WO2023051414A1 - Anticorps ciblant la mésothéline et son utilisation - Google Patents

Anticorps ciblant la mésothéline et son utilisation Download PDF

Info

Publication number
WO2023051414A1
WO2023051414A1 PCT/CN2022/121026 CN2022121026W WO2023051414A1 WO 2023051414 A1 WO2023051414 A1 WO 2023051414A1 CN 2022121026 W CN2022121026 W CN 2022121026W WO 2023051414 A1 WO2023051414 A1 WO 2023051414A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
cells
chain variable
variable region
Prior art date
Application number
PCT/CN2022/121026
Other languages
English (en)
Chinese (zh)
Inventor
李国坤
张静
周亚丽
陈功
郭婷婷
任江涛
Original Assignee
北恒医疗有限公司
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
Priority claimed from CN202210802270.9A external-priority patent/CN116063527A/zh
Application filed by 北恒医疗有限公司 filed Critical 北恒医疗有限公司
Publication of WO2023051414A1 publication Critical patent/WO2023051414A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/64General methods for preparing the vector, for introducing it into the cell or for selecting the vector-containing host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/53Liver

Definitions

  • the invention belongs to the field of immunotherapy. More specifically, the present invention relates to antibodies targeting mesothelin, and their use in the prevention and/or treatment and/or diagnosis of diseases.
  • MSLN Mesothelin, also known as MSLN, is a cell surface glycoprotein encoded by the MSLN gene, which is anchored on the cell surface through glycosylphosphatidylinositol.
  • mesothelin is a differentiation antigen present on normal mesothelial cells. It is rarely expressed in normal tissues, but it is highly expressed in tumors such as mesothelioma, lung cancer, pancreatic cancer, breast cancer, and ovarian cancer. Therefore, mesothelin may become an important target for cancer treatment.
  • MSLN-knockout mice exhibited normal development, reproduction, and blood cell counts. This shows that mesothelin is not necessary for the normal growth, development and reproduction of mice.
  • overexpression of MSLN can activate NF- ⁇ B, MAPK and PI3K pathways, and induce apoptosis or promote cell proliferation, migration and metastasis by inducing the activation and expression of MMP7 and MMP9.
  • Abnormal expression of MSLN plays an important role in tumor cell proliferation, adhesion and drug resistance.
  • the present invention develops a group of antibodies that specifically recognize MSLN, and constructs corresponding chimeric antigen receptor immune cells, providing more options for the treatment of various tumors.
  • the present invention provides an antibody targeting MSLN or an antigen-binding fragment thereof comprising:
  • H-CDR1 selected from SEQ ID NO: 1-4
  • H-CDR2 selected from SEQ ID NO: 5-9
  • (2) light chain variable region which comprises L-CDR1 selected from SEQ ID NO: 14-18, L-CDR2 selected from SEQ ID NO: 19-22 and L selected from SEQ ID NO: 23-27 -CDR3.
  • the antibody or antigen-binding fragment thereof targeting MSLN comprises:
  • Heavy chain variable region which comprises H-CDR1, H-CDR2, H-CDR3 shown in SEQ ID NO: 1, 5, 10 respectively, or shown in SEQ ID NO: 2, 6, 11 respectively H-CDR1, H-CDR2, H-CDR3 shown, or H-CDR1, H-CDR2, H-CDR3 respectively shown in SEQ ID NO: 3, 7, 12, or respectively shown in SEQ ID NO: 4, H-CDR1, H-CDR2, H-CDR3 shown in 8, 13, or H-CDR1, H-CDR2, H-CDR3 shown in SEQ ID NO: 1, 9, 10 respectively; and
  • (2) light chain variable region which comprises L-CDR1, L-CDR2, L-CDR3 respectively as shown in SEQ ID NO: 14, 19, 23, or respectively as shown in SEQ ID NO: 15, 20, 24 L-CDR1, L-CDR2, L-CDR3 shown, or L-CDR1, L-CDR2, L-CDR3 shown respectively as SEQ ID NO: 16, 21, 25, or respectively as shown in SEQ ID NO: 17, L-CDR1, L-CDR2, L-CDR3 shown in 22, 26, or L-CDR1, L-CDR2, L-CDR3 shown in SEQ ID NO: 18, 19, 27 respectively.
  • an antibody of the invention or an antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region selected from the group consisting of SEQ ID NO: 28, 30, 32, 34,
  • the amino acid sequences of 36, 73, 76, 79, 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 have at least 90%, 91%, 92% , 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or with selected from SEQ ID NO: 28, 30, 32, 34, 36, 73, 76, 79 , 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 have one or more amino acids (for example, at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids), preferably a conservative modification of up to 10 amino acids; the light chain variable
  • the modifications are conservative modifications, such as conservative substitutions, additions and deletions of amino acids.
  • the antibody or antigen-binding fragment thereof of the present invention comprises SEQ ID NO: 28, 30, 32, 34, 36, 73, 76, 79, 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 heavy chain variable regions and selected from SEQ ID NO: 29, 31, 33, 35, 37, 74, 77, 80, 83, The light chain variable regions of 86, 89, 92, 95, 98, 101, 104, 107, 110, 113, 116, 119, 122 and 125.
  • an antibody of the invention comprises a heavy chain variable region and a light chain variable region selected from:
  • the antibody or antigen-binding fragment thereof of the invention is selected from the group consisting of SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108,
  • the amino acid sequences of 111, 114, 117, 120, 123 and 126 are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identical , or with an amino acid selected from SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108, 111, 114, 117, 120, 123 and 126
  • the sequences have a modification of one or several amino acids (eg, up to 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids) compared to the sequence.
  • the modifications are conservative modifications, such as conservative substitutions, additions and deletions of amino acids.
  • the amino acid sequence of the antibody or antigen-binding fragment thereof of the present invention is selected from SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108, 111 , 114, 117, 120, 123 and 126.
  • the antibody or antigen-binding fragment thereof of the present invention is a murine antibody, a chimeric antibody, a humanized antibody or a human antibody, preferably a humanized antibody.
  • the invention also provides nucleic acid molecules encoding antibodies or antigen-binding fragments thereof. Therefore, in one embodiment, the nucleic acid molecule encoding said antibody or antigen-binding fragment thereof has at least 90%, 91%, 92%, 93%, 94% of the nucleotide sequence selected from SEQ ID NO: 43-47. %, 95%, 96%, 97%, 98%, 99%, 100% sequence identity, and the encoded antibody or antigen-binding fragment thereof can specifically bind the MSLN antigen.
  • the nucleic acid molecule encoding said antibody or antigen-binding fragment thereof is selected from SEQ ID NO: 43-47.
  • the antibody or antigen-binding fragment thereof of the present invention is a multispecific antibody (preferably a bispecific antibody or a trispecific antibody), which comprises an anti-MSLN antibody or an antigen-binding fragment thereof as described above, and one or A plurality of secondary antibodies or antigen-binding portions thereof that specifically bind to other tumor antigens.
  • the second antibody or antigen binding portion thereof may be in any antibody or antibody fragment form, such as full length antibody, Fab, Fab', F(ab') 2 , Fv, scFv, scFv-scFv, minibody , diabody or sdAb.
  • the present invention also provides a vector comprising a nucleic acid molecule encoding the above-mentioned anti-MSLN antibody or an antigen-binding fragment thereof, and a host cell expressing the anti-MSLN antibody or an antigen-binding fragment thereof.
  • the present invention also provides a recombinant receptor (such as recombinant TCR receptor, chimeric antigen receptor, T cell fusion protein or T cell antigen coupler), which comprises the anti-MSLN antibody or its antigen-binding fragment.
  • a recombinant receptor such as recombinant TCR receptor, chimeric antigen receptor, T cell fusion protein or T cell antigen coupler
  • the recombinant receptor is a chimeric antigen receptor further comprising a transmembrane domain and an intracellular domain comprising one or more co-stimulatory domains and/or primary signaling structures area.
  • the chimeric antigen receptor comprises an anti-MSLN antibody or antigen-binding fragment thereof as provided herein, a CD8 ⁇ or CD28 transmembrane domain, a CD28 and/or 4-1BB co-stimulatory domain, and a CD3 ⁇ primary signal conduction domain.
  • the present invention also provides a nucleic acid molecule encoding the MSLN-targeting recombinant 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 recombinant receptor targeting MSLN as defined above.
  • the engineered immune cells further comprise a second recombinant receptor targeting other tumor antigens, such as a second chimeric antigen receptor or a recombinant TCR receptor.
  • the present invention also provides an antibody conjugate comprising the anti-MSLN antibody or its antigen-binding fragment as defined in the present invention and a second functional structure, wherein the second functional structure is selected from Fc, Radioisotopes, half-life-extending moieties, detectable labels and drugs.
  • the half-life-prolonging structural moiety is selected from the group consisting of albumin binding structure, transferrin binding structure, polyethylene glycol molecule, recombinant polyethylene glycol molecule, human serum albumin, human serum albumin Fragments of proteins and albumin (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, which comprises the anti-MSLN antibody or antigen-binding fragment thereof, antibody conjugate, engineered immune cell or recombinant receptor described in the present invention.
  • the present invention also provides a pharmaceutical composition, which comprises the anti-MSLN antibody or antigen-binding fragment thereof, recombinant receptor, engineered immune cell or antibody conjugate of the present invention, and one or more a pharmaceutically acceptable excipient.
  • the present invention also provides a method for treating and/or preventing and/or diagnosing diseases associated with MSLN expression, comprising administering to a subject the anti-MSLN antibody or its antigen-binding fragment, chimeric Antigen receptors, antibody conjugates, engineered immune cells or pharmaceutical compositions.
  • antibody has the broadest meaning understood by those skilled in the art and includes monoclonal antibodies (including whole antibodies), polyclonal antibodies, multivalent antibodies, multispecific antibodies (such as 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 may be of any class (eg, IgG, IgE, IgM, IgD, IgA, etc.) or subclass (eg, IgG1, IgG2, IgG2a, IgG3, IgG4, IgA1, IgA2, etc.).
  • antibody fragment refers to one or more fragments of an antibody that retain the ability to specifically bind an antigen. It has been shown that the antigen-binding function of antibodies can be performed by fragments of full-length antibodies. Examples of antibody fragments in the present invention include, but are not limited to: Fab, Fab', F(ab') 2 , Fd, Fd', Fv, single chain antibody (scFv), disulfide-linked Fv (sdFv), Linear antibodies, "diabodies” with two antigen binding sites, natural ligands of said antigens or functional fragments thereof, etc.
  • an "antibody” of the present invention encompasses antibody fragments or antigen-binding fragments as defined above.
  • the antibody of the invention is selected from the group consisting of whole antibodies, Fab, Fab', F(ab') 2 , Fd, Fd', Fv, scFv, sdFv, linear antibodies and diabodies.
  • whole antibodies comprise two heavy chains and two light chains disulfide-bonded together, each light chain being disulfide-bonded to a respective heavy chain, in a "Y" configuration.
  • Each heavy chain consists of a heavy chain variable region (VH) and a heavy chain constant region, wherein the heavy chain variable region contains three complementarity determining regions (CDRs): H-CDR1, H-CDR2 and H-CDR3, and the heavy chain is constant
  • the region contains three constant domains: CH1, CH2 and CH3.
  • Each light chain consists of a light chain variable region (VL) and a light chain constant region, wherein the light chain variable region contains three CDRs: L-CDR1, L-CDR2 and L-CDR3, and the light chain constant region contains a constant structure Domain CL.
  • the CDRs are separated by more conserved framework regions (FRs).
  • the variable region of the heavy chain/light chain is responsible for the recognition and binding of the antigen, while the constant region can mediate the binding of the antibody to host tissues or factors, including various cells of the immune system (such as effector cells) and the first stage 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 alignments
  • the Chothia scheme is based on structural information.
  • Both the Kabat and Chothia schemes numbering are based on the sequence lengths of the most common antibody regions where insertions are provided by caret letters (eg "30a") and deletions occur in some antibodies. The two schemes place certain insertions and deletions (indels) at 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 scheme is a compromise between the Kabat and Chothia definitions and is based on the scheme used by Oxford Molecular's AbM antibody modeling software.
  • a "CDR" of a given antibody or region thereof is understood to encompass the CDRs defined by any of the above schemes or other known schemes.
  • a particular CDR eg, CDR3
  • FRs for a given antibody or region thereof are understood to encompass FRs as defined by any of the above schemes or other known schemes.
  • the numbering scheme used herein to define the boundaries of CDRs and FRs adopts the Kabat scheme.
  • Single-chain antibody and “scFv” are used interchangeably herein, and refer to an antibody formed by linking an antibody heavy chain variable region (VH) and a light chain variable region (VL) through a linker.
  • the optimal length and/or amino acid composition of the linker can be selected.
  • the length of the linker can significantly affect the variable domain folding and interaction of scFv. In fact, if shorter linkers (eg, between 5-10 amino acids) are used, intrachain folding can be prevented.
  • linker size and composition see, e.g., Hollinger et al., 1993 Proc Natl Acad. Sci. U.S.A. 90:6444-6448; U.S. Patent Application Publication Nos.
  • WO2006/020258 and WO2007/024715 are hereby incorporated by reference in their entirety.
  • Commonly used linkers such as GSTSGSGKPGSGEGSTKG (SEQ ID NO: 71), GGGGSGGGGSGGGGS (SEQ ID NO: 72).
  • a scFv may comprise VH and VL linked in any order, eg VH-linker-VL or VL-linker-VH.
  • the antibody or antigen-binding fragment thereof of the present invention is a murine antibody, a chimeric antibody, a humanized antibody or a human antibody, preferably a humanized antibody.
  • chimeric antibody refers to an antibody in which a portion of the amino acid sequence of each of the heavy and light chains is homologous to the corresponding sequence in an antibody from a particular species or belonging to a particular class, while the remaining segments of the chains are is homologous to a corresponding sequence in another species or belonging to another class.
  • the variable regions of both the light and heavy chains are derived from the variable regions of antibodies from one species, while the constant regions are homologous to antibody sequences from another species.
  • a distinct advantage of this chimeric format is that the variable regions can be conveniently produced from presently known sources using readily available B cells or hybridomas from non-human hosts, and the constant regions combined therewith from, for example, human cells.
  • variable region has the advantage of being easy to prepare, and the specificity is not affected by the source, while since the constant region is of human origin, the antibody will be more likely to elicit a human immune response when injected than if the constant region is of non-human origin Low.
  • 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.
  • “Humanized forms" of non-human antibodies refer to variants of such non-human antibodies that have been humanized to generally reduce immunogenicity in 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 (e.g., an antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • Humanized antibodies and methods for their preparation 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 the consensus sequences of human antibodies of a particular subgroup of light or heavy chain variable regions ; human mature (somatically mutated) framework regions or human germline framework regions; and framework regions obtained from screening FR libraries.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may comprise amino acid residues not encoded by human germline immunoglobulin sequences (eg, mutations introduced by random or site-directed mutagenesis in vitro or by somatic mutation in vivo).
  • the present invention provides an antibody targeting MSLN or an antigen-binding fragment thereof comprising:
  • H-CDR1 selected from SEQ ID NO: 1-4
  • H-CDR2 selected from SEQ ID NO: 5-9
  • (2) light chain variable region which comprises L-CDR1 selected from SEQ ID NO: 14-18, L-CDR2 selected from SEQ ID NO: 19-22 and L selected from SEQ ID NO: 23-27 -CDR3.
  • the antibody or antigen-binding fragment thereof targeting MSLN comprises:
  • Heavy chain variable region which comprises H-CDR1, H-CDR2, H-CDR3 shown in SEQ ID NO: 1, 5, 10 respectively, or shown in SEQ ID NO: 2, 6, 11 respectively H-CDR1, H-CDR2, H-CDR3 shown, or H-CDR1, H-CDR2, H-CDR3 respectively shown in SEQ ID NO: 3, 7, 12, or respectively shown in SEQ ID NO: 4, H-CDR1, H-CDR2, H-CDR3 shown in 8, 13, or H-CDR1, H-CDR2, H-CDR3 shown in SEQ ID NO: 1, 9, 10 respectively; and
  • (2) light chain variable region which comprises L-CDR1, L-CDR2, L-CDR3 respectively as shown in SEQ ID NO: 14, 19, 23, or respectively as shown in SEQ ID NO: 15, 20, 24 L-CDR1, L-CDR2, L-CDR3 shown, or L-CDR1, L-CDR2, L-CDR3 shown respectively as SEQ ID NO: 16, 21, 25, or respectively as shown in SEQ ID NO: 17, L-CDR1, L-CDR2, L-CDR3 shown in 22, 26, or L-CDR1, L-CDR2, L-CDR3 shown in SEQ ID NO: 18, 19, 27 respectively.
  • an antibody of the invention or an antigen-binding fragment thereof comprises a heavy chain variable region and a light chain variable region selected from the group consisting of SEQ ID NO: 28, 30, 32, 34,
  • the amino acid sequences of 36, 73, 76, 79, 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 have at least 90%, 91%, 92% , 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or with selected from SEQ ID NO: 28, 30, 32, 34, 36, 73, 76, 79 , 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 have one or more amino acids (for example, at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids), preferably a conservative modification of up to 10 amino acids; the light chain variable
  • the modifications are conservative modifications, such as conservative substitutions, additions and deletions of amino acids.
  • the antibody or antigen-binding fragment thereof of the present invention comprises SEQ ID NO: 28, 30, 32, 34, 36, 73, 76, 79, 82, 85, 88, 91, 94, 97, 100, 103, 106, 109, 112, 115, 118, 121 and 124 heavy chain variable regions and selected from SEQ ID NO: 29, 31, 33, 35, 37, 74, 77, 80, 83, The light chain variable regions of 86, 89, 92, 95, 98, 101, 104, 107, 110, 113, 116, 119, 122 and 125.
  • an antibody of the invention comprises a heavy chain variable region and a light chain variable region selected from:
  • the antibody or antigen-binding fragment thereof of the invention is selected from the group consisting of SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108,
  • the amino acid sequences of 111, 114, 117, 120, 123 and 126 are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identical , or with an amino acid selected from SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108, 111, 114, 117, 120, 123 and 126
  • the sequences have a modification of one or several amino acids (eg, up to 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids) compared to the sequence.
  • the modifications are conservative modifications, such as conservative substitutions, additions and deletions of amino acids.
  • the amino acid sequence of the antibody or antigen-binding fragment thereof of the present invention is selected from SEQ ID NO: 38-42, 75, 78, 81, 84, 87, 90, 93, 96, 99, 102, 105, 108, 111 , 114, 117, 120, 123 and 126.
  • the antibody or antigen-binding fragment thereof of the present invention is a murine antibody, a chimeric antibody, a humanized antibody or a human antibody, preferably a humanized antibody.
  • the invention also provides nucleic acid molecules encoding antibodies or antigen-binding fragments thereof. Therefore, in one embodiment, the nucleic acid molecule encoding said antibody or antigen-binding fragment thereof has at least 90%, 91%, 92%, 93%, 94% of the nucleotide sequence selected from SEQ ID NO: 43-47. %, 95%, 96%, 97%, 98%, 99%, 100% sequence identity, and the encoded antibody or antigen-binding fragment thereof can specifically bind the MSLN antigen.
  • the nucleic acid molecule encoding said antibody or antigen-binding fragment thereof is selected from SEQ ID NO: 43-47.
  • conservative modification refers to an amino acid modification that does not significantly affect or alter the binding characteristics of an antibody or antibody fragment comprising the amino acid sequence. These conservative modifications include conservative substitutions, additions and deletions of amino acids. Modifications can be introduced into chimeric antigen receptors of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. A conservative amino acid substitution is one in which an amino acid residue is replaced by an amino acid residue with a similar side chain.
  • Families of amino acid residues with similar side chains have been defined in the art and include basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid, ), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), non-polar side chains (e.g. alanine, valine acid, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g.
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid,
  • uncharged polar side chains e.g. glycine, asparagine, glutamine, serine, threonine, tyros
  • threonine valine, isoleucine
  • aromatic side chains eg, tyrosine, phenylalanine, tryptophan, histidine.
  • Conservative modifications can be selected, for example, on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • sequence identity means the degree to which two (nucleotide or amino acid) sequences in an alignment have the same residue at the same position, and is usually expressed as a percentage. Preferably, identity is determined over the entire length of the sequences being compared. Therefore, two copies of the exact same sequence have 100% identity.
  • 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 Clustal W.
  • the anti-MSLN antibody or antigen-binding fragment thereof of the present invention is multispecific (preferably a bispecific antibody or a trispecific antibody), which further comprises one or more antibodies specifically binding to other antigens. Secondary antibody.
  • multispecific means that the antigen binding protein has polyepitopic specificity (i.e., is capable of specifically binding two, three or more different epitopes on a biomolecule or is capable of specifically binding binds epitopes on two, three or more different biomolecules).
  • bispecific means that an antigen binding protein has two different antigen binding specificities.
  • the second antibody may be in any antibody or antibody fragment format, such as 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: CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25 , CD30, CD33, CD37, CD38, CD40, CD40L, CD44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2C, EGP40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa light chain, TIM3, TSHR, CD19, BAFF-R, CLL-1, EGFRvIII, GPRC5D, tEGFR, GD2, GD3, BCMA, Tn antigen, PSMA, ROR1, FLT3, FAP, TAG72, CD44v6, CEA,
  • nucleic acid nucleic acid, vector, host cell
  • the invention relates to nucleic acid molecules encoding the anti-MSLN antibodies of the invention.
  • a nucleic acid of the 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 anti-MSLN antibody has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% sequence identity, and the encoded anti-MSLN antibody is capable of specifically binding to MSLN (ie, has little binding to non-target antigens).
  • the nucleic acid molecule encoding the anti-MSLN antibody is shown in SEQ ID NO: 43-47.
  • a nucleic acid of the invention may also be in the form of, may be present in and/or may be part of a vector, such as a plasmid, cosmid or YAC.
  • the vector may especially be an expression vector, ie a vector providing for expression of the MSLN 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 and other elements useful or necessary for the expression of the MSLN antibodies of the invention include, but are not limited to, promoters, enhancers, terminators, integrators, selectable markers, leader sequences, reporter genes.
  • the present invention also provides host cells expressing the MSLN antibody, multispecific antibody of the present invention and/or containing the nucleic acid or vector of the present invention.
  • Preferred host cells of the invention are bacterial cells, fungal cells or mammalian cells.
  • Suitable bacterial cells include Gram-negative bacterial strains (such as Escherichia coli strains, Proteus strains, and Pseudomonas strains) and Gram-positive bacterial strains (such as Bacillus Bacillus strains, Streptomyces strains, Staphylococcus strains and Lactococcus strains).
  • Gram-negative bacterial strains such as Escherichia coli strains, Proteus strains, and Pseudomonas strains
  • Gram-positive bacterial strains such as Bacillus Bacillus strains, Streptomyces strains, Staphylococcus strains and Lactococcus strains.
  • Suitable fungal cells include cells of species of Trichoderma, Neurospora, and Aspergillus; or Saccharomyces (e.g., Saccharomyces cerevisiae), fission Schizosaccharomyces (such as Schizosaccharomyces pombe), Pichia (such as Pichia pastoris and Pichia methanolica) and Hansen A cell of a species of Saccharomyces (Hansenula).
  • Saccharomyces e.g., Saccharomyces cerevisiae
  • fission Schizosaccharomyces such as Schizosaccharomyces pombe
  • Pichia such as Pichia pastoris and Pichia methanolica
  • Hansen A cell of a species of Saccharomyces Hansenula
  • 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 cells known in the art for expressing heterologous proteins can also be used in the present invention.
  • the present invention also provides recombinant receptors, such as recombinant TCR receptors, chimeric antigen receptors, T cell fusion proteins or T cell antigen couplers, comprising the anti-MSLN antibodies described above.
  • recombinant receptors such as recombinant TCR receptors, chimeric antigen receptors, T cell fusion proteins or T cell antigen couplers, comprising the anti-MSLN antibodies described above.
  • the present invention also provides a chimeric antigen receptor comprising an anti-MSLN antibody as described above.
  • chimeric antigen receptor refers to an artificially constructed hybrid polypeptide that generally includes a ligand-binding domain (such as an antigen-binding portion of an antibody), a transmembrane domain, Optional co-stimulatory domain and intracellular signaling domain, each domain is connected by a linker.
  • CARs are able to exploit the antigen-binding properties of antibodies to redirect the specificity and reactivity of T cells and other immune cells to a target of choice in a non-MHC-restricted manner.
  • the present invention provides a chimeric antigen receptor comprising an anti-MSLN antibody or an antigen-binding fragment thereof as described above or comprising said anti-MSLN antibody, a transmembrane domain and an intracellular domain, wherein The intracellular domains comprise one or more co-stimulatory domains and/or primary signaling domains.
  • transmembrane domain refers to a polypeptide capable of expressing a chimeric antigen receptor on the surface of an immune cell (such as a lymphocyte, NK cell or NKT cell) and directing a cellular response of the immune cell against a target cell structure.
  • Transmembrane domains 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 suitable for use 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 comprise predominantly hydrophobic residues such as leucine and valine.
  • the transmembrane domain is derived from the CD8 ⁇ chain or CD28, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% of the amino acid sequence shown in SEQ ID NO: 49 or 51. % 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: 50 or 52 Or 99% or 100% sequence identity.
  • the costimulatory domain may be an intracellular functional signaling domain from a costimulatory molecule comprising the entire intracellular portion of said 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 co-stimulatory domains of the invention include, but are not limited to, intracellular regions derived from the following proteins: TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, MSLN, CD8, CD18, CD27, CD28, CD30, CD40, CD54, 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.
  • the costimulatory domain of the CAR of the present invention is from 4-1BB, CD28 or 4-1BB+CD28.
  • the 4-1BB co-stimulatory 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 shown in SEQ ID NO:55.
  • % 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:56 identity.
  • the CD28 co-stimulatory 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 shown in SEQ ID NO:53.
  • 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 with the nucleic acid molecule shown in SEQ ID NO:54 .
  • the term "primary signaling domain” refers to the portion of a protein that transduces effector function signals and directs the cell to perform a given function.
  • the intracellular signaling domains comprised by the chimeric antigen receptors of the present invention may be intracellular domain sequences of T cell receptors and co-receptors, which act together to elicit Signal transduction, and any derivatives or variants of these sequences and any synthetic sequences having the same or similar function.
  • the intracellular signaling domain can contain many immunoreceptor tyrosine-based activation motifs (Immunoreceptor Tyrosine-based Activation Motifs, ITAM).
  • Non-limiting examples of intracellular signaling domains of the present invention include, but are not limited to, intracellular regions of FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD22, MSLN9a, MSLN9b, and CD66d, among others.
  • the signaling domain of the CAR of the present invention may comprise a CD3 ⁇ intracellular region, which has at least 70%, preferably at least 80%, and more preferably at least 90% of the amino acid sequence shown in SEQ ID NO: 57 or 59.
  • %, 95%, 97% or 99% or 100% sequence identity or its coding sequence has at least 70%, preferably at least 80%, more preferably at least 90% of the nucleic acid molecule shown in SEQ ID NO: 58 or 60 , 95%, 97% or 99% or 100% sequence identity.
  • the chimeric antigen receptors of the invention may further comprise a hinge region located 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 is used 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 may be derived in whole or in part from a natural 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 an entirely 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 identical to that shown in SEQ ID NO: 65, 67 or 69
  • 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 and SEQ ID NO: 66, 68 or 70
  • the indicated nucleotide sequences have at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • the CAR of the invention may also 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 stretch of hydrophobic amino acids with a propensity to form a single ⁇ -helix.
  • At the end of the signal peptide there is usually a stretch of amino acids that is recognized and cleaved by the signal peptidase.
  • the signal peptidase can cleave during translocation or after completion to generate a free signal peptide and mature protein. Then, the free signal peptide is digested by specific proteases.
  • Signal peptides that can be used in the present invention are well known to those skilled in the art, such as signal peptides derived from B2M, CD8 ⁇ , IgG1, GM-CSFR ⁇ , and the like.
  • the signal peptide that can be used in the present invention is from B2M or CD8 ⁇ , which has at least 70%, preferably at least 80%, more preferably at least 90%, 95% of the amino acid sequence shown in SEQ ID NO: 61 or 63 , 97% or 99% or 100% sequence identity, or its coding sequence and the nucleic acid molecule shown in SEQ ID NO: 62 or 64 have at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • the CAR comprises an anti-MSLN antibody or antigen-binding fragment thereof as provided herein, a CD8 ⁇ or CD28 transmembrane region, a CD28 and/or 4-1BB co-stimulatory domain, and a CD3 ⁇ intracellular signaling structure area.
  • 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 the MSLN-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 where it 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 sequence at a specific targeting site.
  • An "expression vector” is a vector used for the transcription of heterologous nucleic acid sequences, such as those encoding chimeric antigen receptor polypeptides of the invention, and the translation of their mRNA in a suitable host cell.
  • vectors of the present invention include, but are not limited to, plasmids, viruses (such as retroviruses, lentiviruses, adenoviruses, vaccinia virus, Rous sarcoma virus (RSV, polyoma virus, and adeno-associated virus (AAV), etc. ), 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 acts as the "backbone" of the vector.
  • L vector also usually 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 (such as a multiple cloning site, MCS).
  • the vector may additionally contain a promoter, a multiple PolyA tail (polyA), 3'UTR, enhancer, terminator, insulator, operator, selectable marker, reporter gene, targeting sequence and/or protein purification tag and other elements.
  • the vector is an in vitro transcribed vector.
  • the present invention also provides engineered immune cells expressing the recombinant receptors (eg, chimeric antigen receptors) of the present invention.
  • 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).
  • 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, among others.
  • the immune cells are T cells.
  • the T cells may be any T cells, such as T cells cultured in vitro, such as 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 a site of infection, ascites, pleural effusion, spleen tissue, and tumors. T cells can also be enriched or purified.
  • T cells can be at any developmental stage, including, but not limited to, CD4+CD8+ T cells, CD4+ helper T cells (such as Th1 and Th2 cells), CD8+ T cells (such as cytotoxic T cells), CD4-CD8-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.
  • the engineered immune cells further comprise suppressed or silenced expression of at least one gene selected from: CD52, GR, dCK, TCR/CD3 gene (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, CASP7, F
  • the engineered immune cells further comprise suppressed or silenced expression of at least one gene selected from: 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 chemical agents
  • protease inhibitors to inhibit gene expression Express.
  • genes can also be silenced by mediated DNA fragmentation, for example, by meganucleases, zinc finger nucleases, TALE nucleases, or Cas enzymes in CRISPR systems.
  • the engineered immune cells further comprise a second recombinant receptor targeting other tumor antigens, such as a recombinant TCR receptor or a chimeric antigen receptor.
  • the other tumor antigen targeted by the second recombinant receptor may be selected from, for example, CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33 , CD37, CD38, CD40, CD40L, CD44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2C, EGP40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa light chain, TIM3, TSHR, CD19, GPRC5D, BAFF-R, C
  • a plurality of immune cells is 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 MSLN (e.g., a CAR comprising an anti-MSLN antibody described herein), and another cell is engineered to 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 MSLN.
  • such a multispecific chimeric antigen receptor may comprise a multispecific antibody targeting MSLN, or simultaneously comprise the anti-MSLN antibody of the present invention and antibodies targeting other antigens.
  • the plurality of engineered immune cells may be administered together or separately.
  • the plurality of immune cells can 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 an anti-MSLN antibody as defined in the present invention and a second functional structure, wherein the second functional structure is selected from the group consisting of Fc, radioactive isotopes, and half-life-extending structures moieties, detectable markers and drugs.
  • the present invention provides an antibody conjugate comprising an anti-MSLN antibody as defined in the present invention and 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 monomeric or multimeric, produced by digestion of an intact antibody.
  • the source of immunoglobulin from which native Fc is produced is preferably of human origin.
  • 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.
  • Natural Fc molecules have 1-4 intermolecular disulfides between monomeric subunits depending on class (e.g. IgG, IgA, IgE, IgD, IgM) or subtype (e.g. IgG1, IgG2, IgG3, IgA1, IgGA2) key.
  • An example of a native Fc is a 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.
  • Fc refers to an amino acid sequence that differs from that of a “native” or “wild-type” Fc by virtue of 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 the Fc of a human immunoglobulin, more preferably the Fc of a human IgG1.
  • the present invention provides an antibody conjugate comprising an anti-MSLN antibody as defined herein 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 an anti-MSLN antibody as defined in the present invention and a half-life-extending structural moiety selected from albumin binding structures, transferrin The binding structure of , polyethylene glycol molecules, recombinant polyethylene glycol molecules, human serum albumin, fragments of human serum albumin and albumin-binding albumin (including antibodies).
  • the present invention provides an antibody conjugate comprising an anti-MSLN antibody as defined herein and a detectable label.
  • detectable label means herein a compound that produces a detectable signal.
  • the detectable marker may be an MRI contrast agent, a scintigraphy 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 paprika 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 paprika root peroxidase, glucose-6-phosphatase, ⁇ -galactosidase
  • antibiotics such as kanamycin, ampicillin,
  • the present invention provides an antibody conjugate comprising an anti-MSLN antibody as defined herein and a drug conjugated to said anti-MSLN antibody, such as a cytotoxin or an immunomodulator (i.e., an antibody drug conjugates).
  • a drug conjugated to said anti-MSLN antibody such as a cytotoxin or an immunomodulator (i.e., an antibody drug conjugates).
  • the drug is covalently linked to the antibody, and usually relies on a linker.
  • the drug is a cytotoxin.
  • the drug is an immunomodulator.
  • cytotoxics include, but are not limited to, methotrexate, aminopterin, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, dacarbazine, nitrogen mustard, thiotepa, phentermine Nitrogen mustard, melphalan, carmustine (BSNU), lomustine (CCNU), 1-methylnitrosourea, cyclophosphamide, nitrogen mustard, busulfan, dibromomannitol, chain Zuocin, mitomycin, cis-dichlorodiamine platinum (II) (DDP), cisplatin, carboplatin, zorubicin, doxorubicin, detorubicin, caraminomycin, i Darubicin, epirubicin, mitoxantrone, actinomycin D, bleomycin, calicheamicin, mithromycin, antramycin (AMC), vincristine, vinblastine, Paclitaxe
  • immunomodulators include, but are not limited to, ganciclovir, etanercept, tacrolimus, sirolimus, vorcyclosporine, 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), interferon (such as interferon- ⁇ , interferon interferon-beta and interferon-gamma), stem cell growth factors designated "S1 factor", erythropoietin and thrombopoietin, or combinations thereof.
  • the present invention also provides a detection kit comprising the antibody, antibody conjugate or chimeric antigen receptor described in the present invention.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody of the present invention, recombinant receptors such as chimeric antigen receptors, engineered immune cells or antibody conjugates, and one or more Pharmaceutically acceptable excipients.
  • the term "pharmaceutically acceptable excipient” means pharmacologically and/or physiologically compatible with the subject and the active ingredient (i.e., capable of eliciting the desired therapeutic effect without causing any adverse desired local or systemic effect), which are well known in the art (see, for example, 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, coating agents, adsorbents, anti-adhesive agents, glidants, antioxidants, flavoring agents, coloring agents, Sweeteners, solvents, co-solvents, buffers, chelating agents, surfactants, diluents, wetting agents, preservatives, emulsifiers, coating agents, isotonic agents, absorption delaying agents, stabilizers and tonicity regulators .
  • suitable excipients is known to those skilled in the art for the preparation of the desired pharmaceutical compositions of the present invention.
  • excipients for use in pharmaceutical compositions of the invention include saline, buffered saline, dextrose and water.
  • suitable excipients depends inter alia 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 are suitable for various routes of administration. Typically, administration is accomplished parenterally.
  • Parenteral delivery methods include topical, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.
  • the pharmaceutical composition according to the present invention can also be prepared in various forms, such as solid, liquid, gaseous or lyophilized forms, especially 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 especially adapted to the desired method of administration.
  • Processes known in the present invention for the production of medicaments 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.
  • the pharmaceutical composition according to the invention can also be administered in combination with one or more other agents suitable for the treatment and/or prophylaxis 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, trimetreate 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
  • the present invention also provides a method for treating and/or preventing and/or diagnosing diseases associated with MSLN expression, comprising administering to a subject the above-mentioned antibody, chimeric antigen receptor, antibody-conjugated Drugs, engineered immune cells or pharmaceutical compositions.
  • diseases associated with MSLN expression include, but are not limited to, MSLN-positive adenocarcinoma, mesothelioma, lung cancer, colon cancer, colorectal cancer, breast cancer, ovarian cancer, cervical cancer, gastric cancer, bile duct cancer, gallbladder cancer , esophageal cancer, melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, rectal cancer, Hodgkin's lymphoma, pancreatic cancer, or prostate cancer.
  • Figure 1 Shows the specific binding of five MSLN murine antibodies to CHO-MSLN cells.
  • FIG. 1 Shows CAR expression levels of murine CAR T cells targeting MSLN.
  • Figure 3 Shows the killing effect of murine CAR T cells targeting MSLN on target cell Huh7-Meso and non-target cell A549-CBG at various effect-to-target ratios.
  • Figure 4 Shows the degranulation of murine CAR T cells targeting MSLN after co-culture with target cells A549-CBG-Meso, Hela-luci, Huh7-Meso-luci and non-target cells A549-CBG.
  • Figure 5 Shows the IL2 release level after co-culture of murine CAR T cells targeting MSLN with target cells A549-CBG-Meso, Hela-luci, Huh7-Meso-luci and non-target cells A549-CBG.
  • Figure 6 Shows the IFN ⁇ release levels after co-culture of murine CAR T cells targeting MSLN with target cells A549-CBG-Meso, Hela-luci, Huh7-Meso-luci and non-target cells A549-CBG.
  • Figure 7 Shows the tumor suppressive effect of murine CAR T cells targeting MSLN in mice.
  • Figure 8 Shows CAR expression levels of humanized CAR T cells targeting MSLN.
  • Figure 9 Shows the killing effect of humanized CAR T cells targeting MSLN on target cells Hela cells, Huh7-Meso cells and non-target cells Huh7 cells at various effect-to-target ratios.
  • Figure 10 Degranulation after co-culture of humanized CAR T cells targeting MSLN with target cells Hela cells, A549-CBG-Meso cells, Huh7-Meso cells and non-target cells A549-CBG cells and Huh7 cells effect.
  • Figure 11 shows the release of IL2 after co-culture of humanized CAR T cells targeting MSLN with target cells Hela cells, A549-CBG-Meso cells, Huh7-Meso cells and non-target cells A549-CBG cells and Huh7 cells level.
  • Figure 12 IFN ⁇ release after co-culture of humanized CAR T cells targeting MSLN with target cells Hela cells, A549-CBG-Meso cells, Huh7-Meso cells and non-target cells A549-CBG cells and Huh7 cells level.
  • hMSLN-Fc (Acrobiosystems, Cat. No. MSN-H526x, wherein the amino acid sequence of hMSLN is shown in SEQ ID NO: 48) protein
  • mouse spleen lymphocytes were taken, mixed with SP2/0 myeloma cells, and PEG was added to mediate cell fusion to prepare hybridoma cells.
  • MSLN overexpression cell line (CHO-MSLN cells) was screened for hybridoma clones binding to MSLN by ELISA or flow cytometry.
  • 5 antibody clones that can specifically bind to MSLN were obtained.
  • the 5 clones were sequenced to obtain their amino acid sequence and nucleic acid sequence.
  • the heavy chain variable region VH and light chain variable region VL of each clone were connected through a linker to obtain 5 single-chain antibodies, which were named 001, 002, 003, 004 and 005, and their sequences are shown in Table 1 below.
  • the sequences encoding the following proteins were synthesized and cloned into the pLVX vector (Public Protein/Plasmid Library (PPL), catalog number: PPL00157-4a): CD8 ⁇ signal peptide (SEQ ID No: 63), anti-MSLN single-chain antibody (SEQ ID One of No:38-42), CD8 ⁇ hinge region (SEQ ID No:65), CD8 ⁇ transmembrane region (SEQ ID No:49), 4-1BB intracellular region (SEQ ID No:55) and CD3 ⁇ intracellular region (SEQ ID No:57), and the correct insertion of the target sequence was confirmed by sequencing.
  • PPL Public Protein/Plasmid Library
  • Opti-MEM 3ml Opti-MEM (Gibco, Cat. 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 X-treme GENE HP DNA Transfection Reagent (Roche, Cat. No. 06366236001), mix immediately, incubate at room temperature for 15min, and then add the plasmid/vector/transfection reagent mixture dropwise into the culture flask of 293T cells . Viruses were collected at 24 hours and 48 hours, combined, and ultracentrifuged (25000g, 4°C, 2.5 hours) to obtain concentrated lentiviruses.
  • T cells were activated with DynaBeads CD3/CD28CTSTM (Gibco, Cat. No. 40203D) and cultured at 37°C and 5% CO 2 for 1 day. Then, the concentrated lentivirus was added, and after continuous culture for 3 days, five kinds of CAR-T cells targeting MSLN were obtained, named as BH28-001, BH28-002, BH28-003, BH28-004 and BH28-005CAR-T cells . Unmodified wild-type T cells (NT) were used as controls.
  • the anti-MSLN single-chain antibody in the CAR-T cells prepared by the present invention can be effectively expressed.
  • the CAR T cells of the present invention showed a strong killing effect on the target cell Huh7-Meso, while the killing effect on the non-target cell A549-CBG was weak, indicating that each CAR- T cells can only specifically kill cells expressing MSLN.
  • each CAR-T cell prepared by the present invention showed significantly higher specific degranulation to three target cells, A549-CBG-Meso cells, Hela cells, and Huh7-Meso cells. effect.
  • A549-CBG-Meso cells Hela cells, Huh7-Meso cells
  • non-target cells A549-CBG cells
  • Proportions were added to BH395-CAR T cells, BH398-CAR T cells or NT cells (negative control), and the cell co-culture supernatant was collected after co-cultivation for 18-24 hours.
  • mice Eighteen 7-week-old healthy female NPI mice were divided into 3 groups, 6 mice in each group: NT group (negative control), BH28-003 group, and BH28-004 group.
  • NT group negative control
  • BH28-003 group negative control
  • BH28-004 group On day 0 (D0), 6 ⁇ 10 6 Hela cells were injected into the tail vein of each mouse.
  • D6 6 days later
  • 2x106 NT cells or corresponding CAR-T cells were injected into the tail vein of each mouse according to the grouping situation. The changes in the tumor burden of the mice were assessed weekly, and the results are shown in FIG. 7 .
  • the tumor burden in the mice progressed rapidly throughout the experimental period.
  • the tumor growth of tumor-bearing mice was significantly inhibited.
  • the tumor burden decreased rapidly within a few days after treatment with CAR-T cells until it disappeared, and maintained a very low level of tumor without recurrence throughout the experimental period. This shows that the CAR-T cells of the present invention can effectively kill tumor target cells, showing a significant effect on tumor treatment.
  • the murine antibodies BH28-003 and BH28-004 were humanized, and the specific method was as follows: firstly, searched for the highly similar ones through the IG BLAST database (https://www.ncbi.nlm.nih.gov/igblast/) Humanized antibody sequence, and then replace the FR region in the single-chain antibody with the corresponding human sequence; then replace individual amino acid residues according to the different physical and chemical properties of the amino acid residues, and finally obtain multiple humanized single-chain antibodies.
  • the amino acid sequence is shown in Table 2.
  • CAR-T cells were prepared with humanized antibodies, and CAR-T cells containing anti-MSLN humanized antibodies BH28-3V0, BH28-3V1, BH28-3V2, BH28-3V3, BH28-3V4, BH28- 3V5, BH28-3V6, BH28-3V7, BH28-3V8, BH28-4V0, BH28-4V1, BH28-4V2, BH28-4V3, BH28-4V4, BH28-4V5, BH28-4V6, BH28-4V7, BH28-4V8.
  • Murine CAR-T cells BH28-003, BH28-004, and unmodified wild-type T cells (NT) were used as controls.
  • each target cell Hela cell, Huh7-Meso cell
  • non-target cell Huh7 cell
  • luciferase gene expressing luciferase gene into a 96-well plate at a concentration of 1 ⁇ 10 4 cells/well, and then mix 16:1, 8 :1, 4:1, 2:1 effect-to-target ratio (i.e. the ratio of effector T cells to target cells)
  • NT cells and each CAR T cell were plated in a 96-well plate for co-culture, and after 16-18 hours, the enzyme Fluorescence was measured with a standard instrument. According to the calculation formula: (average fluorescence value of target cells ⁇ average fluorescence value of samples)/average fluorescence value of target cells ⁇ 100%, the killing efficiency was calculated, and the results are shown in FIG. 9 .
  • each CAR T cell of the present invention shows a strong killing effect on target cells Hela cells and Huh7-Meso cells, while killing non-target cells huh7 cells is relatively weak, indicating that Each CAR T cell can only specifically kill cells expressing MSLN.
  • each of the humanized CAR T cells prepared by the present invention showed significantly increased specificity for target cells (A549-CBG-Meso cells, Hela cells, Huh7-Meso cells). No significant increase in degranulation was observed for non-target cells (A549-CBG cells, Huh7 cells).
  • the anti-MSLN murine antibody and the humanized antibody of the present invention can specifically bind to the MSLN protein, and the CAR T cells prepared by using the above-mentioned anti-MSLN murine antibody and the humanized antibody can effectively target Specific killing of tumor target cells.

Abstract

L'invention concerne un anticorps ciblant MSLN, et un anticorps multi-spécifique, un récepteur d'antigène chimère, un conjugué d'anticorps, une composition pharmaceutique et un kit le comprenant, et une utilisation de ceux-ci dans le diagnostic/le traitement/la prévention de maladies associées à l'expression de MSLN.
PCT/CN2022/121026 2021-09-30 2022-09-23 Anticorps ciblant la mésothéline et son utilisation WO2023051414A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202111157860 2021-09-30
CN202111157860.2 2021-09-30
CN202210802270.9 2022-07-07
CN202210802270.9A CN116063527A (zh) 2021-09-30 2022-07-07 靶向间皮素的抗体及其用途

Publications (1)

Publication Number Publication Date
WO2023051414A1 true WO2023051414A1 (fr) 2023-04-06

Family

ID=85781300

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/121026 WO2023051414A1 (fr) 2021-09-30 2022-09-23 Anticorps ciblant la mésothéline et son utilisation

Country Status (1)

Country Link
WO (1) WO2023051414A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150274836A1 (en) * 2012-09-27 2015-10-01 The United States Of America,As Represented By The Secretary,Department Of Health And Human Services Mesothelin antibodies and methods for eliciting potent antitumor activity
CN107840891A (zh) * 2016-09-19 2018-03-27 上海吉倍生物技术有限公司 高亲和力的抗msln抗体及其应用
CN112028996A (zh) * 2020-10-30 2020-12-04 南京北恒生物科技有限公司 靶向bcma的单域抗体及其用途
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

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150274836A1 (en) * 2012-09-27 2015-10-01 The United States Of America,As Represented By The Secretary,Department Of Health And Human Services Mesothelin antibodies and methods for eliciting potent antitumor activity
CN107840891A (zh) * 2016-09-19 2018-03-27 上海吉倍生物技术有限公司 高亲和力的抗msln抗体及其应用
CN112028996A (zh) * 2020-10-30 2020-12-04 南京北恒生物科技有限公司 靶向bcma的单域抗体及其用途
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 (2)

* Cited by examiner, † Cited by third party
Title
KLAMPATSA ASTERO, DIMOU VIVIAN, ALBELDA STEVEN M.: "Mesothelin-targeted CAR-T cell therapy for solid tumors", EXPERT OPINION ON BIOLOGICAL THERAPY,, vol. 21, no. 4, 3 April 2021 (2021-04-03), pages 473 - 486, XP093054525, ISSN: 1471-2598, DOI: 10.1080/14712598.2021.1843628 *
LI, JUN ET AL.: "Research Progress of Mesothelin in Diagnosis and Treatment of Malignant Tumors", ONCOLOGY PROGRESS, vol. 19, no. 9, 10 May 2021 (2021-05-10), XP009545192 *

Similar Documents

Publication Publication Date Title
WO2022095803A1 (fr) Anticorps humanisé ciblant cd7 et son utilisation
US20220233593A1 (en) Combinations of engineered natural killer cells and engineered t cells for immunotherapy
JP2022116230A (ja) 免疫療法用改変細胞
WO2022089353A1 (fr) Anticorps à domaine unique ciblant bcma et son utilisation
WO2022152168A1 (fr) Anticorps ciblant ror1 et son utilisation
WO2018108106A1 (fr) Anticorps humanisé anti-cd19 et cellule effectrice immunitaire ciblant cd19
WO2022222910A1 (fr) Anticorps ciblant gprc5d et son utilisation
WO2022247795A1 (fr) Nanocorps ciblant la claudine 18.2 et son utilisation
WO2022111405A1 (fr) Anticorps ciblant la claudine 18.2 et utilisation associée
WO2022105826A1 (fr) Anticorps ciblant nkg2a et son utilisation
WO2023051414A1 (fr) Anticorps ciblant la mésothéline et son utilisation
KR20230146032A (ko) Tgf-b 신호전달을 조절하기 위한 세포 치료 조성물 및 방법
WO2024001470A1 (fr) Anticorps ciblant b7-h3 et son utilisation
CN115873115A (zh) 一种抗4-1bb单克隆抗体在肿瘤免疫治疗上的应用
WO2022121880A1 (fr) Anticorps humanisé ciblant cd19 et son utilisation
CN116063527A (zh) 靶向间皮素的抗体及其用途
WO2022095801A1 (fr) Anticorps ciblant lir1 et son utilisation
JP2023516595A (ja) Cd20を結合する二量体抗原受容体(dar)
JP2024519335A (ja) がん免疫療法のための投薬レジメン
JP2024501546A (ja) Cd22に特異的なヒト化抗体およびそれを用いたキメラ抗原受容体
TW202334215A (zh) 靶向cldn18.2之抗體、雙特異性抗體及其應用
CN117586410A (zh) 一种结合TNFR2和Claudin18.2的双特异性抗体

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: 22874800

Country of ref document: EP

Kind code of ref document: A1