting agents, emulsifying agents or pH buffering agents may be present in such compositions. The carrier may be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions for ingestion of the pharmaceutical composition by a patient.
투여를 위한 바람직한 형태는, 예로써 주사(injection) 또는 주입(infusion)(예를 들면, 환괴(bolus) 주사 또는 연속적 주입)에 의한 비경구적 투약에 적합한 형태를 포함한다. 생성물이 주입 또는 주사용일 경우에는, 오일 또는 수용성 부형제내의 현탁제, 용액 또는 에멀젼의 형태를 취할 수 있고, 이는 현탁제, 방부제, 안정화제 및/또는 분산제와 같은 처방제들을 포함할 수 있다. 또는, 항체분자는 무수형태일 수 있고, 사용전에 적절한 멸균액으로 재구성될 수 있다.Preferred forms for administration include those suitable for parenteral administration, eg, by injection or infusion (eg, bolus injection or continuous infusion). When the product is intended for infusion or injection, it may take the form of suspensions, solutions or emulsions in oil or water-soluble excipients, which may contain prescription agents such as suspending, preservative, stabilizing and/or dispersing agents. Alternatively, the antibody molecule may be in anhydrous form and reconstituted with an appropriate sterile solution prior to use.
일단 제제화된 경우, 본 발명의 조성물은 환자에게 직접 투여될 수 있다. 치료받을 환자들은 동물일 수 있다. 그러나, 조성물은 인간 환자 투여를 위해 맞추는 것이 바람직하다.Once formulated, the compositions of the present invention can be administered directly to a patient. The patients to be treated may be animals. However, the composition is preferably adapted for administration to human patients.
본 발명의 약제 조성물은 제한은 없지만, 경구, 정맥, 근육내, 동맥내, 골수내, 척추강내, 심실내, 경피(transdermal), 경피(transcutaneous)(예, WO 98/20734 참조), 피하, 복강내, 비강내, 장내, 국소, 혀밑, 질내 또는 직장 경로를 포함하는 어떤 경로에 의해 투여될 수 있다. 본 발명의 약제 조성물을 투여하는 데 하이포스프레이(hypospray)가 사용될 수 있다. 전형적으로, 치료 조성물은 액체 용액 또는 현탁액으로서 주사가능한 물질로서 제조될 수 있다. 또한, 주입전에 액체 부형제내용액 또는 현탁액에 적합한 고체 형태가 제조될 수 있다.The pharmaceutical composition of the present invention is not limited, but oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intraventricular, transdermal, transcutaneous (see, e.g., WO 98/20734), subcutaneous, Administration may be by any route, including intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal routes. A hypospray may be used to administer the pharmaceutical composition of the present invention. Typically, therapeutic compositions may be prepared as injectables as liquid solutions or suspensions. In addition, solid forms suitable for solution or suspension in liquid excipients prior to injection may be prepared.
조성물의 직접적인 전달은 일반적으로 주사, 피하주사, 복강내주사, 정맥내주사, 근육내주사에 의해 이루어질 수 있거나, 또는 조직의 간질(interstitial) 공간으로 전달될 수도 있다. 또한, 조성물은 상처부위로 투여될 수 있다. 투여량 처리는 단일 복용 스케쥴 또는 다중 복용 스케쥴일 수 있다.Direct delivery of the composition may generally be achieved by injection, subcutaneous injection, intraperitoneal injection, intravenous injection, intramuscular injection, or may be delivered to the interstitial space of a tissue. In addition, the composition may be administered to the wound site. Dosage treatment may be a single dose schedule or a multiple dose schedule.
조성물내의 활성성분은 항체분자일 수 있다. 그 자체로, 위장관내에서 분해에 민감할 수 있다. 따라서, 조성물이 위장관을 사용하는 경로에 의해 투여되면, 조성물은, 분해로부터 항체를 보호하지만 일단 위장관으로부터 흡수된 항체를 방출시키는 제제를 함유할 필요가 있을 것이다.The active ingredient in the composition may be an antibody molecule. As such, it may be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is administered by a route using the gastrointestinal tract, the composition will need to contain an agent that protects the antibody from degradation but releases the antibody once absorbed from the gastrointestinal tract.
약제학적으로 허용가능한 담체의 완벽한 논의는 레밍톤 약제학지(Remington's Pharmaceutical Sciences)(Mack Publishing Company, NJ, 1991)를 이용할 수 있다.A complete discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (Mack Publishing Company, NJ, 1991).
CD22를 발현하는 세포에 의해 매개되는 질환의 진단 또는 모니터링Diagnosis or monitoring of diseases mediated by cells expressing CD22
본 발명은 또 다른 관점에서, CD22에 특이적으로 결합하는 항체를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 진단 또는 모니터링용 조성물에 관한 것이다.In another aspect, the present invention relates to a composition for diagnosing or monitoring a disease mediated by a CD22-expressing cell comprising an antibody that specifically binds to CD22.
상기 CD22에 특이적으로 결합하는 항체는 직접적으로 또는 간접적으로 표지될 수 있다. 간접적 표지는 검출가능 표지를 포함하는 2차 항체를 포함하는데, 여기서 2차 항체가 CD22에 특이적으로 결합하는 항체에 결합한다. 다른 간접적 표지는 바이오틴을 포함하는데, 여기서 바이오티닐화된 CD22에 특이적으로 결합하는 항체는 검출가능 표지를 포함하는 아비딘 또는 스트렙트아비딘을 사용하여 검출될 수 있다.The antibody that specifically binds to CD22 may be directly or indirectly labeled. Indirect labels include secondary antibodies comprising a detectable label, wherein the secondary antibody binds to an antibody that specifically binds to CD22. Other indirect labels include biotin, wherein an antibody that specifically binds to biotinylated CD22 can be detected using avidin or streptavidin comprising a detectable label.
적절한 검출가능 표지는 분광분석적, 광화학적, 생화학적, 면역화학적, 전기적, 광학적 또는 화학적 수단에 의해 검출가능한 모든 조성물을 포함한다. 적절한 표지는, 비제한적으로, 자석 비드, 형광염료(예를 들어, 플루오레세인이소티오시안산염, 텍사스 레드, 로다민, 초록 형광 단백질, 적색 형광 단백질, 황색 형광 단백질 등), 방사성 표지(예를 들어, 3H, 125I, 35S, 14C또는 32P), 효소(예를 들어, 겨자무과산화효소, 알칼린 포스파타아제, 루시퍼라아제 및 효소-연결 면역흡착검사(ELISA)에 일반적으로 사용되는 것들) 및 콜로이드성 골드 또는 착색 유리 또는 플라스틱(예를 들면, 폴리스티렌, 폴리프로필렌, 라텍스 등) 비드와 같은 표색계 표지를 포함한다.Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Suitable labels include, but are not limited to, magnetic beads, fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, green fluorescent protein, red fluorescent protein, yellow fluorescent protein, etc.), radioactive labels (e.g., For example, 3 H, 125 I, 35 S, 14 C or 32 P), enzymes (eg, mustard radish peroxidase, alkaline phosphatase, luciferase and enzyme-linked immunosorbent assay (ELISA)) commonly used) and colorimetric labels such as colloidal gold or colored glass or plastic (eg, polystyrene, polypropylene, latex, etc.) beads.
또한, 진단 또는 모니터링을 위해 상기 항체는 형광 단백질로 표지될 수 있으며, 조영제 또는 방사선 동위원소를 포함할 수 있다.In addition, for diagnosis or monitoring, the antibody may be labeled with a fluorescent protein, and may contain a contrast agent or a radioisotope.
본 발명의 CD22에 특이적으로 결합하는 항체를 진단 키트에 이용하는 경우, 상기 항체는 지지체에 고정되어 있으며, 상기 지지체는 마이크로플레이트, 마이크로어레이, 칩, 유리, 비드 또는 입자, 또는 멤브레인 일 수 있다. When the antibody specifically binding to CD22 of the present invention is used in a diagnostic kit, the antibody is immobilized on a support, and the support may be a microplate, microarray, chip, glass, bead or particle, or a membrane.
이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예를 제시한다. 그러나 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 하기 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다.Hereinafter, preferred examples are presented to help the understanding of the present invention. However, the following examples are only provided for easier understanding of the present invention, and the contents of the present invention are not limited by the following examples.
실시예 1: CD22에 특이적으로 결합하는 항체 제조 및 선별Example 1: Preparation and selection of antibodies that specifically bind to CD22
CD22 펩타이드 특이적인 항체를 선별하기위해, CD22와 결합하는 항체를 생산하는 하이브리도마를 제조하여 항체를 선별하였다. To select the CD22 peptide-specific antibody, a hybridoma producing an antibody binding to CD22 was prepared and the antibody was selected.
먼저, CD22 단백질(ACRObiosystems Inc., cat# CD2-H52H8, 미국)을 면역하여 비장세포를 적출하고 마우스 골수증세포와 세포 융합을 통하여 하이브리도마 세포를 제작하였다. First, splenocytes were extracted by immunization with CD22 protein (ACRObiosystems Inc., cat# CD2-H52H8, USA), and hybridoma cells were prepared by cell fusion with mouse myeloma cells.
세포 융합에 이용하는 마우스 골수종 세포는 HGPRT(HypoxanthineGuanidine-Phosphoribosyl-Transferase)를 가지고 있지 않기 때문에 HAT 배지에서는 생존할 수 없으나, 하이브리도마는 비장세포와 융합함으로써 HAT 배지에서 생존할 수 있다. 이를 이용하면 하이브리도마만을 증식시킬 수 있으므로, 통상 하이브리도마를 확립시킬때까지 HAT 배지에서 증식시켰다.Mouse myeloma cells used for cell fusion cannot survive in HAT medium because they do not have HGPRT (HypoxanthineGuanidine-Phosphoribosyl-Transferase), but hybridomas can survive in HAT medium by fusion with splenocytes. Since only hybridomas can be grown using this, it is usually grown in HAT medium until hybridomas are established.
증식된 하이브리도마 중에서 CD22와 결합하는 항체를 생산하는 하이브리도마를 선별하기 위해 한계희석법을 사용하였다. 우선 96웰당 1개 세포 이하가 되도록한 다음, 1개의 세포로부터 증식된 클론에서 얻어진 항체가 CD22와 결합하는지를 ELISA로 확인하고 CD22와 결합하는 클론을 선별하였다. 상기과정을 3회 반복하여 CD22와 결합하는 항체를 생산하는 하이브리도마를 선별하였다. 이와 같은 방법으로 CD22에 결합하는 2종의 항체를 수득하였다.The limiting dilution method was used to select hybridomas producing an antibody binding to CD22 from among the proliferated hybridomas. First, it was made to be less than one cell per 96 well, and then, it was confirmed by ELISA whether the antibody obtained from the clones proliferated from one cell binds to CD22, and clones that bind to CD22 were selected. The above process was repeated three times to select hybridomas producing an antibody binding to CD22. In this way, two types of antibodies that bind to CD22 were obtained.
상기 2종의 항체는 각각 3F11 및 7B5로 명명하였으며, 이들의 염기서열과 아미노산 서열을 분석하였다. 서열분석 결과에 따른 각 항체의 중쇄 가변부위 및 경쇄 가변부위에 대한 서열정보는 하기 표 1 및 표 2에 나타내었으며, 하기 표 1 및 표 2에서 밑줄친 부분은 상보적 결정 부위(complementarity determining region; CDR)를 의미한다.The two antibodies were named 3F11 and 7B5, respectively, and their base and amino acid sequences were analyzed. Sequence information on the heavy chain variable region and the light chain variable region of each antibody according to the sequencing results is shown in Tables 1 and 2 below, and the underlined portions in Tables 1 and 2 below are complementarity determining regions; CDR).
3F11 항체의 서열정보Sequence information of the 3F11 antibody
3F113F11 서열정보sequence information 서열번호SEQ ID NO:
중쇄가변부위 CDR1Heavy chain variable region CDR1 GFTFSNYWGFTFSNYW 서열번호 1SEQ ID NO: 1
중쇄가변부위 CDR2Heavy chain variable region CDR2 IRLKSNNYATIRLKSNNYAT 서열번호 2SEQ ID NO: 2
중쇄가변부위 CDR3Heavy chain variable region CDR3 TSIYYYGRDYAMDYTSIYYYGRDYAMDY 서열번호 3SEQ ID NO: 3
경쇄가변부위 CDR1Light chain variable region CDR1 SSVSSSYSSVSSSY 서열번호 4SEQ ID NO: 4
경쇄가변부위 CDR2Light chain variable region CDR2 STSSTS 서열번호 5SEQ ID NO: 5
경쇄가변부위 CDR3Light chain variable region CDR3 HQYHRSPYTHQYHRSPYT 서열번호 6SEQ ID NO: 6
중쇄가변부위
아미노산서열
heavy chain variable region
amino acid sequence
EVKLVESGGGLVQPGGSMKLSCVAS GFTFSNYW MNWVRQSPEKGLEWVAE IRLKSNNYAT HYAESVKGRFTISRDDSKSSVYLQMNNLRAEDTGIYYC TSIYYYGRDYAMDY WGQGTSVTVSSEVKLVESGGGLVQPGGSMKLSCVAS GFTFSNYW MNWVRQSPEKGLEWVAE IRLKSNNYAT HYAESVKGRFTISRDDSKSSVYLQMNNLRAEDTGIYYC TSIYYYGRDYAMDY WGQGTSVTVSS 서열번호 7SEQ ID NO: 7
경쇄가변부위
아미노산서열
light chain variable region
amino acid sequence
DIVMTQSPAIMSASLGERVTMTCTAS SSVSSSY LHWYQQKPGSSPKLWIY STS NLASGVPARFSGSGSGTSYSLTISSMEAEDAATYYC HQYHRSPYT FGGGTKLEIKDIVMTQSPAIMSASLGERVTMTCTAS SSVSSSY LHWYQQKPGSSPKLWIY STS NLASGVPARFSGSGSGTSYSLTISSMEAEDAATYYC HQYHRSPYT FGGGTKLEIK 서열번호 8SEQ ID NO: 8
중쇄가변부위
염기서열
heavy chain variable region
base sequence
GAGGTGAAGCTGGTGGAGAGCGGCGGCGGCCTGGTGCAGCCCGGCGGCAGCATGAAGCTGAGCTGCGTGGCCAGCGGCTTCACCTTCAGCAACTACTGGATGAACTGGGTGAGGCAGAGCCCCGAGAAGGGCCTGGAGTGGGTGGCCGAGATCAGGCTGAAGAGCAACAACTACGCCACCCACTACGCCGAGAGCGTGAAGGGCAGGTTCACCATCAGCAGGGACGACAGCAAGAGCAGCGTGTACCTGCAGATGAACAACCTGAGGGCCGAGGACACCGGCATCTACTACTGCACCAGCATCTACTACTACGGCAGGGACTACGCCATGGACTACTGGGGCCAGGGCACCAGCGTGACCGTGAGCAGC GAGGTGAAGCTGGTGGAGAGCGGCGGCGGCCTGGTGCAGCCCGGCGGCAGCATGAAGCTGAGCTGCGTGGCCAGCGGCTTCACCTTCAGCAACTACTGGATGAACTGGGTGAGGCAGAGCCCCGAGAAGGGCCTGGAGTGGGTGGCCGAGATCAGGCTGAAGAGCAACAACTACGCCACCCACTACGCCGAGAGCGTGAAGGGCAGGTTCACCATCAGCAGGGACGACAGCAAGAGCAGCGTGTACCTGCAGATGAACAACCTGAGGGCCGAGGACACCGGCATCTACTACTGCACCAGCATCTACTACTACGGCAGGGACTACGCCATGGACTACTGGGGCCAGGGCACCAGCGTGACCGTGAGCAGC 서열번호 9SEQ ID NO: 9
경쇄가변부위
염기서열
light chain variable region
base sequence
GACATCGTGATGACCCAGAGCCCCGCCATCATGAGCGCCAGCCTGGGCGAGAGGGTGACCATGACCTGCACCGCCAGCAGCAGCGTGAGCAGCAGCTACCTGCACTGGTACCAGCAGAAGCCCGGCAGCAGCCCCAAGCTGTGGATCTACAGCACCAGCAACCTGGCCAGCGGCGTGCCCGCCAGGTTCAGCGGCAGCGGCAGCGGCACCAGCTACAGCCTGACCATCAGCAGCATGGAGGCCGAGGACGCCGCCACCTACTACTGCCACCAGTACCACAGGAGCCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGATCAAG GACATCGTGATGACCCAGAGCCCCGCCATCATGAGCGCCAGCCTGGGCGAGAGGGTGACCATGACCTGCACCGCCAGCAGCAGCGTGAGCAGCAGCTACCTGCACTGGTACCAGCAGAAGCCCGGCAGCAGCCCCAAGCTGTGGATCTACAGCACCAGCAACCTGGCCAGCGGCGTGCCCGCCAGGTTCAGCGGCAGCGGCAGCGGCACCAGCTACAGCCTGACCATCAGCAGCATGGAGGCCGAGGACGCCGCCACCTACTACTGCCACCAGTACCACAGGAGCCCCTACACCTTCGGCGGCGGCACCAAGCTGGAGATCAAG 서열번호 10SEQ ID NO: 10
7B5 항체의 서열정보Sequence information of the 7B5 antibody
7B57B5 서열정보sequence information 서열번호SEQ ID NO:
중쇄가변부위 CDR1Heavy chain variable region CDR1 GFSLTRYDGFSLTRYD 서열번호 11SEQ ID NO: 11
중쇄가변부위 CDR2Heavy chain variable region CDR2 MWTGGGTMWTGGGT 서열번호 12SEQ ID NO: 12
중쇄가변부위 CDR3Heavy chain variable region CDR3 VRSYYGSAMDYVRSYYGSAMDY 서열번호 13SEQ ID NO: 13
경쇄가변부위 CDR1Light chain variable region CDR1 DHINNWDHINW 서열번호 14SEQ ID NO: 14
경쇄가변부위 CDR2Light chain variable region CDR2 GATGAT 서열번호 15SEQ ID NO: 15
경쇄가변부위 CDR3Light chain variable region CDR3 QQHWSTPFTQQHWSTPFT 서열번호 16SEQ ID NO: 16
중쇄가변부위
아미노산서열
heavy chain variable region
amino acid sequence
EVQLEESGPGLVAPSQSLSITCTVS GFSLTRYD ISWIRQPPGKGLEWLGV MWTGGGT NYNSAFMSRLSISRDNSKSQVFLKMNSLQTDDTAIYYC VRSYYGSAMDY WGQGTSVTVSS EVQLEESGPGLVAPSQSLSITCTVS GFSLTRYD ISWIRQPPGKGLEWLGV MWTGGGT NYNSAFMSRLSISRDNSKSQVFLKMNSLQTDDTAIYYC VRSYYGSAMDY WGQGTSVTVSS 서열번호 17SEQ ID NO: 17
경쇄가변부위
아미노산서열
light chain variable region
amino acid sequence
DIVMTQSSSYLSVSLGGRVTITCKAS DHINNW LAWYQQKPGNAPRLLIS GAT SLETGVPSRFSGSGSGKDYTLSITSLQTEDVATYYC QQHWSTPFT FGSGTKLEIKDIVMTQSSSYLSVSLGGRVTITCKAS DHINNW LAWYQQKPGNAPRLLIS GAT SLETGVPSRFSGSGSGKDYTLSITSLQTEDVATYYC QQHWSTPFT FGSGTKLEIK 서열번호 18SEQ ID NO: 18
중쇄가변부위
염기서열
heavy chain variable region
base sequence
GAGGTGCAGCTGGAGGAGAGCGGCCCCGGCCTGGTGGCCCCCAGCCAGAGCCTGAGCATCACCTGCACCGTGAGCGGCTTCAGCCTGACCAGGTACGACATCAGCTGGATCAGGCAGCCCCCCGGCAAGGGCCTGGAGTGGCTGGGCGTGATGTGGACCGGCGGCGGCACCAACTACAACAGCGCCTTCATGAGCAGGCTGAGCATCAGCAGGGACAACAGCAAGAGCCAGGTGTTCCTGAAGATGAACAGCCTGCAGACCGACGACACCGCCATCTACTACTGCGTGAGGAGCTACTACGGCAGCGCCATGGACTACTGGGGCCAGGGCACCAGCGTGACCGTGAGCAGCGAGGTGCAGCTGGAGGAGAGCGGCCCCGGCCTGGTGGCCCCCAGCCAGAGCCTGAGCATCACCTGCACCGTGAGCGGCTTCAGCCTGACCAGGTACGACATCAGCTGGATCAGGCAGCCCCCCGGCAAGGGCCTGGAGTGGCTGGGCGTGATGTGGACCGGCGGCGGCACCAACTACAACAGCGCCTTCATGAGCAGGCTGAGCATCAGCAGGGACAACAGCAAGAGCCAGGTGTTCCTGAAGATGAACAGCCTGCAGACCGACGACACCGCCATCTACTACTGCGTGAGGAGCTACTACGGCAGCGCCATGGACTACTGGGGCCAGGGCACCAGCGTGACCGTGAGCAGC 서열번호 19SEQ ID NO: 19
경쇄가변부위
염기서열
light chain variable region
base sequence
GACATCGTGATGACCCAGAGCAGCAGCTACCTGAGCGTGAGCCTGGGCGGCAGGGTGACCATCACCTGCAAGGCCAGCGACCACATCAACAACTGGCTGGCCTGGTACCAGCAGAAGCCCGGCAACGCCCCCAGGCTGCTGATCAGCGGCGCCACCAGCCTGGAGACCGGCGTGCCCAGCAGGTTCAGCGGCAGCGGCAGCGGCAAGGACTACACCCTGAGCATCACCAGCCTGCAGACCGAGGACGTGGCCACCTACTACTGCCAGCAGCACTGGAGCACCCCCTTCACCTTCGGCAGCGGCACCAAGCTGGAGATCAAGGACATCGTGATGACCCAGAGCAGCAGCTACCTGAGCGTGAGCCTGGGCGGCAGGGTGACCATCACCTGCAAGGCCAGCGACCACATCAACAACTGGCTGGCCTGGTACCAGCAGAAGCCCGGCAACGCCCCCAGGCTGCTGATCAGCGGCGCCACCAGCCTGGAGACCGGCGTGCCCAGCAGGTTCAGCGGCAGCGGCAGCGGCAAGGACTACACCCTGAGCATCACCAGCCTGCAGACCGAGGACGTGGCCACCTACTACTGCCAGCAGCACTGGAGCACCCCCTTCACCTTCGGCAGCGGCACCAAGCTGGAGATCAAG 서열번호 20SEQ ID NO: 20
실시예 2: 선별한 항체의 CD22에 대한 특이성 확인 - ELISA 분석Example 2: Confirmation of specificity of the selected antibody for CD22 - ELISA analysis
본 발명에서는 상기 실시예 1에서 확립한 3F11 및 7B5 항체의 CD22에 대한 특이성을 확인하기 위해, ELISA 분석을 수행하였다. In the present invention, in order to confirm the specificity of the 3F11 and 7B5 antibodies established in Example 1 for CD22, ELISA analysis was performed.
먼저, CD22 펩타이드를 코딩하기 위해, CD22-His tag(CD22 extracellular domain; ACRObiosystems Inc.)를 100 ng/웰이 되도록 96-웰 플레이트에 분주한 다음, 4℃에서 하룻밤 동안 반응시켰다. 그 다음, 3% BSA가 포함된 1 X PBST를 처리한 후, 상온에서 30분 동안 블로킹시켰다.First, in order to encode the CD22 peptide, CD22-His tag (CD22 extracellular domain; ACRObiosystems Inc.) was dispensed in a 96-well plate at a concentration of 100 ng/well, and then reacted at 4°C overnight. Then, after treatment with 1 X PBST containing 3% BSA, blocking at room temperature for 30 minutes.
3F11 및 7B5 항체를 각 웰에 처리한 다음, 상온에서 2시간 동안 반응시킨 후, 1 X PBST로 3번 세척하였다. 2차 항체(anti-HRP, 1:10,000)를 처리하여 상온에서 30분 동안 반응시킨 후, 1 X PBST로 3번 세척한 다음, 발색을 위해 TMB를 처리하여 상온에서 5분 동안 반응시켰다. 마지막으로 1N H 2SO 4의 정지액(stop solution)을 처리하여 반응을 종료시킨 다음, 450nm에서 흡광도를 측정하였다.3F11 and 7B5 antibodies were treated in each well, and then reacted at room temperature for 2 hours, and then washed 3 times with 1 X PBST. Secondary antibody (anti-HRP, 1:10,000) was treated and reacted at room temperature for 30 minutes, washed 3 times with 1 X PBST, and then treated with TMB for color development and reacted at room temperature for 5 minutes. Finally, the reaction was terminated by treatment with a stop solution of 1N H 2 SO 4 , and then the absorbance was measured at 450 nm.
ELISA 실험 조건ELISA Experimental Conditions
ImmobilizationImmobilization Corning (Cat#. 3690)Corning (Cat#. 3690)
AntigenAntigen CD22-His tag, 100 ng/wellCD22-His tag, 100 ng/well
Primary AbPrimary Ab 0.4 ㎍/㎖0.4 μg/ml
Secondary Ab-HRPSecondary Ab-HRP 1:10,0001:10,000
TMB substrate incubationTMB substrate incubation 5 min5 min
Measurement filterMeasurement filter 450nm450nm
Plate readerplate reader Infinite F50Infinite F50
ELISA 실험 결과ELISA test results
항체 종류Antibody type OD450 측정값OD450 measurements
3F113F11 1.86161.8616
7B57B5 2.66202.6620
그 결과, 표 4에 나타난 바와 같이, 본 발명에서 선별한 항체가 모두 CD22에 특이적으로 결합하는 것을 확인하였다.As a result, as shown in Table 4, it was confirmed that all of the antibodies selected in the present invention specifically bind to CD22.
실시예 3: 선별한 항체의 CD22에 대한 특이성 확인 - FACS 분석Example 3: Confirmation of specificity for CD22 of the selected antibody - FACS analysis
본 발명에서는 상기 실시예 1에서 확립한 3F11 및 7B5 항체의 CD22에 대한 특이성을 확인하기 위해, FACS 분석을 수행하였다. In the present invention, in order to confirm the specificity of the 3F11 and 7B5 antibodies established in Example 1 for CD22, FACS analysis was performed.
먼저, CD22를 발현하는 비세포림프종 U2932 (B-cell lymphoma U2932 cell)이 1x10 6개와 3F11 및 7B5 항체 1 ㎍ 각각을 30분간 반응시킨 다음, 2차 항체로 표면(surface)을 염색한 후, 유세포분석기로 측정하였다.First, CD22-expressing non-cell lymphoma U2932 (B-cell lymphoma U2932 cell) was reacted with 1×10 6 cells and 1 μg of 3F11 and 7B5 antibodies for 30 minutes, and then the surface was stained with a secondary antibody, followed by flow cytometry. Measured with an analyzer.
양성대조군으로 PE-컨쥬게이션된 항-CD22 항체(PE-conjugated anti-CD22 antibody; Biolegend Inc., cat# 302506, 미국)를 사용하였으며, 2차 항체로는 PE-컨쥬게이션된 항-마우스 IgG 항체(PE-conjugated goat anti-mouse IgG; Biolegend Inc., cat# 405307, 미국)를 사용하였다. As a positive control, a PE-conjugated anti-CD22 antibody (PE-conjugated anti-CD22 antibody; Biolegend Inc., cat# 302506, USA) was used, and as a secondary antibody, a PE-conjugated anti-mouse IgG antibody was used. (PE-conjugated goat anti-mouse IgG; Biolegend Inc., cat# 405307, USA) was used.
그 결과, 도 1에 나타난 바와 같이 3F11 및 7B5 항체 모두 CD22를 발현하는 세포와 특이적으로 결합하는 것을 확인하였다.As a result, as shown in FIG. 1 , it was confirmed that both the 3F11 and 7B5 antibodies specifically bind to CD22-expressing cells.
실시예 4: CD22를 표적으로 하는 키메라 항원 수용체(CAR) 발현 벡터 제작Example 4: Construction of a chimeric antigen receptor (CAR) expression vector targeting CD22
본 발명에서는 상기 실시예 1에서 제조한 3F11 및 7B5 항체를 이용하여, CD22를 표적으로 하는 키메라 항원수용체(CAR)를 발현하는 렌티바이러스 벡터(CD22-CAR 렌티바이러스)를 제조하였다. In the present invention, a lentiviral vector (CD22-CAR lentivirus) expressing a chimeric antigen receptor (CAR) targeting CD22 was prepared using the 3F11 and 7B5 antibodies prepared in Example 1 above.
도 2의 모식도에 나타난 바와 같이, EF1α 프로모터(서열번호 27); 시그널 펩타이드를 코딩하는 폴리뉴클레오타이드(서열번호 28); CD22-결합 도메인을 코딩하는 폴리뉴클레오타이드(서열번호 24로 표시되는 3F11 또는 서열번호 26로 표시되는 7B5); CD8 힌지 부위를 코딩하는 폴리뉴클레오타이드(서열번호 29); 막관통 도메인을 코딩하는 폴리뉴클레오타이드(서열번호 730); 4-1BB(공동자극도메인)을 코딩하는 폴리뉴클레오타이드(서열번호 31); CD3ζ세포내신호전달도메인)을 코딩하는 폴리뉴클레오타이드(서열번호 32); 및 WPRE를 코딩하는 폴리뉴클레오타이드(서열번호 33)로 구성된 CAR DNA를 생체외(in vitro)에서 합성하여 3세대 렌티바이러스 벡터에 삽입하였다. As shown in the schematic diagram of Figure 2, EF1α promoter (SEQ ID NO: 27); a polynucleotide encoding a signal peptide (SEQ ID NO: 28); a polynucleotide encoding a CD22-binding domain (3F11 represented by SEQ ID NO: 24 or 7B5 represented by SEQ ID NO: 26); a polynucleotide encoding the CD8 hinge region (SEQ ID NO: 29); a polynucleotide encoding a transmembrane domain (SEQ ID NO: 730); a polynucleotide encoding 4-1BB (costimulatory domain) (SEQ ID NO: 31); a polynucleotide encoding CD3ζ intracellular signaling domain (SEQ ID NO: 32); And CAR DNA composed of a polynucleotide encoding WPRE (SEQ ID NO: 33) was synthesized in vitro and inserted into a third-generation lentiviral vector.
렌티바이러스 벡터는 pMDLg/pRRE(Addgene, cat# #12251) pMD2.G(Addgene, cat##12259), pRSV-Rev(Addgene, cat##12253)의 세 가지 벡터 함께 Lenti-X 293T 세포로 공동 감염(co-transfection)시킨 다음, CD22-CAR 렌티바이러스를 생산하였다. 공동 감염을 위해 Lipofectamine 3000 transfection kit(Invitrogen, cat# L3000-015)와 Opti-MEM+GlutaMAX(gibco, cat# 51985-034) 배지를 사용하여 세 가지 벡터와 Lenti-X 293T 세포를 6시간 배양하였다.Lentiviral vectors were co-transfected into Lenti-X 293T cells with three vectors, pMDLg/pRRE (Addgene, cat# #12251), pMD2.G (Addgene, cat##12259), and pRSV-Rev (Addgene, cat##12253). After co-transfection, CD22-CAR lentivirus was produced. For co-infection, three vectors and Lenti-X 293T cells were cultured for 6 hours using Lipofectamine 3000 transfection kit (Invitrogen, cat# L3000-015) and Opti-MEM+GlutaMAX (gibco, cat# 51985-034) medium. .
실시예 5: CD22-CAR-T 세포 제조Example 5: CD22-CAR-T cell preparation
본 발명에서는 상기 실시예 4에서 제조한 CD22-CAR 렌티바이러스 벡터를 T 세포에 형질전환시켜 CD22-CAR-T 세포를 제조하였다. In the present invention, the CD22-CAR lentiviral vector prepared in Example 4 was transformed into T cells to prepare CD22-CAR-T cells.
구체적으로, 도 3에 나타난 바와 같이, 혈액에서 말초혈액단핵세포(peripheral blood mononuclear cell, PBMC)를 분리한 다음, T 세포활성화비드(T cell activation bead; Miltenyl Biotec, cat# 130-091-441)를 사용해 T 세포를 활성화시켰다. 활성화된 T 세포에 상기 실시예 4에서 제조한 CD22-CAR 렌티바이러스를 T 세포에 형질 도입시켜 CD22-CAR-T 세포를 제조하였으며, Lenti-boost-p를 사용하여 형질 도입 효율을 증가시켰다. Specifically, as shown in FIG. 3, after separating peripheral blood mononuclear cells (PBMC) from blood, T cell activation beads (T cell activation bead; Miltenyl Biotec, cat# 130-091-441) was used to activate T cells. CD22-CAR-T cells were prepared by transducing the activated T cells with the CD22-CAR lentivirus prepared in Example 4 into T cells, and the transduction efficiency was increased using Lenti-boost-p.
CD22-CAR-T 세포의 CD22 펩타이드 결합능은 유세포분석(Flow Cytometry) 방법을 통해 확인하였다. 상기에서 제조한 CD22-CAR-T세포를 FITC-CD22 단백질과 anti-CD3, anti-CD4, anti-CD8 항체와 반응시킨 후 FACS 기계를 이용해 형광 세기를 측정하였다. 분석과정에서 CD3를 발현하는 세포를 T 세포로하여 T 세포안에서 FITC 발현 정도를 확인하였다.CD22 peptide binding capacity of CD22-CAR-T cells was confirmed by flow cytometry. The CD22-CAR-T cells prepared above were reacted with FITC-CD22 protein and anti-CD3, anti-CD4, and anti-CD8 antibodies, and then fluorescence intensity was measured using a FACS machine. In the course of the analysis, CD3 expressing cells were used as T cells, and the level of FITC expression in T cells was confirmed.
그 결과, 도 4에 나타난 바와 같이, CD22 펩타이드 증가에 따라, CD22와 결합하는 CD4 또는 CD8를 발현하는 CD22-CAR-T 세포가 증가하는 것을 확인하였다. 이는 본 발명에서 제조한 CD22-CAR-T 세포가 효과적으로 CD22와 결합하는 것을 의미한다.As a result, as shown in FIG. 4 , it was confirmed that CD22-CAR-T cells expressing CD4 or CD8 binding to CD22 increased as the CD22 peptide increased. This means that the CD22-CAR-T cells prepared in the present invention effectively bind to CD22.
실시예 6: CD22 펩타이드에 의한 CD22-CAR-T 세포 활성화 확인Example 6: Confirmation of CD22-CAR-T cell activation by CD22 peptide
본 발명에서는 상기 실시예 5에서 제조한 CD22-CAR-T 세포가 CD22 펩타이드에 의해 활성화되는지 확인하기 위해, 표적세포의 존재 하에 CD22-CAR-T 세포에 의한 IFNγ 발현 정도를 확인하였다.In the present invention, in order to confirm that the CD22-CAR-T cells prepared in Example 5 are activated by the CD22 peptide, the level of IFNγ expression by the CD22-CAR-T cells in the presence of target cells was checked.
표적세포는 CD22를 발현하지 않는 K562 세포(ATCC, cat#CCL-243) 및 CD22를 발현하는 U2932 세포(DSMZ, cat#ACC-633)를 이용하였으며, CD22-CAR-T 세포와 표적세포를 2:1, 1:1. 0.5:1, 0:1 비율로 일정시간 반응시킨 후 surface & intra antibody로 염색하여 FACS 측정하였다 (CD22 protein, INF-r, CD107a, CD3, CD4, CD8 염색). 0:1(CAR-T only)를 기준으로 표적세포와 반응시킨 CD22-CAR-T의 IFNγ 발현 정도를 확인하였다.As target cells, K562 cells that do not express CD22 (ATCC,  cat#CCL-243) and U2932 cells that express CD22 (DSMZ, cat#ACC-633) were used. :1, 1:1. After reacting for a certain period of time at a ratio of 0.5:1, 0:1, surface & intra antibody staining was performed for FACS measurement (CD22 protein, INF-r, CD107a, CD3, CD4, CD8 staining). The level of IFNγ expression of CD22-CAR-T reacted with target cells was confirmed on the basis of 0:1 (CAR-T only).
그 결과 도 5a 및 도 5b에 나타난 바와 같이, CD22를 발현하지 않는 K562 세포에서는 T 세포가 활성화되지 않은 반면, CD22를 발현하는 U2932 세포의 존재 하에 T 세포가 활성화되어 IFNγ 발현이 증가하는 것을 확인하였다.As a result, as shown in FIGS. 5a and 5b, T cells were not activated in K562 cells that do not express CD22, whereas T cells were activated in the presence of U2932 cells expressing CD22, thereby increasing IFNγ expression. .
실시예 7: CD22 발현 세포에 대한 CD22-CAR-T 세포의 사멸 효과 확인Example 7: Confirmation of the killing effect of CD22-CAR-T cells on CD22-expressing cells
본발명에서는 CD22-CAR-T 세포에 의한 표적세포의 사멸효과를 확인하였다. In the present invention, the killing effect of target cells by CD22-CAR-T cells was confirmed.
표적세포로 CD22를 발현하지 않는 K562 세포와 CD22를 발현하는 U2932 세포를 이용하였으며, CD22-CAR-T 세포와 1:4, 1:2, 1:1, 1:0.5 및 1:0.25 비율이 되도록 혼합한 다음, 루미네센스(CytoTox-Glo Cytotoxicity Assay, Promega, cat# G9291)를 측정하였다. 측정한 값으로 하기 수학식 1을 이용하여 세포 사멸 정도를 계산하였다. As target cells, K562 cells, which do not express CD22, and U2932 cells, which express CD22, were used so that the ratios of CD22-CAR-T cells were 1:4, 1:2, 1:1, 1:0.5 and 1:0.25. After mixing, luminescence (CytoTox-Glo Cytotoxicity Assay, Promega, cat# G9291) was measured. The degree of cell death was calculated using Equation 1 below as the measured value.
[수학식 1][Equation 1]
% Cytotoxicity = [(Experimental - Effector Spontaneous - Target Spontaneous) / (Target Maximum - Target Spontaneous)] X 100% Cytotoxicity = [(Experimental - Effector Spontaneous - Target Spontaneous) / (Target Maximum - Target Spontaneous)] X 100
Experimental: 표적세포 및 CAR-T 세포 복합 배양의 배지로부터 도출된 발광(Luminescence)값Experimental: Luminescence value derived from the medium of the target cell and CAR-T cell complex culture
Effector Spontaneous: CAR-T 세포만의 배지로부터 도출된 발광값Effector Spontaneous: Luminescence value derived from the medium of CAR-T cells only
Target Spontaneous: 표적세포만의 배지로부터 도출된 발광값Target Spontaneous: Luminescence value derived from the medium of target cells only
Target Maximum: 표적세포의 100% 용해(용해시약 (Lysis Reagent) 이용)로부터 도출된 발광값Target Maximum: Luminescence value derived from 100% lysis of target cells (using Lysis Reagent)
그 결과, 도 6에 나타난 바와 같이 본 발명의 CD22-CAR-T 세포는 CD22를 발현하는 U2932 세포를 특이적으로 사멸시키는 것을 확인하였다.As a result, as shown in FIG. 6 , it was confirmed that the CD22-CAR-T cells of the present invention specifically killed U2932 cells expressing CD22.
본 발명에서 선별한 항체는 CD22를 발현하는 세포를 특이적으로 인식하는 것을 확인하였으며, 상기 확립된 항체를 이용하여 CD22를 표적으로 하는 키메라 항원 수용체(CAR) 및 CAR-T 세포는 CD22와 효과적으로 결합할 뿐만 아니라, CD22와 결합한 CAR-T 세포의 활성화가 이루어진 것을 확인하였다. It was confirmed that the antibody selected in the present invention specifically recognizes CD22-expressing cells, and using the established antibody, the chimeric antigen receptor (CAR) and CAR-T cells targeting CD22 effectively bind to CD22 In addition, it was confirmed that the activation of CAR-T cells bound to CD22 was made.
또한, 본 발명의 CAR-T 세포는 CD22를 발현하는 세포를 효과적으로 사멸시키는 것을 확인하였으므로, 본 발명의 CD22에 특이적인 항체, CD22를 표적으로 하는 키메라 항원 수용체 및 CAR-T 세포는 CD22 발현과 관련된 질환 예방 또는 치료용 조성물로 유용하게 활용할 수 있다. In addition, since it was confirmed that the CAR-T cells of the present invention effectively kill CD22-expressing cells, the CD22-specific antibody, the chimeric antigen receptor targeting CD22, and CAR-T cells of the present invention are related to CD22 expression. It can be usefully used as a composition for preventing or treating diseases.

Claims (14)

  1. (a) 서열번호 1의 아미노산으로 표시되는 CDR1 영역, 서열번호 2의 아미노산으로 표시되는 CDR2 영역 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역, 서열번호 5의 아미노산으로 표시되는 CDR2 영역 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위; 또는 (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and the amino acid of SEQ ID NO: 4 a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 5 and a CDR3 region represented by the amino acid of SEQ ID NO: 6; or
    (b) 서열번호 11의 아미노산으로 표시되는 CDR1 영역, 서열번호 12의 아미노산으로 표시되는 CDR2 영역 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역, 서열번호 15의 아미노산으로 표시되는 CDR2 영역 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위로 구성된 CD22에 특이적으로 결합하는 항체 또는 이의 단편.(b) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid of SEQ ID NO: 16.
  2. 제1항에 있어서, 상기 (a) 항체는 서열번호 7의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 8의 아미노산으로 표시되는 경쇄 가변부위로,According to claim 1, wherein (a) the antibody has a heavy chain variable region represented by the amino acid of SEQ ID NO: 7 and a light chain variable region represented by the amino acid of SEQ ID NO: 8,
    상기 (b) 항체는 서열번호 17의 아미노산으로 표시되는 중쇄 가변부위 및 서열번호 18의 아미노산으로 표시되는 경쇄 가변부위로 구성되는 것을 특징으로 하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편.The (b) antibody is an antibody or fragment thereof that specifically binds to CD22, characterized in that it consists of a heavy chain variable region represented by the amino acid of SEQ ID NO: 17 and a light chain variable region represented by the amino acid of SEQ ID NO: 18.
  3. 제1항 또는 제2항의 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 코딩하는 폴리뉴클레오타이드.A polynucleotide encoding an antibody or fragment thereof that specifically binds to the CD22 of claim 1 or 2 .
  4. 제1항 또는 제2항의 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 코딩하는 폴리뉴클레오타이드 포함하는 벡터.A vector comprising a polynucleotide encoding an antibody or fragment thereof that specifically binds to the CD22 of claim 1 or 2.
  5. 제4항의 벡터로 형질전환된 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 생산하는 재조합 세포.A recombinant cell producing an antibody or fragment thereof that specifically binds to CD22 transformed with the vector of claim 4 .
  6. CD22-결합 도메인; CD22-binding domain;
    막관통 도메인(transmembrane domain); transmembrane domain;
    공동자극 도메인(costimulatory domain); 및 costimulatory domain; and
    세포 내 신호전달 도메인(intracellular signal transduction domain)을 포함하는 키메릭 항원 수용체(chimeric antigen receptor: CAR)로,A chimeric antigen receptor (CAR) comprising an intracellular signal transduction domain,
    상기 CD22-결합 도메인은 (a) 서열번호 1의 아미노산으로 표시되는 CDR1 영역, 서열번호 2의 아미노산으로 표시되는 CDR2 영역 및 서열번호 3의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 4의 아미노산으로 표시되는 CDR1 영역, 서열번호 5의 아미노산으로 표시되는 CDR2 영역 및 서열번호 6의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편; 또는The CD22-binding domain comprises (a) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 1, the CDR2 region represented by the amino acid of SEQ ID NO: 2 and the CDR3 region represented by the amino acid of SEQ ID NO: 3 and SEQ ID NO: An antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region represented by the amino acid of 4, a CDR2 region represented by the amino acid of SEQ ID NO: 5, and a CDR3 region represented by the amino acid of SEQ ID NO: 6. ; or
    (b) 서열번호 11의 아미노산으로 표시되는 CDR1 영역, 서열번호 12의 아미노산으로 표시되는 CDR2 영역 및 서열번호 13의 아미노산으로 표시되는 CDR3 영역을 포함하는 중쇄 가변부위 및 서열번호 14의 아미노산으로 표시되는 CDR1 영역, 서열번호 15의 아미노산으로 표시되는 CDR2 영역 및 서열번호 16의 아미노산으로 표시되는 CDR3 영역을 포함하는 경쇄 가변 부위를 포함하는 CD22에 특이적으로 결합하는 항체 또는 이의 단편인 것을 특징으로 하는 키메릭 항원 수용체.(b) a heavy chain variable region comprising the CDR1 region represented by the amino acid of SEQ ID NO: 11, the CDR2 region represented by the amino acid of SEQ ID NO: 12 and the CDR3 region represented by the amino acid of SEQ ID NO: 13 and the amino acid of SEQ ID NO: 14 A key characterized in that it is an antibody or fragment thereof that specifically binds to CD22 comprising a light chain variable region comprising a CDR1 region, a CDR2 region represented by the amino acid of SEQ ID NO: 15, and a CDR3 region represented by the amino acid of SEQ ID NO: 16 Merrick antigen receptor.
  7. 제6항에 있어서, 상기 막관통 도메인은 CD8α, CD4, CD28, CD137, CD80, CD86, CD152 및 PD1로 구성된 군에서 선택되는 단백질이며,7. The method of claim 6, wherein the transmembrane domain is a protein selected from the group consisting of CD8α, CD4, CD28, CD137, CD80, CD86, CD152 and PD1,
    공동자극 도메인은 CD28, 4-1BB, OX-40 및 ICOS로 구성된 군에서 선택되는 단백질이고, the costimulatory domain is a protein selected from the group consisting of CD28, 4-1BB, OX-40 and ICOS,
    상기 신호전달 도메인은 CD3ζ인 것을 특징으로 하는 키메릭 항원 수용체. The signaling domain is a chimeric antigen receptor, characterized in that CD3ζ.
  8. 제6항에 있어서, 상기 CD22-결합 도메인의 C 말단 및 막관통 도메인의 N 말단 사이에 힌지 부위(hinge region)가 추가로 포함되는 것을 특징으로 하는 키메릭 항원 수용체.The chimeric antigen receptor according to claim 6, wherein a hinge region is further included between the C terminus of the CD22-binding domain and the N terminus of the transmembrane domain.
  9. 제6항 내지 제8항 중 어느 한 항의 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드.A polynucleotide encoding the chimeric antigen receptor (CAR) of any one of claims 6-8.
  10. 제6항 내지 제8항 중 어느 한 항의 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드를 포함하는 벡터.A vector comprising a polynucleotide encoding the chimeric antigen receptor (CAR) of any one of claims 6 to 8.
  11. 제6항 내지 제8항 중 어느 한 항의 키메릭 항원 수용체(CAR)를 코딩하는 폴리뉴클레오타이드 또는 상기 폴리뉴클레오타이드를 포함하는 벡터를 포함하는 면역 이펙터 세포.An immune effector cell comprising a polynucleotide encoding the chimeric antigen receptor (CAR) of any one of claims 6 to 8 or a vector comprising the polynucleotide.
  12. 제1항 또는 제2항의 CD22에 특이적으로 결합하는 항체 또는 이의 단편; An antibody or fragment thereof that specifically binds to the CD22 of claim 1 or 2;
    또는 제11항의 면역 이펙터 세포를 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 예방 또는 치료용 약학적 조성물.Or a pharmaceutical composition for preventing or treating a disease mediated by a cell expressing CD22, including the immune effector cell of claim 11 .
  13. 제12항에 있어서, 상기 CD22를 발현하는 세포에 의해 매개되는 질환은 림프종, 비호치킨 림프종(non-Hogkins lymphoma: NHL), 공격적 NHL, 재발성 공격적 NHL, 재발성 지연성 NHL, 불응성 NHL, 불응성 지연성 NHL, 만성 림프성 백혈병(chronic lymphocytic leukemia: CLL), 소형 림프성 림프종, 백혈병, 모발성 세포 백혈병(hairy cell leukemia: HCL), 급성 림프성 백혈병(acute lymphocytic leukemia: ALL), 버킷트 림프종 및 외투 세포 림프종로 구성된 군에서 선택되는 것을 특징으로 하는 CD22를 발현하는 세포에 의해 매개되는 질환의 예방 또는 치료용 약학적 조성물.13. The method of claim 12, wherein the disease mediated by cells expressing CD22 is lymphoma, non-Hogkins lymphoma (NHL), aggressive NHL, relapsed aggressive NHL, relapsed delayed NHL, refractory NHL, Refractory delayed NHL, chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma, leukemia, hairy cell leukemia (HCL), acute lymphocytic leukemia (ALL), Burkitt A pharmaceutical composition for the prophylaxis or treatment of a disease mediated by cells expressing CD22, characterized in that it is selected from the group consisting of tree lymphoma and mantle cell lymphoma.
  14. 제1항 또는 제2항의 CD22에 특이적으로 결합하는 항체 또는 이의 단편을 포함하는 CD22를 발현하는 세포에 의해 매개되는 질환의 진단 또는 모니터링용 조성물.A composition for diagnosing or monitoring a disease mediated by cells expressing CD22, comprising an antibody or fragment thereof that specifically binds to CD22 according to claim 1 or 2.
PCT/KR2021/004566 2020-05-19 2021-04-12 Cd22-specific antibody and use thereof WO2021235697A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20200059647 2020-05-19
KR10-2020-0059647 2020-05-19
KR1020200169497A KR20210143097A (en) 2020-05-19 2020-12-07 Antibody specific for CD22 and uses thereof
KR10-2020-0169497 2020-12-07

Publications (1)

Publication Number Publication Date
WO2021235697A1 true WO2021235697A1 (en) 2021-11-25

Family

ID=78700384

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/004566 WO2021235697A1 (en) 2020-05-19 2021-04-12 Cd22-specific antibody and use thereof

Country Status (2)

Country Link
KR (1) KR20210143097A (en)
WO (1) WO2021235697A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024000259A1 (en) * 2022-06-29 2024-01-04 上海吉倍生物技术有限公司 Antibody specifically binding to cd22, preparation method therefor and use thereof on bispecific cart

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101552735B1 (en) * 2006-12-01 2015-09-14 메다렉스, 엘.엘.시. 22 human antibodies that bind cd22 and uses thereof
US9518115B2 (en) * 2014-02-25 2016-12-13 Immunomedics, Inc. Humanized anti-CD22 antibody
KR20180030656A (en) * 2015-07-16 2018-03-23 유씨비 바이오파마 에스피알엘 Antibody molecules that bind to CD22
US20190085082A1 (en) * 2016-03-15 2019-03-21 Cancer Research Technology Limited Antibodies and related molecules and uses thereof
WO2019126756A1 (en) * 2017-12-22 2019-06-27 Teneobio, Inc. Heavy chain antibodies binding to cd22

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101552735B1 (en) * 2006-12-01 2015-09-14 메다렉스, 엘.엘.시. 22 human antibodies that bind cd22 and uses thereof
US9518115B2 (en) * 2014-02-25 2016-12-13 Immunomedics, Inc. Humanized anti-CD22 antibody
KR20180030656A (en) * 2015-07-16 2018-03-23 유씨비 바이오파마 에스피알엘 Antibody molecules that bind to CD22
US20190085082A1 (en) * 2016-03-15 2019-03-21 Cancer Research Technology Limited Antibodies and related molecules and uses thereof
WO2019126756A1 (en) * 2017-12-22 2019-06-27 Teneobio, Inc. Heavy chain antibodies binding to cd22

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024000259A1 (en) * 2022-06-29 2024-01-04 上海吉倍生物技术有限公司 Antibody specifically binding to cd22, preparation method therefor and use thereof on bispecific cart

Also Published As

Publication number Publication date
KR20210143097A (en) 2021-11-26

Similar Documents

Publication Publication Date Title
WO2021235696A1 (en) Cd22-specific antibody and use thereof
WO2018124766A2 (en) Chimeric antigen receptor and natural killer cells expressing same
WO2020111913A1 (en) Anti-4-1bb antibody and use thereof
WO2021256724A1 (en) Chimeric antigen receptor targeting bcma and use thereof
WO2019225787A1 (en) Anti-b7-h3 antibody and use thereof
WO2015058573A1 (en) Monoclonal antibody for antagonizing and inhibiting binding of programmed death (pd-1) to ligand thereof and coding sequence and use thereof
WO2019112347A2 (en) Antibody or antigen binding fragment thereof for specifically recognizing b cell malignancy, chimeric antigen receptor comprising same, and use thereof
WO2020032784A1 (en) Chimeric antigen receptor binding to hla-dr, and car-t cell
WO2017030370A1 (en) Chimeric antibody receptor to which anti-cotinine antibody is linked, and use thereof
WO2020005003A1 (en) Monoclonal antibody specifically binding to lag-3 and use thereof
US20230242661A1 (en) Engineered immune cell expressing nk inhibitory molecule and use thereof
WO2021210939A1 (en) Anti-her2 affibody, and switchable chimeric antigen receptor using same as switch molecule
WO2022030730A1 (en) Anti-mesothelin chimeric antigen receptor specifically binding to mesothelin
WO2021235697A1 (en) Cd22-specific antibody and use thereof
WO2021153979A1 (en) Fusion protein comprising anti-taa antibody, anti-pd-l1 antibody, and il-2, and uses thereof
WO2023277361A1 (en) Mesothelin-specific antibodies and use thereof
WO2021060914A1 (en) Anti-claudin-3 chimeric antigen receptor
KR20230011959A (en) Engineered Immune Cells for Allogeneic Transplantation
WO2021246637A1 (en) Antibody specific to cd22, and use thereof
WO2022181992A1 (en) Cd22-specific humanized antibody and use thereof
WO2022231032A1 (en) Anti-cntn4-specific antibodies and use thereof
WO2022124764A1 (en) Antibody specific for cd47 and use thereof
WO2021182929A1 (en) Bcma-specific antibody and chimeric antigen receptor
WO2022124765A1 (en) Gpc3-specific antibody and use thereof
WO2023090704A1 (en) Antibody specific for humanized cd22 and chimeric antigen receptor using same

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: 21808284

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: 21808284

Country of ref document: EP

Kind code of ref document: A1