WO2022095803A1 - 靶向cd7的人源化抗体及其用途 - Google Patents

靶向cd7的人源化抗体及其用途 Download PDF

Info

Publication number
WO2022095803A1
WO2022095803A1 PCT/CN2021/127570 CN2021127570W WO2022095803A1 WO 2022095803 A1 WO2022095803 A1 WO 2022095803A1 CN 2021127570 W CN2021127570 W CN 2021127570W WO 2022095803 A1 WO2022095803 A1 WO 2022095803A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
cells
humanized antibody
chain variable
Prior art date
Application number
PCT/CN2021/127570
Other languages
English (en)
French (fr)
Inventor
周亚丽
姜小燕
陈功
任江涛
贺小宏
王延宾
韩露
Original Assignee
南京北恒生物科技有限公司
江苏浦珠生物医药科技有限公司
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 南京北恒生物科技有限公司, 江苏浦珠生物医药科技有限公司 filed Critical 南京北恒生物科技有限公司
Priority to JP2023524926A priority Critical patent/JP2023547425A/ja
Priority to EP21888500.2A priority patent/EP4219554A4/en
Priority to US18/034,170 priority patent/US20230399398A1/en
Priority to KR1020237012746A priority patent/KR20230098158A/ko
Publication of WO2022095803A1 publication Critical patent/WO2022095803A1/zh

Links

Images

Classifications

    • 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
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • 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
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention belongs to the field of immunotherapy. More specifically, the present invention relates to humanized antibodies targeting CD7, and their use in the prevention and/or treatment and/or diagnosis of diseases.
  • CD7 is a cell surface glycoprotein with a molecular weight of about 40 kDa, which belongs to the immunoglobulin superfamily. CD7 is expressed on most T cells, NK cells, myeloid cells, T cell acute lymphoblastic leukemia/lymphoma, acute myeloid leukemia and chronic myeloid leukemia. It has been reported that the CD7 molecule acts as a costimulatory signal during T cell activation through binding to its ligand K12/SECTM1.
  • T-ALL T-cell acute lymphoblastic leukemia
  • the present invention aims to provide a humanized antibody targeting CD7 and its use in disease prevention and/or treatment and/or diagnosis.
  • the present invention provides a CD7 humanized antibody comprising a light chain variable region and a heavy chain variable region, wherein the light chain variable region comprises the CDRs shown in SEQ ID NO: 1 -L1, CDR-L2 as shown in SEQ ID NO: 2, CDR-L3 as shown in SEQ ID NO: 3, the heavy chain variable region comprising CDR-H1 as shown in SEQ ID NO: 4, CDR-H2 as set forth in SEQ ID NO:5, CDR-H3 as set forth in SEQ ID NO:6, and the light chain variable region is selected from the group consisting of set forth in SEQ ID NO:8, 11, 14 and 17
  • the amino acid sequence of the heavy chain variable region is at least 90% identical to the amino acid sequence selected from the group consisting of SEQ ID NOs: 9, 12, 15 and 18.
  • the CD7 humanized antibody comprises a light chain variable region selected from the group consisting of SEQ ID NOs: 8, 11, 14 and 17 and a light chain variable region selected from the group consisting of SEQ ID NOs: 9, 12, 15 and 18 heavy chain variable region.
  • amino acid sequence of the CD7 humanized antibody is selected from the group consisting of SEQ ID Nos: 10, 13, 16 and 19.
  • the present invention also provides nucleic acid molecules encoding the aforementioned CD7 humanized antibodies.
  • the nucleic acid molecule encoding the CD7 humanized antibody has at least 90%, 91%, 92%, 93%, 94% of the nucleotide sequence selected from the group consisting of SEQ ID NOs: 20-23 , 95%, 96%, 97%, 98%, 99%, 100% sequence identity, and the CD7 humanized antibody encoded by it can specifically bind to CD7 antigen.
  • the nucleic acid molecule encoding the CD7 humanized antibody is selected from SEQ ID NOs: 20-23.
  • the present invention also provides a multispecific antibody (preferably a bispecific or trispecific antibody) comprising the CD7 humanized antibody as described above and one or more antibodies that specifically bind to other antigens the second antibody or antigen-binding portion thereof.
  • a multispecific antibody preferably a bispecific or trispecific antibody
  • the second antibody or antigen-binding portion thereof may be in the form of any antibody or antibody fragment, such as a full-length antibody, Fab, Fab', (Fab') 2 , Fv, scFv, scFv-scFv, minibody, Diabodies or sdAbs.
  • the present invention also provides a vector comprising a nucleic acid molecule encoding the above-mentioned CD7 humanized antibody or multispecific antibody, and a host cell expressing the CD7 humanized antibody or multispecific antibody.
  • the present invention also provides a chimeric antigen receptor comprising the CD7 humanized antibody of the present invention, a transmembrane domain and an intracellular signaling domain.
  • the chimeric antigen receptor further comprises one or more costimulatory domains.
  • the chimeric antigen receptor comprises a CD7 humanized antibody as provided herein or a multispecific antibody comprising the CD7 humanized antibody, a CD8 ⁇ transmembrane region, a CD28 or a 4-1BB costimulatory domain , and the CD3 ⁇ intracellular signaling domain.
  • the present invention also provides a nucleic acid molecule encoding a CD7-targeting chimeric antigen receptor as defined above, and a vector comprising said nucleic acid molecule.
  • the present invention also provides cells, preferably immune cells, such as T cells, NK cells, NKT cells, macrophages, dendritic cells, comprising a CD7-targeting chimeric antigen receptor as defined above.
  • immune cells such as T cells, NK cells, NKT cells, macrophages, dendritic cells, comprising a CD7-targeting chimeric antigen receptor as defined above.
  • the present invention also provides an antibody conjugate comprising the CD7 humanized antibody as defined in the present invention and a second functional structure, wherein the second functional structure is selected from Fc, radioisotopes, Structural moieties, detectable labels and drugs that prolong half-life.
  • the half-life extending moiety is selected from: the half-life extending moiety is selected from the binding structure of albumin, the binding structure of transferrin, polyethylene glycol molecules, recombinant polyethylene glycol molecules, Human serum albumin, fragments of human serum albumin, and albumin polypeptides (including antibodies) that bind human serum albumin.
  • the detectable label is selected from the group consisting of fluorophores, chemiluminescent compounds, bioluminescent compounds, enzymes, antibiotic resistance genes, and contrast agents.
  • the drug is selected from cytotoxins and immunomodulators.
  • the present invention also provides a detection kit comprising the humanized antibody, multispecific antibody, antibody conjugate or chimeric antigen receptor of the present invention.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the humanized antibody, chimeric antigen receptor, multispecific antibody or antibody conjugate of the present invention, and one or more pharmaceutical agents acceptable excipients.
  • the present invention also provides a method of treating and/or preventing and/or diagnosing a disease associated with CD7 expression, comprising administering to a subject a humanized antibody, chimeric antigen receptor, Multispecific antibodies, antibody conjugates or pharmaceutical compositions.
  • antibody has the broadest meaning as understood by those skilled in the art, and includes monoclonal antibodies (including whole antibodies), polyclonal antibodies, multivalent antibodies, multispecific antibodies (eg, bispecific antibodies) ), and antibody fragments or synthetic polypeptides carrying one or more CDR sequences capable of exhibiting the desired biological activity.
  • the antibodies of the invention can be of any class (eg, IgG, IgE, IgM, IgD, IgA, etc.) or subclass (eg, IgGl, IgG2, IgG2a, IgG3, IgG4, IgAl, IgA2, etc.).
  • an intact antibody comprises two heavy chains and two light chains linked together by disulfide bonds, each light chain being disulfide bonded to its respective heavy chain in a "Y"-shaped structure.
  • Each heavy chain comprises a heavy chain variable region (VH) and a heavy chain constant region, wherein the heavy chain variable region comprises three complementarity determining regions (CDRs): CDR-H1, CDR-H2 and CDR-H3, the heavy chain constant
  • the region contains three constant domains: CH1, CH2 and CH3.
  • Each light chain includes a light chain variable region (VL) and a light chain constant region, wherein the light chain variable region includes three CDRs: CDR-L1, CDR-L2 and CDR-L3, and the light chain constant region includes a constant structure Domain CL.
  • the CDRs are separated by more conserved framework regions (FRs).
  • FRs conserved framework regions
  • the variable region of the heavy/light chain is responsible for antigen recognition and binding, while the constant region mediates the binding of antibodies to host tissues or factors, including various cells of the immune system (such as effector cells) and the first of the classical complement system. one component.
  • the boundaries of a given CDR or FR may vary depending on the protocol used for identification.
  • the Kabat scheme is based on structural alignment
  • the Chothia scheme is based on structural information.
  • Both Kabat and Chothia's scheme numbering is based on the most common antibody region sequence lengths, where insertions are provided by intervening letters (eg "30a") and deletions occur in some antibodies. These two schemes place certain insertions and deletions ("indels”) in different positions, resulting in different numbers.
  • the Contact scheme is based on the analysis of complex crystal structures and is similar in many respects to the Chothia numbering scheme.
  • the AbM protocol is a compromise between the Kabat and Chothia definitions and is based on the protocol used by Oxford Molecular's AbM antibody modeling software.
  • CDRs of a given antibody or regions thereof (eg, variable regions thereof) encompass the CDRs defined by any of the above schemes or other known schemes.
  • CDR a particular CDR (eg, CDR3) is designated to contain a given amino acid sequence
  • FRs of a given antibody or region thereof encompass the FRs defined by any of the above schemes or other known schemes.
  • a “humanized” antibody refers to an antibody in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all FR amino acid residues are derived from human FRs.
  • a "humanized form" of a non-human antibody refers to a variant of said non-human antibody that has undergone humanization to generally reduce immunogenicity to humans, while retaining the specificity and affinity of the parent non-human antibody.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (eg, an antibody from which the CDR residues are derived), eg, to restore or improve antibody specificity or affinity.
  • Humanized antibodies and methods of making them are well known to those skilled in the art, see eg, Almagro and Fransson, Front. Biosci. 13:1619-1633 (2008).
  • Human framework regions that can be used for humanization include, but are not limited to: framework regions selected using a "best fit" approach; framework regions derived from consensus sequences of human antibodies of a particular subset of light or heavy chain variable regions ; human mature (somatic mutation) framework regions or human germline framework regions; and framework regions obtained by screening FR libraries.
  • antibody fragment or "antigen-binding portion” encompasses only a portion of an intact antibody, typically comprising the antigen-binding site of the intact antibody and thus retaining the ability to bind antigen.
  • antibody fragments in the present invention include, but are not limited to: Fab, Fab', F(ab')2, Fd fragment, Fd', Fv fragment, scFv, disulfide-linked Fv (sdFv), heavy Chain variable region (VH) or light chain variable region (VL), linear antibodies, "diabodies” with two antigen binding sites, single domain antibodies, nanobodies, natural ligands for said antigen or functional fragments, etc. Accordingly, "antibodies” of the present invention encompass antibody fragments as defined above.
  • the CD7 humanized antibody of the invention is an scFv.
  • Single-chain antibody and “scFv” are used interchangeably herein and refer to an antibody comprising an antibody heavy chain variable region (VH) and light chain variable region (VL) linked by a linker.
  • the optimal length and/or amino acid composition of the linker can be selected.
  • the length of the linker significantly affects the variable region folding and interaction of scFv. In fact, intrachain folding can be prevented if shorter linkers are used (eg between 5-10 amino acids).
  • linker size and composition see, eg, Hollinger et al., 1993 Proc Natl Acad. Sci. U.S.A.
  • the scFv can comprise VH and VL linked in any order, eg, VH-linker-VL or VL-linker-VH.
  • the present invention provides a CD7 humanized antibody comprising a light chain variable region and a heavy chain variable region, wherein the light chain variable region comprises the CDRs shown in SEQ ID NO: 1 -L1, CDR-L2 as shown in SEQ ID NO: 2, CDR-L3 as shown in SEQ ID NO: 3, the heavy chain variable region comprising CDR-H1 as shown in SEQ ID NO: 4, CDR-H2 as set forth in SEQ ID NO:5, CDR-H3 as set forth in SEQ ID NO:6, and said light chain variable region with amino acids selected from the group consisting of SEQ ID NOs:8, 11, 14 and 17
  • the sequence has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% sequence identity, said heavy chain variable region and SEQ ID
  • the amino acid sequences shown in NO: 9, 12, 15 and 18 have at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%,
  • the CD7 humanized antibody comprises a light chain variable region selected from the group consisting of SEQ ID NOs: 8, 11, 14 and 17 and a light chain variable region selected from the group consisting of SEQ ID NOs: 9, 12, 15 and 18 The heavy chain variable region shown.
  • amino acid sequence of the CD7 humanized antibody is set forth in SEQ ID Nos: 10, 13, 16 and 19.
  • sequence identity refers to the degree to which two (nucleotide or amino acid) sequences have identical residues at the same positions in an alignment, and is usually expressed as a percentage. Preferably, identity is determined over the entire length of the sequences being compared. Therefore, two copies with the exact same sequence are 100% identical.
  • sequence identity can be determined using several algorithms can be used to determine sequence identity, such as Blast (Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402), Blast2 (Altschul et al. (1990) J. Mol. Biol. 215: 403-410), Smith-Waterman (Smith et al. (1981) J. Mol. Biol. 147: 195-197) and ClustalW.
  • the present invention also provides a multispecific antibody (preferably a bispecific or trispecific antibody) comprising a CD7 humanized antibody as above, which further comprises one or more first antibodies that specifically bind to other antigens. Secondary antibodies.
  • multispecific refers to an antigen binding protein that has polyepitopic specificity (ie, is capable of specifically binding to two, three or more different epitopes on a biomolecule or capable of specific binds epitopes on two, three or more different biomolecules).
  • bispecific means that an antigen binding protein has two different antigen binding specificities.
  • the secondary antibody may be in any antibody or antibody fragment format, eg, a full-length antibody, Fab, Fab', (Fab') 2 , Fv, scFv, scFv-scFv, minibody, diabody, or sdAb.
  • the second antibody targets an antigen selected from the group consisting of BCMA, CD4, CD5, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD25, CD33, CD37, CD38 , CD40, CD40L, CD46, CD52, CD54, CD74, CD80, CD126, CD138, B7, MUC-1, Ia, HM1.24, HLA-DR, tenascin, angiogenic factor, VEGF, PIGF, ED-B Fibronectin, oncogene, oncogene product, CD66a-d, necrotic antigen, Ii, IL-2, T101, TAC, IL-6, ROR1, TRAIL-R1(DR4), TRAIL-R2(DR5), tEGFR, Her2, L1-CAM, mesothelin, CEA, hepatitis B surface antigen, antifolate receptor, CD24, CD30, CD44, EGFR, E
  • the present invention relates to nucleic acid molecules encoding the CD7 humanized antibodies or multispecific antibodies of the present invention.
  • the nucleic acid of the present invention may be RNA, DNA or cDNA.
  • the nucleic acid of the invention is a substantially isolated nucleic acid.
  • the nucleic acid molecule encoding the CD7 humanized antibody has at least 90%, 91%, 92%, 93%, 94%, 95% of the nucleotide sequence selected from the group consisting of SEQ ID NOs: 20-23 %, 96%, 97%, 98%, 99%, 100% sequence identity, and the CD7 humanized antibody encoded by it can specifically bind to CD7 antigen.
  • the nucleic acid molecule encoding the CD7 humanized antibody is shown in SEQ ID NOs: 20-23.
  • the nucleic acid of the present invention may also be in the form of, may be present in and/or be part of a vector, such as a plasmid, cosmid or YAC.
  • the vector may in particular be an expression vector, ie a vector that provides for expression of the CD7 humanized antibody in vitro and/or in vivo (ie in a suitable host cell, host organism and/or expression system).
  • the expression vector typically comprises at least one nucleic acid molecule of the invention operably linked to one or more suitable expression control elements (eg, promoters, enhancers, terminators, etc.). Selection of such regulatory elements and their sequences for expression in a particular host is well known to those skilled in the art. Specific examples of regulatory elements and other elements useful or necessary for the expression of the CD7 humanized antibodies of the invention include, but are not limited to, promoters, enhancers, terminators, integration factors, selectable markers, leader sequences, reporter genes.
  • the present invention also provides host cells expressing the CD7 humanized antibodies, multispecific antibodies of the present invention and/or containing the nucleic acids or vectors of the present invention.
  • Preferred host cells of the present invention are bacterial cells, fungal cells or mammalian cells.
  • Suitable bacterial cells include gram-negative bacterial strains (eg, Escherichia coli, Proteus, and Pseudomonas strains) and gram-positive bacterial strains (eg, Bacillus Bacillus strains, Streptomyces strains, Staphylococcus strains and Lactococcus strains).
  • gram-negative bacterial strains eg, Escherichia coli, Proteus, and Pseudomonas strains
  • gram-positive bacterial strains eg, Bacillus Bacillus strains, Streptomyces strains, Staphylococcus strains and Lactococcus strains.
  • Suitable fungal cells include cells of species of Trichoderma, Neurospora and Aspergillus; or Saccharomyces (eg Saccharomyces cerevisiae), Saccharomyces (such as Schizosaccharomyces pombe), Pichia (such as Pichia pastoris and Pichia methanolica) and Hansen A cell of a species of the genus Hansenula.
  • Saccharomyces eg Saccharomyces cerevisiae
  • Saccharomyces such as Schizosaccharomyces pombe
  • Pichia such as Pichia pastoris and Pichia methanolica
  • Suitable mammalian cells include, for example, HEK293 cells, CHO cells, BHK cells, HeLa cells, COS cells, and the like.
  • amphibian cells insect cells, plant cells, and any other cell in the art for expressing heterologous proteins may also be used in the present invention.
  • the present invention also provides a recombinant receptor comprising a CD7 humanized antibody as described above, eg, a recombinant TCR receptor or a chimeric antigen receptor.
  • a recombinant receptor comprising a CD7 humanized antibody as described above, eg, a recombinant TCR receptor or a chimeric antigen receptor.
  • the present invention also provides a chimeric antigen receptor comprising the CD7 humanized antibody as described above.
  • chimeric antigen receptor refers to an artificially constructed hybrid polypeptide that generally includes a ligand binding domain (eg, an antigen binding portion of an antibody), a transmembrane domain, Optional co-stimulatory domains and intracellular signaling domains, each of which is linked by a linker.
  • CARs can exploit the antigen-binding properties of antibodies to redirect the specificity and reactivity of T cells and other immune cells to selected targets in a non-MHC-restricted manner.
  • the present invention provides a chimeric antigen receptor comprising a CD7 humanized antibody as described above or a multispecific antibody comprising the CD7 humanized antibody, a transmembrane domain and an intracellular signaling domain.
  • transmembrane domain refers to a polypeptide that enables expression of a chimeric antigen receptor on the surface of immune cells (eg, lymphocytes, NK cells, or NKT cells) and directs the cellular response of the immune cells against target cells structure.
  • immune cells eg, lymphocytes, NK cells, or NKT cells
  • the transmembrane domain can be natural or synthetic, and can be derived from any membrane-bound or transmembrane protein.
  • the transmembrane domain is capable of signaling when the chimeric antigen receptor binds to the target antigen.
  • Transmembrane domains particularly useful in the present invention may be derived from, for example, TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ chain, CD3 ⁇ subunit, CD3 ⁇ subunit, CD3 ⁇ subunit, CD3 ⁇ subunit, CD45, CD4, CD5, CD8 ⁇ , CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137, CD154 and functional fragments thereof.
  • the transmembrane domain may be synthetic and may contain predominantly hydrophobic residues such as leucine and valine.
  • the transmembrane domain is derived from the CD8 alpha chain or CD28, which is at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% of the amino acid sequence shown in SEQ ID NO: 24 or 26 % or 99% or 100% sequence identity, or its coding sequence has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% with the nucleic acid molecule shown in SEQ ID NO: 25 or 27 or 99% or 100% sequence identity.
  • intracellular signaling domain refers to the portion of a protein that transduces effector function signals and directs cells to perform specified functions.
  • the chimeric antigen receptors of the invention comprise intracellular signaling domains that may be sequences of the intracellular regions of T cell receptors and co-receptors that act together upon antigen receptor binding to elicit Signaling, as well as any derivatives or variants of these sequences and any synthetic sequences with the same or similar function.
  • the intracellular signaling domain can contain a number of immunoreceptor tyrosine-based activation motifs (ITAM).
  • ITAM immunoreceptor tyrosine-based activation motifs
  • intracellular signaling domains of the invention include, but are not limited to, intracellular regions of FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD22, CD79a, CD79b, and CD66d, among others.
  • the signaling domain of the CAR of the present invention may comprise the CD3 ⁇ intracellular region, which is at least 70%, preferably at least 80%, more preferably at least 90% identical to the amino acid sequence shown in SEQ ID NO: 32 or 34 %, 95%, 97% or 99% or 100% sequence identity, or its coding sequence is at least 70%, preferably at least 80%, more preferably at least 90% identical to the nucleic acid molecule set forth in SEQ ID NO: 33 or 35 , 95%, 97% or 99% or 100% sequence identity.
  • the chimeric antigen receptors of the invention may further comprise a hinge region between the antibody and the transmembrane domain.
  • the term "hinge region” generally refers to any oligopeptide or polypeptide that functions to link a transmembrane domain to an antibody. Specifically, the hinge region serves to provide greater flexibility and accessibility to the antibody.
  • the hinge region may comprise up to 300 amino acids, preferably 10 to 100 amino acids and most preferably 25 to 50 amino acids.
  • the hinge region can be derived in whole or in part from a native molecule, such as in whole or in part from the extracellular region of CD8, CD4 or CD28, or in whole or in part from an antibody constant region.
  • the hinge region may be a synthetic sequence corresponding to a naturally occurring hinge sequence, or may be a fully synthetic hinge sequence.
  • the hinge region comprises a CD8 ⁇ , CD28, Fc ⁇ RIII ⁇ receptor, IgG4 or IgG1 hinge region portion, more preferably a CD8 ⁇ , CD28 or IgG4 hinge, which is set forth in SEQ ID NO: 40, 42 or 44
  • the amino acid sequence has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity, or its coding sequence is identical to that of SEQ ID NO:41, 43 or 45
  • the nucleotide sequences shown have at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • the chimeric antigen receptor may also comprise one or more costimulatory domains.
  • a costimulatory domain may be an intracellular functional signaling domain from a costimulatory molecule, comprising the entire intracellular portion of the costimulatory molecule, or a functional fragment thereof.
  • a "costimulatory molecule” refers to a cognate binding partner that specifically binds to a costimulatory ligand on a T cell, thereby mediating a costimulatory response (eg, proliferation) of the T cell.
  • Costimulatory molecules include, but are not limited to, MHC class 1 molecules, BTLA and Toll ligand receptors.
  • Non-limiting examples of costimulatory domains of the invention include, but are not limited to, costimulatory signaling domains derived from the following proteins: TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11 , CD2, CD7, CD8, CD18(LFA-1), CD27, CD28, CD30, CD40, CD54(ICAM), CD83, CD134(OX40), CD137(4-1BB), CD270(HVEM), CD272(BTLA) , CD276 (B7-H3), CD278 (ICOS), CD357 (GITR), DAP10, LAT, NKG2C, SLP76, PD-1, LIGHT, TRIM and ZAP70.
  • costimulatory signaling domains derived from the following proteins: TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11 , CD2, CD7
  • the costimulatory domain of the CAR of the present invention is from 4-1BB, CD28 or 4-1BB+CD28.
  • the 4-1BB costimulatory domain is at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% of the amino acid sequence set forth in SEQ ID NO:30 % sequence identity, or its coding sequence has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence with the nucleic acid molecule shown in SEQ ID NO:31 identity.
  • the CD28 costimulatory domain has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% of the amino acid sequence set forth in SEQ ID NO:28 Sequence identity, or its coding sequence, has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the nucleic acid molecule set forth in SEQ ID NO: 29 .
  • the CAR of the present invention may further comprise a signal peptide such that when it is expressed in a cell such as a T cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface.
  • the core of the signal peptide may contain a long segment of hydrophobic amino acids that has a tendency to form a single alpha-helix.
  • signal peptidases At the end of the signal peptide, there is usually a segment of amino acids that is recognized and cleaved by signal peptidases.
  • the signal peptidase can cleave during or after translocation to generate the free signal peptide and mature protein. Then, the free signal peptide is digested by specific proteases.
  • Signal peptides useful in the present invention are well known to those skilled in the art, eg, signal peptides derived from B2M, CD8 ⁇ , IgG1, GM-CSFR ⁇ , and the like.
  • the signal peptide useful in the present invention is from B2M or CD8 ⁇ , which is at least 70%, preferably at least 80%, more preferably at least 90%, 95% of the amino acid sequence shown in SEQ ID NO: 36 or 38 , 97% or 99% or 100% sequence identity, or its coding sequence has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • the CAR comprises a CD7 humanized antibody as provided herein or a multispecific antibody comprising the CD7 humanized antibody, a CD8 ⁇ transmembrane region, a CD28 or a 4-1BB costimulatory domain, and CD3 ⁇ intracellular signaling domain.
  • the CAR may further comprise a signal peptide from B2M, CD8 ⁇ , IgG1 or GM-CSFR ⁇ .
  • the present invention also provides a nucleic acid molecule encoding a CD7-targeting chimeric antigen receptor as defined above, and a vector comprising said nucleic acid molecule.
  • vector is a nucleic acid molecule used as a vehicle for the transfer of (exogenous) genetic material into a host cell, in which the nucleic acid molecule can eg be replicated and/or expressed.
  • Vectors generally include targeting vectors and expression vectors.
  • a "targeting vector” is a medium that delivers an isolated nucleic acid to the interior of a cell by, for example, homologous recombination or a hybrid recombinase using a specific targeting sequence at the site.
  • An "expression vector” is a vector used for the transcription of heterologous nucleic acid sequences, such as those encoding the chimeric antigen receptor polypeptides of the invention, in suitable host cells and the translation of their mRNAs.
  • Suitable carriers for use in the present invention are known in the art and many are commercially available.
  • the vectors of the present invention include, but are not limited to, plasmids, viruses (eg, retroviruses, lentiviruses, adenoviruses, vaccinia virus, Rous sarcoma virus (RSV, polyoma virus, and adeno-associated virus (AAV)), and the like ), phages, phagemids, cosmids, and artificial chromosomes (including BACs and YACs).
  • the vector itself is usually a nucleic acid molecule, usually a DNA sequence containing an insert (transgene) and a larger sequence that serves as the "backbone" of the vector.
  • the VL vector typically also contains an origin of autonomous replication in the host cell (if stable expression of the polynucleotide is desired), a selectable marker, and a restriction enzyme cleavage site (eg, a multiple cloning site, MCS).
  • the vector may additionally contain a promoter, multiple elements such as polyadenylated tails (polyA), 3'UTRs, enhancers, terminators, insulators, operons, selectable markers, reporter genes, targeting sequences and/or protein purification tags.
  • polyA polyadenylated tails
  • the vector is an in vitro transcribed vector.
  • the present invention also provides engineered immune cells expressing the CAR of the present invention.
  • the term "immune cell” refers to any cell of the immune system that has one or more effector functions (eg, cytotoxic cell killing activity, secretion of cytokines, induction of ADCC and/or CDC).
  • the immune cells can be T cells, macrophages, dendritic cells, monocytes, NK cells, and/or NKT cells.
  • the immune cells are derived from stem cells, such as adult stem cells, embryonic stem cells, cord blood stem cells, progenitor cells, bone marrow stem cells, induced pluripotent stem cells, totipotent stem cells, or hematopoietic stem cells, and the like.
  • the immune cells are T cells.
  • the T cells can be any T cells, such as T cells cultured in vitro, eg, primary T cells, or T cells from T cell lines cultured in vitro, such as Jurkat, SupT1, etc., or T cells obtained from a subject. Examples of subjects include humans, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from a variety of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from sites of infection, ascites, pleural effusion, spleen tissue, and tumors. T cells can also be concentrated or purified.
  • T cells can be at any stage of development, including, but not limited to, CD4+/CD8+ T cells, CD4+ helper T cells (eg, Th1 and Th2 cells), CD8+ T cells (eg, cytotoxic T cells), tumor-infiltrating cells, memory T cells, naive T cells, ⁇ -T cells, ⁇ -T cells, etc.
  • the immune cells are human T cells.
  • T cells can be obtained from the blood of a subject using a variety of techniques known to those of skill in the art, such as Ficoll separation.
  • Nucleic acid sequences encoding chimeric antigen receptors can be introduced into immune cells using conventional methods known in the art (eg, by transduction, transfection, transformation, etc.).
  • Transfection is the process of introducing a nucleic acid molecule or polynucleotide, including a vector, into a target cell.
  • An example is RNA transfection, the process of introducing RNA (eg, in vitro transcribed RNA, ivtRNA) into a host cell.
  • RNA transfection the process of introducing RNA (eg, in vitro transcribed RNA, ivtRNA) into a host cell.
  • the term is mainly used for non-viral methods in eukaryotic cells.
  • transduction is generally used to describe virus-mediated transfer of nucleic acid molecules or polynucleotides.
  • Transfection of animal cells typically involves opening transient pores or "holes" in the cell membrane to allow uptake of material.
  • Transfection can be performed using calcium phosphate, by electroporation, by cell extrusion, or by mixing cationic lipids with materials to create liposomes that fuse with cell membranes and deposit their cargo inside.
  • Exemplary techniques for transfecting eukaryotic host cells include lipid vesicle-mediated uptake, heat shock-mediated uptake, calcium phosphate-mediated transfection (calcium phosphate/DNA co-precipitation), microinjection, and electroporation. perforation.
  • transformation is used to describe the non-viral transfer of nucleic acid molecules or polynucleotides (including vectors) into bacteria, but also into non-animal eukaryotic cells (including plant cells).
  • transformation is the genetic alteration of a bacterial or non-animal eukaryotic cell, which is produced by the direct uptake of the cell membrane from its surroundings and subsequent incorporation of exogenous genetic material (nucleic acid molecules). Conversion can be achieved by manual means.
  • the cells or bacteria must be in a competent state.
  • techniques can include heat shock-mediated uptake, fusion of bacterial protoplasts with intact cells, microinjection, and electroporation.
  • the engineered immune cells further comprise suppressed or silenced expression of at least one gene selected from the group consisting of CD52, GR, dCK, TCR/CD3 genes (such as TRAC, TRBC, CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ ), MHC-related genes (HLA-A, HLA-B, HLA-C, B2M, HLA-DPA, HLA-DQ, HLA-DRA, TAP1, TAP2, LMP2, LMP7, RFX5, RFXAP, RFXANK, CIITA) and immune checkpoint genes such as PD1, LAG3, TIM3, CTLA4, PPP2CA, PPP2CB, PTPN6, PTPN22, PDCD1, HAVCR2, BTLA, CD160, TIGIT, CD96, CRTAM, TNFRSF10B, TNFRSF10A , CASP8, CASP10, CASP3, CASP6, CASP
  • the engineered immune cells further comprise suppressed or silenced expression of at least one gene selected from the group consisting of TRAC, TRBC, HLA-A, HLA-B, HLA-C, B2M, RFX5, RFXAP, RFXANK, CIITA, PD1, LAG3, TIM3, CTLA4, more preferably TRAC, TRBC, HLA-A, HLA-B, HLA-C, B2M, RFX5, RFXAP, RFXANK, CIITA.
  • RNA decoys RNA decoys
  • RNA aptamers siRNA, shRNA/miRNA, trans dominant negative protein (TNP), chimeric/antibody conjugates, chemokine ligands, anti-infective cellular proteins
  • intracellular antibodies sFv
  • nucleoside analogs NRTI
  • non-nucleoside analogs NRTI
  • integrase inhibitors oligonucleotides, dinucleotides and chemicals
  • protease inhibitors to inhibit gene expression Express.
  • gene silencing can also be achieved by mediating DNA fragmentation, eg, by meganucleases, zinc finger nucleases, TALE nucleases, or Cas enzymes in the CRISPR system.
  • a plurality of immune cells are provided, each immune cell engineered to express one or more chimeric antigen receptors.
  • one immune cell is engineered to express a chimeric antigen receptor that binds and/or targets CD7 (eg, a CAR comprising a CD7 humanized antibody described herein), and another Cells are engineered to express chimeric antigen receptors that bind and/or target other antigens.
  • immune cells may also express multispecific chimeric antigen receptors that target one or more antigens including CD7.
  • a multispecific chimeric antigen receptor may comprise a multispecific antibody targeting CD7, or a combination of CD7 humanized antibodies of the present invention and antibodies targeting other antigens.
  • the plurality of engineered immune cells can be administered together or separately.
  • the plurality of immune cells may be in the same composition or in different compositions. Exemplary compositions of cells include those described in the following sections of this application.
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and a second functional structure, wherein the second functional structure is selected from Fc, radioisotopes, extended half-life Structural moieties, detectable labels and drugs.
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and an Fc.
  • Fc is used to define the C-terminal region of an immunoglobulin heavy chain, which includes native Fc and variant Fc.
  • Native Fc refers to a molecule or sequence comprising a non-antigen-binding fragment, whether in monomeric or multimeric form, produced by digestion of an intact antibody.
  • the source of immunoglobulins for the production of native Fc is preferably derived from humans.
  • Native Fc fragments are composed of monomeric polypeptides that can be linked in dimeric or multimeric form by covalent linkages (eg, disulfide bonds) and non-covalent linkages.
  • native Fc the disulfide-linked dimer produced by papain digestion of IgG (see Ellison et al. (1982) Nucleic Acids Res. 10:4071-9).
  • native Fc the disulfide-linked dimer produced by papain digestion of IgG (see Ellison et al. (1982) Nucleic Acids Res. 10:4071-9).
  • native Fc as used herein generally refers to monomeric, dimeric and multimeric forms.
  • “Variant Fc” refers to an amino acid sequence that differs from that of a “native” or “wild-type” Fc due to at least one "amino acid modification” as defined herein, also referred to as an "Fc variant".
  • “Fc” also includes single-chain Fc (scFc), ie, a single-chain Fc consisting of two Fc monomers linked by a polypeptide linker, which is capable of naturally folding into a functional dimeric Fc region.
  • the Fc is preferably a human immunoglobulin Fc, more preferably a human IgGl Fc.
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and a radioisotope.
  • radioisotopes useful in the present invention include, but are not limited to, At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 , 99m Tc, 123 I, 18 F and 68 Ga.
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and a half-life extending structural moiety selected from the group consisting of albumin binding structures, trans Binding structures of ferritin, polyethylene glycol molecules, recombinant polyethylene glycol molecules, human serum albumin, fragments of human serum albumin, and albumin polypeptides (including antibodies) that bind human serum albumin.
  • a half-life extending structural moiety selected from the group consisting of albumin binding structures, trans Binding structures of ferritin, polyethylene glycol molecules, recombinant polyethylene glycol molecules, human serum albumin, fragments of human serum albumin, and albumin polypeptides (including antibodies) that bind human serum albumin.
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and a detectable label.
  • detectable label herein means a compound that produces a detectable signal.
  • the detectable label can be an MRI contrast agent, a scintigraphic contrast agent, an X-ray imaging contrast agent, an ultrasound contrast agent, an optical imaging contrast agent.
  • detectable labels examples include fluorophores (such as fluorescein, Alexa, or cyanine), chemiluminescent compounds (such as luminol), bioluminescent compounds (such as luciferase or alkaline phosphatase), enzymes (such as Root peroxidase, glucose-6-phosphatase, ⁇ -galactosidase), antibiotics (such as kanamycin, ampicillin, chloramphenicol, tetracycline, etc.) resistance genes and contrast agents (such as nanoparticles or gadolinium).
  • fluorophores such as fluorescein, Alexa, or cyanine
  • chemiluminescent compounds such as luminol
  • bioluminescent compounds such as luciferase or alkaline phosphatase
  • enzymes such as Root peroxidase, glucose-6-phosphatase, ⁇ -galactosidase
  • antibiotics such as kanamycin, ampicillin, chloramphenicol, t
  • the present invention provides an antibody conjugate comprising a CD7 humanized antibody as defined in the present invention and a drug, such as a cytotoxin or an immunomodulator, conjugated to the CD7 humanized antibody ( That is, antibody drug conjugates).
  • a drug such as a cytotoxin or an immunomodulator
  • the drug is covalently attached to the antibody and usually relies on a linker.
  • the drug is a cytotoxin.
  • the drug is an immunomodulatory agent.
  • cytotoxins include, but are not limited to, methotrexate, aminopterin, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, dacarbazine, nitrogen mustard, thiotepa, benzidine Nitrogen mustard, melphalan, carmustine (BSNU), lomustine (CCNU), 1-methylnitrosourea, cyclophosphamide, nitrogen mustard, busulfan, dibromomannitol, chain Zorcine, mitomycin, cis-dichlorodiamineplatinum (II) (DDP), cisplatin, carboplatin, zorubicin, doxorubicin, detorubicin, caminomycin, iota Darubicin, epirubicin, mitoxantrone, actinomycin D, bleomycin, calicheamicin, glaremycin, atramycin (AMC), vincristine, vinblastine,
  • immunomodulators include, but are not limited to, ganciclovir, etanercept, tacrolimus, sirolimus, vortexporine, cyclosporine, rapamycin, cyclophosphamide, azathioprine , mycophenolate mofetil, methotrexate, glucocorticoids and their analogs, cytokines, stem cell growth factors, lymphotoxins, tumor necrosis factor (TNF), hematopoietic factors, interleukins (such as IL-1, IL-2, IL-3, IL-6, IL-10, IL-12, IL-18 and IL-21), colony stimulating factors such as G-CSF and (GM-CSF), interferons such as interferon-alpha, interferon interferon-beta and interferon-gamma), stem cell growth factors designated "S1 factor", erythropoietin and thrombopoietin, or a combination thereof.
  • TNF tumor nec
  • the present invention also provides a detection kit comprising the humanized antibody, multispecific antibody, antibody conjugate or chimeric antigen receptor of the present invention.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the humanized antibody, chimeric antigen receptor, multispecific antibody or antibody conjugate of the present invention, and one or more pharmaceutical agents acceptable excipients.
  • the term "pharmaceutically acceptable excipient” means pharmacologically and/or physiologically compatible with the subject and the active ingredient (ie, capable of eliciting the desired therapeutic effect without causing any inconvenience desired local or systemic effect) carriers and/or excipients, which are well known in the art (see, e.g., Remington's Pharmaceutical Sciences. Edited by Gennaro AR, 19th ed. Pennsylvania: Mack Publishing Company, 1995).
  • Examples of pharmaceutically acceptable excipients include, but are not limited to, fillers, binders, disintegrants, coatings, adsorbents, antiadherents, glidants, antioxidants, flavoring agents, colorants, Sweeteners, solvents, co-solvents, buffers, chelating agents, surfactants, diluents, wetting agents, preservatives, emulsifiers, coating agents, isotonic agents, absorption delaying agents, stabilizers and tonicity modifiers . It is known to those skilled in the art to select suitable excipients to prepare the desired pharmaceutical compositions of the present invention.
  • excipients for use in the pharmaceutical compositions of the present invention include saline, buffered saline, dextrose and water.
  • suitable excipients depends, among other things, on the active agent used, the disease to be treated and the desired dosage form of the pharmaceutical composition.
  • compositions according to the present invention may be suitable for administration by various routes. Typically, administration is accomplished parenterally.
  • Parenteral delivery methods include topical, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.
  • compositions according to the invention can also be prepared in various forms, such as solid, liquid, gaseous or lyophilized forms, in particular ointments, creams, transdermal patches, gels, powders, tablets, solutions, gaseous In the form of aerosols, granules, pills, suspensions, emulsions, capsules, syrups, elixirs, extracts, tinctures or liquid extracts, or in a form particularly suitable for the desired method of administration.
  • Processes known in the present invention for the manufacture of pharmaceuticals may include, for example, conventional mixing, dissolving, granulating, dragee-making, milling, emulsifying, encapsulating, entrapping, or lyophilizing processes.
  • Pharmaceutical compositions comprising immune cells such as those described herein are typically provided in solution and preferably comprise a pharmaceutically acceptable buffer.
  • compositions according to the present invention may also be administered in combination with one or more other agents suitable for the treatment and/or prevention of the disease to be treated.
  • agents suitable for combination include known anticancer drugs such as cisplatin, maytansine derivatives, rachelmycin, calicheamicin, docetaxel, etoposide , gemcitabine, ifosfamide, irinotecan, melphalan, mitoxantrone, sorfimer sodium photofrin II, temozolomide, topotecan, trimetate glucuronate, auristatin E, vincristine and doxorubicin; peptide cytotoxins such as ricin, diphtheria toxin, Pseudomonas bacterial exotoxin A, DNase and RNase; radionuclides such as iodine 131, rhenium 186, indium 111, iridium 90, bismuth 210 and 213, act
  • the present invention also provides a method of treating and/or preventing and/or diagnosing a disease associated with CD7 expression, comprising administering to a subject a humanized antibody, chimeric antigen receptor, Multispecific antibodies, antibody conjugates or pharmaceutical compositions.
  • diseases associated with CD7 expression include non-solid tumors (such as hematological tumors, eg, leukemias and lymphomas) and solid tumors.
  • Hematological neoplasms are cancers of the blood or bone marrow, including but not limited to acute leukemias (such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia and myeloblastoid, promyelocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemia (such as chronic myeloid (myeloid) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma Chikin's lymphoma (painless and high-grade forms), multiple myeloma, Waldenstrom's macroglobulinemia, myelodys
  • a solid tumor is an abnormal mass of tissue that does not usually contain cysts or areas of fluid, which can be benign or malignant.
  • Different types of solid tumors are named after the cell type that forms them (such as sarcomas, carcinomas, and lymphomas).
  • Examples of solid tumors include, but are not limited to, fibrosarcoma, myxosarcoma, liposarcoma, mesothelioma, pancreatic cancer, ovarian cancer, peritoneal, omentum and mesenteric cancers, pharyngeal cancer, prostate cancer, rectal cancer, kidney cancer, skin cancer, Small bowel cancer, melanoma, kidney cancer, laryngeal cancer, soft tissue cancer, stomach cancer, testicular cancer, colon cancer, esophagus cancer, cervical cancer, acinar rhabdomyosarcoma, bladder cancer, bone cancer, brain cancer, breast cancer, anal cancer , cancer of the eye, intrahepatic bile duct, joint,
  • the disease associated with CD7 expression is preferably selected from the group consisting of acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), T lymphoblastic lymphoma (T-LBL), early pre-T lymphoblastoma Leukemia (ETP-ALL) and extranodal NK/T cell lymphoma.
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • T-LBL T lymphoblastic lymphoma
  • EDP-ALL early pre-T lymphoblastoma Leukemia
  • extranodal NK/T cell lymphoma extranodal NK/T cell lymphoma.
  • Figure 1 Shows scFv expression levels of CAR-T cells containing CD7 humanized antibody.
  • Figure 2 shows the killing effect of CAR-T cells containing CD7 humanized antibody on target cells.
  • Figure 3 Shows the level of cytokine release after co-culture of CAR-T cells containing CD7 humanized antibody with target cells.
  • Clone m189 comprises CDR-L1 as SEQ ID NO: 1, CDR-L2 as SEQ ID NO: 2, CDR-L3 as SEQ ID NO: 3, CDR-L3 as SEQ ID NO: 4 CDR-H1, CDR-H2 shown in SEQ ID NO: 5, CDR-H3 shown in SEQ ID NO: 6, and its amino acid sequence is shown in SEQ ID NO: 7.
  • the specific method for preparing humanized antibodies is as follows: firstly, through Ig BLAST (http://www.ncbi.nlm.nih.gov/igblast/) and IMGT (Immune Gene Database IMGT: http://www.imgt.org) Sequence similarity searches were performed respectively, and in the search results, antibodies with higher sequence similarity to both light and heavy chains were selected as humanized antibody templates. Then, the heavy chain CDRs and light chain CDRs in the murine CD7 scFv were grafted into the framework of the humanized antibody template with high sequence similarity, and then backmutated to ensure the affinity and specificity of the humanized antibody. The amino acid sequence of CD7 humanized single-chain antibody (scFv) was finally obtained as shown in Table 1 below.
  • amino acid sequence of anti-CD7scFv contained in hCAR7-1CAR is shown in SEQ ID NO:10; the amino acid sequence of anti-CD7scFv contained in hCAR7-2CAR is shown in SEQ ID NO:13; the amino acid sequence of anti-CD7scFv contained in hCAR7-3CAR As shown in SEQ ID NO: 16; the amino acid sequence of the anti-CD7 scFv contained in hCAR7-4CAR is shown in SEQ ID NO: 19.
  • Opti-MEM 3ml Opti-MEM (Gibco, Item No. 31985-070) to a sterile tube to dilute the above plasmid, and then add the packaging vector psPAX2 (Addgene, Cat. No. 12260) and the envelope vector pMD2.G (Addgene, Cat. No. 12259). Then, add 120ul of X-treme GENE HP DNA transfection reagent (Roche, Cat. No. 06366236001), mix immediately, incubate at room temperature for 15 minutes, and then add the plasmid/vector/transfection reagent mixture dropwise to the culture flask of 293T cells . Viruses were collected at 24 hours and 48 hours, pooled, and ultracentrifuged (25000 g, 4°C, 2.5 hours) to obtain concentrated lentiviruses.
  • CD7 is also expressed on T cells
  • the inventors knocked out CD7 in T cells through the CRISP/Cas9 system. After 1 day of culture, T cells were activated with DynaBeads CD3/CD28 CTS TM (Gibco, Cat. No. 40203D) and cultured at 37°C and 5% CO2 for an additional day. Then, the concentrated lentivirus was added and cultured for 3 days to obtain CAR T cells expressing different CD7 humanized scFvs. Unmodified wild-type T cells were used as negative controls (NT).
  • Biotin-SP Long spacer AffiniPure Goat Anti-Mouse IgG, F(ab') 2 Fragment Specific (min X Hu, Bov, Hrs Sr Prot) (jackson immunoresearch, Cat. No. 115-065-072) was used as primary antibody, APC Streptavidin (BD Pharmingen, Cat. No. 554067) or PE Streptavidin (BD Pharmingen, Cat. No. 554061) was used as a secondary antibody for the detection of hCAR7-1 T cells, hCAR7-2 T cells, hCAR7-3 T cells and hCAR7-4 by flow cytometry The expression levels of scFv, the results are shown in Figure 1.
  • CD7 humanized scFv in the CAR T cells prepared by the present invention can be effectively expressed.
  • Example 3 Killing effect and cytokine release of CAR T cells on target cells
  • T cells kill target cells, the number of target cells decreases.
  • T cells were co-cultured with target cells expressing luciferase, the number of target cells was decreased, and the secreted luciferase was also decreased.
  • Luciferase catalyzes the conversion of luciferin to oxidative luciferin, and during this oxidation, bioluminescence is produced, and the intensity of this luminescence will depend on the level of luciferase expressed by the target cells. Therefore, the detected fluorescence intensity can reflect the killing ability of T cells to target cells.
  • the CAR T cells of the present invention can specifically kill target cells.
  • ELISA enzyme-linked immunosorbent assay
  • Target cells were plated in 96-well plates at 1x105 /well, then CAR T and NT cells (negative control) were co-cultured with target cells at a ratio of 1:1, and the cell co-culture supernatant was collected 18-24 hours later .
  • the 96-well plate was coated with the capture antibody Purified anti-human IFN- ⁇ Antibody (Biolegend, Cat. No. 506502) and incubated at 4°C overnight, then the antibody solution was removed, and 250 ⁇ L of PBST containing 2% BSA (sigma, Cat. No. V900933-1kg) was added. 0.1% Tween in 1X PBS) for 2 hours at 37°C. Plates were then washed 3 times with 250 [mu]L of PBST (1XPBS with 0.1% Tween). 50 ⁇ L of cell co-culture supernatant or standards were added to each well and incubated at 37° C.
  • the cytokine release water of the CAR T cells of the present invention is significantly higher than that of the control NT cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

本发明提供靶向CD7的人源化抗体,以及包含它的多特异性抗体、嵌合抗原受体、抗体偶联物、药物组合物和试剂盒,以及它们在诊断/治疗/预防与CD7表达相关的疾病中的用途。

Description

靶向CD7的人源化抗体及其用途 技术领域
本发明属于免疫治疗领域。更具体地,本发明涉及靶向CD7的人源化抗体,及其在预防和/或治疗和/或诊断疾病中的用途。
背景技术
CD7是一种细胞表面糖蛋白,分子量为约40kDa,属于免疫球蛋白超家族的一员。CD7在大多数的T细胞、NK细胞、髓细胞、T细胞急性淋巴细胞白血病/淋巴瘤、急性粒细胞性白血病和慢性粒细胞白血病中表达。据报道,CD7分子通过与其配体K12/SECTM1的结合在T细胞活化期间起到共刺激信号的作用。此外,据报道,破坏小鼠T祖细胞中的CD7分子依然会产生正常的T细胞发育和体内平衡,表明CD7似乎对T细胞的发育和功能不会产生关键性的影响,这使其成为治疗T细胞急性淋巴细胞白血病(T-ALL)的非常合适的治疗靶点。实际上,CD7作为治疗白血病和淋巴瘤的细胞毒性分子的靶标已被广泛研究。
本发明旨在提供一种靶向CD7的人源化抗体,及其在疾病预防和/或治疗和/或诊断中的用途。
发明内容
在第一个方面,本发明提供一种CD7人源化抗体,其包含轻链可变区和重链可变区,其中所述轻链可变区包含如SEQ ID NO:1所示的CDR-L1、如SEQ ID NO:2所示的CDR-L2、如SEQ ID NO:3所示的CDR-L3,所述重链可变区包含如SEQ ID NO:4所示的CDR-H1、如SEQ ID NO:5所示的CDR-H2、如SEQ ID NO:6所示的CDR-H3,并且所述轻链可变区与选自SEQ ID NO:8、11、14 和17所示的氨基酸序列具有至少90%同一性,所述重链可变区与选自SEQ ID NO:9、12、15和18的氨基酸序列具有至少90%同一性。
在一个实施方案中,所述CD7人源化抗体包含选自SEQ ID NO:8、11、14和17所示的轻链可变区和选自SEQ ID NO:9、12、15和18的重链可变区。
在一个实施方案中,所述CD7人源化抗体的氨基酸序列选自SEQ ID NO:10、13、16和19。
本发明还提供编码上述CD7人源化抗体的核酸分子。因此,在一个实施方案中,编码所述CD7人源化抗体的核酸分子与选自SEQ ID NO:20-23的核苷酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、100%序列同一性,并且其编码的CD7人源化抗体能够特异性结合CD7抗原。优选地,编码所述CD7人源化抗体的核酸分子选自SEQ ID NO:20-23。
在另一个方面,本发明还提供一种多特异性抗体(优选双特异性抗体或三特异性抗体),其包含如上所述的CD7人源化抗体和一个或多个与其他抗原特异性结合的第二抗体或其抗原结合部分。
在一个实施方案中,第二抗体或其抗原结合部分可以具有任何抗体或抗体片段形式,例如全长抗体、Fab、Fab'、(Fab') 2、Fv、scFv、scFv-scFv、微抗体、双抗体或sdAb。
本发明还提供包含编码上述CD7人源化抗体或多特异性抗体的核酸分子的载体,以及表达所述CD7人源化抗体或多特异性抗体的宿主细胞。
在另一个方面,本发明还提供一种嵌合抗原受体,其包含本发明所述的CD7人源化抗体、跨膜结构域和胞内信号传导结构域。优选地,所述嵌合抗原受体还包含一个或多个共刺激结构域。更优选地,所述嵌合抗原受体包含如本文所提供CD7人源化抗体或含有所述CD7人源化抗体的多特异性抗体、CD8α跨膜区、CD28或4-1BB共刺激结构域,和CD3ζ胞内信号传导结构域。
本发明还提供编码如上所定义的靶向CD7的嵌合抗原受体的核 酸分子,以及包含所述核酸分子的载体。
本发明还提供包含如上所定义的靶向CD7的嵌合抗原受体的细胞,优选免疫细胞,例如T细胞、NK细胞、NKT细胞、巨噬细胞、树突细胞。
在另一个方面,本发明还提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和第二功能性结构,其中所述第二功能性结构选自Fc、放射性同位素、延长半衰期的结构部分、可检测标记物和药物。
在一个实施方案中,所述延长半衰期的结构部分选自:所述延长半衰期的结构部分选自白蛋白的结合结构、转铁蛋白的结合结构、聚乙二醇分子、重组聚乙二醇分子、人血清白蛋白、人血清白蛋白的片段和结合人血清白蛋白的白多肽(包括抗体)。在一个实施方案中,可检测标记物选自荧光团、化学发光化合物、生物发光化合物、酶、抗生素抗性基因和造影剂。在一个实施方案中,所述药物选自细胞毒素和免疫调节剂。
在另一个方面,本发明还提供一种检测试剂盒,其包含本发明所述的人源化抗体、多特异性抗体、抗体偶联物或嵌合抗原受体。
在另一个方面,本发明还提供一种药物组合物,其包含本发明所述的人源化抗体、嵌合抗原受体、多特异性抗体或抗体偶联物,和一种或多种药学上可接受的赋形剂。
在另一个方面,本发明还提供一种治疗和/或预防和/或诊断与CD7表达相关的疾病的方法,包括向受试者施用如上所述的人源化抗体、嵌合抗原受体、多特异性抗体、抗体偶联物或药物组合物。
发明详述
除非另有说明,否则本文中所使用的所有科学技术术语的含义与本发明所属领域的普通技术人员通常所了解的相同。
CD7人源化抗体
如本文所用,术语“抗体”具有本领域技术人员所理解的最广 泛的含义,并且包括单克隆抗体(包含完整抗体)、多克隆抗体、多价抗体、多特异性抗体(例如双特异性抗体)、和能够表现期望的生物活性的携带一个或多个CDR序列的抗体片段或合成多肽。本发明所述抗体可为任何种类(例如IgG、IgE、IgM、IgD、IgA等)或亚类(例如IgG1、IgG2、IgG2a、IgG3、IgG4、IgA1、IgA2等)。
通常,完整抗体包括通过二硫键连接在一起的两条重链和两条轻链,每条轻链通过二硫键被连至各自的重链,呈“Y”形结构。每条重链包含重链可变区(VH)和重链恒定区,其中重链可变区包含三个互补决定区(CDR):CDR-H1、CDR-H2和CDR-H3,重链恒定区包含三个恒定结构域:CH1、CH2和CH3。每条轻链包含轻链可变区(VL)和轻链恒定区,其中轻链可变区包含三个CDR:CDR-L1、CDR-L2和CDR-L3,轻链恒定区包含一个恒定结构域CL。在重链/轻链可变区中,CDR被更保守的框架区(FR)隔开。重链/轻链的可变区负责与抗原的识别和结合,恒定区则可以介导抗体与宿主组织或因子的结合,包括免疫系统的各种细胞(例如效应细胞)和经典补体系统的第一组分。
可以使用许多本领域熟知的编号方案容易地确定给定CDR或FR的精确氨基酸序列边界,这些方案包括:Kabat等人(1991),“Sequences ofProteins of Immunological Interest,”第5版Public Health Service,NationalInstitutes of Health,贝塞斯达,马里兰州(“Kabat”编号方案);Al-Lazikani等人,(1997)JMB273,927-948(“Chothia”编号方案);MacCallum等人,J.Mol.Biol.262:732-745(1996),“Antibody-antigen interactions:Contact analysis and binding sitetopography,”J.Mol.Biol.262,732-745”(“Contact”编号方案);Lefranc MP等人,“IMGTunique numbering for immunoglobulin and T cell receptor variable domains andIg superfamily V-like domains,”Dev Comp Immunol,2003年1月;27(1):55-77(“IMGT”编号方案);Honegger A和Plückthun A,“Yet another numbering scheme forimmunoglobulin  variable domains:an automatic modeling and analysis tool,”JMol Biol,2001年6月8日;309(3):657-70(“Aho”编号方案);和Martin等人,“Modelingantibody hypervariable loops:a combined algorithm,”PNAS,1989,86(23):9268-9272(“AbM”编号方案)。
给定CDR或FR的边界可能取决于用于鉴定的方案而不同。例如,Kabat方案是基于结构比对,而Chothia方案是基于结构信息。Kabat和Chothia方案的编号都是基于最常见的抗体区域序列长度,其中通过插入字母提供插入(例如“30a”)并且在一些抗体中出现缺失。这两种方案将某些插入和缺失(“插入缺失(indel)”)放置在不同的位置,从而产生不同的编号。Contact方案是基于对复杂晶体结构的分析,并且在许多方面与Chothia编号方案相似。AbM方案是介于Kabat与Chothia定义之间的折衷,其基于Oxford Molecular的AbM抗体建模软件所使用的方案。
因此,除非另有规定,否则应当理解,给定抗体或其区域(如其可变区)的“CDR”涵盖由任何上述方案或其他已知方案所定义的CDR。例如,在指定特定的CDR(例如CDR3)含有给定氨基酸序列的情况下,应理解,这样的CDR还可以具有由任何上述方案或其他已知方案所定义的相应CDR(例如CDR3)的序列。同样,除非另有规定,否则应当理解给定抗体或其区域(如其可变区)的FR涵盖由任何上述方案或其他已知方案所定义的FR。
如本文所用,“人源化”抗体是指其中所有或基本上所有CDR氨基酸残基源自非人CDR并且所有或基本上所有FR氨基酸残基源自人FR。非人抗体的“人源化形式”是指所述非人抗体的变体,其经历人源化以通常降低对人的免疫原性,同时保留亲本非人抗体的特异性和亲和力。在一些实施方案中,人源化抗体中的一些FR残基被来自非人抗体(例如,衍生CDR残基的抗体)的相应残基取代,例如以恢复或改善抗体特异性或亲和力。
人源化抗体及其制备方法是本领域技术人员熟知的,参见例如Almagro和Fransson,Front.Biosci.13:1619-1633(2008)。可用于人源化 的人类框架区包括但不限于:使用“最佳拟合”方法选择的框架区;源自轻链或重链可变区的特定亚组的人类抗体的共有序列的框架区;人类成熟(体细胞突变)框架区或人类种系框架区;以及筛选FR文库得到的框架区。
如本文所用,术语“抗体片段”或“抗原结合部分”仅包含完整抗体的一部分,一般包含完整抗体的抗原结合位点并因此保留结合抗原的能力。本发明中的抗体片段的实例包括但不限于:Fab、Fab'、F(ab')2、Fd片段、Fd′、Fv片段、scFv、二硫键-连接的Fv(sdFv)、抗体的重链可变区(VH)或轻链可变区(VL)、线性抗体、具有两个抗原结合位点的“双体”、单结构域抗体、纳米抗体、所述抗原的天然配体或其功能性片段等。因此,本发明的“抗体”涵盖如上定义的抗体片段。
在一个实施方案中,本发明的CD7人源化抗体是scFv。“单链抗体”和“scFv”在本文中可互换使用,是指由抗体重链可变区(VH)和轻链可变区(VL)通过接头连接而成的抗体。可以选择接头的最佳长度和/或氨基酸组成。接头的长度会明显影响scFv的可变区折叠和相互作用情况。事实上,如果使用较短的接头(例如在5-10个氨基酸之间),则可以防止链内折叠。关于接头的大小和组成的选择,参见例如,Hollinger等人,1993Proc Natl Acad.Sci.U.S.A.90:6444-6448;美国专利申请公布号2005/0100543、2005/0175606、2007/0014794;以及PCT公布号WO2006/020258和WO2007/024715,其全文通过引用并入本文。scFv可以包含以任何顺序连接的VH和VL,例如VH-接头-VL或VL-接头-VH。
在一个实施方案中,本发明提供一种CD7人源化抗体,其包含轻链可变区和重链可变区,其中所述轻链可变区包含如SEQ ID NO:1所示的CDR-L1、如SEQ ID NO:2所示的CDR-L2、如SEQ ID NO:3所示的CDR-L3,所述重链可变区包含如SEQ ID NO:4所示的CDR-H1、如SEQ ID NO:5所示的CDR-H2、如SEQ ID NO:6所 示的CDR-H3,并且所述轻链可变区与选自SEQ ID NO:8、11、14和17的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、100%的序列同一性,所述重链可变区与SEQ ID NO:9、12、15和18所示的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、100%的序列同一性。
在一个实施方案中,所述CD7人源化抗体包含选自SEQ ID NO:8、11、14和17所示的轻链可变区和选自SEQ ID NO:9、12、15和18所示的重链可变区。
在一个实施方案中,所述CD7人源化抗体的氨基酸序列如SEQ ID NO:10、13、16和19所示。
如本文所用,术语“序列同一性”表示两个(核苷酸或氨基酸)序列在比对中在相同位置处具有相同残基的程度,并且通常表示为百分数。优选地,同一性在被比较的序列的整体长度上确定。因此,具有完全相同序列的两个拷贝具有100%同一性。本领域技术人员知晓,可以使用一些算法来确定序列同一性,例如Blast(Altschul等(1997)Nucleic Acids Res.25:3389-3402)、Blast2(Altschul等(1990)J.Mol.Biol.215:403-410)、Smith-Waterman(Smith等(1981)J.Mol.Biol.147:195-197)和ClustalW。
在在一个方面,本发明还提供包含如上所CD7人源化抗体的多特异性抗体(优选双特异性抗体或三特异性抗体),其还包含一个或多个与其他抗原特异性结合的第二抗体。
如本文所用,术语“多特异性”是指抗原结合蛋白具有多表位特异性(即,能够特异性结合一个生物分子上的两个、三个或更多个不同的表位或能够特异性结合两个、三个或更多个不同的生物分子上的表位)。如本文所用,术语“双特异性”表示抗原结合蛋白具有两种不同的抗原结合特异性。
在一个实施方案中,第二抗体可以具有任何抗体或抗体片段形式,例如全长抗体、Fab、Fab'、(Fab') 2、Fv、scFv、scFv-scFv、微 抗体、双抗体或sdAb。
因此,在一个实施方案中,所述第二抗体靶向选自以下的抗原:BCMA、CD4、CD5、CD8、CD14、CD15、CD19、CD20、CD21、CD22、CD23、CD25、CD33、CD37、CD38、CD40、CD40L、CD46、CD52、CD54、CD74、CD80、CD126、CD138、B7、MUC-1、Ia、HM1.24、HLA-DR、腱生蛋白、血管生成因子、VEGF、PIGF、ED-B纤连蛋白、致癌基因、致癌基因产物、CD66a-d、坏死抗原、Ii、IL-2、T101、TAC、IL-6、ROR1、TRAIL-R1(DR4)、TRAIL-R2(DR5)、tEGFR、Her2、L1-CAM、间皮素、CEA、乙型肝炎表面抗原、抗叶酸受体、CD24、CD30、CD44、EGFR、EGP-2、EGP-4、EPHa2、ErbB2、ErbB3、ErbB4、ErbB二聚体、EGFR vIII、FBP、FCRL5、FCRH5、胎儿乙酰胆碱受体、GD2、GD3、G蛋白偶联受体C型家族5D(GPRC5D)、HMW-MAA、IL-22R-α、IL-13R-α2、kdr、κ轻链、Lewis Y、L1-细胞粘附分子(L1-CAM)、黑色素瘤相关抗原(MAGE)-A1、MAGE-A3、MAGE-A6、黑色素瘤优先表达抗原(PRAME)、生存素、EGP2、EGP40、TAG72、B7-H6、IL-13受体a2(IL-13Ra2)、CA9、CD171、G250/CAIX、HLA-A1、HLA-A2、NY-ESO-1、PSCA、叶酸受体-a、CD44v6、CD44v7/8、avb6整合素、8H9、NCAM、VEGF受体、5T4、胎儿AchR、NKG2D配体、双抗原、与通用标签相关的抗原、癌症-睾丸抗原、MUC1、MUC16、NY-ESO-1、MART-1、gp100、癌胚胎抗原、VEGF-R2、癌胚抗原(CEA)、前列腺特异性抗原、PSMA、Her2/neu、雌激素受体、孕酮受体、肝配蛋白B2、CD123、c-Met、GD-2、O-乙酰化GD2(OGD2)、CE7、Wilms肿瘤1(WT-1)、细胞周期蛋白、细胞周期蛋白A2、CCL-1、hTERT、MDM2、CYP1B、WT1、活素、AFP、p53、细胞周期蛋白(D1)、CS-1、BAFF-R、TACI、CD56、TIM-3、CD123、L1-细胞粘附分子、MAGE-A1、MAGEA3、细胞周期蛋白(如细胞周期蛋白A1(CCNA1))和/或病原体特异性抗原、生物素化分子、由HIV、HCV、HBV和/或其他病原体表达的分子;和/或新表位或新抗原。
核酸、载体、宿主细胞
在另一方面中,本发明涉及编码本发明的CD7人源化抗体或多特异性抗体的核酸分子。本发明的核酸可为RNA、DNA或cDNA。根据本发明的一个实施方案,本发明的核酸是基本上分离的核酸。
在一个实施方案中,编码所述CD7人源化抗体的核酸分子与选自SEQ ID NO:20-23的核苷酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、100%序列同一性,并且其编码的CD7人源化抗体能够特异性结合CD7抗原。优选地,编码所述CD7人源化抗体的核酸分子如SEQ ID NO:20-23所示。
本发明的核酸也可呈载体形式,可存在于载体中和/或可为载体的一部分,该载体例如质粒、粘端质粒或YAC。载体可尤其为表达载体,即可提供CD7人源化抗体在体外和/或体内(即在适合宿主细胞、宿主有机体和/或表达系统中)表达的载体。该表达载体通常包含至少一种本发明的核酸分子,其可操作地连接至一个或多个适合的表达调控元件(例如启动子、增强子、终止子等)。对所述调控元件及其序列进行选择以便在特定宿主中表达是本领域技术人员熟知的。对本发明的CD7人源化抗体的表达有用或必需的调控元件及其他元件的具体实例包括但不限于启动子、增强子、终止子、整合因子、选择标记物、前导序列、报告基因。
在另一方面中,本发明还提供表达本发明的CD7人源化抗体、多特异性抗体和/或含有本发明的核酸或载体的宿主细胞。本发明的优选宿主细胞为细菌细胞、真菌细胞或哺乳动物细胞。
适合的细菌细胞包括革兰氏阴性细菌菌株(例如大肠杆菌(Escherichia coli)菌株、变形杆菌属(Proteus)菌株及假单胞菌属(Pseudomonas)菌株)及革兰氏阳性细菌菌株(例如芽孢杆菌属(Bacillus)菌株、链霉菌属(Streptomyces)菌株、葡萄球菌属(Staphylococcus)菌株及乳球菌属(Lactococcus)菌株)的细胞。
适合的真菌细胞包括木霉属(Trichoderma)、脉孢菌属(Neurospora)及曲菌属(Aspergillus)的物种的细胞;或者包括酵母属 (Saccharomyces)(例如酿酒酵母(Saccharomyces cerevisiae))、裂殖酵母属(Schizosaccharomyces)(例如粟酒裂殖酵母(Schizosaccharomyces pombe))、毕赤酵母属(Pichia)(例如巴斯德毕赤酵母(Pichiapastoris)及嗜甲醇毕赤酵母(Pichia methanolica))及汉森酵母属(Hansenula)的物种的细胞。
适合的哺乳动物细胞包括例如HEK293细胞、CHO细胞、BHK细胞、HeLa细胞、COS细胞等。
然而,本发明也可使用两栖类细胞、昆虫细胞、植物细胞及本领域中用于表达异源蛋白的任何其他细胞。
嵌合抗原受体
在另一方面,本发明还提供包含如上所述的CD7人源化抗体的重组受体,例如重组TCR受体或嵌合抗原受体。优选地,本发明还提供包含如上所述的CD7人源化抗体的嵌合抗原受体。
如本文所用,术语“嵌合抗原受体”或“CAR”是指人工构建的杂合多肽,该杂合多肽一般包括配体结合结构域(例如抗体的抗原结合部分)、跨膜结构域、任选的共刺激结构域和细胞内信号传导结构域,各个结构域之间通过接头连接。CAR能够利用抗体的抗原结合特性以非MHC限制性的方式将T细胞和其它免疫细胞的特异性和反应性重定向至所选择的靶标。
在一个实施方案中,本发明提供一种嵌合抗原受体,其包含如上所述的CD7人源化抗体或含有所述CD7人源化抗体的多特异性抗体、跨膜结构域和胞内信号传导结构域。
如本文所用,术语“跨膜结构域”是指能够使嵌合抗原受体在免疫细胞(例如淋巴细胞、NK细胞或NKT细胞)表面上表达,并且引导免疫细胞针对靶细胞的细胞应答的多肽结构。跨膜结构域可以是天然或合成的,也可以源自任何膜结合蛋白或跨膜蛋白。当嵌合抗原受体与靶抗原结合时,跨膜结构域能够进行信号传导。特别适用于本发明中的跨膜结构域可以源自例如TCRα链、TCRβ链、TCRγ链、TCRδ链、CD3ζ亚基、CD3ε亚基、CD3γ亚基、CD3 δ亚基、CD45、CD4、CD5、CD8α、CD9、CD16、CD22、CD33、CD28、CD37、CD64、CD80、CD86、CD134、CD137、CD154及其功能性片段。或者,跨膜结构域可以是合成的并且可以主要地包含疏水性残基如亮氨酸和缬氨酸。优选地,所述跨膜结构域源自CD8α链或CD28,其与SEQ ID NO:24或26所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:25或27所示的核酸分子具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。
如本文所用,术语“胞内信号传导结构域”是指转导效应子功能信号并指导细胞进行指定功能的蛋白质部分。在一个实施方案中,本发明的嵌合抗原受体包含的胞内信号传导结构域可以是T细胞受体和共受体的胞内区序列,其在抗原受体结合以后一同起作用以引发信号传导,以及这些序列的任何衍生物或变体和具有相同或相似功能的任何合成序列。胞内信号传导结构域可以包含许多免疫受体酪氨酸激活基序(Immunoreceptor Tyrosine-based Activation Motifs,ITAM)。本发明的胞内信号传导结构域的非限制性施例包括但不限于FcRγ、FcRβ、CD3γ、CD3δ、CD3ε、CD3ζ、CD22、CD79a、CD79b和CD66d等的胞内区。在优选的实施方式中,本发明CAR的信号传导结构域可以包含CD3ζ胞内区,其与SEQ ID NO:32或34所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:33或35所示的核酸分子具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。
在一个实施方案中,本发明的嵌合抗原受体还可以包含位于抗体和跨膜结构域之间的铰链区。如本文所用,术语“铰链区”一般是指作用为连接跨膜结构域至抗体的任何寡肽或多肽。具体地,铰链区用来为抗体提供更大的灵活性和可及性。铰链区可以包含最多达300个氨基酸,优选10至100个氨基酸并且最优选25至50个氨 基酸。铰链区可以全部或部分源自天然分子,如全部或部分源自CD8、CD4或CD28的胞外区,或全部或部分源自抗体恒定区。或者,铰链区可以是对应于天然存在的铰链序列的合成序列,或可以是完全合成的铰链序列。在优选的实施方式中,所述铰链区包含CD8α、CD28、FcγRIIIα受体、IgG4或IgG1的铰链区部分,更优选CD8α、CD28或IgG4铰链,其与SEQ ID NO:40、42或44所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:41、43或45所示的核苷酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。
在一个实施方案中,嵌合抗原受体还可以包含一个或多个共刺激结构域。共刺激结构域可以是来自共刺激分子的细胞内功能性信号传导结构域,其包含所述共刺激分子的整个细胞内部分,或其功能片段。“共刺激分子”是指在T细胞上与共刺激配体特异性结合,由此介导T细胞的共刺激反应(例如增殖)的同源结合配偶体。共刺激分子包括但不限于1类MHC分子、BTLA和Toll配体受体。本发明的共刺激结构域的非限制性施例包括但不限于源自以下蛋白质的共刺激信号传导结构域:TLR1、TLR2、TLR3、TLR4、TLR5、TLR6、TLR7、TLR8、TLR9、TLR10、CARD11、CD2、CD7、CD8、CD18(LFA-1)、CD27、CD28、CD30、CD40、CD54(ICAM)、CD83、CD134(OX40)、CD137(4-1BB)、CD270(HVEM)、CD272(BTLA)、CD276(B7-H3)、CD278(ICOS)、CD357(GITR)、DAP10、LAT、NKG2C、SLP76、PD-1、LIGHT、TRIM以及ZAP70。优选地,本发明CAR的共刺激结构域来自4-1BB、CD28或4-1BB+CD28。在一个实施方案中,4-1BB共刺激结构域与SEQ ID NO:30所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:31所示的核酸分子具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。在一个实施方案中,CD28共刺 激结构域与SEQ ID NO:28所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:29所示的核酸分子具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。
在一个实施方案中,本发明的CAR还可以包含信号肽,使得当其在细胞例如T细胞中表达时,新生蛋白质被引导至内质网并随后引导至细胞表面。信号肽的核心可以含有长的疏水性氨基酸区段,其具有形成单个α-螺旋的倾向。在信号肽的末端,通常有被信号肽酶识别和切割的氨基酸区段。信号肽酶可以在移位期间或完成后切割,以产生游离信号肽和成熟蛋白。然后,游离信号肽被特定蛋白酶消化。可用于本发明的信号肽是本领域技术人员熟知的,例如衍生自B2M、CD8α、IgG1、GM-CSFRα等的信号肽。在一个实施方案中,可用于本发明的信号肽来自B2M或CD8α,其与SEQ ID NO:36或38所示的氨基酸序列具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性,或其编码序列与SEQ ID NO:37或39所示的核酸分子具有至少70%,优选至少80%,更优选至少90%、95%、97%或99%或100%的序列同一性。
在一个实施方案中,所述CAR含有如本文所提供的CD7人源化抗体或含有所述CD7人源化抗体的多特异性抗体、CD8α跨膜区、CD28或4-1BB共刺激结构域,和CD3ζ胞内信号传导结构域。在该实施方案中,所述CAR还可以进一步包含来自B2M、CD8α、IgG1或GM-CSFRα的信号肽。
本发明还提供编码如上所定义的靶向CD7的嵌合抗原受体的核酸分子,以及包含所述核酸分子的载体。
如本文所用,术语“载体”是用作将(外源)遗传材料转移到宿主细胞中的媒介核酸分子,在该宿主细胞中所述核酸分子可以例如复制和/或表达。载体一般包括靶向载体和表达载体。“靶向载体”是 通过例如同源重组或使用特异性靶向位点处序列的杂合重组酶将分离的核酸递送至细胞内部的介质。“表达载体”是用于异源核酸序列(例如编码本发明的嵌合抗原受体多肽的那些序列)在合适的宿主细胞中的转录以及它们的mRNA的翻译的载体。可用于本发明的合适载体是本领域已知的,并且许多可商购获得。在一个实施方案中,本发明的载体包括但不限于质粒、病毒(例如逆转录病毒、慢病毒、腺病毒、牛痘病毒、劳氏肉瘤病毒(RSV、多瘤病毒和腺相关病毒(AAV)等)、噬菌体、噬菌粒、粘粒和人工染色体(包括BAC和YAC)。载体本身通常是核酸分子,通常是包含插入物(转基因)的DNA序列和作为载体“骨架”的较大序列。工程化载体通常还包含在宿主细胞中自主复制的起点(如果需要多核苷酸的稳定表达)、选择标记和限制酶切割位点(如多克隆位点,MCS)。载体可另外包含启动子、多聚腺苷酸尾(polyA)、3’UTR、增强子、终止子、绝缘子、操纵子、选择标记、报告基因、靶向序列和/或蛋白质纯化标签等元件。在一个具体的实施方案中,所述载体是体外转录的载体。
工程化免疫细胞
在一个方面,本发明还提供表达本发明所述CAR的工程化免疫细胞。
如本文所用,术语“免疫细胞”是指免疫系统的具有一种或多种效应子功能(例如,细胞毒性细胞杀伤活性、分泌细胞因子、诱导ADCC和/或CDC)的任何细胞。例如,免疫细胞可以是T细胞、巨噬细胞、树突状细胞、单核细胞、NK细胞和/或NKT细胞。在一个实施方案中,免疫细胞衍生自干细胞,例如成体干细胞、胚胎干细胞、脐带血干细胞、祖细胞、骨髓干细胞、诱导多能干细胞、全能干细胞或造血干细胞等。优选地,免疫细胞是T细胞。T细胞可以是任何T细胞,如体外培养的T细胞,例如原代T细胞,或者来自体外培养的T细胞系例如Jurkat、SupT1等的T细胞,或获得自受试者的T细胞。受试者的实例包括人、狗、猫、小鼠、大鼠及其转基因物种。T细胞可以从多种来源获得,包括外周血单核细胞、骨髓、 淋巴结组织、脐血、胸腺组织、来自感染部位的组织、腹水、胸膜积液、脾组织及肿瘤。T细胞也可以被浓缩或纯化。T细胞可以处于任何发育阶段,包括但不限于,CD4+/CD8+T细胞、CD4+辅助T细胞(例如Th1和Th2细胞)、CD8+T细胞(例如,细胞毒性T细胞)、肿瘤浸润细胞、记忆T细胞、幼稚T细胞、γδ-T细胞、αβ-T细胞等。在一个优选的实施方案中,免疫细胞是人T细胞。可以使用本领域技术人员已知的多种技术,如Ficoll分离从受试者的血液获得T细胞。
采用本领域已知的常规方法(如通过转导、转染、转化等)可以将编码嵌合抗原受体的核酸序列引入免疫细胞。“转染”是将核酸分子或多核苷酸(包括载体)引入靶细胞的过程。一个例子是RNA转染,即将RNA(比如体外转录的RNA,ivtRNA)引入宿主细胞的过程。该术语主要用于真核细胞中的非病毒方法。术语“转导”通常用于描述病毒介导的核酸分子或多核苷酸的转移。动物细胞的转染通常涉及在细胞膜中打开瞬时的孔或“洞”,以允许摄取材料。可以使用磷酸钙、通过电穿孔、通过细胞挤压或通过将阳离子脂质与材料混合以产生与细胞膜融合并将它们的运载物沉积入内部的脂质体,进行转染。用于转染真核宿主细胞的示例性技术包括脂质囊泡介导的摄取、热休克介导的摄取、磷酸钙介导的转染(磷酸钙/DNA共沉淀)、显微注射和电穿孔。术语“转化”用于描述核酸分子或多核苷酸(包括载体)向细菌中、也向非动物真核细胞(包括植物细胞)中的非病毒转移。因此,转化是细菌或非动物真核细胞的基因改变,其通过细胞膜从其周围直接摄取并随后并入外源遗传材料(核酸分子)而产生。转化可以通过人工手段实现。为了发生转化,细胞或细菌必须处于感受态的状态。对于原核转化,技术可包括热休克介导的摄取、与完整细胞的细菌原生质体融合、显微注射和电穿孔。将核酸或载体引入免疫细胞后,本领域技术人员可以通过常规技术对所得免疫细胞进行扩增和活化。
在一个实施方案中,为减少移植物抗宿主病的风险,所述工程 化免疫细胞还包含至少一种选自以下的基因的表达被抑制或沉默:CD52、GR、dCK、TCR/CD3基因(例如TRAC、TRBC、CD3γ、CD3δ、CD3ε、CD3ζ)、MHC相关基因(HLA-A、HLA-B、HLA-C、B2M、HLA-DPA、HLA-DQ、HLA-DRA、TAP1、TAP2、LMP2、LMP7、RFX5、RFXAP、RFXANK、CIITA)和免疫检查点基因,如PD1、LAG3、TIM3、CTLA4、PPP2CA、PPP2CB、PTPN6、PTPN22、PDCD1、HAVCR2、BTLA、CD160、TIGIT、CD96、CRTAM、TNFRSF10B、TNFRSF10A、CASP8、CASP10、CASP3、CASP6、CASP7、FADD、FAS、TGFBRII、TGFRBRI、SMAD2、SMAD3、SMAD4、SMAD10、SKI、SKIL、TGIF1、IL10RA、IL10RB、HMOX2、IL6R、IL6ST、EIF2AK4、CSK、PAG1、SIT、FOXP3、PRDM1、BATF、GUCY1A2、GUCY1A3、GUCY1B2和GUCY1B3。优选地,所述工程化免疫细胞还包含至少一种选自以下的基因的表达被抑制或沉默:TRAC、TRBC、HLA-A、HLA-B、HLA-C、B2M、RFX5、RFXAP、RFXANK、CIITA、PD1、LAG3、TIM3、CTLA4,更优选TRAC、TRBC、HLA-A、HLA-B、HLA-C、B2M、RFX5、RFXAP、RFXANK、CIITA。
抑制基因表达或使基因沉默的方法是本领域技术人员熟知的。例如,可以使用反义RNA、RNA诱饵、RNA适体、siRNA、shRNA/miRNA、反式显性阴性蛋白(TNP)、嵌合/抗体偶联物、趋化因子配体、抗感染性细胞蛋白、细胞内抗体(sFv)、核苷类似物(NRTI)、非核苷类似物(NNRTI)、整合酶抑制剂(寡核苷酸、二核苷酸和化学剂)和蛋白酶抑制剂来抑制基因的表达。另外,也可以通过例如大范围核酸酶、锌指核酸酶、TALE核酸酶或CRISPR系统中的Cas酶介导DNA断裂,从而使基因沉默。
在一个实施方案中,提供多种免疫细胞,每种免疫细胞被改造为表达一种或多种嵌合抗原受体。例如,在一些实施方案中,将一种免疫细胞改造为表达结合和/或靶向CD7的嵌合抗原受体(例如包含本发明所述CD7人源化抗体的CAR),并且将另一种细胞改造为表达结合和/或靶向其他抗原的嵌合抗原受体。在一个实施方案中, 免疫细胞也可以表达多特异性嵌合抗原受体,其靶向包括CD7在内的一种或多种抗原。例如,这种多特异性嵌合抗原受体可以包含靶向CD7的多特异性抗体,或者同时包含本发明所述的CD7人源化抗体和靶向其他抗原的抗体。在此类实施方案中,所述多种工程化免疫细胞可以一起或单独施用。在一个实施方案中,所述多种免疫细胞可以在同一组合物中或在不同组合物中。细胞的示例性组合物包括本申请以下章节中所描述的组合物。
抗体偶联物
在一个方面,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和第二功能性结构,其中所述第二功能性结构选自Fc、放射性同位素、延长半衰期的结构部分、可检测标记物和药物。
在一个实施方案中,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和Fc。如本文所用,术语“Fc”用于定义免疫球蛋白重链的C末端区,其包括天然Fc和变体Fc。“天然Fc”是指包含通过消化完整抗体产生的、无论是单体形式或是多聚体形式的非抗原结合片段的分子或序列。产生天然Fc的免疫球蛋白源优选来源于人类。天然Fc片段由可以通过共价连接(例如二硫键)和非共价连接而连接为二聚体或多聚体形式的单体多肽构成。根据类别(例如IgG、IgA、IgE、IgD、IgM)或亚型(例如IgG1、IgG2、IgG3、IgA1、IgGA2)的不同,天然Fc分子单体亚基之间具有1-4个分子间二硫键。天然Fc的一个实例是通过用木瓜蛋白酶消化IgG产生的二硫键连接的二聚体(参见Ellison等(1982),Nucleic Acids Res.10:4071-9)。本文所用的术语“天然Fc”一般是指单体、二聚体和多聚体形式。“变体Fc”是指由于至少一个本文定义的“氨基酸修饰”而与“天然”或“野生型”Fc的氨基酸序列不同的氨基酸序列,也称为“Fc变体”。因此,“Fc”也包括单链Fc(scFc),即,由多肽接头连接的两个Fc单体组成的单链Fc,其能够自然折叠成功能性二聚体Fc区域。在一个实施方案中,所述Fc 优选是人免疫球蛋白的Fc,更优选是人IgG1的Fc。
在一个实施方案中,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和放射性同位素。可用于本发明的放射性同位素的实例包括但不限于At 211、I 131、I 125、Y 90、Re 186、Re 188、Sm 153、Bi 212、P 32、Pb 21299mTc、 123I、 18F和 68Ga。
在一个实施方案中,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和延长半衰期的结构部分,所述延长半衰期的结构部分选自白蛋白的结合结构、转铁蛋白的结合结构、聚乙二醇分子、重组聚乙二醇分子、人血清白蛋白、人血清白蛋白的片段和结合人血清白蛋白的白多肽(包括抗体)。
在一个实施方案中,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和可检测标记物。术语“可检测标记物”在本文中意指产生可检测信号的化合物。例如,可检测标记物可以是MRI造影剂、闪烁扫描造影剂、X射线成像造影剂、超声造影剂、光学成像造影剂。可检测标记物的实施例包括荧光团(如荧光素、Alexa或花青)、化学发光化合物(如鲁米诺)、生物发光化合物(如荧光素酶或碱性磷酸酶)、酶(如辣根过氧化物酶、葡萄糖-6-磷酸酶、β-半乳糖苷酶)、抗生素(例如卡那霉素、氨苄霉素、氯霉素、四环素等)抗性基因和造影剂(如纳米颗粒或钆)。本领域技术人员可以根据所用的检测系统选择合适的可检测标记物。
在一个实施方案中,本发明提供一种抗体偶联物,其包含本发明所定义的CD7人源化抗体和与所述CD7人源化抗体偶联的药物,例如细胞毒素或免疫调节剂(即,抗体药物偶联物)。通常药物通过共价与抗体连接,并且通常依赖于接头。在一个实施方案中,所述药物是细胞毒素。在另一个实施方案中,所述药物是免疫调节剂。细胞毒素的实例包括但不限于甲氨蝶呤、氨基蝶呤、6-巯基嘌呤、6-硫鸟嘌呤、阿糖胞苷、5-氟尿嘧啶、达卡巴嗪、氮芥、噻替派、苯丁酸氮芥、美法仑、卡莫司汀(BSNU)、洛莫司汀(CCNU)、1-甲基亚硝基脲、环磷酰胺、氮芥、白消安、二溴甘露醇、链佐星、丝裂霉素、 顺-二氯二胺铂(II)(DDP)、顺铂、卡铂、佐柔比星、多柔比星、地托比星、卡米诺霉素、伊达比星、表柔比星、米托蒽醌、放线菌素D、博来霉素、刺孢霉素、光辉霉素、安曲霉素(AMC)、长春新碱、长春花碱、紫杉醇、蓖麻毒素、假单胞菌外毒素、吉西他滨、细胞松弛素B、短杆菌肽D、溴乙锭、依米丁、依托泊苷、替尼泊苷、秋水仙素、二羟基蒽二酮、1-脱氢睾酮、糖皮质激素、普鲁卡因、丁卡因、利多卡因、普萘洛尔、嘌呤霉素、丙卡巴肼、羟基脲、天冬酰胺酶、皮质类固醇、米托坦(O,P'-(DDD))、干扰素,以及它们的组合。免疫调节剂的实例包括但不限于更昔洛韦、依那西普、他克莫司、西罗莫司、伏环孢素、环孢灵、雷帕霉素、环磷酰胺、硫唑嘌呤、霉酚酸酯、甲氨蝶呤、糖皮质素及其类似物、细胞因子、干细胞生长因子、淋巴毒素、肿瘤坏死因子(TNF)、造血因子、白介素(例如IL-1、IL-2、IL-3、IL-6、IL-10、IL-12、IL-18及IL-21)、集落刺激因子(例如G-CSF及(GM-CSF)、干扰素(例如干扰素-α、干扰素-β及干扰素-γ)、命名为“S1因子”的干细胞生长因子、红细胞生成素和血小板生成素,或其组合。
试剂盒和药物组合物
在另一个方面,本发明还提供一种检测试剂盒,其包含本发明所述的人源化抗体、多特异性抗体、抗体偶联物或嵌合抗原受体。
在另一个方面,本发明还提供一种药物组合物,其包含本发明所述的人源化抗体、嵌合抗原受体、多特异性抗体或抗体偶联物,和一种或多种药学上可接受的赋形剂。
如本文所用,术语“药学上可接受的赋型剂”是指在药理学和/或生理学上与受试者和活性成分相容(即,能够引发所需的治疗效果而不会引起任何不希望的局部或全身作用)的载体和/或赋形剂,其是本领域公知的(参见例如Remington's Pharmaceutical Sciences.Edited by Gennaro AR,19th ed.Pennsylvania:Mack Publishing Company,1995)。药学上可接受的赋型剂的实例包括但不限于填充剂、粘合剂、崩解剂、包衣剂、吸附剂、抗粘附剂、助流剂、抗氧 化剂、调味剂、着色剂、甜味剂、溶剂、共溶剂、缓冲剂、螯合剂、表面活性剂、稀释剂、润湿剂、防腐剂、乳化剂、包覆剂、等渗剂、吸收延迟剂、稳定剂和张力调节剂。本领域技术人员已知选择合适的赋型剂以制备本发明期望的药物组合物。用于本发明的药物组合物中的示例性赋型剂包括盐水、缓冲盐水、葡萄糖和水。通常,合适的赋形剂的选择尤其取决于所使用的活性剂、待治疗的疾病和药物组合物的期望剂型。
根据本发明的药物组合物可适用于多种途径施用。通常,通过胃肠外完成施用。胃肠外递送方法包括局部、动脉内、肌内、皮下、髓内、鞘内、心室内、静脉内、腹膜内、子宫内、阴道内、舌下或鼻内施用。
根据本发明的药物组合物也可以制备成各种形式,如固态、液态、气态或冻干形式,特别可以是软膏、乳膏、透皮贴剂、凝胶、粉末、片剂、溶液、气雾剂、颗粒、丸剂、混悬剂、乳剂、胶囊、糖浆、酏剂、浸膏剂、酊剂或流浸膏提取物的形式,或者是特别适用于所需施用方法的形式。本发明已知的用于生产药物的过程可包括例如常规混合、溶解、制粒、制糖衣、研磨、乳化、包封、包埋或冻干过程。包含例如本文所述的免疫细胞的药物组合物通常以溶液形式提供,并且优选包含药学上可接受的缓冲剂。
根据本发明的药物组合物还可以与一种或多种适用于治疗和/或预防待治疗疾病的其它药剂组合施用。适用于组合的药剂的优选实例包括已知的抗癌药物,比如顺铂、美登素衍生物、雷查霉素(rachelmycin)、卡里奇霉素(calicheamicin)、多西紫杉醇、依托泊苷、吉西他滨、异环磷酰胺、伊立替康、美法仑、米托蒽醌、sorfimer卟啉钠II(sorfimer sodiumphotofrin II)、替莫唑胺、拓扑替康、葡萄糖醛酸曲美沙特(trimetreate glucuronate)、奥利斯他汀E(auristatin E)、长春新碱和阿霉素;肽细胞毒素,比如蓖麻毒素、白喉毒素、假单胞菌细菌外毒素A、DNA酶和RNA酶;放射性核素,比如碘131、铼186、铟111、铱90、铋210和213、锕225和砹213;前药,比 如抗体定向的酶前药;免疫刺激剂,比如血小板因子4、黑色素瘤生长刺激蛋白等;抗体或其片段,比如抗CD3抗体或其片段,补体活化剂,异种蛋白结构域,同种蛋白结构域,病毒/细菌蛋白结构域和病毒/细菌肽。此外,本发明的药物组合物也可以与其他一种或多种治疗方法,例如化疗、放疗组合使用。
治疗/预防/诊断用途
在另一个方面,本发明还提供一种治疗和/或预防和/或诊断与CD7表达相关的疾病的方法,包括向受试者施用如上所述的人源化抗体、嵌合抗原受体、多特异性抗体、抗体偶联物或药物组合物。
在一个实施方案中,与CD7表达相关的疾病包括非实体瘤(诸如血液学肿瘤,例如白血病和淋巴瘤)和实体瘤。血液学肿瘤是血液或骨髓的癌症,包括但不限于急性白血病(诸如急性淋巴细胞白血病、急性髓细胞白血病、急性骨髓性白血病和成髓细胞性、前髓细胞性、粒-单核细胞型、单核细胞性和红白血病)、慢性白血病(诸如慢性髓细胞(粒细胞性)白血病、慢性骨髓性白血病和慢性淋巴细胞白血病)、真性红细胞增多症、淋巴瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤(无痛和高等级形式)、多发性骨髓瘤、瓦尔登斯特伦氏巨球蛋白血症、骨髓增生异常综合征、多毛细胞白血病、伯基特淋巴瘤、弥漫性大细胞淋巴瘤、套细胞淋巴瘤、T淋巴母细胞性白血病/淋巴瘤(T-ALL/LBL)、早期前T淋巴母细胞白血病(ETP-ALL)、结外NK/T细胞淋巴瘤、小淋巴细胞淋巴瘤(SLL)和脊髓发育不良。实体瘤是通常不包含囊肿或液体区的组织的异常肿块,其可以是良性或恶性的。不同类型的实体瘤以形成它们的细胞类型命名(诸如肉瘤、癌和淋巴瘤)。实体瘤的实例包括但不限于纤维肉瘤、粘液肉瘤、脂肪肉瘤间皮瘤、胰腺癌、卵巢癌,腹膜、大网膜和肠系膜癌、咽癌、前列腺癌、直肠癌、肾癌、皮肤癌、小肠癌、黑素瘤、肾癌,喉癌、软组织癌、胃癌、睾丸癌、结肠癌,食道癌,宫颈癌、腺泡型横纹肌肉瘤、膀胱癌、骨癌、脑癌、乳腺癌、肛门癌、眼癌、肝内胆管癌、关节癌、颈癌、胆囊癌、胸膜癌、鼻癌、中耳癌、口腔癌、外 阴癌、甲状腺癌和输尿管癌。
在一个实施方案中,与CD7表达相关的疾病优选选自急性淋巴细胞白血病(ALL)、急性髓细胞白血病(AML)、T淋巴母细胞性淋巴瘤(T-LBL)、早期前T淋巴母细胞白血病(ETP-ALL)和结外NK/T细胞淋巴瘤。
下面将参考附图并结合实例来详细说明本发明。需要说明的是,本领域的技术人员应该理解本发明的附图及其实施例仅仅是为了例举的目的,并不能对本发明构成任何限制。在不矛盾的情况下,本申请中的实施例及实施例中的特征可以相互组合。
附图说明
图1:示出了包含CD7人源化抗体的CAR-T细胞的scFv表达水平。
图2:示出了包含CD7人源化抗体的CAR-T细胞对靶细胞的杀伤效果。
图3:示出了包含CD7人源化抗体的CAR-T细胞与靶细胞共培养后的细胞因子释放水平。
具体实施方式
实施例1.制备CD7人源化抗体
基于鼠源CD7scFv(克隆m189)制备人源化抗体。克隆m189包含如SEQ ID NO:1所示的CDR-L1、如SEQ ID NO:2所示的CDR-L2、如SEQ ID NO:3所示的CDR-L3、如SEQ ID NO:4所示的CDR-H1、如SEQ ID NO:5所示的CDR-H2、如SEQ ID NO:6所示的CDR-H3,其氨基酸序列如SEQ ID NO:7所示。制备人源化抗体的具体方法如下:首先通过Ig BLAST(http://www.ncbi.nlm.nih.gov/igblast/)和IMGT(免疫基因数据库IMGT:http://www.imgt.org)分别进行序列相似性搜索,并在搜索的结果中,选择与轻链和重链均有较高序列相似性的抗体作为人源 化抗体模板。然后,将鼠源CD7scFv中的重链CDR和轻链CDR移植到序列相似度较高的人源化抗体模板的框架中,而后做回复突变,以保证人源化抗体的亲和力和特异性。最终得到CD7人源化单链抗体(scFv)氨基酸序列如下表1所示。
表1.人源化CD7scFv的序列
  h189-1 h189-2 h189-3 h189-4
VL(氨基酸) SEQ ID NO:8 SEQ ID NO:11 SEQ ID NO:14 SEQ ID NO:17
VH(氨基酸) SEQ ID NO:9 SEQ ID NO:12 SEQ ID NO:15 SEQ ID NO:18
scFv(氨基酸) SEQ ID NO:10 SEQ ID NO:13 SEQ ID NO:16 SEQ ID NO:19
scFv(核苷酸) SEQ ID NO:20 SEQ ID NO:21 SEQ ID NO:22 SEQ ID NO:23
实施例2.制备包含CD7人源化抗体的CAR-T细胞
合成编码以下蛋白的序列,并将其克隆至pLVX载体(Public Protein/Plasmid Library(PPL),货号:PPL00157-4a):CD8α信号肽(SEQ ID NO:38)、CD7人源化scFv(选自SEQ ID NO:10、13、16、19)、CD8α铰链区(SEQ ID NO:40)、CD8α跨膜区(SEQ ID NO:24)、4-1BB胞内区(SEQ ID NO:30)、CD3ζ胞内信号传导结构域(SEQ ID NO:32),并通过测序确认目标序列的正确插入。其中,hCAR7-1CAR包含的抗CD7scFv的氨基酸序列如SEQ ID NO:10所示;hCAR7-2CAR包含的抗CD7scFv的氨基酸序列如SEQ ID NO:13所示;hCAR7-3CAR包含的抗CD7scFv的氨基酸序列如SEQ ID NO:16所示;hCAR7-4CAR包含的抗CD7scFv的氨基酸序列如SEQ ID NO:19所示。
在无菌管中加入3ml Opti-MEM(Gibco,货号31985-070)稀释上述质粒后,再根据质粒:病毒包装载体:病毒包膜载体=4:2:1的比例加入包装载体psPAX2(Addgene,货号12260)和包膜载体pMD2.G(Addgene,货号12259)。然后,加入120ul X-treme GENE HP DNA转染试剂(Roche,货号06366236001),立即混匀,于室温下孵育15min,然后将质粒/载体/转染试剂混合物逐滴加入到293T细胞的培 养瓶中。在24小时和48小时收集病毒,将其合并后,超速离心(25000g,4℃,2.5小时)获得浓缩的慢病毒。
由于CD7也在T细胞上表达,因此为了避免CAR-T细胞之间的互相杀伤,发明人通过CRISP/Cas9系统敲除了T细胞中的CD7。培养1d后,用DynaBeads CD3/CD28 CTS TM(Gibco,货号40203D)激活T细胞,并在37℃和5%CO2下继续培养1天。然后,加入浓缩的慢病毒,持续培养3天后,获得表达不同CD7人源化scFv的CAR T细胞。未经修饰的野生型T细胞用作阴性对照(NT)。
使用Biotin-SP(long spacer)AffiniPure Goat Anti-Mouse IgG,F(ab') 2Fragment Specific(min X Hu,Bov,Hrs Sr Prot)(jackson immunoresearch,货号115-065-072)作为一抗,APC Streptavidin(BD Pharmingen,货号554067)或PE Streptavidin(BD Pharmingen,货号554061)作为二抗,通过流式细胞仪检测hCAR7-1 T细胞、hCAR7-2 T细胞、hCAR7-3 T细胞和hCAR7-4上的scFv的表达水平,结果如图1所示。
可以看出,本发明制备的CAR T细胞中的CD7人源化scFv均可以有效表达。
实施例3:CAR T细胞对靶细胞的杀伤效果和细胞因子释放
3.1 CAR-T细胞对靶细胞的杀伤效果
当T细胞对靶细胞有杀伤时,靶细胞的数量就会减少。将T细胞和带有可表达荧光素酶的靶细胞共培养后,靶细胞数量减少的同时,分泌的荧光素酶也会随之减少。荧光素酶可以催化荧光素转化为氧化性荧光素,而在此氧化过程中,会产生生物发光,并且这种发光的强度将取决于靶细胞表达的荧光素酶的水平。因此,检测的荧光强度能够反应T细胞对靶细胞的杀伤能力。
为了检测CAR-T细胞对靶细胞的杀伤能力,首先以1x10 4/孔将携带荧光素基因的Jurkat靶细胞铺入96孔板中,然后以1:1、0.5:1和0.25:1的效靶比(即效应T细胞与靶细胞之比)将CAR T细胞和 NT细胞铺入到96孔板进行共培养,16-18小时后利用酶标仪测定荧光值。根据计算公式:(靶细胞荧光均值-样品荧光均值)/靶细胞荧光均值×100%,计算得到杀伤效率,结果如图2所示。
可以看出,与NT相比,本发明的CAR T细胞对靶细胞均有特异性杀伤。
3.2 CAR-T细胞的细胞因子释放
T细胞杀伤靶细胞时,靶细胞数量减少的同时也会释放细胞因子。根据以下步骤,使用酶联免疫吸附法(ELISA)来测定本发明的CAR T细胞杀伤靶细胞时细胞因子IFNγ的释放水平。
(1)收集细胞共培养上清液
以1x10 5/孔将靶细胞铺于96孔板中,然后以1:1的比例将CAR T和NT细胞(阴性对照)与靶细胞共培养,18-24小时后收集细胞共培养上清液。
(2)ELISA检测上清中IFNγ分泌量
使用捕获抗体Purified anti-human IFN-γAntibody(Biolegend,货号506502)包被96孔板4℃孵育过夜,然后移除抗体溶液,加入250μL含有2%BSA(sigma,货号V900933-1kg)的PBST(含0.1%吐温的1XPBS)溶液,37℃孵育2小时。然后用250μL PBST(含0.1%吐温的1XPBS)清洗板3次。每孔加入50μL细胞共培养上清液或标准品,并在37℃孵育1小时,然后用250μL PBST(含0.1%吐温的1XPBS)清洗板3次。然后向各孔分别加入50μL检测抗体Anti-Interferon gamma抗体[MD-1](Biotin)(abcam,货号ab25017),在37℃孵育1小时后,用250μL PBST(含0.1%吐温的1XPBS)清洗板3次。再加入HRP Streptavidin(Biolegend,货号405210),在37℃孵育30分钟后,弃上清液,加入250μL PBST(含0.1%吐温的1XPBS),清洗5次。向各孔加入50μL TMB底物溶液。使反应在室温下于暗处发生30分钟,之后向各孔中加入50μL 1mol/L H 2SO 4以停止反应。在停止反应的30分钟内,使用酶标仪检测450nm处吸光度,并根据标准曲线(根据标准品的读值和浓度绘制)计算 细胞因子的含量,结果如图3所示。
可以看出,本发明的CAR T细胞的细胞因子释放水显著高于对照的NT细胞。
需要说明的是,以上仅为本发明的优选实施例而已,并不用于限制本发明,对于本领域的技术人员来说,本发明可以有各种更改和变化。本领域技术人员理解的是,凡在本发明的精神和原则之内,所作的任何修改、等同替换、改进等,均应包含在本发明的保护范围之内。

Claims (19)

  1. 一种靶向CD7的人源化抗体,其包含轻链可变区和重链可变区,其中所述轻链可变区包含如SEQ ID NO:1所示的CDR-L1、如SEQ ID NO:2所示的CDR-L2、如SEQ ID NO:3所示的CDR-L3,所述重链可变区包含如SEQ ID NO:4所示的CDR-H1、如SEQ ID NO:5所示的CDR-H2、如SEQ ID NO:6所示的CDR-H3,并且所述轻链可变区与选自SEQ ID NO:8、11、14和17所示的氨基酸序列具有至少90%同一性,所述重链可变区与选自SEQ ID NO:9、12、15和18的氨基酸序列具有至少90%同一性。
  2. 权利要求1所述的人源化抗体,其中所述CD7人源化抗体包含选自SEQ ID NO:8、11、14和17所示的轻链可变区和选自SEQ ID NO:9、12、15和18的重链可变区。
  3. 权利要求1所述的人源化抗体,其中所述CD7人源化抗体的氨基酸序列选自SEQ ID NO:10、13、16和19。
  4. 一种核酸分子,其编码权利要求1-3任一项所述的人源化抗体。
  5. 权利要求4所述的核酸分子,其与选自SEQ ID NO:20-23的核苷酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、100%序列同一性,并且其编码的CD7人源化抗体能够特异性结合CD7抗原。
  6. 一种多特异性抗体,其包含权利要求1-3任一项所述的人源化抗体和一个或多个与其他抗原特异性结合的第二抗体或其抗原结合部分。
  7. 权利要求6所述的多特异性抗体,其中所述第二抗体或其抗原结合部分选自全长抗体、Fab、Fab'、(Fab') 2、Fv、scFv、scFv-scFv、微抗体、双抗体或sdAb。
  8. 一种载体,其包含编码权利要求1-3任一项所述的人源化抗体或权利要求6或7所述的多特异性抗体的核酸分子。
  9. 一种宿主细胞,其表达权利要求1-3任一项所述的人源化抗体或权利要求6或7所述的多特异性抗体。
  10. 一种嵌合抗原受体,其包含权利要求1-3任一项所述的人源化抗体或权利要求6或7所述的多特异性抗体、跨膜结构域和胞内信号传导结构域。
  11. 权利要求10所述的嵌合抗原受体,其还包含选自CD28或4-1BB的共刺激结构域。
  12. 权利要求11所述的嵌合抗原受体,其包含权利要求1-3任一项所述的人源化抗体或权利要求6或7所述的多特异性抗体、CD8α跨膜区、CD28或4-1BB共刺激结构域,和CD3ζ胞内信号传导结构域。
  13. 一种工程化免疫细胞,其包含权利要求10-12任一项所述的嵌合抗原受体。
  14. 权利要求13所述的工程化免疫细胞,其选自T细胞、NK细胞、NKT细胞、巨噬细胞、树突细胞。
  15. 一种抗体偶联物,其包含权利要求1-3任一项所述的人源化抗体或权利要求6或7所述的多特异性抗体,和第二功能性结构,其中所述第二功能性结构选自Fc、放射性同位素、延长半衰期的结构部分、可检测标记物和药物。
  16. 权利要求15所述的抗体偶联物,其中所述延长半衰期的结构部分选自:白蛋白的结合结构、转铁蛋白的结合结构、聚乙二醇分子、重组聚乙二醇分子、人血清白蛋白、人血清白蛋白的片段和结合人血清白蛋白的白多肽;所述可检测标记物选自荧光团、化学发光化合物、生物发光化合物、酶、抗生素抗性基因和造影剂;所述药物选自细胞毒素和免疫调节剂。
  17. 一种检测试剂盒,其包含权利要求1-3任一项所述的人源化抗体、权利要求6或7所述的多特异性抗体、权利要求10-12任一项所述的嵌合抗原受体,或权利要求15或16所述的抗 体偶联物。
  18. 一种药物组合物,包含权利要求1-3任一项所述的人源化抗体、权利要求6或7所述的多特异性抗体、权利要求10-12任一项所述的嵌合抗原受体、或权利要求15或16所述的抗体偶联物,和一种或多种药学上可接受的赋形剂。
  19. 权利要求1-3任一项所述的人源化抗体、权利要求6或7所述的多特异性抗体、权利要求10-12任一项所述的嵌合抗原受体、权利要求15或16所述的抗体偶联物或权利要求18所述的药物组合物在制备用于治疗和/或预防和/或诊断与CD7表达相关的疾病的药物中的用途。
PCT/CN2021/127570 2020-11-03 2021-10-29 靶向cd7的人源化抗体及其用途 WO2022095803A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2023524926A JP2023547425A (ja) 2020-11-03 2021-10-29 Cd7を標的にするヒト化抗体及びその使用
EP21888500.2A EP4219554A4 (en) 2020-11-03 2021-10-29 HUMANIZED CD7-TARGETED ANTIBODY AND USE THEREOF
US18/034,170 US20230399398A1 (en) 2020-11-03 2021-10-29 Cd7-targeting humanized antibody and use thereof
KR1020237012746A KR20230098158A (ko) 2020-11-03 2021-10-29 Cd7 표적의 인간화 항체 및 이의 용도

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011208669.1 2020-11-03
CN202011208669.1A CN112300282A (zh) 2020-11-03 2020-11-03 靶向cd7的人源化抗体及其用途

Publications (1)

Publication Number Publication Date
WO2022095803A1 true WO2022095803A1 (zh) 2022-05-12

Family

ID=74332451

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/127570 WO2022095803A1 (zh) 2020-11-03 2021-10-29 靶向cd7的人源化抗体及其用途

Country Status (6)

Country Link
US (1) US20230399398A1 (zh)
EP (1) EP4219554A4 (zh)
JP (1) JP2023547425A (zh)
KR (1) KR20230098158A (zh)
CN (1) CN112300282A (zh)
WO (1) WO2022095803A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115991776A (zh) * 2022-10-26 2023-04-21 上海驯鹿生物技术有限公司 一种靶向cd7的全人源抗体及其应用
CN116003631A (zh) * 2022-11-15 2023-04-25 江苏元晟生物科技有限公司 靶向cd7的人源化抗体及其应用
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2024040194A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering
WO2024119157A1 (en) 2022-12-02 2024-06-06 Sana Biotechnology, Inc. Lipid particles with cofusogens and methods of producing and using the same

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112300282A (zh) * 2020-11-03 2021-02-02 南京北恒生物科技有限公司 靶向cd7的人源化抗体及其用途
US20240082306A1 (en) * 2021-02-03 2024-03-14 Bioheng Therapeutics Limited Novel chimeric antigen receptor and use thereof
CN113214402B (zh) * 2021-06-03 2022-06-07 上海交通大学 分离的抗原结合蛋白及其应用
CN115501330A (zh) * 2021-06-07 2022-12-23 南京北恒生物科技有限公司 用于细胞免疫治疗的方法和组合物
CN113603778A (zh) * 2021-08-06 2021-11-05 上海优卡迪生物医药科技有限公司 一种封闭抗体及其在制备靶向t细胞表达抗原的car-t细胞中的应用
WO2023051414A1 (zh) * 2021-09-30 2023-04-06 北恒医疗有限公司 靶向间皮素的抗体及其用途
WO2023155852A1 (zh) * 2022-02-17 2023-08-24 上海优替济生生物医药有限公司 经修饰的免疫效应细胞及其用途
WO2023185256A1 (zh) * 2022-03-29 2023-10-05 中国医学科学院血液病医院(中国医学科学院血液学研究所) 特异性结合cd7的抗体及其在制备嵌合抗原受体中的应用
CN116970082B (zh) * 2023-09-22 2023-12-12 汇智生物技术(苏州)有限公司 人源cd7单克隆抗体及其制备方法和应用

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050100543A1 (en) 2003-07-01 2005-05-12 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
US20050175606A1 (en) 2001-04-11 2005-08-11 Hua-Liang Huang Cyclic single-chain trispecific antibody
WO2006020258A2 (en) 2004-07-17 2006-02-23 Imclone Systems Incorporated Novel tetravalent bispecific antibody
US20070014794A1 (en) 1995-03-01 2007-01-18 Genentech, Inc. Method for making heteromultimeric polypeptides
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
WO2017213979A1 (en) * 2016-06-06 2017-12-14 St. Jude Children's Research Hospital Anti-cd7 chimeric antigen receptor and method of use thereof
CN109652379A (zh) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 Cd7嵌合抗原受体修饰的nk-92mi细胞及其应用
CN110268049A (zh) * 2016-11-22 2019-09-20 新加坡国立大学 用于t细胞恶性肿瘤免疫疗法的cd7表达阻滞剂和嵌合抗原受体
WO2020102589A1 (en) * 2018-11-14 2020-05-22 Medisix Therapeutics Pte Ltd. Two-gene vectors for generating car-t cells and uses thereof
CN112300282A (zh) * 2020-11-03 2021-02-02 南京北恒生物科技有限公司 靶向cd7的人源化抗体及其用途

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI560199B (en) * 2010-08-31 2016-12-01 Sanofi Sa Peptide or peptide complex binding to α2 integrin and methods and uses involving the same
US10858448B2 (en) * 2015-02-02 2020-12-08 I2 Pharmaceuticals, Inc. Anti-surrogate light chain antibodies
CN111196852A (zh) * 2018-11-16 2020-05-26 四川科伦博泰生物医药股份有限公司 抗tigit抗体及其用途
GB201905552D0 (en) * 2019-04-18 2019-06-05 Kymab Ltd Antagonists

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070014794A1 (en) 1995-03-01 2007-01-18 Genentech, Inc. Method for making heteromultimeric polypeptides
US20050175606A1 (en) 2001-04-11 2005-08-11 Hua-Liang Huang Cyclic single-chain trispecific antibody
US20050100543A1 (en) 2003-07-01 2005-05-12 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
WO2006020258A2 (en) 2004-07-17 2006-02-23 Imclone Systems Incorporated Novel tetravalent bispecific antibody
WO2007024715A2 (en) 2005-08-19 2007-03-01 Abbott Laboratories Dual variable domain immunoglobin and uses thereof
WO2017213979A1 (en) * 2016-06-06 2017-12-14 St. Jude Children's Research Hospital Anti-cd7 chimeric antigen receptor and method of use thereof
CN110268049A (zh) * 2016-11-22 2019-09-20 新加坡国立大学 用于t细胞恶性肿瘤免疫疗法的cd7表达阻滞剂和嵌合抗原受体
WO2020102589A1 (en) * 2018-11-14 2020-05-22 Medisix Therapeutics Pte Ltd. Two-gene vectors for generating car-t cells and uses thereof
CN109652379A (zh) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 Cd7嵌合抗原受体修饰的nk-92mi细胞及其应用
CN112300282A (zh) * 2020-11-03 2021-02-02 南京北恒生物科技有限公司 靶向cd7的人源化抗体及其用途

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
"Antibody-antigen interactions: Contact analysis and binding sitetopography", J. MOL. BIOL., vol. 262, pages 732 - 745
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING COMPANY
AL-LAZIKANI ET AL., JMB, vol. 273, 1997, pages 927 - 948
ALMAGROFRANSSON, FRONT. BIOSCI, vol. 13, 2008, pages 1619 - 1633
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
DATABASE PROTEIN 13 December 1995 (1995-12-13), ANONYMOUS : "sFv antibody, partial [Mus musculus]", XP055928515, retrieved from NCBI Database accession no. AAA83267 *
ELLISON ET AL., NUCLEIC ACIDS RES., vol. 10, 1982, pages 4071 - 9
HOLLINGER ET AL., PROC NATL ACAD. SCI. U.S.A., vol. 90, 1993, pages 6444 - 6448
HONEGGER APLUCKTHUN A: "Yet another numbering scheme for immunoglobulin variable domains: an automatic modeling and analysis tool", JMOL BIOL, vol. 309, no. 3, 8 June 2001 (2001-06-08), pages 657 - 70, XP004626893, DOI: 10.1006/jmbi.2001.4662
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
LEFRANC MP ET AL.: "IMGT unique numbering for immunoglobulin and T cell receptor variable domains and Ig superfamily V-like domains", DEV COMP IMMUNOL, vol. 27, no. 1, January 2003 (2003-01-01), pages 55 - 77, XP055585227, DOI: 10.1016/S0145-305X(02)00039-3
MACCALLUM ET AL., J. MOL. BIOL, vol. 262, 1996, pages 732 - 745
MARTIN ET AL.: "Modeling antibody hypervariable loops: a combined algorithm", PNAS, vol. 86, no. 23, 1989, pages 9268 - 9272, XP000165667, DOI: 10.1073/pnas.86.23.9268
See also references of EP4219554A4
SMITH ET AL., J. MOL. BIOL., vol. 147, 1981, pages 195 - 197

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2024040194A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering
WO2024040195A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering
CN115991776A (zh) * 2022-10-26 2023-04-21 上海驯鹿生物技术有限公司 一种靶向cd7的全人源抗体及其应用
CN116003631A (zh) * 2022-11-15 2023-04-25 江苏元晟生物科技有限公司 靶向cd7的人源化抗体及其应用
CN116003631B (zh) * 2022-11-15 2024-01-26 江苏元晟生物科技有限公司 靶向cd7的人源化抗体及其应用
WO2024119157A1 (en) 2022-12-02 2024-06-06 Sana Biotechnology, Inc. Lipid particles with cofusogens and methods of producing and using the same

Also Published As

Publication number Publication date
JP2023547425A (ja) 2023-11-10
EP4219554A1 (en) 2023-08-02
KR20230098158A (ko) 2023-07-03
US20230399398A1 (en) 2023-12-14
EP4219554A4 (en) 2024-05-22
CN112300282A (zh) 2021-02-02

Similar Documents

Publication Publication Date Title
WO2022095803A1 (zh) 靶向cd7的人源化抗体及其用途
WO2021147928A1 (zh) 包含嵌合抗原受体的免疫细胞及其用途
WO2022089353A1 (zh) 靶向bcma的单域抗体及其用途
WO2022152168A1 (zh) 靶向ror1的抗体及其用途
WO2021147929A1 (zh) 多链嵌合抗原受体及其用途
EP4219693A1 (en) Chimeric antigen receptor targeting cd7 and use thereof
WO2022222910A1 (zh) 靶向gprc5d的抗体及其用途
WO2022247795A1 (zh) 靶向Claudin18.2的纳米抗体及其用途
WO2022111405A1 (zh) 靶向Claudin18.2的抗体及其用途
WO2021249462A1 (zh) 表达nk抑制性分子的工程化免疫细胞及其用途
WO2022105826A1 (zh) 靶向nkg2a的抗体及其用途
JP2023545907A (ja) 新規共刺激ドメインを含むキメラ抗原受容体及びその使用
CN116063527A (zh) 靶向间皮素的抗体及其用途
WO2022121880A1 (zh) 靶向cd19的人源化抗体及其用途
WO2024001470A1 (zh) 靶向b7-h3的抗体及其用途
WO2023051414A1 (zh) 靶向间皮素的抗体及其用途
WO2022095801A1 (zh) 靶向lir1的抗体及其用途
US20240131158A1 (en) Cell therapy compositions and methods for modulating tgf-b signaling
JP2023516595A (ja) Cd20を結合する二量体抗原受容体(dar)
JP2018522564A (ja) 抗グリピカン3抗体およびその使用

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: 21888500

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2023524926

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2021888500

Country of ref document: EP

Effective date: 20230427

NENP Non-entry into the national phase

Ref country code: DE