WO2023045934A1 - Utilisation de t-car bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes - Google Patents

Utilisation de t-car bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes Download PDF

Info

Publication number
WO2023045934A1
WO2023045934A1 PCT/CN2022/119980 CN2022119980W WO2023045934A1 WO 2023045934 A1 WO2023045934 A1 WO 2023045934A1 CN 2022119980 W CN2022119980 W CN 2022119980W WO 2023045934 A1 WO2023045934 A1 WO 2023045934A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
acid sequence
use according
car
Prior art date
Application number
PCT/CN2022/119980
Other languages
English (en)
Chinese (zh)
Inventor
汪文
胡广
蔡松柏
李大可
操昌普
谢白露
吴明
徐夏君
姚正
郭静静
庞昊
Original Assignee
南京驯鹿医疗技术有限公司
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 南京驯鹿医疗技术有限公司 filed Critical 南京驯鹿医疗技术有限公司
Publication of WO2023045934A1 publication Critical patent/WO2023045934A1/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
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • 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
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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

Definitions

  • This application relates to the field of biomedicine, in particular to the application of CAR-T cells targeting BCMA in the preparation of drugs for the treatment of autoimmune diseases.
  • Antibody-mediated idiopathic inflammatory disease of the nervous system is an autoimmune disease in which autoimmune cells and molecules attack the nervous system as the main pathogenic mechanism.
  • pathogens that act on the nervous system autoantigens are collectively called nervous system autoantibodies, and antibody-mediated nervous system idiopathic inflammatory diseases can occur in the central nervous system, peripheral nervous system, and nerve-muscle junctions.
  • NMO Neuromyelitis optica
  • NMO sectrum disorder NMO sectrum disorder
  • AQP4-Ab is the most important pathogenic antibody of NMOSD. A large number of basic research and clinical research It has been confirmed that the antibody can cause damage to the central nervous system of animals and humans, and its diagnostic specificity can be as high as more than 90%. The positive rate of AQP4-Ab in NMOSD patients is between 40% and 90%.
  • BCMA B cell maturation antigen, B cell maturation antigen
  • B cell maturation antigen is a cell surface receptor mainly expressed in long-lived plasma cells (effector B cells), germinal center B cells and human memory B cells. BCMA detaches from the cell surface to form free BCMA (sBCMA) in peripheral blood. Studies have shown that the level of sBCMA is correlated with the level of IgG. So far, the drugs marketed in China include hormones, immunosuppressants, etc., and patients frequently relapse after using them. There is no effective therapy for the treatment of neuromyelitis optica. Drugs used for off-label diseases are not effective. Relapse and refractory treatment are still a difficult problem for the majority of medical workers. Therefore, it is necessary and urgent to find new treatments.
  • the application provides the use of immune effector cells in the preparation of drugs for the prevention and/or treatment of autoimmune diseases
  • the immune effector cells include a chimeric antigen receptor (CAR) targeting BCMA, and the immune effector cells can effectively use
  • autoimmune diseases such as neuromyelitis optica spectrum diseases.
  • the application provides the use of immune effector cells in the preparation of drugs for the prevention and/or treatment of autoimmune diseases, the immune effector cells comprising a chimeric antigen receptor (CAR), wherein the CAR comprises a BCMA-targeted Antigen binding domain, transmembrane domain, co-stimulatory domain and intracellular signaling domain, wherein the antigen binding domain is a fully human antigen binding domain.
  • CAR chimeric antigen receptor
  • the autoimmune disease comprises an autoimmune inflammatory disease.
  • the inflammatory disease comprises neuritis.
  • the inflammatory disease comprises a demyelinating disease.
  • the inflammatory disease comprises an inflammatory disease of the central nervous system.
  • the inflammatory disease comprises an inflammatory demyelinating disease of the central nervous system.
  • the inflammatory disease comprises optic neuritis.
  • the inflammatory disease comprises myelitis.
  • the inflammatory disease comprises Neuromyelitis Optic Spectrum Disease (NMOSD).
  • NOSD Neuromyelitis Optic Spectrum Disease
  • the inflammatory disease comprises AQP4 (aquaporin 4)-Ab positive neuromyelitis optica spectrum disorder.
  • the autoimmune disease comprises an AQP4-Ab-mediated autoimmune disease.
  • the autoimmune disease comprises an AQP4-Ab positive disease and/or condition.
  • the AQP4-Ab positive disease and/or condition comprises central nervous system demyelinating disease.
  • the AQP4-Ab positive disease and/or condition comprises an AQP4-Ab positive neuromyelitis optica spectrum disorder.
  • the autoimmune disease comprises Neuromyelitis Optic Spectrum Disease (NMOSD).
  • NOSD Neuromyelitis Optic Spectrum Disease
  • the neuromyelitis optica spectrum disorder is relapsed refractory NMOSD.
  • the antigen binding domain comprises HCDR1, HCDR2 and HCDR3, the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:9, and the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:10, Said HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 11.
  • the antigen binding domain comprises LCDR1, LCDR2 and LCDR3, wherein the LCDR1 comprises the amino acid sequence shown in SEQ ID NO: 17, and the LCDR2 comprises the amino acid sequence shown in SEQ ID NO: 18 (AAS)
  • the amino acid sequence of the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:19.
  • the antigen binding domain comprises a heavy chain variable region comprising SEQ ID NO: 1
  • the antigen binding domain comprises a light chain variable region comprising SEQ ID NO: 1
  • the antigen binding domain comprises the amino acid sequence shown in SEQ ID NO:43.
  • the antigen binding domain comprises an antibody or fragment thereof.
  • the antigen binding domain comprises a scFv.
  • the antigen binding domain is capable of binding BCMA.
  • the transmembrane domain comprises a transmembrane domain derived from a protein selected from the group consisting of the alpha, beta or zeta chain of a T cell receptor, CD28, CD3e, CD45, CD4, CD5, CD8a , CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, and CD154.
  • the transmembrane domain comprises the amino acid sequence shown in SEQ ID NO:27.
  • the co-stimulatory domain comprises a polypeptide selected from the group consisting of CD28, 4-1BB, OX-40, and ICOS.
  • the co-stimulatory domain comprises the amino acid sequence set forth in SEQ ID NO:29 or SEQ ID NO:31.
  • the intracellular signaling domain comprises a signaling domain from CD3 ⁇ .
  • the intracellular signaling domain comprises the amino acid sequence shown in SEQ ID NO:33.
  • the CAR further comprises a hinge region linking the antigen binding domain and the transmembrane domain.
  • the hinge region comprises the amino acid sequence shown in SEQ ID NO:25.
  • the CAR is also linked to a signal peptide.
  • the signal peptide comprises the amino acid sequence shown in SEQ ID NO:3.
  • the CAR is also linked to a cleavage peptide.
  • the cleaved peptide comprises an amino acid sequence from a T2A peptide.
  • the cleaved peptide comprises the amino acid sequence shown in SEQ ID NO:35.
  • the CAR comprises the amino acid sequence shown in SEQ ID NO:49 or SEQ ID NO:51.
  • the immune effector cells comprise T cells and/or natural killer (NK) cells.
  • the medicament comprises optionally a pharmaceutically acceptable carrier.
  • the present application also provides an administration method, which comprises administering the immune effector cells described in the present application to a subject in need.
  • the administration method can be used for the prevention and/or treatment of autoimmune diseases.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.25 ⁇ 10 6 to 1 ⁇ 10 6 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.25 ⁇ 10 6 to 0.5 ⁇ 10 6 cells/kg.
  • the administration method comprises administering the immune effector cells to the subject at a dose of about 0.5 ⁇ 10 6 to 1 ⁇ 10 6 cells/kg
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.25 x 106 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.5 x 106 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 1 x 106 cells/kg.
  • the method of administration comprises intravenous injection.
  • the immune effector cells are administered once.
  • the method of administration comprises treating the subject prior to infusion of the immune effector cells.
  • the treating of the subject comprises lymphoid depleting treatment.
  • said lymphoid depletion treatment is initiated about 4 days prior to infusion of said immune effector cells.
  • the period of the lymphoid depletion treatment is about 3 days.
  • the method of administration comprises a 3-day lymphodepletion treatment beginning 4 days prior to infusion of the immune effector cells.
  • the lymphoid depletion treatment comprises infusing the subject with cyclophosphamide prior to infusion of the immune effector cells.
  • the infusion dose of cyclophosphamide is about 500 mg/m 2 /day.
  • said lymphoid depletion treatment comprises infusion of said cyclophosphamide 4 days, 3 days, or 2 days prior to infusion of said immune effector cells.
  • the method of administration comprises administering fludarabine to the subject following infusion of the cyclophosphamide.
  • the fludarabine is administered at a dose of about 30 mg/m 2 .
  • the fludarabine is administered for more than about 30 minutes.
  • the lymphoid depletion treatment comprises receiving an infusion of cyclophosphamide at about 500 mg/m 2 /day 4 days, 3 days, and 2 days prior to the infusion of the immune effector cells, and infusion of cyclophosphamide Administration of about 30 mg/ m2 of fludarabine over about 30 minutes immediately following the amide.
  • the present application provides a pharmaceutical kit, which includes: 1) immune effector cells, the immune effector cells comprising a chimeric antigen receptor (CAR), wherein the CAR comprises an antigen-binding domain targeting BCMA, transmembrane domain, co-stimulatory domain and intracellular signal transduction domain, wherein the antigen binding domain is a fully human antigen binding domain; and 2) a depleting reagent.
  • CAR chimeric antigen receptor
  • the depleting agent includes cyclophosphamide, and/or fludarabine.
  • the immune effector cells in the pharmaceutical kit comprise CAR
  • the antigen binding domain of the CAR comprises HCDR1, HCDR2 and HCDR3
  • the HCDR1 comprises amino acids shown in SEQ ID NO:9
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO: 10
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 11.
  • the nucleic acid sequence encoding the HCDR1 comprises the sequence shown in SEQ ID NO:12
  • the nucleic acid sequence encoding the HCDR2 comprises the sequence shown in SEQ ID NO:13
  • the nucleic acid sequence encoding the HCDR3 Comprising the sequence shown in SEQ ID NO:14.
  • the immune effector cells in the pharmaceutical kit comprise a CAR
  • the antigen binding domain of the CAR comprises LCDR1, LCDR2 and LCDR3, wherein the LCDR1 comprises SEQ ID NO: 17 Amino acid sequence, the LCDR2 comprises the amino acid sequence shown in SEQ ID NO: 18, and the LCDR3 comprises the amino acid sequence shown in SEQ ID NO: 19.
  • the nucleic acid sequence encoding the LCDR1 comprises the sequence shown in SEQ ID NO:20
  • the nucleic acid sequence encoding the LCDR2 comprises the sequence shown in SEQ ID NO:21 (gctgcatcc)
  • encoding the LCDR3 The nucleic acid sequence comprises the sequence shown in SEQ ID NO:22.
  • the immune effector cells in the pharmaceutical kit comprise a CAR
  • the antigen binding domain of the CAR comprises a heavy chain variable region
  • the heavy chain variable region comprises SEQ ID NO: 7 Amino acid sequence shown.
  • the immune effector cells in the pharmaceutical kit comprise a CAR
  • the antigen binding domain of the CAR comprises a light chain variable region comprising SEQ ID NO: 15 Amino acid sequence shown.
  • the immune effector cells in the pharmaceutical kit comprise CAR, and the antigen binding domain of the CAR comprises the amino acid sequence shown in SEQ ID NO:43.
  • Figure 1A-1B shows the results of AQP4-IgG concentration changes in serum and cerebrospinal fluid of NMOSD patients after reinfusion of BCMA CAR-T cells described in this application.
  • Figure 2 shows the results of serum sBCMA detection of NMOSD patients after reinfusion of BCMA CAR-T cells described in this application.
  • Figures 3A-3B show the results of the BCMA CAR-T VCN-time curve and the BCMA CAR-T cell concentration-time curve described in this application.
  • chimeric antigen receptor Chimeric Antigen Receptor, CAR
  • CAR Chimeric Antigen Receptor
  • TAA chimeric Antigen receptor T cells
  • the CAR can be a genetically engineered chimeric protein capable of redirecting the cytotoxicity of immune effector cells to B cells, which combines antibody-based antigen (such as BCMA) specificity with T cell receptor activation
  • B cells which combines antibody-based antigen (such as BCMA) specificity with T cell receptor activation
  • the intracellular domains are grouped together.
  • T cells genetically modified to express CAR can specifically recognize and eliminate malignant cells expressing target antigens.
  • BCMA and “B-cell maturation antigen” are used interchangeably, a member of the tumor necrosis factor receptor family.
  • the BCMA may be human BCMA, whose GenBank accession number is BAB60895.1.
  • BCMA is a type III transmembrane protein with a cysteine-rich domain (CRD) characteristic of TNFR family members in the extracellular domain (ECD), which forms a ligand-binding motif.
  • CCD cysteine-rich domain
  • ECD extracellular domain
  • BCMA proteins may also include fragments of BCMA, such as the extracellular domain and fragments thereof.
  • BCMA binding domain generally refers to a domain that can specifically bind to a BCMA protein or a fragment thereof.
  • the BCMA extracellular binding domain may comprise a chimeric antigen receptor capable of specifically binding to a human BCMA polypeptide expressed on a B cell, an anti-BCMA antibody or an antigen-binding fragment thereof.
  • binding domain "binding domain”, “extracellular domain”, “extracellular binding domain”, “antigen-specific binding domain” and “extracellular antigen-specific binding domain” are used interchangeably in this application used, and a CAR having the ability to specifically bind a target antigen of interest (eg, BCMA) is provided.
  • BCMA binding domains may be of natural, synthetic, semi-synthetic or recombinant origin.
  • an antibody generally refers to a polypeptide molecule capable of specifically recognizing and/or neutralizing a specific antigen.
  • an antibody may comprise an immunoglobulin composed of at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, and includes any molecule comprising an antigen-binding portion thereof.
  • the term “antibody” includes monoclonal antibodies, antibody fragments or antibody derivatives, including but not limited to human antibodies, humanized antibodies, chimeric antibodies, single domain antibodies (e.g., dAb), single chain antibodies (e.g., scFv), As well as antibody fragments (eg, Fab, Fab' and (Fab)2 fragments) that bind to the antigen.
  • antibody also includes all recombinant forms of antibodies, such as antibodies expressed in prokaryotic cells, unglycosylated antibodies, and any antigen-binding antibody fragments and derivatives thereof.
  • Each heavy chain can be composed of a heavy chain variable region (VH) and a heavy chain constant region.
  • Each light chain can be composed of a light chain variable region (VL) and a light chain constant region.
  • the VH and VL regions can be further distinguished into hypervariable regions called complementarity determining regions (CDRs), which are interspersed in more conserved regions called framework regions (FRs).
  • CDRs complementarity determining regions
  • Each VH and VL may consist of three CDR and four FR regions, which may be arranged in the following order from amino-terminus to carboxy-terminus: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the variable regions of the heavy and light chains contain binding domains that interact with the antigen.
  • the constant regions of the antibodies mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (eg, effector cells) and the first component (Clq) of the classical complement system.
  • single-chain antibody may be an antibody formed by linking the heavy chain variable region and the light chain variable region through a linker peptide.
  • transmembrane domain (Transmembrane Domain) generally refers to the domain in CAR that passes through the cell membrane, which is connected to the intracellular signal transduction domain and plays a role in transmitting signals.
  • co-stimulatory domain generally refers to an intracellular domain that can provide immune co-stimulatory molecules, which are cell surface molecules required for an effective response of lymphocytes to antigens.
  • the costimulatory domain may include the costimulatory domain of CD28, and may also include the costimulatory domain of the TNF receptor family, such as the costimulatory domain of OX40 and 4-1BB.
  • hinge region generally refers to the structure used to connect the antigen recognition region and the transmembrane region.
  • HA-tag generally refers to a protein tag based on human influenza virus hemagglutinin (Human influenza hemagglutinin) antigen. short amino acid sequence. After the HA-tag sequence is joined to one end of the target protein by means of molecular biology, the specific antibody against the HA-tag can be used to bind the recombinant protein, which is beneficial to the carrying out of experiments such as immunohistochemistry (IHC) and Western Blotting (see Schembri, Laura et al. The HA tag is cleaved and loses immunoreactivity during apoptosis. Nature Methods. February 2007, 4(2):107-108).
  • IHC immunohistochemistry
  • Western Blotting see Schembri, Laura et al. The HA tag is cleaved and loses immunoreactivity during apoptosis. Nature Methods. February 2007, 4(2):107-108).
  • intracellular signaling domain generally refers to the component of CAR located in intracellular signal transduction, which includes a signaling domain and a domain that specifically binds to the receptor component, for example: its It may be selected from CD3 ⁇ intracellular domain, CD28 intracellular domain, CD28 intracellular domain, 4-1BB intracellular domain and OX40 intracellular domain.
  • signal peptide generally refers to a short (5-30 amino acids in length) peptide chain that directs the transfer of newly synthesized proteins to the secretory pathway.
  • cleaved peptide refers to self-cleaved 2A peptide, which can realize the function of cleaved protein through ribosome jumping instead of proteolytic hydrolysis, which may include T2A, F2A and P2A, etc.
  • the term "marker detection signal” generally refers to a gene, protein or other molecule with known function or sequence that can function as a specific marker and emit a detectable signal.
  • the label detection signal can be a fluorescent protein, such as: GFP, RFP, YFP and the like.
  • the marker detection signal may be EGFRt.
  • the term "EGFRt” refers to the gene encoding a truncated human epidermal growth factor receptor polypeptide that lacks the distal membrane EGF-binding domain and cytoplasmic signaling tail, but retains extracellular epitopes recognized by anti-EGFR antibodies.
  • EGFRt can be used as a non-immunogenic selection tool as well as a tracking marker for the function of genetically modified cells. In this application, it can be used as a marker molecule for CAR-T cells, and it can be used to clear CAR-T cells in vivo if necessary through the cetuximab mediated ADCC pathway (see US8802374B2).
  • Kozak sequence generally refers to the (gcc)gccRccAUGG sequence shared in eukaryotic mRNA, which plays an important role in the initiation of the translation process and is recognized by the ribosome as a translation initiation site (see De Angioletti M et al. A novel silent beta-thalassaemia mutation, the first in the Kozak sequence. Br J Haematol. 2004, 124(2):224–31.).
  • sequence identity generally refers to the degree to which sequences are identical on a nucleotide-by-nucleotide or amino-acid-by-amino acid basis over a comparison window.
  • Percent sequence identity can be calculated by comparing two optimally aligned sequences over a comparison window and determining the presence of identical nucleic acid bases (e.g., A, T, C, G, I) in the two sequences ) or the same amino acid residue (for example, Ala, Pro, Ser, Thr, Gly, Val, Leu, Ile, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gln, Cys and Met) Number of positions To obtain the number of matching positions, the number of matching positions was divided by the total number of positions in the comparison window (ie, window size) and the result multiplied by 100 to yield the percent sequence identity.
  • Optimal alignment for purposes of determining percent sequence identity can be achieved in various ways known in the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared or over the sequence region of interest.
  • variant in relation to an antibody is used in this application to refer to an antibody that has been substituted by at least 1, such as 1-30, or 1-20 or 1-10, such as 1 or 2 or 3 or 4 or 5 amino acids, Antibodies with deletions and/or insertions of amino acid alterations to regions of the antibody of interest (e.g., heavy chain variable region or light chain variable region or heavy chain CDR region or light chain CDR region), wherein the variant substantially retains the antibody prior to the alteration Biological properties of molecules.
  • this application encompasses variants of any of the antibodies described in this application.
  • the antibody variant retains at least 60%, 70%, 80%, 90%, or 100% of the biological activity (eg, antigen binding ability) of the pre-altered antibody.
  • the alterations do not result in the antibody variant losing binding to the antigen, but optionally may confer properties such as increased affinity for the antigen and different effector functions.
  • the antibody heavy chain variable region or light chain variable region, or each CDR region can be altered individually or in combination.
  • the amino acid changes may be amino acid substitutions, such as conservative substitutions.
  • the antibody variant has at least 80%, 85%, 90% or 95% or 99% or more amino acid identity to the parent antibody over the antibody sequence region of interest.
  • immune effector cells generally refers to immune cells involved in clearing foreign antigens and performing effector functions in an immune response.
  • immune effector cells For example, plasma cells, cytotoxic T cells, NK cells, APSC pluripotent cells, mast cells, etc.
  • pharmaceutically acceptable carrier generally refers to a pharmaceutically acceptable formulation carrier, solution or additive that enhances the properties of the formulation.
  • additives are well known to those skilled in the art.
  • the term “comprising” or “comprising” means including stated elements, integers or steps, but not excluding any other elements, integers or steps.
  • the term “comprising” or “comprises” is used, unless otherwise specified, the situation consisting of the mentioned elements, integers or steps is also covered.
  • an antibody variable region that "comprises” a particular sequence it is also intended to encompass an antibody variable region that consists of that particular sequence.
  • the term "about” generally refers to a range of 0.5%-10% above or below the specified value, such as 0.5%, 1%, 1.5%, 2%, 2.5%, above or below the specified value. 3%, 3.5%, 4%, 4.5%, 5%, 5.5%, 6%, 6.5%, 7%, 7.5%, 8%, 8.5%, 9%, 9.5%, or 10%.
  • autoimmune disease is used interchangeably with “autoimmune disease” and generally refers to any disease and/or condition in which the body mounts an immune system response to some component of its own tissues.
  • the immune system loses its ability to recognize a tissue or system in the body as itself, targeting and attacking it as if it were foreign.
  • the autoimmune disease may include autoimmune inflammatory disease.
  • inflammatory disease generally includes immune-mediated inflammatory processes, e.g., components of the immune system in the body that cause, mediate, or otherwise contribute to an inflammatory response, contributing to a disease or disorder.
  • the term "AQP4-Ab positive” can be used interchangeably with “AQP4 antibody positive” and "AQP4-IgG positive”, and generally refers to the detection of AQP4-Ab by one or more detection means.
  • an in vitro diagnostic kit enzyme-linked immunoassay
  • the positive judgment value can be determined independently through experiments according to each kit.
  • the AQP4-Ab positive may include normal or pathologically significant references established in the clinic. For example, it can be judged whether it is AQP4-Ab positive according to the positive determination part in the instruction manual of the ElisaRSRTM AQP4 Ab Version kit.
  • AQP4-Ab-mediated diseases and/or disorders generally refers to diseases and/or disorders associated with AQP4-Ab expression.
  • the disease and/or condition can be caused by AQP4-Ab binding to AQP4, which binding can induce complement-dependent cytotoxicity and/or antibody-dependent cytotoxicity.
  • the binding can result in inflammation, oligodendrocyte damage demyelination, and/or neuronal loss.
  • the term "neuromyelitis optica spectrum disorder” can be used interchangeably with “NMOSD”, and generally refers to an autoimmune disease of the central nervous system, which can include two groups of symptoms of the optic nerve and spinal cord.
  • NMOSD a autoimmune disease of the central nervous system
  • the term can cover AQP4-Ab positive NMOSD as well as AQP4-Ab negative NMOSD.
  • AQP4-Ab and “AQP4-IgG” generally refer to anti-AQP4 antibodies.
  • the present application provides the use of immune effector cells comprising chimeric antigen receptors in the preparation of medicaments for preventing and/or treating autoimmune diseases.
  • the immune effector cells comprise an antigen binding domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain.
  • the present application provides immune effector cells for preventing and/or treating autoimmune diseases.
  • the present application provides a method for preventing and/or treating an autoimmune disease, which comprises administering the immune effector cells described in the present application to a subject in need.
  • the autoimmune disease may include autoimmune inflammatory disease.
  • the inflammatory disease may include neuritis.
  • the inflammatory diseases may include demyelinating diseases.
  • the inflammatory diseases may include central nervous system inflammatory diseases.
  • the inflammatory disease may include central nervous system inflammatory demyelinating disease.
  • the inflammatory disease may include optic neuritis.
  • the inflammatory disease may include myelitis.
  • the inflammatory disease may comprise neuromyelitis optica (NMO).
  • the inflammatory disease may comprise Neuromyelitis Optic Spectrum Disease (NMOSD).
  • the inflammatory disease may comprise AQP4 (aquaporin 4)-Ab positive neuromyelitis optica spectrum disorder.
  • the autoimmune disease may include AQP4-Ab-mediated autoimmune disease.
  • the autoimmune disease may include AQP4-Ab positive diseases and/or disorders.
  • the AQP4-Ab-positive diseases and/or disorders may include central nervous system demyelinating diseases.
  • the AQP4-Ab-positive diseases and/or disorders may comprise AQP4-Ab-positive neuromyelitis optica spectrum disorders.
  • the autoimmune disease may comprise neuromyelitis optica spectrum disease (NMOSD).
  • NOSD neuromyelitis optica spectrum disease
  • a method of reducing the titer of AQP4-Ab in AQP4-Ab positive subjects in need of treatment for NMOSD is provided.
  • a method of reducing the occurrence and/or progression of NMOSD-related diseases and/or disorders in a subject in need of treatment for NMOSD is provided.
  • Neuromyelitis optica also known as Devic's disease or Devic's syndrome, is an autoimmune inflammatory disease in which a person's own immune system attacks the optic nerve and spinal cord. The disease causes inflammation of the optic nerve (optic neuritis) and spinal cord (myelitis).
  • Neuromyelitis optica is an acute or subacute inflammatory demyelinating disease of the central nervous system, usually an antibody-mediated idiopathic inflammatory disease of the nervous system.
  • NMO sectrum disorder is marked by the presence of NMO-IgG antibodies in serum, covering NMO and NMO-related diseases.
  • AQP4-Ab is the most important pathogenic antibody of NMOSD. A large number of basic and clinical studies have confirmed that this antibody can cause damage to the central nervous system of animals and humans, and its diagnostic specificity can be as high as 90%. NMOSD patients The positive rate of AQP4-Ab was between 40% and 90%.
  • AQP4-Ab is the pathogenic antibody of NMOSD.
  • AQP4-IgG in serum and plasma cells producing AQP4-IgG infiltrate into the central nervous system, causing AQP4-IgG to bind to aquaporin 4 (AQP4) channels on astrocytes.
  • AQP4 aquaporin 4
  • the immune effector cells may comprise a chimeric antigen receptor, and the chimeric antigen receptor may comprise an antigen binding domain, a transmembrane domain, a co-stimulatory domain and an intracellular signaling domain.
  • the antigen-binding domain of the CAR may include a fully human antigen-binding domain.
  • the extracellular domain of the CAR may comprise the single-chain scFv antibody of the present invention.
  • the single chain antibody may be linked to the transmembrane domain via a hinge region, such as the CD8 hinge.
  • the CAR can be used to transduce immune effector cells (such as T cells) and expressed on the cell surface.
  • the chimeric antigen receptor may comprise a BCMA binding domain, a transmembrane domain, a co-stimulatory domain and an intracellular signaling domain.
  • the BCMA-binding domain may comprise an antibody or fragment thereof that specifically binds BCMA.
  • the antigen binding domain may comprise heavy chain complementarity determining region 1 (HCDR1), heavy chain complementarity determining region 2 (HCDR2) and heavy chain complementarity determining region 3 (HCDR3), the amino acids of the HCDR1-3
  • the antibody may comprise light chain complementarity determining region 1 (LCDR1), light chain complementarity determining region 2 (LCDR2) and light chain complementarity determining region 3 (LCDR3), the LCDR1
  • the amino acid sequence of -3 is shown in SEQ ID No:17-19.
  • the antigen-binding domain may comprise a heavy chain variable region, and the amino acid sequence of the heavy chain variable region is shown in SEQ ID NO:7.
  • the antibody may comprise a light chain variable region, and the amino acid sequence of the light chain variable region is shown in SEQ ID NO: 15.
  • the antigen-binding domain may comprise a single-chain antibody.
  • the antigen binding domain may comprise the amino acid sequence shown in SEQ ID NO: 43 or a functional variant thereof.
  • the single chain antibody may comprise the amino acid sequence shown in SEQ ID NO:43.
  • the CAR described in the present application may include a transmembrane domain, which may comprise a polypeptide from a protein selected from the group consisting of ⁇ , ⁇ or ⁇ chains of T cell receptors, CD28, CD3e, CD45, CD4, CD5 , CD8a, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, and CD154.
  • the transmembrane domain may comprise the amino acid sequence shown by xx or a functional variant thereof.
  • the transmembrane domain of the present application may include CD8a, the sequence of which is shown in SEQ ID NO:27.
  • the co-stimulatory domain may comprise a polypeptide from a protein selected from CD28, 4-1BB, OX40 and ICOS.
  • the co-stimulatory domain may comprise the amino acid sequence shown in SEQ ID NO: 29 or SEQ ID NO: 31 or a functional variant thereof.
  • the CAR described herein can include an intracellular signaling domain, which can comprise a signaling domain from CD3 ⁇ .
  • the intracellular signaling domain may comprise the amino acid sequence shown in SEQ ID NO: 33 or a functional variant thereof.
  • the CAR described in the present application may include a hinge region, and the hinge region may connect the antibody and the transmembrane domain.
  • the hinge region may comprise the amino acid sequence shown in SEQ ID NO: 25 or a functional variant thereof.
  • the CAR described in the present application may also include an HA-tag, and the HA-tag may be located at the N-terminal of the CAR.
  • the HA-tag may comprise the amino acid sequence shown in SEQ ID NO: 5 or a functional variant thereof.
  • the expression of the CAR described in this application can be detected by using an anti-HA antibody to specifically bind it and used to enrich CAR-T cells for functional research.
  • the CAR described in the present application may include a signal peptide, and the signal peptide may include the amino acid sequence shown in SEQ ID NO: 3 or a functional variant thereof.
  • the signal peptide can be a CD8a signal peptide, the sequence of which is shown in SEQ ID NO:3.
  • the CAR may include CAR0037, CAR0085, CAR0087.
  • the CAR can also be linked to a cleavage peptide.
  • the cleaved peptide may comprise an amino acid sequence derived from a T2A peptide.
  • the cleaved peptide may comprise the amino acid sequence shown in SEQ ID NO: 35 or a functional variant thereof.
  • the cleaved peptide can be T2A, the sequence of which is shown in SEQ ID NO:35.
  • the CAR may include CAR0037, CAR0087.
  • the CAR can also be connected with a marker detection signal, and the marker detection signal can be located at the C-terminal of the CAR.
  • the label detection signal can be a fluorescent protein, which can be selected from the following group: GFP, RFP and YFP.
  • the expression of CAR molecules can be evaluated indirectly by detecting the signal of GFP.
  • the CAR may include CAR0037, the marker detection signal sequence of which is shown in SEQ ID NO:37.
  • the marker detection signal may be EGFRt.
  • the CAR may include CAR0087, the marker detection signal sequence of which is shown in SEQ ID NO:39.
  • the vector expressing the CAR may include the coding sequence of Kozak, the nucleotide sequence of which is shown in SEQ ID NO: 2 (gccgccacc).
  • the vector expressing the CAR may include the coding sequence of Kozak.
  • the CAR may include CAR0037, CAR0085, CAR0087.
  • the CAR may comprise the amino acid sequence shown in SEQ ID NO: 49 or SEQ ID NO: 51 or a functional variant thereof.
  • the CAR can be selected from CAR0037, the sequence of which is shown in SEQ ID NO:49.
  • the CAR can be selected from CAR0085, whose sequence is shown in SEQ ID NO:51; the CAR can be selected from CAR0087, whose sequence is shown in SEQ ID NO:51.
  • the CAR described in the present application may sequentially include a BCMA-binding domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain from the N-terminus.
  • the CAR may include a BCMA binding domain, and the sequence of the BCMA binding domain is shown in SEQ ID NO:43.
  • the BCMA binding domain may include HCDR1-3, its sequence is shown in sequence as SEQ ID NO:9-11; and, the BCMA binding domain may include LCDR1-3, its sequence is as shown in SEQ ID NO:17- 19.
  • the CAR may include CAR0037 or the CAR described in this application having the same LCDR1-3 and HCDR1-3.
  • the BCMA-binding domain can include a heavy chain variable region, whose sequence is shown in SEQ ID NO:7; and, the BCMA-binding domain can also include a light chain variable region, whose sequence is as shown in SEQ ID NO:15 shown.
  • the CAR may include CAR0037 or the CAR described in the present application having the same light chain variable region and heavy chain variable region as CAR0037.
  • a connecting peptide may also be included between the light chain variable region and the heavy chain variable region, the sequence of which is shown in SEQ ID NO:23.
  • the CAR may include CAR0037 or the CAR described in the present application having the same linking peptide as CAR0037.
  • the transmembrane domain may comprise a transmembrane domain from CD8a, and its sequence may be as shown in SEQ ID NO:27.
  • the CAR may include CAR0037 or the CAR described in the present application having the same transmembrane domain as CAR0037.
  • the co-stimulatory domain may comprise a co-stimulatory structure from CD28, and its sequence may be as shown in SEQ ID NO:29.
  • the CAR may include CAR0037 or the CAR described in the present application having the same costimulatory domain as CAR0037.
  • the intracellular signaling domain may comprise a signaling domain from CD3 ⁇ , the sequence of which is shown in SEQ ID NO:33.
  • the CAR may include CAR0037 or the CAR described in the present application having the same intracellular signal transduction domain as CAR0037.
  • the CAR may also include a hinge region, which may be located at the C-terminus of the BCMA-binding domain and at the N-terminus of the transmembrane domain, the sequence of which is shown in SEQ ID NO: 25.
  • the CAR may include CAR0037 or the CAR described in this application having the same hinge region as CAR0037.
  • the CAR can also include an HA-tag, and the HA-tag can be located at the N-terminus of the BCMA-binding domain, and its sequence is shown in SEQ ID NO:5.
  • the CAR may include CAR0037 or the CAR described in this application having the same HA-tag as it.
  • the CAR can also be connected with a signal peptide, which can be located at the N-terminus of the CAR, and its sequence can be as shown in SEQ ID NO:3.
  • the CAR can also be linked to a cleavage peptide, such as T2A.
  • the cleavage peptide may be located at the C-terminus of the intracellular signal transduction domain, and its sequence may be as shown in SEQ ID NO:35.
  • the CAR can also be linked with a label detection signal, which can be located at the C-terminus of the CAR (or, the cleaved peptide).
  • the marker detection signal can be selected from the following groups: GFP, RFP and YFP, the sequence of which is shown in SEQ ID NO:37.
  • the CAR described in this application can be CAR0037, the amino acid sequences of its LCDR1-3 are shown in SEQ ID NO:17, SEQ ID NO:18 (AAS) and SEQ ID NO:19 respectively; the amino acid sequence of VL is shown in SEQ ID NO:19.
  • the amino acid sequence of HCDR1-3 is shown in SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 respectively;
  • the amino acid sequence of VH is shown in SEQ ID NO:7;
  • VH and The sequence of the connecting peptide between VL is shown in SEQ ID NO:23; its hinge region is shown in SEQ ID NO:25; its transmembrane domain is shown in SEQ ID NO:27; its co-stimulatory domain is CD28
  • the co-stimulatory domain is shown in SEQ ID NO:29; its CD3 ⁇ intracellular signaling domain is shown in SEQ ID NO:33;
  • the CAR0037 can also include a cleavage peptide shown in SEQ ID NO:35, And the GFP marker detection signal shown in SEQ ID NO:37;
  • the CAR0037 can also comprise the KOZAK sequence shown in SEQ ID NO:1 (AAT), the CD8a signal peptide shown in SEQ ID NO:3, such as HA-tag shown in S
  • the CAR described in this application can be CAR0085, the amino acid sequences of its LCDR1-3 are shown in SEQ ID NO:17, SEQ ID NO:18 (AAS) and SEQ ID NO:19 respectively; the amino acid sequence of VL is shown in SEQ ID NO:19.
  • the amino acid sequence of HCDR1-3 is shown in SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 respectively;
  • the amino acid sequence of VH is shown in SEQ ID NO:7;
  • VH and The sequence of the connecting peptide between VL is shown in SEQ ID NO:23; its hinge region is shown in SEQ ID NO:25; its transmembrane domain is shown in SEQ ID NO:27; its co-stimulatory domain is 4 -1BB co-stimulatory domain, as shown in SEQ ID NO:31;
  • Its CD3 ⁇ intracellular signaling domain is as shown in SEQ ID NO:33;
  • the vector expressing CAR0085 can also include SEQ ID NO:1 (AAT ) shown in the KOZAK sequence; the CAR0085 is shown in the CD8a signal peptide of SEQ ID NO:3; the CAR0085 can also include the cleavage peptide shown in SEQ ID NO:35, and as shown in SEQ ID NO:39
  • the CAR described in this application can be CAR0087, the amino acid sequences of its LCDR1-3 are shown in SEQ ID NO:17, SEQ ID NO:18 (AAS) and SEQ ID NO:19 respectively; the amino acid sequence of VL is shown in SEQ ID NO:19.
  • the amino acid sequence of HCDR1-3 is shown in SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:11 respectively;
  • the amino acid sequence of VH is shown in SEQ ID NO:7;
  • VH and The sequence of the connecting peptide between VL is shown in SEQ ID NO:23; its hinge region is shown in SEQ ID NO:25; its transmembrane domain is shown in SEQ ID NO:27; its co-stimulatory domain is 4 -1BB co-stimulatory domain, as shown in SEQ ID NO:31;
  • Its CD3 ⁇ intracellular signaling domain is as shown in SEQ ID NO:33;
  • the vector expressing CAR0087 can also include SEQ ID NO:2 (gccgccacc ) the KOZAK sequence shown in ); the CAR0087 can also include the CD8a signal peptide shown in SEQ ID NO:3.
  • NMOSD can be evaluated with reference to the EDSS neurological scale score, the number of active lesions on MRI, the annual recurrence rate and the number of inactive lesions, the annual recurrence rate, and the patient-free ratio.
  • the measured values of serum AQP4-Ab titers of NMOSD patients before and after treatment can be analyzed by descriptive statistics method, and the changes from the baseline can be used to evaluate the treatment effect.
  • the AQP4-Ab titer in the subject's serum can be detected by ELISA method to measure the change of AQP4-Ab in the subject's serum.
  • the medicament for preventing and/or treating autoimmune diseases may comprise the immune effector cells described in the present application and optionally a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may include buffers, antioxidants, preservatives, low molecular weight polypeptides, proteins, hydrophilic polymers, amino acids, sugars, chelating agents, counterions, metal complexes and/or non-ionic surface Active agents, etc.
  • the drug can be formulated for intravenous injection.
  • the present application provides an administration method, the method comprising administering the immune effector cells described in the present application to a subject in need.
  • the subject in need includes a subject suffering from an autoimmune disease.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.25 ⁇ 10 6 to 1 ⁇ 10 6 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.25 x 106 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.5 x 106 cells/kg. In certain embodiments, the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.395 x 106 cells/kg. In certain embodiments, the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.425 x 106 cells/kg. In certain embodiments, the method of administering comprises administering the immune effector cells to the subject at a dose of about 0.495 x 106 cells/kg.
  • the method of administering comprises administering the immune effector cells to the subject at a dose of about 1 x 106 cells/kg.
  • the administration method may comprise intravenous injection.
  • the number of administration of the immune effector cells may be one time.
  • the method of administration comprises treating the subject prior to infusion of the immune effector cells.
  • the treating of the subject comprises lymphoid depleting treatment.
  • said lymphoid depletion treatment is initiated about 4 days prior to infusion of said immune effector cells.
  • the period of the lymphoid depletion treatment is about 3 days.
  • the method of administration comprises a 3-day lymphodepletion treatment beginning 4 days prior to infusion of the immune effector cells.
  • the lymphoid depletion treatment comprises infusing the subject with cyclophosphamide prior to infusion of the immune effector cells.
  • the infusion dose of cyclophosphamide is about 500 mg/m 2 /day.
  • said lymphoid depletion treatment comprises infusion of said cyclophosphamide 4 days, 3 days, or 2 days prior to infusion of said immune effector cells.
  • the method of administration comprises administering fludarabine to the subject following infusion of the cyclophosphamide.
  • the fludarabine is administered at a dose of about 30 mg/m 2 .
  • the fludarabine is administered for more than about 30 minutes.
  • the lymphoid depletion treatment comprises receiving an infusion of cyclophosphamide at about 500 mg/m 2 /day 4 days, 3 days, or 2 days prior to infusion of the immune effector cells, and Immediately thereafter, administer about 30 mg/ m2 of fludarabine over about 30 minutes.
  • the present application also provides a pharmaceutical kit, which includes: 1) immune effector cells, the immune effector cells comprise a chimeric antigen receptor (CAR), wherein the CAR comprises an antigen-binding antigen targeting BCMA domain, transmembrane domain, co-stimulatory domain and intracellular signaling domain, wherein the antigen-binding domain is a fully human antigen-binding domain; and a depleting reagent.
  • CAR chimeric antigen receptor
  • the rinsing agent may include cyclophosphamide and/or fludarabine.
  • the immune effector cells in the pharmaceutical kit may comprise CAR, and the antigen binding domain of the CAR may comprise HCDR1, HCDR2 and HCDR3, and the HCDR1 may comprise SEQ ID NO:9 Amino acid sequence, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO: 10, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO: 11.
  • the immune effector cells in the pharmaceutical kit may comprise CAR, and the antigen binding domain of the CAR may comprise LCDR1, LCDR2 and LCDR3, wherein the LCDR1 may comprise SEQ ID NO: 17
  • the amino acid sequence of the LCDR2 can include the amino acid sequence shown in SEQ ID NO: 18 (AAS), and the LCDR3 can include the amino acid sequence shown in SEQ ID NO: 19.
  • the immune effector cells in the pharmaceutical kit may comprise CAR, and the antigen binding domain of the CAR may comprise a heavy chain variable region, and the heavy chain variable region may comprise SEQ ID NO: The amino acid sequence shown in 7.
  • the immune effector cells in the pharmaceutical kit may comprise a CAR, and the antigen binding domain of the CAR may comprise a light chain variable region, and the light chain variable region may comprise SEQ ID NO: The amino acid sequence shown in 15.
  • the immune effector cells in the pharmaceutical kit may comprise CAR, and the antigen-binding domain of the CAR may comprise the amino acid sequence shown in SEQ ID NO:43.
  • the lentiviral vector PLVX-EF1alpha-IRES-Puro was double digested with NotI and MluI, and the vector fragment was recovered.
  • the candidate scFv plasmid PXL0026 (its nucleotide sequence is SEQ ID NO: 44) was amplified by PCR, and the NotI restriction site (including protective bases), CD8a signal peptide, HA -label; hinge region, transmembrane region, CD28 co-stimulatory factor, CD3 ⁇ intracellular signaling domain gene synthesis, PCR amplification; T2A cleavage peptide and eGFP PCR amplified from plasmid pMy-BirA-T2A-eGFP, 3 Carry MluI restriction site and protective base at the ' end; then use overlap PCR to obtain a PCR fragment with a NotI restriction site at the 5' end and a MluI site at the 3' end, and perform NotI and MluI
  • the CAR plasmid numbered PXL0085 was obtained in a similar way, the lentiviral vector PLVX-EF1alpha-IRES-Puro was double-digested with NotI and MluI, and the vector fragment was recovered.
  • the candidate scFv plasmid PXL0026 was amplified by PCR, and the NotI restriction site (including protective bases), CD8a signal peptide, hinge region, transmembrane region, and 4-1BB co-stimulatory factor ( Its nucleotide sequence is SEQ ID NO: 32), CD3 ⁇ intracellular signaling domain gene synthesis, PCR amplification; then use overlap PCR to obtain the 5' end band NotI restriction site 3' end band MluI site For the PCR fragment, NotI and MluI double enzyme digestion was performed on the fragment, and the fragment was recovered. T4 connection was constructed to obtain the CAR plasmid numbered PXL0085 (the nucleotide sequence of the CAR molecular part of CAR0085 is shown in SEQ ID NO: 52).
  • the CAR plasmid numbered PXL0087 was obtained in a similar way, the lentiviral vector PLVX-EF1alpha-IRES-Puro was double digested with NotI and MluI, and the vector fragment was recovered.
  • the candidate scFv plasmid PXL0026 was amplified by PCR, and the NotI restriction site (including protective bases), CD8a signal peptide, hinge region, transmembrane region, and 4-1BB co-stimulatory factor ( Its nucleotide sequence is SEQ ID NO:32), CD3 ⁇ intracellular signaling domain, T2A cleavage peptide, EGFRt (its nucleotide sequence is SEQ ID NO:40) gene synthesis, PCR amplification; Then use overlap A PCR fragment with a NotI restriction site at the 5' end and a MluI site at the 3' end was obtained by PCR, and the fragment was double digested with NotI and MluI and recovered. T4 connection was constructed to obtain the CAR plasmid numbered PXL0087 (the nucleotide sequence of the CAR molecular part of CAR0087 is shown in SEQ ID NO: 52).
  • PBMCs were isolated from peripheral blood collected from healthy donors, and T cells were further sorted using CD3 MicroBeads.
  • the sorted T cells were activated using CD3/CD28 Dynabeads, and transduced with LV0007, LV0020, and LV0021 lentiviruses.
  • the density of T cells during transduction was about 1.5 ⁇ 106 cells/ml.
  • a medium change was performed on the transduced T cells.
  • LV0007-CAR-T, LV0020-CAR-T and LV0021-CAR-T cells were obtained.
  • the preparation process of CAR-T cells is shown in the table below. The product must pass the quality inspection before it can be released. Before the release of each batch of finished products, the quality control department must collect and evaluate all the test records related to the batch of finished products. In the form of frozen suspension, filled in specific infusion bags for storage and transportation.
  • the preparation process of CAR-T cells is shown in Table 1.
  • Embodiment 2 Lymph clearing (lymphocyte removal) pretreatment
  • the pretreatment process for clearing stranguria is as follows: intravenous infusion of 500 mg/m 2 cyclophosphamide, once a day for 3 consecutive days, each time over 30 minutes, and immediately after the completion of cyclophosphamide intravenous infusion, intravenous infusion of 30 mg/m 2 Fludarabine, once a day for 3 consecutive days, each time more than 30 minutes.
  • intravenous infusion of 500 mg/m 2 cyclophosphamide once a day for 3 consecutive days, each time over 30 minutes, and immediately after the completion of cyclophosphamide intravenous infusion, intravenous infusion of 30 mg/m 2 Fludarabine, once a day for 3 consecutive days, each time more than 30 minutes.
  • subject Neu-003 as an example, the process of clearing stranguria is described in detail, and the pretreatment process of clearing stranguria is shown in Table 2. The degreasing treatment of other subjects was carried out in a similar manner.
  • BCMA CAR-T cells were administered to 3 subjects (Neu-003, Neu-004, Neu-008) at a dose of 0.5 ⁇ 10 6 CAR+ cells/kg, and 8 subjects (Neu-007, Neu-009, Neu-010, Neu-012, Neu-013, Neu-014, Neu-015, Neu-016) Administer 1.0 ⁇ 10 6 CAR+ cells/kg of BCMA CAR-T cells (containing the sequence of CAR0087) .
  • the BCMA CAR-T cells were infused intravenously two days after the end of the depletion preconditioning, and the number of infusions was once.
  • the timing of venous blood collection for changes in serum AQP4-IgG titer before and after treatment is as follows: screening period (D42-D20), twice a week from the screening period to before clearing the leprosy, 12 to 5 days before cell reinfusion (time window: no later than 1 minute before delineation), 1 day before cell reinfusion (time window: before reinfusion), 3 days (time window 8 hours), 7 days (time window 12 hours), 10 days (time window ⁇ 12 hours ), 14 days (time window ⁇ 1 day), 21 days (time window ⁇ 1 day), 28 days (time window ⁇ 3 days), 56 days (time window ⁇ 3 days), 84 days (time window ⁇ 7 days ), every 3 months (time window ⁇ 14 days) within 2 years after 84 days.
  • CSF AQP4-IgG collection time D12 to D5, 14 days (time window ⁇ 1 day), 28 days (time window ⁇ 3 days), 84 days (time window ⁇ 7 days), 84 days later Every 3 months (time window ⁇ 14 days) within 2 years and at the time of group visit.
  • the second-generation aquaporin 4 (AQP4) autoantibody ELISA detection kit (source: RSR company, batch number: 2KAQE66) was used for detection, and the absorbance value (OD value) was read at 450nm and 405nm on a microplate reader, and the OD value was obtained by fitting Values were compared with a standard curve to calculate the concentration levels of AQP4-IgG.
  • Descriptive statistics were used to analyze the measured values of AQP4-IgG concentrations in serum and cerebrospinal fluid of NMOSD patients before and after treatment.
  • the serum AQP4-IgG concentration-time curves of NMOSD patients were drawn at each time point.
  • the serum AQP4-IgG levels of 2 subjects (Neu-004 and Neu-008) in the 0.5 ⁇ 10 6 CAR-T cell/kg dose group were still within the normal range 6 months after cell reinfusion, and Neu- The serum AQP4-IgG level of 003 subjects rose significantly and was close to the baseline level before reinfusion.
  • the serum AQP4-IgG levels of 8 subjects in the 1.0 ⁇ 10 6 CAR-T cell/kg dose group were still within or near the normal value (3u/mL) at the cut-off date, maintaining a downward trend without rebounding.
  • Figure 1B shows the change trend of AQP4-IgG in cerebrospinal fluid of 11 patients.
  • the results showed that, except for the AQP4-IgG level in the cerebrospinal fluid of the Neu-004 subject, which was significantly lower than the baseline, the AQP4-IgG level in the cerebrospinal fluid of other subjects did not show a significant upward or downward trend.
  • the subjects were AQP4-IgG positive relapsed refractory NMOSD patients.
  • Three subjects were given a dose of 0.5 ⁇ 10 6 CAR+ cells/kg and eight subjects (Neu-007, Neu-009, Neu-010 , Neu-012, Neu-013, Neu-014, Neu-015, Neu-016) administered BCMA CAR-T cells (containing the sequence of CAR0087) at a dose of 1.0 ⁇ 10 6 CAR+ cells/kg.
  • the BCMA CAR-T cells were infused intravenously two days after the end of the depletion preconditioning, and the number of infusions was once.
  • the collection time of peripheral blood free BCMA (sBCMA) before and after treatment is as follows: D12 to D5, 7 days (time window 12 hours), 14 days (time window 1 day), 21 days (time window 1 day) day), 28 days (time window 3 days), 56 days (time window ⁇ 7 days), 84 days (time window 7 days), 84 days to 2 years every 3 months (time window 14 days) once and out of the group When visiting.
  • the Human BCMA/TNFRSF17 DuoSet ELISA detection kit (source: R&R company, article number: CAT#DY193) was used for detection, and the absorbance value (OD value) was read at 450nm and 570nm on the SpectraMax i3 microplate reader, and 570nm was used as the corrected wavelength. Fit the OD value to the standard curve to calculate the concentration level of sBCMA.
  • the sBCMA results and changing trends of the 11 subjects are shown in Figure 2.
  • the sBCMA concentration level was high before cell reinfusion, and the sBCMA level dropped sharply within 1 month after reinfusion with the expansion of CAR-T cells.
  • Limit (3.2ng/mL), suggesting that CT103A can significantly reduce sBCMA in patients, indirectly indicating its ability to rapidly clear plasma cells.
  • 0.5 ⁇ 10 6 CAR-T cells/kg dose group and 1.0 ⁇ 10 6 CAR-T cells/kg dose group had similar decrease in peripheral blood sBCMA.
  • Three months after the reinfusion the sBCMA levels in the peripheral blood of 3 subjects in the 0.5 ⁇ 10 6 CAR-T cell/kg dose group rebounded.
  • the sBCMA level of the first 7 subjects in the 1.0 ⁇ 10 6 CAR-T cell/kg dose group remained below the detection limit at the cut-off date (the longest observation time was 84 days after reinfusion), and the last 1 subject
  • the sBCMA level of the patient (Neu-016) has dropped to near the detection limit (the longest observation time is 14 days).
  • Embodiment 5 Pharmacokinetics (PK) experiment and result
  • the concentration of BCMA CAR-T cells was detected by flow cytometry, and the main reagents and instruments involved were Anti-CD45 (PercP-labeled) and Anti-CD3, FITC-labeled BCMA protein from Acro BIOSYSTEMS, and Agilent’s Novocyte flow cytometer.
  • the concentration of BCMA CAR-T cells is presented as the percentage (%) of CAR-T in lymphocytes.
  • the vector copy number (Vector copy number, VCN) of BCMA CAR-T cells is detected by droplet digital PCR (ddPCR).
  • the main reagents and instruments involved are: Genomic DNA extraction kit from Qiagen, Thermo Fisher's NanoDrop Nucleic Acid Quantitative Instrument, BIO-RAD's droplet generator oil, droplet generator card and pad, ddPCR Supermix for Probes (no dUTP), droplet reading oil and QX200 droplet digital PCR system.
  • each BCMA CAR-T cell CD3 + CAR + cell
  • Sampling time 12 to 5 days before the reinfusion of BCMA CAR-T cells (time window: before clearing lymph pretreatment), 1 day after reinfusion (time window ⁇ 8 hours), 3 days (time window ⁇ 8 hours), 5 days Days (time window ⁇ 12 hours), 7 days (time window ⁇ 12 hours), 10 days (time window ⁇ 12 hours), 14 days (time window ⁇ 1 day), 21 days (time window ⁇ 1 day), 28 days
  • Blood samples were collected every 3 months (time window ⁇ 14 days) after 84 days (time window ⁇ 14 days) until CAR -2 years after the peak value was detected, no VCN was detected by droplet digital PCR (ddPCR), the disease worsened, and the group was discharged or reinfused.
  • ddPCR droplet digital PCR
  • 1 day after reinfusion means 24 hours after reinfusion time
  • 3 days after reinfusion means 72 hours after reinfusion time, and so on for subsequent visits. Try to conduct blood routine sampling at the same time as PK sampling, and the time between blood routine sampling and PK blood collection should not exceed 24 hours at most.
  • Plasma drug concentration-time data analysis draw the concentration-time curves of BCMA CAR-T cells in peripheral blood and VCN-time curves at each time point; T concentrations and VCN values.
  • PK parameter analysis Calculate the arithmetic mean, standard deviation coefficient of variation, median, and maximum value of PK parameters (C max , T max , AUC 0 ⁇ 28d , and AUC 0 ⁇ last , etc.) through VCN and BCMA CAR-T cell concentrations , Minimum, Geometric Mean, and Geometric Mean Coefficient of Variation.
  • Figure 3A-3B shows the results of BCMA CAR-T cell concentration and VCN changes, 3 cases of 0.5 ⁇ 10 6 CAR-T cells/kg dose group and 8 cases of 1.0 ⁇ 10 6 CAR-T cells/kg dose group
  • Blood CAR-T cells showed similar PK characteristics, that is, after administration, the concentration of CAR-T cells and the level of VCN rose rapidly at first, reached a peak within 14 days, and then declined slowly.
  • CAR VCN could still be detected after 89 days of reinfusion of BCMA CAR-T cells of this application.
  • the above data indicate that the BCMA CAR-T cells of the present application expand well in the subject and have a long duration.
  • the PK parameters are shown in Table 3 and Table 4, the 0.5 ⁇ 10 6 CAR-T cell/kg dose group and the 1.0 ⁇ 10 6 CAR-T cell/kg dose group were moderately increased in peripheral blood CAR-T cell expansion.
  • the peak time (Tmax) was the 10th day after reinfusion.
  • the median Cmax of BCMA CAR-T cell concentrations in the two dose groups were 77.78% and 85.853%, respectively, the median AUC0-28 were 650.402days ⁇ % and 950.484days ⁇ %, and the median AUC0-last was 738.203days ⁇ % and 1128.923days ⁇ %.
  • the median Cmax of VCN in the two dose groups were 55300Copies/ ⁇ g DNA and 78025.5Copies/ ⁇ g DNA, respectively, and the median AUC0-28 were 303732.7Days ⁇ Copies/ ⁇ g DNA and 462986.75Days ⁇ Copies/ ⁇ g DNA, and the median AUC0- last were 385535.8Days ⁇ Copies/ ⁇ g DNA and 462986.75Days ⁇ Copies/ ⁇ g DNA respectively.
  • the median Cmax, AUC0-28 and AUC0-last of the 1.0 ⁇ 10 6 CAR-T cells/kg dose group were higher than those of the 0.5 ⁇ 10 6 CAR-T cells/kg dose group.
  • Embodiment 6 Inflammation factor detection method and result
  • the concentration of ferritin was detected by particle-enhanced immunoturbidimetry, the concentration of IL-6 was detected by electrochemiluminescence three-antigen sandwich turbidimetry, the concentration of C-reactive protein was detected by immune projection turbidimetry, and the concentration of C-reactive protein was detected by electrochemiluminescence three-antigen sandwich turbidimetry. Measure procalcitonin concentration.
  • the results of inflammatory factors in patients with Neu-003 are shown in Table 5.
  • the highest concentration of ferritin after cell reinfusion was 654.6ug/mL
  • the highest concentration of IL-6 after cell reinfusion was 31.11pg/mL
  • the concentration of C-reactive protein in cells The highest concentration after reinfusion was 16.7mg/L, and the concentration of procalcitonin after cell reinfusion was lower than that before reinfusion, and the concentration of various inflammatory factors did not increase after administration, which usually caused side effects.
  • the results showed that BCMA CAR - Less risk of cytokine release syndrome from T cell therapy.

Landscapes

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

Abstract

L'invention concerne l'utilisation d'une cellule effectrice immunitaire dans la préparation d'un médicament pour la prévention et/ou le traitement de maladies auto-immunes. La cellule effectrice immunitaire contient un récepteur antigénique chimérique (CAR), le CAR contenant un domaine de liaison à l'antigène ciblant BCMA, un domaine transmembranaire, un domaine co-stimulateur et un domaine de signalisation intracellulaire, et le domaine de liaison à l'antigène étant un domaine de liaison à l'antigène entièrement humain.
PCT/CN2022/119980 2021-09-22 2022-09-20 Utilisation de t-car bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes WO2023045934A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/CN2021/119720 WO2023044633A1 (fr) 2021-09-22 2021-09-22 Application de car-t bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes
CNPCT/CN2021/119720 2021-09-22

Publications (1)

Publication Number Publication Date
WO2023045934A1 true WO2023045934A1 (fr) 2023-03-30

Family

ID=85719765

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2021/119720 WO2023044633A1 (fr) 2021-09-22 2021-09-22 Application de car-t bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes
PCT/CN2022/119980 WO2023045934A1 (fr) 2021-09-22 2022-09-20 Utilisation de t-car bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/119720 WO2023044633A1 (fr) 2021-09-22 2021-09-22 Application de car-t bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes

Country Status (1)

Country Link
WO (2) WO2023044633A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019149250A1 (fr) * 2018-02-01 2019-08-08 南京驯鹿医疗技术有限公司 Récepteur antigénique chimérique (car) se liant à bcma et ses applications
WO2019149269A1 (fr) * 2018-02-01 2019-08-08 信达生物制药(苏州)有限公司 Fragment variable à chaîne unique anti-antigène de maturation des lymphocytes b (bcma) entièrement humain, et application associée
CN112062851A (zh) * 2019-06-11 2020-12-11 南京驯鹿医疗技术有限公司 靶向bcma嵌合抗原受体的抗体及其应用
WO2021163329A1 (fr) * 2020-02-12 2021-08-19 Celgene Corporation Thérapie anti-bcma dans des troubles auto-immuns

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2015292590B2 (en) * 2014-07-24 2020-01-16 2Seventy Bio, Inc. BCMA chimeric antigen receptors
JP2020507605A (ja) * 2017-02-17 2020-03-12 フレッド ハッチンソン キャンサー リサーチ センター Bcma関連癌および自己免疫疾患の治療のための併用療法
JP6968389B2 (ja) * 2018-02-01 2021-11-17 南京馴鹿医療技術有限公司 Bcmaに結合するキメラ抗原受容体(car)及びその応用
CN111454358A (zh) * 2019-01-18 2020-07-28 四川科伦博泰生物医药股份有限公司 一种嵌合抗原受体及其应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019149250A1 (fr) * 2018-02-01 2019-08-08 南京驯鹿医疗技术有限公司 Récepteur antigénique chimérique (car) se liant à bcma et ses applications
WO2019149269A1 (fr) * 2018-02-01 2019-08-08 信达生物制药(苏州)有限公司 Fragment variable à chaîne unique anti-antigène de maturation des lymphocytes b (bcma) entièrement humain, et application associée
CN112062851A (zh) * 2019-06-11 2020-12-11 南京驯鹿医疗技术有限公司 靶向bcma嵌合抗原受体的抗体及其应用
WO2021163329A1 (fr) * 2020-02-12 2021-08-19 Celgene Corporation Thérapie anti-bcma dans des troubles auto-immuns

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DERDELINCKX JUDITH, REYNDERS TATJANA, WENS INEZ, COOLS NATHALIE, WILLEKENS BARBARA: "Cells to the Rescue: Emerging Cell-Based Treatment Approaches for NMOSD and MOGAD", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 22, no. 15, 1 January 2021 (2021-01-01), pages 7925, XP093055494, DOI: 10.3390/ijms22157925 *
TRAUB JAN, HUSSEINI LEILA, WEBER MARTIN S.: "B Cells and Antibodies as Targets of Therapeutic Intervention in Neuromyelitis Optica Spectrum Disorders", PHARMACEUTICALS, vol. 14, no. 1, pages 37, XP093033172, DOI: 10.3390/ph14010037 *
ZHAO WANHONG, MENG SHAN: "Advances in BCMA Targeted Chimeric Antigen Receptor Modified T Cells Therapy for Multiple Myeloma", LINCHUANG XUEYEXUE ZAZHI = JOURNAL OF CLINICAL HEMATOLOGY, HUAZHONG KEJI DAXUE TONGJI YIXUEYUAN FUSHU XIEHE YIYUAN, CN, vol. 31, no. 7, 31 December 2018 (2018-12-31), CN , pages 495 - 498, XP093055491, ISSN: 1004-2806, DOI: 10.13201/j.issn.1004-2806.2018.07.002 *

Also Published As

Publication number Publication date
WO2023044633A1 (fr) 2023-03-30

Similar Documents

Publication Publication Date Title
US20210179735A1 (en) Trispecific binding proteins and methods of use
JP7011574B2 (ja) Flt3及びcd3に対する抗体構築物
US20190225702A1 (en) Innate immune cell trispecific binding proteins and methods of use
WO2020043184A1 (fr) Anticorps bifonctionnel anti-pd-1 et anti-vegfa, composition pharmaceutique et utilisation associées
DK2427212T3 (en) ANTI-CD100 ANTIBODIES AND PROCEDURES FOR USE THEREOF
JP2018524997A (ja) Egfrviii及びcd3に結合する二重特異性抗体構築物
WO2018160671A1 (fr) Inhibiteurs de point de contrôle ciblés et méthodes d'utilisation
US12006365B2 (en) Human anti-semaphorin 4D antibody
US11584799B2 (en) Anti-CD30 antibodies and methods for treating CD30+ cancer
JP2018508214A (ja) Cd20結合分子およびその使用方法
CN109952319B (zh) 对gp100具有特异性的TCR-抗CD3 scFv融合蛋白的定量用药方案
KR20190137077A (ko) T 세포 관여 항체 구축물을 포함하는 저 pH 약제학적 조성물
US10683358B2 (en) Human TNFRSF25 antibody
JP2019514871A (ja) 骨髄性白血病の治療方法で使用するための、cd33とcd3に結合する二重特異性構築物の投与の方法
US20230312738A1 (en) System and method for the development of cd30 bispecific antibodies for immunotherapy of cd30+ malignancies
JP2024501809A (ja) Ceacam5およびcd47に対する二重特異性抗体
JP2022553129A (ja) ポリオウイルス受容体(pvr)に対する抗体およびその使用
WO2023169583A1 (fr) Préparation et application d'une molécule d'engagement de cellule bispécifique construite sur la base de pep42
WO2023045934A1 (fr) Utilisation de t-car bcma dans la préparation d'un médicament pour le traitement de maladies auto-immunes
US20240002530A1 (en) Anti-psma antibodies and methods of use
KR20230031814A (ko) 항-세라마이드 항체
WO2020047818A1 (fr) Anticorps bispécifique et son procédé d'utilisation
US20030180799A1 (en) Antibodies against plasma cells
WO2024109657A1 (fr) Anticorps anti-ccr8 et son utilisation
CA3206413A1 (fr) Anticorps diriges contre cd112r et leurs utilisations

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: 22871980

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE