CN115786268A - Engineered immune cells and uses thereof - Google Patents

Engineered immune cells and uses thereof Download PDF

Info

Publication number
CN115786268A
CN115786268A CN202111060954.8A CN202111060954A CN115786268A CN 115786268 A CN115786268 A CN 115786268A CN 202111060954 A CN202111060954 A CN 202111060954A CN 115786268 A CN115786268 A CN 115786268A
Authority
CN
China
Prior art keywords
cell
leu
engineered immune
antigen
gly
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202111060954.8A
Other languages
Chinese (zh)
Inventor
邢芸
任江涛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Bioheng Biotech Co Ltd
Original Assignee
Nanjing Bioheng Biotech Co Ltd
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 Nanjing Bioheng Biotech Co Ltd filed Critical Nanjing Bioheng Biotech Co Ltd
Priority to CN202111060954.8A priority Critical patent/CN115786268A/en
Priority to PCT/CN2022/114484 priority patent/WO2023035947A1/en
Publication of CN115786268A publication Critical patent/CN115786268A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • 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
    • 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
    • A61K39/464436Cytokines
    • A61K39/464442Chemokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • 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
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • 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

Abstract

The present invention relates to an engineered immune cell expressing a cell surface molecule that specifically recognizes an antigen, exogenous IL7 and CXCL9. The invention also provides the use of the engineered immune cells in the treatment of cancer, infection or autoimmune disease. Compared with the traditional engineered immune cell, the engineered immune cell has obviously improved tumor killing activity.

Description

Engineered immune cells and uses thereof
Technical Field
The present invention is in the field of immunotherapy. More specifically, the invention relates to an engineered immune cell expressing a cell surface molecule that specifically recognizes an antigen, exogenous IL7 and CXCL9. More preferably, the cell surface molecule specifically recognizing an antigen is a chimeric antigen receptor.
Background
In recent years, adoptive cell therapy, an emerging immunotherapy, has shown great advantages in the field of tumor therapy. Such therapies typically require that cells be first engineered, for example by gene editing and/or transduction techniques, to carry exogenous proteins such as chimeric antigen receptors, recombinant T cell receptors, etc., and then expanded in vitro and returned to the patient. Currently, these therapies have shown good efficacy against hematological tumors, but their efficacy has not been satisfactory for solid tumors, one of the reasons being the inability to effectively transport the engineered cells to the tumor site (i.e., the immunosuppressive tumor microenvironment).
Thus, there remains a need for improved cell therapies to counteract the inhibitory effects of the tumor microenvironment, while recruiting other immune effector cells to the tumor site to potentiate the anti-tumor effect.
Disclosure of Invention
In a first aspect, the invention provides a novel engineered immune cell that expresses a cell surface molecule that specifically recognizes an antigen, and exogenous IL7 and CXCL9.
In one embodiment, the cell surface molecule specifically recognizing an antigen is a chimeric antigen receptor, a T cell fusion protein or a T cell antigen coupler, preferably a chimeric antigen receptor.
In one embodiment, the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor comprising an antigen binding region, a transmembrane domain, and an intracellular domain comprising a costimulatory domain and/or a primary signaling domain. Wherein the antigen-antigen binding region may be selected from IgG, fab ', F (ab ') 2, fd ', fv, scFv, sdFv, linear antibody, single domain antibody, nanobody, diabody, anticalin and DARPIN. Preferably, the antigen-antigen binding region is selected from the group consisting of scFv, fab, single domain antibody and nanobody.
In one embodiment, the cell surface molecule specifically recognizing an antigen binds to one or more targets selected from the group consisting of: <xnotran> CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33, CD37, CD38, CD40, CD40 3245 zxft 3245 44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2 3732 zxft 3732 40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa , TIM3, TSHR, CD19, BAFF-3963 zxft 3963-1, EGFRvIII, tEGFR, GD2, GD3, BCMA, tn , PSMA, ROR1, FLT3, FAP, TAG72, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, IL-llRa, IL-22Ra, IL-2, , PSCA, PRSS21, VEGFR2, lewisY, PDGFR- β, SSEA-4, AFP, folate α, erbB2 (Her 2/neu), erbB3, erbB4, MUC1, MUC16, EGFR, CS1, NCAM, claudin18.2, c-Met, prostase, PAP, ELF2M, ephrin B2, IGF-I , CAIX, LMP2, gpl00, bcr-abl, , ephA2, fucosyl GMl, sLe, GM3, TGS5, HMWMAA, o- -GD2, folate β, TEM7 4325 zxft 4325 6, GPRC5 3536 zxft 3536 61, ALK, , PLAC1, globoH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6 3926 zxft 3926 51E2, TARP, WT1, NY-ESO-1, LAGE-la, MAGE-A1, MAGE-A3, MAGE-A6, , HPV E6, E7, ETV6-AML, 17, XAGE1, tie 2, MAD-CT-1, MAD-CT-2, fos 1, p53, p53 , PSA, , PCTA-l/Galectin 8, melanA/MARTl, ras , hTERT, , ML-IAP, </xnotran> TMPRSS2 ETS fusion gene, NA17, PAX3, androgen receptor, progesterone receptor, cyclin Bl, MYCN, rhoC, TRP-2, CYP1B 1, BORIS, SART3, PAX5, OY-TES 1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxylesterase, mut hsp70-2, CD79a, CD79B, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, IGLL1, PD1, PDL2, TGF β, APRIL, NKG2D, NKG D ligand, and/or a biotinylated molecule specific for antigen, biotinylated molecule, molecule expressed by HIV, HBV, HCV and/or other pathogens; and/or a neoepitope or neoantigen.
In one embodiment, the transmembrane domain is selected from the transmembrane domains of the following proteins: TCR α chain, TCR β chain, TCR γ chain, TCR δ chain, CD3 δ subunit, CD3 ε subunit, CD3 γ subunit, CD3 δ subunit, CD45, CD4, CD5, CD8 α, CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. Preferably, the transmembrane domain is selected from the transmembrane domains of CD8 α, CD4, CD28 and CD 278.
In one embodiment, the primary signaling domain is an intracellular region of a protein selected from the group consisting of: fcR γ, fcR β, CD3 γ, CD3 δ, CD3 epsilon, CD3 δ, CD22, CD79a, CD79b, and CD66d. Preferably, the primary signaling domain comprises a CD3 ζ intracellular region.
In one embodiment, the co-stimulatory domain comprises one or more intracellular regions of a protein selected from the group consisting of: CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD7, CD8, CD18, CD27, CD28, CD30, CD40, CD54, CD83, CD134 (OX 40), CD137 (4-1 BB), CD270 (HVEM), CD272 (BTLA), CD276 (B7-H3), CD278 (ICOS), CD357 (GITR), DAP10, DAP12, LAT, NKG2 8978 zx8978, PD-1, LIGHT, TRIM, ZAP70 and combinations thereof. Preferably, the co-stimulatory domain is selected from the intracellular domains of CD27, CD28, CD134, CD137, DAP10, DAP12 or CD278 or a combination thereof.
In one embodiment, the immune cell is selected from a T cell, a macrophage, a dendritic cell, a monocyte, an NK cell, or an NKT cell. Preferably, the T cell is a CD4+ CD8+ T cell, a CD4+ helper T cell, a CD8+ T cell, a CD4-CD8-T cell, a tumor infiltrating cell, a memory T cell, a naive T cell, a γ δ -T cell, or an α β -T cell.
In one embodiment, the expression or activity of exogenous CXCL9 and/or IL7 is constitutive expression. In another embodiment, the exogenous CXCL9 and/or IL7 expression or activity is conditional expression. For example, conditional expression can be achieved by operably linking an exogenous gene to an inducible, repressible, or tissue-specific promoter.
In one embodiment, CXCL9 and/or IL7 can be operably linked to a localization domain that can localize the exogenous gene of the invention to a specific cellular location for expression, e.g., a cell membrane. In one embodiment, exogenous genes of the invention, e.g., CXCL9 and/or IL7, are operably linked to a transmembrane domain, thereby anchoring expression at the surface of an engineered immune cell.
In a second aspect, the invention provides a nucleic acid molecule comprising a nucleic acid sequence encoding a cell surface molecule that specifically recognizes an antigen, a nucleic acid sequence encoding CXCL9 and a nucleic acid sequence encoding IL7. Preferably, the cell surface molecule specifically recognizing an antigen is a chimeric antigen receptor, a T cell fusion protein or a T cell antigen coupler, more preferably a chimeric antigen receptor.
The invention also provides a vector comprising the nucleic acid molecule described above. In particular, the vector is selected from the group consisting of plasmids, retroviruses, lentiviruses, adenoviruses, vaccinia viruses, rous Sarcoma Viruses (RSV), polyoma viruses, and adeno-associated viruses (AAV). In some embodiments, the vector further comprises elements such as an origin of autonomous replication in immune cells, a selectable marker, a restriction enzyme cleavage site, a promoter, a poly A tail (polyA), a 3'UTR, a 5' UTR, an enhancer, a terminator, an insulator, an operator, a selectable marker, a reporter gene, a targeting sequence, and/or a protein purification tag. In a specific embodiment, the vector is an in vitro transcription vector.
In one embodiment, the invention also provides a pharmaceutical composition comprising an engineered immune cell, nucleic acid molecule or vector of the invention, and one or more pharmaceutically acceptable excipients.
In a third aspect, the invention also provides a method of treating a subject having cancer, an infection or an autoimmune disease, comprising administering to the subject an effective amount of an immune cell, a nucleic acid molecule, a vector or a pharmaceutical composition according to the invention.
In a fourth aspect, the invention also provides a combination therapy comprising: (1) An engineered immune cell expressing CXCL9 and a composition comprising exogenous IL 7; (2) Engineered immune cells expressing IL7 and compositions comprising exogenous CXCL 9; or (3) an engineered immune cell that expresses a cell surface molecule that specifically recognizes an antigen, and a composition comprising exogenous IL7 and CXCL9.
Drawings
FIG. 1: CAR expression levels of CAR-T cells determined by flow cytometry.
FIG. 2: expression level of IL7 by CAR-T cells determined by ELISA.
FIG. 3: expression level of CXCL9 by CAR-T cells determined by ELISA.
FIG. 4 is a schematic view of: (iii) IFN- γ release levels after co-culture of CAR-T cells with target and non-target cells, respectively.
FIG. 5: weight change profile of mice after treatment of pancreatic cancer in mice with CAR-T cells.
FIG. 6: tumor growth curves in mice after treatment of mouse pancreatic cancer with CAR-T cells.
Detailed Description
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
Cell surface molecules that specifically recognize antigens
In a first aspect, the invention provides a novel engineered immune cell that expresses a cell surface molecule that specifically recognizes an antigen, and exogenous IL7 and CXCL9.
As used herein, the term "cell surface molecule that specifically recognizes an antigen" refers to a molecule expressed on the surface of a cell that is capable of specifically binding to a target molecule (e.g., an antigen). Such surface molecules typically comprise an antigen-binding region capable of specifically binding to an antigen, a transmembrane domain that anchors the surface molecule to the cell surface, and an intracellular domain responsible for signaling. Examples of common such surface molecules include, for example, T Cell Receptors (TCRs), chimeric Antigen Receptors (CARs), T cell fusion proteins (TFPs), or T cell antigen couplers (TACs).
As used herein, the term "T cell receptor" or "TCR" is a characteristic marker of the T cell surface that binds to CD3 in a non-covalent bond to form a complex. Antigen presenting cells present antigenic peptides to T cells via major histocompatibility complex Molecules (MHC) and bind to the TCR complex to induce a series of intracellular signaling. TCRs are composed of six peptide chains that form heterodimers, which are generally classified into α β type and γ δ type. Each peptide chain includes a constant region and a variable region, wherein the variable region is responsible for binding to specific antigens and MHC molecules.
As used herein, the term "chimeric antigen receptor" or "CAR" refers to an artificially constructed hybrid polypeptide generally comprising an antigen binding region (e.g., an antigen-binding portion of an antibody), a transmembrane domain, and an intracellular domain (comprising a costimulatory domain and/or a primary signaling domain) connected by a linker. CARs are able to redirect the specificity and reactivity of T cells and other immune cells to selected targets in a non-MHC-restricted manner using the antigen-binding properties of monoclonal antibodies. non-MHC-restricted antigen recognition gives CAR cells the ability to recognize antigens independent of antigen processing, thus bypassing the major mechanism of tumor escape. Furthermore, when expressed in T cells, the CAR advantageously does not dimerize with the alpha and beta chains of the endogenous T Cell Receptor (TCR).
As used herein, the term "T cell fusion protein" or "TFP" refers to a recombinant polypeptide derived from components of a TCR, typically consisting of TCR subunits and antigen-binding regions linked thereto, and expressed on the cell surface. Wherein the TCR subunit comprises at least a portion of a TCR extracellular domain, a transmembrane domain, a TCR intracellular signaling domain.
As used herein, the term "T cell antigen coupler" or "TAC" includes three functional domains: 1 a tumor targeting domain comprising a single chain antibody, designed ankyrin repeat protein (DARPin), or other targeting group; 2 an extracellular domain, a single chain antibody that binds to CD3, thereby bringing the TAC receptor into proximity with the TCR receptor; 3 transmembrane region and the intracellular region of the CD4 co-receptor, wherein the intracellular region is linked to the protein kinase LCK, catalyzes the phosphorylation of the Immunoreceptor Tyrosine Activation Motif (ITAM) of the TCR complex as an initial step in T cell activation.
As used herein, "antigen binding region" refers to any structure or functional variant thereof that can bind to an antigen. The antigen binding region can be an antibody structure including, but not limited to, monoclonal antibodies, polyclonal antibodies, recombinant antibodies, human antibodies, humanized antibodies, murine antibodies, chimeric antibodies, and functional fragments thereof. For example, antigen binding regions include, but are not limited to, igG, fab ', F (ab ') 2, fd ', fv, scFv, sdFv, linear antibody, single domain antibody, nanobody, diabody, anticalin, DARPIN, and the like, preferably selected from Fab, scFv, sdAb, and nanobody. In the present invention, the antigen binding region may be monovalent or bivalent, and may be a monospecific, bispecific or multispecific antibody. In another embodiment, the antigen binding region can also be a specific binding polypeptide or receptor structure for a particular protein, such as PD1, PDL2, TGF β, APRIL and NKG2D.
The term "functional variant" or "functional fragment" refers to a variant that substantially comprises the amino acid sequence of a parent but contains at least one amino acid modification (i.e., substitution, deletion, or insertion) as compared to the parent amino acid sequence, provided that the variant retains the biological activity of the parent amino acid sequence. In one embodiment, the amino acid modification is preferably a conservative modification.
As used herein, the term "conservative modification" refers to an amino acid modification that does not significantly affect or alter the binding characteristics of an antibody or antibody fragment containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into the chimeric antigen receptors of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are those in which an amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including 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), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine tryptophan, histidine). Conservative modifications may be selected, for example, based on similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
Thus, a "functional variant" or "functional fragment" has at least 75%, preferably at least 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to a parent amino acid sequence and retains the biological activity, e.g., binding activity, of the parent amino acid.
As used herein, the term "sequence identity" refers to the degree to which two (nucleotide or amino acid) sequences have the same residue at the same position in an alignment, and is typically expressed as a percentage. Preferably, identity is determined over the entire length of the sequences being compared. Thus, two copies of an identical sequence have 100% identity. One skilled in the art will recognize that several algorithms can be used to determine sequence identity using standard parameters, such as Blast (Altschul et al (1997) Nucleic Acids Res.25: 3389-3402), blast2 (Altschul et al (1990) J.mol.biol.215: 403-410), smith-Waterman (Smith et al (1981) J.mol.biol.147: 195-197), and ClustalW.
The choice of antigen-binding region depends on the cell surface marker on the target cell to be identified that is associated with a particular disease state, e.g., a tumor-specific antigen or a tumor-associated antigen. Thus, in one embodiment, the antigen binding region of the invention binds to one or more targets selected from the group consisting of: <xnotran> CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33, CD37, CD38, CD40, CD40 3245 zxft 3245 44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2 3732 zxft 3732 40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa , TIM3, TSHR, CD19, BAFF-3963 zxft 3963-1, EGFRvIII, tEGFR, GD2, GD3, BCMA, tn , PSMA, ROR1, FLT3, FAP, TAG72, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, IL-llRa, IL-22Ra, IL-2, , PSCA, PRSS21, VEGFR2, lewisY, PDGFR- β, SSEA-4, AFP, folate α, erbB2 (Her 2/neu), erbB3, erbB4, MUC1, MUC16, EGFR, CS1, NCAM, claudin18.2, c-Met, prostase, PAP, ELF2M, ephrin B2, IGF-I , CAIX, LMP2, gpl00, bcr-abl, , ephA2, fucosyl GMl, sLe, GM3, TGS5, HMWMAA, o- -GD2, folate β, TEM7 4325 zxft 4325 6, GPRC5 3536 zxft 3536 61, ALK, , PLAC1, globoH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6 3926 zxft 3926 51E2, TARP, WT1, NY-ESO-1, LAGE-la, MAGE-A1, MAGE-A3, MAGE-A6, , HPV E6, E7, ETV6-AML, 17, XAGE1, tie 2, MAD-CT-1, MAD-CT-2, fos 1, p53, p53 , PSA, , PCTA-l/Galectin 8, melanA/MARTl, ras , hTERT, , ML-IAP, </xnotran> TMPRSS2 ETS fusion gene, NA17, PAX3, androgen receptor, progesterone receptor, cyclin Bl, MYCN, rhoC, TRP-2, CYP1B 1, BORIS, SART3, PAX5, OY-TES 1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxylesterase, mut hsp70-2, CD79a, CD79B, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, IGLL1, PD1, PDL2, TGF β, APRIL, NKG2D, NKG D ligand, and/or a biotinylated molecule specific for antigen, biotinylated molecule, molecule expressed by HIV, HBV, HCV and/or other pathogens; and/or a neoepitope or neoantigen. Depending on the antigen to be targeted, the CAR of the invention may be designed to include an antigen binding region specific for that antigen. Preferably, the target is selected from the group consisting of CD7, CD19, CD20, CD22, CD30, CD33, CD38, CD123, CD138, CD171, MUC1, AFP, folate receptor alpha, CEA, PSCA, PSMA, her2, EGFR, IL13Ra2, GD2, NKG2D, claudin18.2, ROR1, EGFRvIII, CS1, BCMA, GPRC5D, mesothelin, and any combination thereof. For example, if CD19 is the antigen to be targeted, CD19 antibodies may be used as the antigen binding region of the invention.
As used herein, the term "transmembrane domain" refers to a polypeptide structure that enables a chimeric antigen receptor to be expressed on the surface of an immune cell (e.g., a lymphocyte, NK cell, or NKT cell) and to direct the cellular response of the immune cell against a target cell. The transmembrane domain may be natural or synthetic, and may be derived from any membrane-bound or transmembrane protein. Transmembrane domains particularly suitable for use in the present invention may be derived from, for example, TCR α chain, TCR β chain, TCR γ chain, TCR δ chain, CD3 δ subunit, CD3 ε subunit, CD3 γ subunit, CD3 δ subunit, CD45, CD4, CD5, CD8 α, CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137, CD154 and functional fragments thereof. Alternatively, the transmembrane domain may be synthetic and may contain predominantly hydrophobic residues such as leucine and valine. Preferably, the transmembrane domain is derived from CD28 having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence shown in SEQ ID NO. 3; or from CD8 alpha, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity with the amino acid sequence shown in SEQ ID NO. 4 or 5.
In one embodiment, the chimeric antigen receptor of the present invention may further comprise a hinge region located between the antigen binding region and the transmembrane domain. As used herein, the term "hinge region" generally refers to any oligopeptide or polypeptide that functions to connect a transmembrane domain to an antigen binding region. In particular, the hinge region serves to provide greater flexibility and accessibility to the antigen binding region. 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 native molecule, such as in whole or in part from the extracellular region of CD8, fc γ RIII α receptor, igG4, igG1, CD4 or CD28, or in whole or in part from an antibody constant region. Alternatively, the hinge region may be a synthetic sequence corresponding to a naturally occurring hinge sequence, or may be a fully synthetic hinge sequence. In a preferred embodiment, the hinge region comprises a CD28 hinge having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 15; or comprises a CD8 a hinge having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence as set forth in SEQ ID NO 16 or 17; or an IgG4 hinge having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence depicted in SEQ ID NO 18.
As used herein, the term "intracellular domain" refers to a portion of a protein that transduces effector function signals and directs a cell to a specified function, which comprises a costimulatory domain and/or a primary signaling domain. The intracellular domain is responsible for intracellular signaling after binding of the antigen at the antigen binding region, resulting in activation of the immune cell and immune response.
In one embodiment, the chimeric antigen receptor of the invention comprises a primary signaling domain, which may be the cytoplasmic sequences of the T cell receptor and co-receptor that work together to trigger primary signaling after antigen receptor binding, as well as any derivatives or variants of these sequences and any synthetic sequences with the same or similar function. The primary signaling domain may comprise a number of immunoreceptor tyrosine activation motifs. Non-limiting examples of primary signaling domains of the invention include, but are not limited to, those derived from FcR γ, fcR β, CD3 γ, CD3 δ, CD3 e, CD3 δ, CD22, CD79a, CD79b, and CD66d. In a preferred embodiment, the primary signalling domain of a CAR of the invention may comprise a CD3 ζ intracellular region having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 9, 10 or 11.
In one embodiment, the chimeric antigen receptor of the present invention comprises one or more co-stimulatory domains. The co-stimulatory domain may be an intracellular functional signaling domain from a co-stimulatory molecule, which comprises the entire intracellular portion of the co-stimulatory molecule, or a functional fragment thereof. "costimulatory molecule" refers to a cognate binding partner that specifically binds to a costimulatory ligand on a T cell, thereby mediating a costimulatory response (e.g., 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: CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD7, CD8, CD18, CD27, CD28, CD30, CD40, CD54, CD83, CD134 (OX 40), CD137 (4-1 BB), CD270 (HVEM), CD272 (BTLA), CD276 (B7-H3), CD278 (ICOS), CD357 (GITR), DAP10, DAP12, LAT, NKG2 8978 zx8978, PD-1, LIGHT, TRIFT and ZAP70. Preferably, the co-stimulatory domain of the CAR of the invention is from 4-1BB, CD28, CD27, OX40, ICOS, DAP10, DAP12 or a combination thereof. In one embodiment, the CAR of the invention comprises a CD28 co-stimulatory domain having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6; and/or comprises a 4-1BB co-stimulatory domain having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence depicted in SEQ ID NO 7 or 8.
In one embodiment, the CAR of the invention may further comprise a signal peptide such that when it is expressed in a cell, for example a T cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface. Signal peptides useful in the present invention are well known to those skilled in the art, such as those derived from B2M, CD α, igG1, GM-CSFR α, and the like. In one embodiment, the signal peptide useful in the present invention is a B2M signal peptide having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence shown in SEQ ID NO. 12; or a CD8 alpha signal peptide having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence shown in SEQ ID NO 13 or 14.
Exogenous gene
In addition to cell surface molecules that specifically recognize antigens, the engineered immune cells of the invention also express exogenous IL7 and CXCL9.
CXCL9 is one of the members of the CXC subfamily, is expressed mainly on T cells and NK cells, and plays an important role in autoimmune diseases, tumor therapy, and allogeneic transplantation in the body. CXCL9 has been reported to promote the polarization of effector Th1 and Th17 cells, thereby enhancing immune responses and anti-tumor effects.
In one embodiment, CXCL9 for use in the invention can be wild-type CXCL9, a variant or functional fragment thereof having the same or similar biological function as wild-type CXCL9, or a functional fragment thereof. Specifically, CXCL9 is identical to SEQ ID NO:25 or 27 has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity, or the coding sequence for CXCL9 shares sequence identity with SEQ ID NO:24 or 26 has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity and comparable activity to wild-type CXCL9.
In one embodiment, the IL7 used in the present invention may be a wild-type IL7, a variant or functional fragment thereof having the same or similar biological function as wild-type IL7. Specifically, the peptide has a sequence similar to SEQ ID NO:21 or 23, or the coding sequence for IL7 has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO:22 or 24, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
The expression of exogenous genes in the present invention, such as CXCL9 and/or IL7, may be constitutive expression or conditional expression. In one embodiment, expression of exogenous CXCL9 and/or IL7 is conditional. For example, the exogenous gene of the present invention may be operably linked to an inducible, repressible or tissue-specific promoter, as desired, to regulate the expression level of the introduced exogenous gene at a particular time or in a particular tissue, cell type. In one embodiment, the promoter is an inducible promoter, i.e., a promoter that initiates transcription only in the presence of a particular environmental condition, developmental condition, or inducer. In another embodiment, the promoter is a repressible promoter, i.e., expression of the foreign gene in the cell is inhibited or not expressed in the presence of a repressor specific for the repressible promoter.
In one embodiment, CXCL9 and/or IL7 can be operably linked to a localization domain that can localize the exogenous gene of the invention for expression at a particular cellular location, e.g., cell membrane, etc. Localization domains include, but are not limited to, nuclear localization signals, leader peptides, transmembrane domains, and the like. In one embodiment, the exogenous genes CXCL9 and/or IL7 of the invention are operably linked to a transmembrane domain, thereby anchoring surface expression in engineered immune cells.
Nucleic acids and vectors
The invention also provides a nucleic acid molecule comprising a nucleic acid sequence encoding a cell surface molecule that specifically recognizes an antigen, a nucleic acid sequence encoding CXCL9 and a nucleic acid sequence encoding IL7.
In one embodiment, the cell surface molecule specifically recognizing an antigen is a T cell receptor or a chimeric antigen receptor, preferably a chimeric antigen receptor. The definition of chimeric antigen receptor is as described above.
As used herein, the term "nucleic acid molecule" includes sequences of ribonucleotides and deoxyribonucleotides, such as modified or unmodified RNA or DNA, each in linear or circular form in single-and/or double-stranded form, or mixtures thereof (including hybrid molecules). Thus, nucleic acids according to the invention include DNA (such as dsDNA, ssDNA, cDNA), RNA (such as dsRNA, ssRNA, mRNA, ivtRNA), combinations or derivatives thereof (such as PNA). Preferably, the nucleic acid is DNA or RNA, more preferably mRNA.
The invention also provides a vector comprising a nucleic acid according to the invention. Wherein the nucleic acid sequence encoding a cell surface molecule specifically recognizing the antigen, the nucleic acid sequence encoding CXCL9 and the nucleic acid sequence encoding IL7 can be located in one or more vectors. When in a vector, the nucleic acid sequences may be operably linked by a 2A peptide.
As used herein, the term "vector" is a vector nucleic acid molecule used as a vehicle for transferring (foreign) genetic material into a host cell where it can, for example, be replicated and/or expressed.
Vectors generally include targeting vectors and expression vectors. A "targeting vector" is a medium for delivering an isolated nucleic acid to the interior of a cell, for example, by homologous recombination or by using a hybrid recombinase that targets sequences at a site specifically. An "expression vector" is a vector for the transcription of heterologous nucleic acid sequences (such as those encoding the chimeric antigen receptor polypeptides of the invention) in a suitable host cell and the translation of their mRNA. Suitable carriers for use in the present invention are known in the art and many are commercially available. In one embodiment, vectors of the invention include, but are not limited to, plasmids, viruses (e.g., retroviruses, oncolytic viruses, lentiviruses, adenoviruses, vaccinia viruses, rous sarcoma viruses, polyoma viruses, and adeno-associated viruses (AAV), etc.), bacteriophages, phagemids, cosmids, and artificial chromosomes (including BAC and YACs). The vector itself is usually a nucleotide sequence, usually a DNA sequence comprising an insert (transgene) and a larger sequence that serves as the "backbone" of the vector. Engineered vectors typically also contain 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 (e.g., a multiple cloning site, MCS). The vector may additionally comprise elements such as a promoter, poly A tail (polyA), 3' UTR, enhancer, terminator, insulator, operator, selectable marker, reporter gene, targeting sequence and/or protein purification tag. In a specific embodiment, the vector is an in vitro transcription vector.
Engineered immune cells
The invention also provides an engineered immune cell comprising a nucleic acid or vector of the invention. In other words, the engineered immune cells of the invention express a cell surface molecule that specifically recognizes an antigen, exogenous IL7 and CXCL9.
As used herein, the term "immune cell" refers to any cell of the immune system that has one or more effector functions (e.g., cytotoxic cell killing activity, secretion of cytokines, induction of ADCC and/or CDC). For example, the immune cell may be a T cell, macrophage, dendritic cell, monocyte, NK cell and/or NKT cell, or an immune cell obtained from a stem cell source such as iPSC, ESC or the like. Preferably, the immune cell is a T cell. The T cell may be any T cell, such as an in vitro cultured T cell, e.g., a primary T cell, or a T cell from an in vitro cultured T cell line, e.g., jurkat, supT1, etc., or a T cell 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 the site of infection, ascites, pleural effusion, spleen tissue, and tumors. T cells may also be concentrated or purified. The T cells may be at any developmental stage, including, but not limited to, CD4+ CD8+ T cells, CD4+ helper T cells (e.g., th1 and Th2 cells), CD8+ T cells (e.g., cytotoxic T cells), CD4-CD8-T cells, tumor infiltrating cells, memory T cells, naive T cells, γ δ -T cells, α β -T cells, and the like. In a preferred embodiment, the immune cell is a human T cell. T cells can be obtained from the blood of a subject using a variety of techniques known to those skilled in the art, such as Ficoll isolation.
In one embodiment, the immune cell of the invention further comprises suppressed or silenced expression of at least one endogenous gene selected from the group consisting of: CD52, GR, TCR alpha, TCR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD247 delta, HLA-I, HLA-II, B2M, immune checkpoint genes such as PD1, CTLA-4, LAG3 and TIM3. More particularly, expression of at least a TCR component (including TCR α, TCR β genes) or a CD3 component (including CD3 γ, CD3 δ, CD3 epsilon, CD247 ζ) in the immune cell is inhibited or silenced. This strategy is particularly useful for avoiding graft versus host disease (GvHD). Methods of inhibiting or silencing a gene are known in the art, e.g., by mediating DNA cleavage by meganucleases, zinc finger nucleases, TALEN nucleases or Cas enzymes in CRISPR systems, thereby knocking out the gene; or by shRNA, RNAi, or the like.
Pharmaceutical compositions and combination therapies
The invention also provides a pharmaceutical composition comprising the engineered immune cell, nucleic acid molecule or vector of the invention as an active agent, and one or more pharmaceutically acceptable excipients. Thus, the invention also encompasses the use of said nucleic acid molecule, vector or engineered immune cell for the preparation of a pharmaceutical composition.
As used herein, the term "pharmaceutically acceptable excipient" refers to carriers and/or excipients that are pharmacologically and/or physiologically compatible with the subject and active ingredient (i.e., capable of eliciting a desired therapeutic effect without causing any undesirable local or systemic effects) and are well known in the art (see, e.g., remington's pharmaceutical sciences. Edited by Gennaro AR,19th ed. Pennsylvania mack Publishing company, 1995). Examples of pharmaceutically acceptable excipients include, but are not limited to, fillers, binders, disintegrants, coatings, adsorbents, anti-adherents, glidants, antioxidants, flavoring agents, colorants, sweeteners, solvents, co-solvents, buffers, chelating agents, surfactants, diluents, wetting agents, preservatives, emulsifiers, coating agents, isotonizing agents, absorption delaying agents, stabilizers, and tonicity adjusting agents. The selection of suitable excipients to prepare the desired pharmaceutical compositions of the present invention is known to those skilled in the art. Exemplary excipients for use in the pharmaceutical compositions of the present invention include saline, buffered saline, dextrose, and water. In general, the selection of suitable excipients depends, inter alia, on the active agent used, the disease to be treated and the desired dosage form of the pharmaceutical composition.
The pharmaceutical composition according to the present invention may be suitable for administration by various routes. Typically, administration is accomplished parenterally. Methods of parenteral delivery include topical, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.
The pharmaceutical compositions according to the invention may also be administered in combination with one or more other agents suitable for the treatment and/or prevention of the diseases to be treated.
The present invention also provides a combination therapy comprising: (1) An engineered immune cell expressing CXCL9 and a composition comprising exogenous IL 7; (2) Engineered immune cells expressing IL7 and compositions comprising exogenous CXCL 9; or (3) an engineered immune cell that expresses a cell surface molecule that specifically recognizes an antigen, and a composition comprising exogenous IL7 and CXCL9.
Therapeutic applications
The invention also provides a method of treating a subject having cancer, an infection, or an autoimmune disease, comprising administering to the subject an effective amount of a nucleic acid molecule, vector, engineered immune cell, or pharmaceutical composition according to the invention. Thus, the invention also encompasses the use of said nucleic acid molecule, vector, engineered immune cell for the preparation of a medicament for the treatment of cancer, infection or autoimmune disease.
In one embodiment, the method of treatment comprises administering to a subject an effective amount of an immune cell and/or pharmaceutical composition of the invention.
In one embodiment, the immune cell is an autologous or allogeneic cell, preferably a T cell, macrophage, dendritic cell, monocyte, NK cell and/or NKT cell, more preferably a T cell, NK cell or NKT cell.
As used herein, the term "autologous" means that any material derived from an individual will be reintroduced into the same individual at a later time. As used herein, the term "allogeneic" refers to any material derived from a different animal or patient of the same species as the individual into which the material is introduced. When the genes at one or more loci are different, two or more individuals are considered allogeneic to each other. In some cases, genetic differences in allogenic material from individuals of the same species may be sufficient for antigen interactions to occur.
As used herein, the term "subject" is a mammal. The mammal may be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples. Mammals other than humans can be advantageously used as subjects representing animal models of cancer. Preferably, the subject is a human.
In one embodiment, the cancer is a cancer associated with expression of a target to which an antigen binding region binds, such as a hematologic tumor or a solid tumor. For example, the cancer includes, but is not limited to: <xnotran> , , , , , , , , CNS , , , , , , , , , , , , ( ), (GBM), , , , , , , ( , , ), ( ), , , , ( , , ), , , , , , , , , , , , , , , , , B ( / (NHL), (SL) NHL, / NHL, NHL, NHL, NHL, NHL, NHL), B (B-LBL), , AIDS , Waldenstrom , (CLL), </xnotran> Acute Lymphocytic Leukemia (ALL), B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), B-cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell tumor, burkitt's lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, chronic Myelogenous Leukemia (CML), malignant lymphoproliferative disease, MALT lymphoma, hairy cell leukemia, marginal zone lymphoma, multiple myeloma, myelodysplasia, plasmacytic lymphoma, pre-leukemic, plasmacytoid dendritic cell tumor, and post-transplant lymphoproliferative disorder (PTLD); and other diseases associated with target expression. Preferably, the diseases that can be treated with the engineered immune cells or the pharmaceutical compositions of the invention are selected from: leukemia, lymphoma, multiple myeloma, brain glioma, pancreatic cancer, gastric cancer, liver cancer, breast cancer, esophageal cancer, thyroid cancer, prostate cancer, bone cancer, lung cancer, etc.
In one embodiment, the infection includes, but is not limited to, infections caused by viruses, bacteria, fungi, and parasites.
In one embodiment, the autoimmune disease includes, but is not limited to, type I diabetes, celiac disease, graves 'disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, addison's disease, sjogren's syndrome, hashimoto's thyroiditis, myasthenia gravis, vasculitis, pernicious anemia, and systemic lupus erythematosus, among others.
In one embodiment, the method further comprises administering to the subject one or more additional chemotherapeutic agents, biologies, drugs or treatments. In this embodiment, the chemotherapeutic agent, biological agent, drug or treatment is selected from the group consisting of radiation therapy, surgery, antibody agents and/or small molecules and any combination thereof.
The invention will be described in detail below with reference to the accompanying drawings and examples. It should be noted that the drawings and their embodiments of the present invention are for illustrative purposes only and are not to be construed as limiting the invention. The embodiments and features of the embodiments in the present application may be combined with each other without contradiction.
Detailed Description
Example 1 preparation of CAR-T cells
1.1 construction of retroviral plasmids
MSCV-mCD19-CAR plasmid was constructed comprising the coding sequences for CD19-scFv (SEQ ID NO: 2), CD8 a hinge region (SEQ ID NO: 17), CD8 a transmembrane region (SEQ ID NO: 5), 41BB costimulatory domain (SEQ ID NO: 7) and CD3 ζ intracellular region (SEQ ID NO: 11).
MSCV-mCD19-CAR-IL7 plasmid was constructed, which further comprises the coding sequence of T2A (SEQ ID NO: 19) and IL7 (SEQ ID NO: 23) on the basis of MSCV-mCD19-CAR plasmid.
MSCV-mCD19-CAR-CXCL9 plasmid was constructed, which further comprises the coding sequence of T2A (SEQ ID NO: 19) and CXCL9 (SEQ ID NO: 25) on the basis of MSCV-mCD19-CAR plasmid.
1.2. Preparation of retrovirus
In T175 flasks at 30X 10 6 Density of Individual cells/flask 293T cells were seeded in 30ml DMEM medium containing 10% foetal calf serum at 37 ℃ 5% 2 The cells were cultured overnight in an incubator for virus packaging.
To a sterile tube, 3ml of Opti-MEM (Gibco, cat # 31985-070), 45. Mu.g of the prepared retroviral plasmid and 15. Mu.g of the packaging vector pCL-Eco (Shanghai cereal noon Biotech Co., ltd., cat # P3029) were added. Then 120 mu l X-treme GENE HP DNA transfection reagent (Roche, cat # 06366236001) was addedI.e., mixed well and incubated at room temperature for 15min. The plasmid/vector/transfection reagent mixture was then added dropwise to a pre-prepared 293T cell culture flask at 37 ℃ 5% 2 Incubated under conditions overnight. The culture was collected 72 hours after transfection, and centrifuged (2000 g,4 ℃,10 minutes) to obtain a retrovirus supernatant.
1.3 preparation of CAR-T cells
T lymphocytes were isolated from mouse spleen and CTS with DynaBeads CD3/CD28 TM (Gibco, cat # 40203D) activates T cells, then CO at 37 ℃ and 5% 2 The culture was performed for 1 day.
At a rate of 3X 10 per hole 6 Density of individual cells/mL activated T cells were seeded into 24-well plates previously coated overnight with RetroNectin, then 500 μ L of retroviral supernatant was added and complete medium was supplemented to 2mL.
The 24-well plate was placed in a centrifuge for centrifugation infection at 2000g for 2h at 32 ℃. Then, the 24-well plate was immediately placed in a CO2 incubator at 37 ℃ for static culture. The next day, the fresh medium was changed and the cell density was adjusted to 1X 10 6 Individual cells/mL. Three days after infection, cells were collected for subsequent analysis. The collected cells are mCD19-CAR cells, mCD19-CAR + CXCL9 cells, mCD19-CAR + IL7 cells and mCD19-CAR + IL7+ CXCL9 cells.
Example 2 detection of expression of CAR-T cells
2.1 expression levels of cell surface CAR
2X 10 of the product prepared in example 1 were taken out 5 Individual CAR-T cells, treated with Goat Anti-Rat IgG (H)&L) Biotin (BioVision, cat # 6910-250) as a primary antibody and APC Streptavidin (BD Pharmingen, cat # 554067) as a secondary antibody, the expression level of CAR on CAR T cells was examined by flow cytometry, and the results are shown in FIG. 1. It can be seen that CAR was efficiently expressed in all CAR-T cells compared to untreated NT cells.
2.2 Expression level of IL7
The supernatant of CAR-T cells was collected and the level of IL7 secretion in the cells was determined using the Mouse IL-7DuoSet ELISA kit (R & D Systems, cat # DY 407) according to the manufacturer's recommendations and the results are shown in FIG. 2. It can be seen that both mCD19-CAR + IL7 cells and mCD19-CAR + IL7+ CXCL9 cells can efficiently express IL7.
2.3 Expression level of CXCL9
The supernatant of CAR-T cells was collected and the level of CXCL9 secretion in the cells was measured using the Mouse CXCL9 DuoSet ELISA kit (R & D Systems, cat # DY 479) according to the manufacturer's recommendations and the results are shown in figure 3. It can be seen that both mCD19-CAR + CXCL9 cells and mCD19-CAR + IL7+ CXCL9 cells can efficiently express CXCL9.
Example 3 detection of IFN- γ secretion levels in CAR-T cells
In 96-hole round bottom plate with 2X 10 5 NT cells, CD19-CAR cells, mCD19-CAR + IL7 cells, mCD19-CAR + CXCL9 cells and mCD19-CAR + IL7+ CXCL9 cells were added at a concentration of 100. Mu.l each. Then 1X 10 in each well 4 At a concentration of 100. Mu.l of each cell, either Panc02-mCD19 target cells or Panc02 non-target cells were added. After incubation at 37 ℃ for 24h, culture supernatants were collected. According to the manufacturer's recommendations, use the Mouse IFN-gamma DuoSet ELISA kit (R)&D, cat No. DY 485) the expression level of IFN-. Gamma.in the culture supernatant was determined.
The results of the detection are shown in FIG. 4. It can be seen that no IFN- γ release was detected in non-target cells Panc02, but only significantly elevated IFN- γ levels were detected after co-culture with target cells Panc02-CD19, and that the NT cells did not express IFN- γ, indicating that the killing of CAR-T cells in this example is specific. In addition, the level of IFN- γ was significantly higher for mCD19-CAR + CXCL9 cells and mCD19-CAR + IL7 cells than for CD19-CAR cells, indicating that both the additionally expressed IL7 and CXCL9 genes alone significantly increased the killing activity of CAR-T cells. Furthermore, the inventors have also unexpectedly found that CAR-T cells expressing the IL7+ CXCL9 combination have significantly higher IFN- γ levels than CAR-T cells expressing either CXCL9 or IL7 alone, indicating that IL7 and CXCL9 can produce a synergistic effect, further enhancing the killing activity of CAR-T cells.
Example 4 demonstration of tumor-inhibiting Effect of CAR-T cells
In healthy C57BL/6 miceThe axillary part of the left forelimb is inoculated with 5 multiplied by 10 under the skin 5 And the Panc02-mCD19 pancreatic cancer cells. Mice inoculated with pancreatic cancer cells were randomly divided into 3 groups of 5 mice each. When the tumor volume grows to 100mm 3 At the same time, 1X 10 injections were administered into each group of mice via the tail vein 6 Individual NT cells, mCD19-CAR + IL7 cells, mCD19-CAR + CXCL9 cells or mCD19-CAR + IL7+ CXCL9 cells. Mice were monitored for changes in body weight and tumor volume until the end of the experiment.
The body weight changes of the mice are shown in fig. 5. It can be seen that there was no significant difference in body weight of mice in each group compared to the control group after CAR-T cell administration, indicating that CAR-T cell administration did not have a significant toxic side effect on the mice.
Changes in tumor volume in mice are shown in figure 6. It can be seen that expression of IL7 alone can enhance the tumor-suppressing effect of CAR-T cells, while CXCL9 alone has a tumor-suppressing effect comparable to that of conventional CAR-T cells, indicating that CXCL9 does not promote CAR-T cells as well as IL7. Furthermore, the inventors have also unexpectedly found that the in vivo anti-tumor effect of mCD19-CAR + IL7+ CXCL9 cells is significantly better than that of CAR-T cells expressing only IL7 or CXCL9, indicating that the additionally expressed IL7+ CXCL9 can produce a synergistic effect with CAR-T cells, enhancing the anti-tumor effect of the latter.
The above results indicate that the combination of co-expressing CXCL9 and IL7 is effective in enhancing the inhibitory effect of CAR-expressing engineered immune cells on tumor cells.
It should be noted that the above-mentioned embodiments are merely preferred examples of the present invention, and the present invention is not limited thereto. It will be understood by those skilled in the art that any modification, equivalent replacement, or improvement made without departing from the spirit and principle of the present invention shall fall within the protection scope of the present invention.
Sequence listing
<110> Nanjing Beijing Heng Biotechnology Ltd
<120> engineered immune cells and uses thereof
<130> BHCN46
<160> 27
<170> SIPOSequenceListing 1.0
<210> 1
<211> 242
<212> PRT
<213> Artificial Sequence(Artificial Sequence)
<220>
<223> CD19 scFv
<400> 1
Asp Ile Gln Met Thr Gln Thr Thr Ser Ser Leu Ser Ala Ser Leu Gly
1 5 10 15
Asp Arg Val Thr Ile Ser Cys Arg Ala Ser Gln Asp Ile Ser Lys Tyr
20 25 30
Leu Asn Trp Tyr Gln Gln Lys Pro Asp Gly Thr Val Lys Leu Leu Ile
35 40 45
Tyr His Thr Ser Arg Leu His Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Tyr Ser Leu Thr Ile Ser Asn Leu Glu Gln
65 70 75 80
Glu Asp Ile Ala Thr Tyr Phe Cys Gln Gln Gly Asn Thr Leu Pro Tyr
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Thr Gly Gly Gly Gly Ser
100 105 110
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Glu Val Lys Leu Gln Glu
115 120 125
Ser Gly Pro Gly Leu Val Ala Pro Ser Gln Ser Leu Ser Val Thr Cys
130 135 140
Thr Val Ser Gly Val Ser Leu Pro Asp Tyr Gly Val Ser Trp Ile Arg
145 150 155 160
Gln Pro Pro Arg Lys Gly Leu Glu Trp Leu Gly Val Ile Trp Gly Ser
165 170 175
Glu Thr Thr Tyr Tyr Asn Ser Ala Leu Lys Ser Arg Leu Thr Ile Ile
180 185 190
Lys Asp Asn Ser Lys Ser Gln Val Phe Leu Lys Met Asn Ser Leu Gln
195 200 205
Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Ala Lys His Tyr Tyr Tyr Gly
210 215 220
Gly Ser Tyr Ala Met Asp Tyr Trp Gly Gln Gly Thr Ser Val Thr Val
225 230 235 240
Ser Ser
<210> 2
<211> 238
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCD19 scFv
<400> 2
Asp Ile Gln Met Thr Gln Ser Pro Ala Ser Leu Ser Thr Ser Leu Gly
1 5 10 15
Glu Thr Val Thr Ile Gln Cys Gln Ala Ser Glu Asp Ile Tyr Ser Gly
20 25 30
Leu Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ser Pro Gln Leu Leu Ile
35 40 45
Tyr Gly Ala Ser Asp Leu Gln Asp Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Gln Tyr Ser Leu Lys Ile Thr Ser Met Gln Thr
65 70 75 80
Glu Asp Glu Gly Val Tyr Phe Cys Gln Gln Gly Leu Thr Tyr Pro Arg
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Leu Lys Gly Gly Gly Gly Ser
100 105 110
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Glu Val Gln Leu Gln Gln
115 120 125
Ser Gly Ala Glu Leu Val Arg Pro Gly Thr Ser Val Lys Leu Ser Cys
130 135 140
Lys Val Ser Gly Asp Thr Ile Thr Phe Tyr Tyr Met His Phe Val Lys
145 150 155 160
Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile Gly Arg Ile Asp Pro Glu
165 170 175
Asp Glu Ser Thr Lys Tyr Ser Glu Lys Phe Lys Asn Lys Ala Thr Leu
180 185 190
Thr Ala Asp Thr Ser Ser Asn Thr Ala Tyr Leu Lys Leu Ser Ser Leu
195 200 205
Thr Ser Glu Asp Thr Ala Thr Tyr Phe Cys Ile Tyr Gly Gly Tyr Tyr
210 215 220
Phe Asp Tyr Trp Gly Gln Gly Val Met Val Thr Val Ser Ser
225 230 235
<210> 3
<211> 27
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD28 transmembrane Domain
<400> 3
Phe Trp Val Leu Val Val Val Gly Gly Val Leu Ala Cys Tyr Ser Leu
1 5 10 15
Leu Val Thr Val Ala Phe Ile Ile Phe Trp Val
20 25
<210> 4
<211> 25
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD8 alpha transmembrane domain
<400> 4
Ile Tyr Ile Trp Ala Pro Leu Ala Gly Thr Cys Gly Val Leu Leu Leu
1 5 10 15
Ser Leu Val Ile Thr Leu Tyr Cys Lys
20 25
<210> 5
<211> 21
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCD8 a transmembrane domain
<400> 5
Ile Trp Ala Pro Leu Ala Gly Ile Cys Val Ala Leu Leu Leu Ser Leu
1 5 10 15
Ile Ile Thr Leu Ile
20
<210> 6
<211> 41
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD28 Co-stimulatory Domain
<400> 6
Arg Ser Lys Arg Ser Arg Leu Leu His Ser Asp Tyr Met Asn Met Thr
1 5 10 15
Pro Arg Arg Pro Gly Pro Thr Arg Lys His Tyr Gln Pro Tyr Ala Pro
20 25 30
Pro Arg Asp Phe Ala Ala Tyr Arg Ser
35 40
<210> 7
<211> 40
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> 4-1BB Co-stimulatory Domain
<400> 7
Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met Arg
1 5 10 15
Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe Pro
20 25 30
Glu Glu Glu Glu Gly Gly Cys Glu
35 40
<210> 8
<211> 42
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> m4-1BB co-stimulatory domain
<400> 8
Arg Lys Lys Phe Pro His Ile Phe Lys Gln Pro Phe Lys Lys Thr Thr
1 5 10 15
Gly Ala Ala Gln Glu Glu Asp Ala Cys Ser Cys Arg Cys Pro Gln Glu
20 25 30
Glu Glu Gly Gly Gly Gly Gly Tyr Glu Leu
35 40
<210> 9
<211> 114
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> intracellular domain of CD3 ζ
<400> 9
Leu Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Gln Gln
1 5 10 15
Gly Gln Asn Gln Leu Phe Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu
20 25 30
Phe Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly
35 40 45
Lys Pro Gln Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu
50 55 60
Gln Lys Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly
65 70 75 80
Glu Arg Arg Arg Gly Lys Gly His Asp Gly Leu Phe Gln Gly Leu Ser
85 90 95
Thr Ala Thr Lys Asp Thr Phe Asp Ala Leu His Met Gln Ala Leu Pro
100 105 110
Pro Arg
<210> 10
<211> 113
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> intracellular domain of CD3 ζ
<400> 10
Leu Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Gln Gln
1 5 10 15
Gly Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu
20 25 30
Tyr Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly
35 40 45
Lys Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln
50 55 60
Lys Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu
65 70 75 80
Arg Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr
85 90 95
Ala Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro
100 105 110
Arg
<210> 11
<211> 109
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCD3 ζ intracellular domain
<400> 11
Ser Arg Ser Ala Glu Thr Ala Ala Asn Leu Gln Asp Pro Asn Gln Leu
1 5 10 15
Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr Asp Val Leu Glu
20 25 30
Lys Lys Arg Ala Arg Asp Pro Glu Met Gly Gly Lys Gln Gln Arg Arg
35 40 45
Arg Asn Pro Gln Glu Gly Val Tyr Asn Ala Leu Gln Lys Asp Lys Met
50 55 60
Ala Glu Ala Tyr Ser Glu Ile Gly Thr Lys Gly Glu Arg Arg Arg Gly
65 70 75 80
Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala Thr Lys Asp
85 90 95
Thr Tyr Asp Ala Leu His Met Gln Thr Leu Ala Pro Arg
100 105
<210> 12
<211> 20
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> B2M Signal peptide
<400> 12
Met Ser Arg Ser Val Ala Leu Ala Val Leu Ala Leu Leu Ser Leu Ser
1 5 10 15
Gly Leu Glu Ala
20
<210> 13
<211> 21
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD8 alpha signal peptide
<400> 13
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro
20
<210> 14
<211> 24
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCD8 alpha signal peptide
<400> 14
Met Ala Ser Pro Leu Thr Arg Phe Leu Ser Leu Asn Leu Leu Leu Leu
1 5 10 15
Gly Glu Ser Ile Ile Leu Gly Ser
20
<210> 15
<211> 39
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD28 hinge region
<400> 15
Ile Glu Val Met Tyr Pro Pro Pro Tyr Leu Asp Asn Glu Lys Ser Asn
1 5 10 15
Gly Thr Ile Ile His Val Lys Gly Lys His Leu Cys Pro Ser Pro Leu
20 25 30
Phe Pro Gly Pro Ser Lys Pro
35
<210> 16
<211> 45
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> CD8 alpha hinge region
<400> 16
Thr Thr Thr Pro Ala Pro Arg Pro Pro Thr Pro Ala Pro Thr Ile Ala
1 5 10 15
Ser Gln Pro Leu Ser Leu Arg Pro Glu Ala Cys Arg Pro Ala Ala Gly
20 25 30
Gly Ala Val His Thr Arg Gly Leu Asp Phe Ala Cys Asp
35 40 45
<210> 17
<211> 45
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCD8 α hinge region
<400> 17
Thr Thr Thr Lys Pro Val Leu Arg Thr Pro Ser Pro Val His Pro Thr
1 5 10 15
Gly Thr Ser Gln Pro Gln Arg Pro Glu Asp Cys Arg Pro Arg Gly Ser
20 25 30
Val Lys Gly Thr Gly Leu Asp Phe Ala Cys Asp Ile Tyr
35 40 45
<210> 18
<211> 12
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> IgG4 hinge region
<400> 18
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5 10
<210> 19
<211> 18
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> T2A
<400> 19
Glu Gly Arg Gly Ser Leu Leu Thr Cys Gly Asp Val Glu Glu Asn Pro
1 5 10 15
Gly Pro
<210> 20
<211> 402
<212> DNA
<213> Artificial sequence(Artificial Sequence)
<220>
<223> hIL-7
<400> 20
atgttccatg tttcttttag gtatatcttt ggacttcctc ccctgatcct tgttctgttg 60
ccagtagcat catctgattg tgatattgaa ggtaaagatg gcaaacaata tgagagtgtt 120
ctaatggtca gcatcgatca attattggac agcatgaaag aaattggtag caattgcctg 180
aataatgaat ttaacttttt taaaagacat atctgtgatg ctaataaggt taaaggaaga 240
aaaccagctg ccctgggtga agcccaacca acaaagagtt tggaagaaaa taaatcttta 300
aaggaacaga aaaaactgaa tgacttgtgt ttcctaaaga gactattaca agagataaaa 360
acttgttgga ataaaatttt gatgggcact aaagaacact ga 402
<210> 21
<211> 133
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> hIL-7
<400> 21
Met Phe His Val Ser Phe Arg Tyr Ile Phe Gly Leu Pro Pro Leu Ile
1 5 10 15
Leu Val Leu Leu Pro Val Ala Ser Ser Asp Cys Asp Ile Glu Gly Lys
20 25 30
Asp Gly Lys Gln Tyr Glu Ser Val Leu Met Val Ser Ile Asp Gln Leu
35 40 45
Leu Asp Ser Met Lys Glu Ile Gly Ser Asn Cys Leu Asn Asn Glu Phe
50 55 60
Asn Phe Phe Lys Arg His Ile Cys Asp Ala Asn Lys Val Lys Gly Arg
65 70 75 80
Lys Pro Ala Ala Leu Gly Glu Ala Gln Pro Thr Lys Ser Leu Glu Glu
85 90 95
Asn Lys Ser Leu Lys Glu Gln Lys Lys Leu Asn Asp Leu Cys Phe Leu
100 105 110
Lys Arg Leu Leu Gln Glu Ile Lys Thr Cys Trp Asn Lys Ile Leu Met
115 120 125
Gly Thr Lys Glu His
130
<210> 22
<211> 465
<212> DNA
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mIL-7
<400> 22
atgttccatg tttcttttag atatatcttt ggaattcctc cactgatcct tgttctgctg 60
cctgtcacat catctgagtg ccacattaaa gacaaagaag gtaaagcata tgagagtgta 120
ctgatgatca gcatcgatga attggacaaa atgacaggaa ctgatagtaa ttgcccgaat 180
aatgaaccaa acttttttag aaaacatgta tgtgatgata caaaggaagc tgcttttcta 240
aatcgtgctg ctcgcaagtt gaagcaattt cttaaaatga atatcagtga agaattcaat 300
gtccacttac taacagtatc acaaggcaca caaacactgg tgaactgcac aagtaaggaa 360
gaaaaaaacg taaaggaaca gaaaaagaat gatgcatgtt tcctaaagag actactgaga 420
gaaataaaaa cttgttggaa taaaattttg aagggcagta tataa 465
<210> 23
<211> 154
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mIL-7
<400> 23
Met Phe His Val Ser Phe Arg Tyr Ile Phe Gly Ile Pro Pro Leu Ile
1 5 10 15
Leu Val Leu Leu Pro Val Thr Ser Ser Glu Cys His Ile Lys Asp Lys
20 25 30
Glu Gly Lys Ala Tyr Glu Ser Val Leu Met Ile Ser Ile Asp Glu Leu
35 40 45
Asp Lys Met Thr Gly Thr Asp Ser Asn Cys Pro Asn Asn Glu Pro Asn
50 55 60
Phe Phe Arg Lys His Val Cys Asp Asp Thr Lys Glu Ala Ala Phe Leu
65 70 75 80
Asn Arg Ala Ala Arg Lys Leu Lys Gln Phe Leu Lys Met Asn Ile Ser
85 90 95
Glu Glu Phe Asn Val His Leu Leu Thr Val Ser Gln Gly Thr Gln Thr
100 105 110
Leu Val Asn Cys Thr Ser Lys Glu Glu Lys Asn Val Lys Glu Gln Lys
115 120 125
Lys Asn Asp Ala Cys Phe Leu Lys Arg Leu Leu Arg Glu Ile Lys Thr
130 135 140
Cys Trp Asn Lys Ile Leu Lys Gly Ser Ile
145 150
<210> 24
<211> 381
<212> DNA
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCXCL9
<400> 24
atgaagtccg ctgttctttt cctcttgggc atcatcttcc tggagcagtg tggagttcga 60
ggaaccctag tgataaggaa tgcacgatgc tcctgcatca gcaccagccg aggcacgatc 120
cactacaaat ccctcaaaga cctcaaacag tttgccccaa gccccaattg caacaaaact 180
gaaatcattg ctacactgaa gaacggagat caaacctgcc tagatccgga ctcggcaaat 240
gtgaagaagc tgatgaaaga atgggaaaag aagatcagcc aaaagaaaaa gcaaaagagg 300
gggaaaaaac atcaaaagaa catgaaaaac agaaaaccca aaacacccca aagtcgtcgt 360
cgttcaagga agactacata a 381
<210> 25
<211> 126
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCXCL9
<400> 25
Met Lys Ser Ala Val Leu Phe Leu Leu Gly Ile Ile Phe Leu Glu Gln
1 5 10 15
Cys Gly Val Arg Gly Thr Leu Val Ile Arg Asn Ala Arg Cys Ser Cys
20 25 30
Ile Ser Thr Ser Arg Gly Thr Ile His Tyr Lys Ser Leu Lys Asp Leu
35 40 45
Lys Gln Phe Ala Pro Ser Pro Asn Cys Asn Lys Thr Glu Ile Ile Ala
50 55 60
Thr Leu Lys Asn Gly Asp Gln Thr Cys Leu Asp Pro Asp Ser Ala Asn
65 70 75 80
Val Lys Lys Leu Met Lys Glu Trp Glu Lys Lys Ile Ser Gln Lys Lys
85 90 95
Lys Gln Lys Arg Gly Lys Lys His Gln Lys Asn Met Lys Asn Arg Lys
100 105 110
Pro Lys Thr Pro Gln Ser Arg Arg Arg Ser Arg Lys Thr Thr
115 120 125
<210> 26
<211> 378
<212> DNA
<213> Artificial sequence(Artificial Sequence)
<220>
<223> hCXCL9
<400> 26
atgaagaaaa gtggtgttct tttcctcttg ggcatcatct tgctggttct gattggagtg 60
caaggaaccc cagtagtgag aaagggtcgc tgttcctgca tcagcaccaa ccaagggact 120
atccacctac aatccttgaa agaccttaaa caatttgccc caagcccttc ctgcgagaaa 180
attgaaatca ttgctacact gaagaatgga gttcaaacat gtctaaaccc agattcagca 240
gatgtgaagg aactgattaa aaagtgggag aaacaggtca gccaaaagaa aaagcaaaag 300
aatgggaaaa aacatcaaaa aaagaaagtt ctgaaagttc gaaaatctca acgttctcgt 360
caaaagaaga ctacataa 378
<210> 27
<211> 125
<212> PRT
<213> Artificial sequence(Artificial Sequence)
<220>
<223> mCXCL9
<400> 27
Met Lys Lys Ser Gly Val Leu Phe Leu Leu Gly Ile Ile Leu Leu Val
1 5 10 15
Leu Ile Gly Val Gln Gly Thr Pro Val Val Arg Lys Gly Arg Cys Ser
20 25 30
Cys Ile Ser Thr Asn Gln Gly Thr Ile His Leu Gln Ser Leu Lys Asp
35 40 45
Leu Lys Gln Phe Ala Pro Ser Pro Ser Cys Glu Lys Ile Glu Ile Ile
50 55 60
Ala Thr Leu Lys Asn Gly Val Gln Thr Cys Leu Asn Pro Asp Ser Ala
65 70 75 80
Asp Val Lys Glu Leu Ile Lys Lys Trp Glu Lys Gln Val Ser Gln Lys
85 90 95
Lys Lys Gln Lys Asn Gly Lys Lys His Gln Lys Lys Lys Val Leu Lys
100 105 110
Val Arg Lys Ser Gln Arg Ser Arg Gln Lys Lys Thr Thr
115 120 125

Claims (21)

1. An engineered immune cell expressing a cell surface molecule that specifically recognizes an antigen and exogenous IL7 and CXCL9.
2. The engineered immune cell of claim 1, wherein the CXCL9 is identical to SEQ ID NO:25 or 27, or a coding sequence thereof which is at least 90% identical to the amino acid sequence set forth in SEQ ID NO:24 or 26 have at least 90% identity; wherein said IL7 is substantially identical to SEQ ID NO:21 or 23, or a coding sequence thereof which shares at least 90% identity with the amino acid sequence set forth in SEQ ID NO:22 or 24 have at least 90% identity.
3. The engineered immune cell of any one of claims 1-2, wherein the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor, a T cell fusion protein, or a T cell antigen coupler.
4. The engineered immune cell of claim 3, wherein the chimeric antigen receptor comprises an antigen binding region, a transmembrane domain, and an intracellular domain comprising a costimulatory domain and/or a primary signaling domain.
5. The engineered immune cell of claim 4, wherein the antigen binding region is selected from the group consisting of IgG, fab ', F (ab ') 2, fd ', fv, scFv, sdFv, linear antibody, single domain antibody, nanobody, diabody, anticalin, and DARPIN antigen.
6. The engineered immune cell of claim 4, wherein the antigen-antigen binding region binds to one or more targets selected from the group consisting of: <xnotran> CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33, CD37, CD38, CD40, CD40 3245 zxft 3245 44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2 3732 zxft 3732 40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa , TIM3, TSHR, CD19, BAFF-3963 zxft 3963-1, EGFRvIII, tEGFR, GD2, GD3, BCMA, tn , PSMA, ROR1, FLT3, FAP, TAG72, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, IL-llRa, IL-22Ra, IL-2, , PSCA, PRSS21, VEGFR2, lewisY, PDGFR- β, SSEA-4, AFP, folate α, erbB2 (Her 2/neu), erbB3, erbB4, MUC1, MUC16, EGFR, CS1, NCAM, claudin18.2, c-Met, prostase, PAP, ELF2M, ephrin B2, IGF-I , CAIX, LMP2, gpl00, bcr-abl, , ephA2, fucosyl GMl, sLe, GM3, TGS5, HMWMAA, o- -GD2, folate β, TEM7 4325 zxft 4325 6, GPRC5 3536 zxft 3536 61, ALK, , PLAC1, globoH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6 3926 zxft 3926 51E2, TARP, WT1, NY-ESO-1, LAGE-la, MAGE-A1, MAGE-A3, MAGE-A6, , HPV E6, E7, ETV6-AML, 17, XAGE1, tie 2, MAD-CT-1, MAD-CT-2, fos 1, p53, p53 , PSA, , PCTA-l/Galectin 8, melanA/MARTl, ras , hTERT, , ML-IAP, </xnotran> TMPRSS2 ETS fusion gene, NA17, PAX3, androgen receptor, progesterone receptor, cyclin Bl, MYCN, rhoC, TRP-2, CYP1B 1, BORIS, SART3, PAX5, OY-TES 1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxylesterase, mut hsp70-2, CD79a, CD79B, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, IGLL1, PD1, PDL2, TGF β, APRIL, NKG2D, NKG D ligand, and/or a biotinylated molecule specific for antigen, biotinylated molecule, molecule expressed by HIV, HBV, HCV and/or other pathogens; and/or a neoepitope or neoantigen.
7. The engineered immune cell of claim 4, wherein the transmembrane domain is selected from the transmembrane domains of the following proteins: TCR α chain, TCR β chain, TCR γ chain, TCR δ chain, CD3 δ subunit, CD3 epsilon subunit, CD3 γ subunit, CD3 δ subunit, CD45, CD4, CD5, CD8 α, CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137 and CD154.
8. The engineered immune cell of any one of claims 4-7, wherein the primary signaling domain is selected from the intracellular regions of: fcR γ, fcR β, CD3 γ, CD3 δ, CD3 e, CD3 δ, CD22, CD79a, CD79b, and CD66d.
9. The engineered immune cell of claim 4, wherein the co-stimulatory domain comprises one or more intracellular regions of a protein selected from the group consisting of: CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD7, CD8, CD18, CD27, CD28, CD30, CD40, CD54, CD83, CD134 (OX 40), CD137 (4-1 BB), CD270 (HVEM), CD272 (BTLA), CD276 (B7-H3), CD278 (ICOS), CD357 (GITR), DAP10, DAP12, LAT, NKG2 8978 zx8978, PD-1, LIGHT, TRIM or ZAP70.
10. The engineered immune cell of any one of claims 1-9, wherein the immune cell is selected from a T cell, a macrophage, a dendritic cell, a monocyte, an NK cell, or an NKT cell.
11. The engineered immune cell of claim 10, wherein the T cell is a CD4+ CD8+ T cell, a CD4+ helper T cell, a CD8+ T cell, a CD4-CD8-T cell, a tumor infiltrating cell, a memory T cell, a naive T cell, a γ δ -T cell, or an α β -T cell.
12. The engineered immune cell of any one of claims 1-11, wherein the expression of CXCL9 and/or IL7 is conditional or constitutive expression.
13. The engineered immune cell of any one of claims 1-12, wherein the CXCL9 and/or IL7 is operably linked to a localization domain.
14. A nucleic acid molecule comprising a nucleic acid sequence encoding a cell surface molecule that specifically recognizes an antigen, a nucleic acid sequence encoding CXCL9 and a nucleic acid sequence encoding IL7.
15. The nucleic acid molecule of claim 14, wherein the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor, a T cell fusion protein, or a T cell antigen coupler.
16. A vector comprising the nucleic acid molecule of any one of claims 14-15.
17. The vector of claim 16, wherein the vector is selected from the group consisting of a plasmid, a retrovirus, a lentivirus, an adenovirus, a vaccinia virus, a Rous Sarcoma Virus (RSV), a polyoma virus, and an adeno-associated virus (AAV).
18. A pharmaceutical composition comprising an engineered immune cell according to any one of claims 1-13, a nucleic acid molecule according to any one of claims 14-15 or a vector according to any one of claims 16-17, and one or more pharmaceutically acceptable excipients.
19. Use of the engineered immune cell of any one of claims 1-13, the nucleic acid molecule of any one of claims 14-15, the vector of any one of claims 16-17, or the pharmaceutical composition of claim 18 in the manufacture of a medicament for treating a subject having cancer, an infection, or an autoimmune disease.
20. The use of claim 19, wherein the cancer is a hematological tumor or a solid tumor.
21. A combination therapy comprising: (1) An engineered immune cell expressing CXCL9 and a composition comprising exogenous IL 7; (2) Engineered immune cells expressing IL7 and compositions comprising exogenous CXCL 9; or (3) engineered immune cells and a composition comprising exogenous IL7 and CXCL 9; wherein the engineered immune cell expresses a cell surface molecule that specifically recognizes an antigen.
CN202111060954.8A 2021-09-10 2021-09-10 Engineered immune cells and uses thereof Pending CN115786268A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202111060954.8A CN115786268A (en) 2021-09-10 2021-09-10 Engineered immune cells and uses thereof
PCT/CN2022/114484 WO2023035947A1 (en) 2021-09-10 2022-08-24 Engineered immune cell and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111060954.8A CN115786268A (en) 2021-09-10 2021-09-10 Engineered immune cells and uses thereof

Publications (1)

Publication Number Publication Date
CN115786268A true CN115786268A (en) 2023-03-14

Family

ID=85417074

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111060954.8A Pending CN115786268A (en) 2021-09-10 2021-09-10 Engineered immune cells and uses thereof

Country Status (2)

Country Link
CN (1) CN115786268A (en)
WO (1) WO2023035947A1 (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110272873A (en) * 2018-03-15 2019-09-24 北京卡替医疗技术有限公司 A kind of immune cell therapy drug comprising chemotactic type neoantigen identity T cell
CN113286874A (en) * 2018-10-12 2021-08-20 美商生物细胞基因治疗有限公司 Chimeric Antigen Receptor (CARs) compositions and methods of use
BR112022007952A2 (en) * 2019-10-28 2022-10-04 Noile Immune Biotech Inc DRUG TO TREAT CANCER, COMBINATION DRUG, DRUG COMPOSITION, IMMUNE RESPONSIVE CELL, NUCLEIC ACID DELIVERY VEHICLE AND PRODUCT
CN112063588A (en) * 2020-08-13 2020-12-11 南京北恒生物科技有限公司 Engineered immune cells and uses thereof
CN112625142B (en) * 2021-02-03 2023-05-09 郑州大学第一附属医院 CXCL9 modified CAR-T structure and application thereof

Also Published As

Publication number Publication date
WO2023035947A1 (en) 2023-03-16

Similar Documents

Publication Publication Date Title
US20240009311A1 (en) Engineered immune cell and use thereof
EP3025719B1 (en) Combination immunotherapy of antigen-recognizing receptors and hematopoietic cells for the treatment of diseases
CN111849910B (en) Engineered immune cells and uses thereof
CN112029001B (en) Chimeric antigen receptors targeting NK activating receptors
CN113321737A (en) Novel chimeric antigen receptor and use thereof
CN111849913B (en) Engineered immune cells and uses thereof
CN114853900A (en) Novel chimeric antigen receptor and use thereof
WO2022223049A1 (en) Engineered immune cell and use thereof
EP4194472A1 (en) Chimeric antigen receptor comprising novel co-stimulatory domain and use thereof
CN115786268A (en) Engineered immune cells and uses thereof
WO2023051361A1 (en) Engineered immune cell and use thereof
WO2023071811A1 (en) Engineered immune cell and use thereof
WO2023083003A1 (en) Engineered immune cell and use thereof
WO2023025009A1 (en) Engineered immune cell and use thereof
WO2023016284A1 (en) Engineered immune cell and use thereof
WO2021004400A1 (en) Immune cell expressing cd3 antibody-receptor complex and use thereof
CN115785279A (en) Chimeric antigen receptor comprising novel costimulatory domains and uses thereof
CN115521917A (en) Engineered immune cells and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination