WO2023155852A1 - 经修饰的免疫效应细胞及其用途 - Google Patents

经修饰的免疫效应细胞及其用途 Download PDF

Info

Publication number
WO2023155852A1
WO2023155852A1 PCT/CN2023/076567 CN2023076567W WO2023155852A1 WO 2023155852 A1 WO2023155852 A1 WO 2023155852A1 CN 2023076567 W CN2023076567 W CN 2023076567W WO 2023155852 A1 WO2023155852 A1 WO 2023155852A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
seq
acid sequence
sequence shown
domain
Prior art date
Application number
PCT/CN2023/076567
Other languages
English (en)
French (fr)
Inventor
赵阳兵
朱庚振
刘晓军
Original Assignee
上海优替济生生物医药有限公司
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 上海优替济生生物医药有限公司 filed Critical 上海优替济生生物医药有限公司
Publication of WO2023155852A1 publication Critical patent/WO2023155852A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • 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
    • 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

Definitions

  • This application relates to the field of biomedicine, in particular to a modified immune effector cell and its application.
  • T lymphocytes can be engineered to express T cell receptors (TCRs) [1-3] or chimeric antigen receptors (CARs) [4,5] that recognize tumor antigens to kill tumors for the treatment of cancer and other disease.
  • T lymphocytes engineered with CARs specific for B cell markers such as CD19 have shown tremendous clinical responses in hematological malignancies, effective immunotherapy in solid tumors has proven challenging, mainly due to Immune evasion caused by the complex and dynamic tumor microenvironment (TME) induces hypofunction and exhaustion of T cells, limiting the anti-tumor immune response [6] .
  • TAE complex and dynamic tumor microenvironment
  • CD28 is considered the most prominent co-stimulatory molecule for optimal T cell clonal expansion, differentiation and effector function. CD28 engagement lowers T cell activation threshold and leads to enhancement of TCR signaling events, which contributes to potent cytokine production (through enhanced transcriptional activity and stabilization of messenger RNA), cell cycle progression, survival, regulation of metabolism and T cell responses are necessary. This is because CD28 is a key player in the organization of the immunological synapse (IS), which reinforces intimate contacts between T cells and APCs.
  • IS immunological synapse
  • T cells can be introduced into T cells genetically to enhance their effector function, persistence, and antitumor activity [8-10] .
  • T cells expressing co-stimulatory receptors or ligands are expected to function poorly in neoplastic lesions. Insufficient [11] .
  • the PD1-CD28 switch receptor a fusion protein consisting of the extracellular portion of PD1 and the intracellular portion of CD28, enhances T cell function both in vitro and in animal models of tumors [12-16] .
  • the introduction of PD1-CD28 chimeric switch receptors may be the key to rescue T cells from A more efficient and effective approach to tumor immunosuppression [17,18] .
  • T-ALL T-cell acute lymphoblastic leukemia
  • CAR chimeric antigen receptor
  • CD7 overexpression in T-cell malignancies may be an attractive target for T-ALL immunotherapy.
  • its presence on normal T cells means that expression of the CD7 CAR on these cells is not feasible, as it would prove cannibalistic. Therefore, it is urgent to develop safe and effective autologous CD7-CAR T cells for the treatment of T-ALL and AML.
  • T-ALL T-cell acute lymphoblastic leukemia
  • CAR chimeric antigen receptor
  • CD7 overexpression in T-cell malignancies may be an attractive target for T-ALL immunotherapy.
  • its presence on normal T cells means that expression of the CD7 CAR on these cells is not feasible, as it would prove cannibalistic.
  • Whether to design and prepare sgRNA that precisely and specifically targets the CD7 gene has become a key technology for CRISPR/cas9 to specifically knock out the CD7 gene. Therefore, it is urgent to develop safe and effective autologous CD7-CAR T cells for the treatment of T-ALL and AML.
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • peats highly conserved repeats
  • spacers spacers
  • the length of the repeats is usually 21-48bp, and the repeats are separated by 26-72bp spacers separated.
  • CRISPR specifically edits target sequences through complementary recognition of crRNA and tracrRNA and target sequences.
  • tracrRNA and crRNA are expressed as a chimeric guide RNA (single guide RNA, sgRNA), and the CRISPR-Cas9 system is simplified into two components of Cas9 protein and sgRNA, making the CRISPR-Cas9 system simple to construct, high in efficiency, Low cost and other advantages make it the most suitable choice for genome editing technology.
  • sgRNA single guide RNA
  • Circular RNA is a class of end-to-end non-coding RNA produced by special alternative splicing. circRNA is a covalently closed circular structure without 5' and 3' polarity, and cannot be degraded by ribonucleases. Studies have confirmed that circRNA plays an important role in the occurrence and development of diseases. Some circRNAs can be used as translation templates, and some circRNAs can affect the expression of parental genes through cis or trans effects.
  • the present application provides a modified immune effector cell expressing a set of chimeric protein molecules called lymphocyte Cell-APCs co-stimulators (Lymphocytes-APCs Co-stimulators, LACO-Stim), including: a. Membrane fusion protein composed of extracellular CD40 ligand (CD40L) and intracellular CD28, or fusion protein targeting intracellular CD28 scFv of CD40; b. soluble fusion protein consisting of extracellular CD40L and scFv against CD28; c. bispecific antibody against CD40 and CD28.
  • a Membrane fusion protein composed of extracellular CD40 ligand (CD40L) and intracellular CD28, or fusion protein targeting intracellular CD28 scFv of CD40
  • b soluble fusion protein consisting of extracellular CD40L and scFv against CD28
  • c bispecific antibody against CD40 and CD28.
  • LACO-stim molecules When co-introduced into human T lymphocytes with CARs or TCRs, LACO-stim molecules showed strong effects on increasing T cell anti-tumor function, stimulating and maturing APCs, macrophages, and myeloid-derived cells.
  • T cells expressing LACO-stimulatory molecules have the ability to overcome the inhibitory effects of TEM, such as that of PD1/PD-L1, Treg, and TGF- ⁇ .
  • LACO-stim can coordinate the interaction between T lymphocytes and APCs, promote the epitope diffusion ability of APCs, and further enhance the antitumor activity.
  • the application provides a modified immune effector cell comprising a nucleic acid molecule encoding a fusion protein comprising a first domain that activates an antigen presenting cell (APC) and a second domain that activates an immune effector cell domains, wherein (i) the first domain comprises (a) an activating receptor ligand or receptor binding fragment thereof that binds APC, or (b) an activating receptor antibody or an antigen binding fragment thereof that binds APC; and (ii) The second domain comprises (a) a costimulatory ligand or receptor-binding fragment thereof of an immune effector cell, (b) an antibody or antigen-binding fragment thereof that binds a costimulatory receptor of an immune effector cell, or (c) an immune effector cell co-stimulatory receptors or functional fragments thereof.
  • APC antigen presenting cell
  • first domain and the second domain are linked by a linker.
  • the linker comprises a peptide linker
  • the antibody or antigen-binding fragment thereof is a scFv.
  • the APCs are selected from the group consisting of dendritic cells, macrophages, myeloid-derived suppressor cells, monocytes, B cells, T cells, and Langerhans cells.
  • the activating receptor of APC is selected from CD40, CD80, CD86, CD91, DEC-205 and DC-SIGN.
  • the first domain comprises ligands that bind CD40, CD80, CD86, CD91, DEC-205, DC-SIGN or receptor binding fragments thereof.
  • the first domain comprises a receptor binding fragment of CD40 ligand (CD40L).
  • said first domain comprises an antibody that binds to said APC activating receptor or Antigen-binding fragments.
  • the first knot domain is an anti-CD40 antibody or an antigen-binding fragment thereof.
  • the immune effector cells are selected from the group consisting of T cells, NK cells, NKT cells, macrophages, neutrophils and granulocytes.
  • the second domain comprises an intracellular domain of a costimulatory receptor.
  • co-stimulatory receptor is selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 and CD43.
  • co-stimulatory receptor is CD28 or 4-1BB.
  • the second domain is a co-stimulatory ligand for immune effector cells or a receptor-binding fragment thereof.
  • the costimulatory ligand is selected from CD58, CD70, CD83, CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL- 4R, GITR-L, TIM-4, CD153, CD48, CD160, CD200R and CD44.
  • the second domain is an antibody or antigen-binding fragment thereof that binds a co-stimulatory receptor.
  • co-stimulatory receptor is selected from CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 and CD43.
  • co-stimulatory receptor is CD28 or 4-1BB.
  • first domain and the second domain are selected from the following combinations:
  • the first domain comprises CD40L or a receptor binding fragment thereof, and the second domain comprises a CD28 or 4-1BB intracellular domain;
  • the first domain comprises CD40L or a receptor binding fragment thereof, and the second domain comprises an anti-CD28 antibody or an antigen binding fragment thereof;
  • the first domain comprises an anti-CD40 antibody or antigen-binding fragment thereof, and the second domain comprises a CD28 intracellular domain;
  • said first domain comprises an anti-CD40 antibody or antigen-binding fragment thereof
  • the second domain comprises an anti-CD28 antibody or antigen-binding fragment thereof
  • said first domain comprises CD40L or a receptor binding fragment thereof and the second domain comprises a CD28 costimulatory ligand or receptor binding fragment thereof;
  • said first domain comprises an anti-CD40 antibody or antigen-binding fragment thereof and the second domain comprises CD28 Costimulatory ligands or receptor-binding fragments thereof.
  • the fusion protein comprises the amino acid sequence shown in any one of SEQ ID NO:234 to SEQ ID NO:250.
  • the immune effector cells comprise engineered immune effector cells.
  • the engineered immune effector cells include CAR-T cells, CAR-NK or TCR-T cells.
  • the modified immune effector cells have reduced surface expression of CD7 and express an anti-CD7 CAR compared to corresponding immune cells.
  • the engineered immune effector cells include anti-CD7 CAR-T cells.
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • the expression of CD7 in the modified immune effector cells is absent or suppressed.
  • the immune effector cells do not induce cannibalism.
  • the first domain comprises (a) an activating receptor ligand or a receptor-binding fragment thereof that binds APC, or (b) an activating receptor antibody or an antigen-binding fragment thereof that binds APC
  • the second domain comprises (a) a costimulatory ligand or receptor-binding fragment thereof of an immune effector cell, (b) an antibody or antigen-binding fragment thereof that binds a costimulatory receptor of an immune effector cell, or (c ) costimulatory receptors or functional fragments thereof of immune effector cells.
  • the expression of CD7 in the CAR-T cells is absent or suppressed.
  • the CAR-T cells do not induce cannibalism.
  • the present application provides a pharmaceutical combination comprising immune effector cells and a fusion protein
  • the fusion protein comprises a first domain for activating antigen-presenting cells (APC) and a second domain for activating immune effector cells
  • the first domain comprises (a) an activating receptor ligand or receptor binding fragment thereof that binds APC, or (b) an activating receptor antibody or an antigen binding fragment thereof that binds APC
  • a second Domains include (a) costimulatory ligands or receptor-binding fragments thereof for immune effector cells, or (b) antibodies or antigen-binding fragments thereof that bind costimulatory receptors for immune effector cells.
  • first domain and the second domain are linked by a linker.
  • the linker comprises a peptide linker
  • the antibody or antigen-binding fragment thereof is a scFv.
  • the APCs are selected from the group consisting of dendritic cells, macrophages, myeloid-derived suppressor cells, monocytes, B cells, T cells, and Langerhans cells.
  • the activating receptor of APC is selected from CD40, CD80, CD86, CD91, DEC-205 and DC-SIGN.
  • the first domain comprises ligands that bind CD40, CD80, CD86, CD91, DEC-205, DC-SIGN or receptor binding fragments thereof.
  • the first domain comprises a receptor binding fragment of CD40 ligand (CD40L).
  • said first domain comprises an antibody or antigen-binding fragment thereof that binds to said activated receptor of APC.
  • the immune effector cells are selected from the group consisting of T cells, NK cells, NKT cells, macrophages, neutrophils and granulocytes.
  • the second domain comprises an intracellular domain of a costimulatory receptor.
  • co-stimulatory receptor is selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 and CD43.
  • co-stimulatory receptor is CD28.
  • the second domain is a co-stimulatory ligand for immune effector cells or a receptor-binding fragment thereof.
  • the costimulatory ligand is selected from CD58, CD70, CD83, CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL- 4R, GITR-L, TIM-4, CD153, CD48, CD160, CD200R and CD44.
  • the second domain is an antibody or antigen-binding fragment thereof that binds a co-stimulatory receptor.
  • co-stimulatory receptor is selected from CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 and CD43.
  • co-stimulatory receptor is CD28 or 4-1BB.
  • first domain and the second domain are selected from the following combinations:
  • the first domain comprises CD40L or a receptor binding fragment thereof
  • the second domain comprises CD28 or 4-1BB costimulatory ligand or a receptor binding fragment thereof
  • the first domain comprises CD40L or a receptor binding fragment thereof, and the second domain comprises an anti-CD28 antibody or an antigen binding fragment thereof;
  • said first domain comprises an anti-CD40 antibody or antigen-binding fragment thereof, and the second domain comprises a CD28 costimulatory ligand or receptor-binding fragment thereof;
  • said first domain comprises an anti-CD40 antibody or antigen-binding fragment thereof and the second domain comprises an anti-CD28 antibody or antigen-binding fragment thereof.
  • the fusion protein comprises the amino acid sequence shown in any one of SEQ ID NO:237 to SEQ ID NO:241.
  • the immune effector cells comprise engineered immune effector cells.
  • the engineered immune effector cells include CAR-T cells, CAR-NK or TCR-T cells.
  • the pharmaceutical combination reduces surface expression of CD7 and expresses an anti-CD7 CAR compared to corresponding immune cells.
  • the engineered immune effector cells include anti-CD7 CAR-T cells.
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • the immune effector cells do not induce cannibalism.
  • the present application provides a pharmaceutical composition, which comprises the modified immune effector cells described in the present application or the pharmaceutical combination described in the present application, and a pharmaceutically acceptable carrier.
  • the present application provides the use of the modified immune effector cells described in the present application, the pharmaceutical combination described in the present application or the pharmaceutical composition described in the present application in the preparation of medicines, and the medicines are used to treat tumors.
  • the tumor includes hematological tumors and solid tumors.
  • the tumor comprises a CD7 expressing tumor.
  • the tumor comprises a CD7-positive hematologic malignancy.
  • the tumor comprises a T cell malignancy.
  • T-cell malignancy comprises acute T-lymphoblastic leukemia (T-ALL), acute myeloid leukemia, or NK/T-cell lymphoma.
  • T-ALL acute T-lymphoblastic leukemia
  • NK/T-cell lymphoma NK/T-cell lymphoma
  • the present application provides a method for treating tumors, the method comprising administering the modified immune effector cells described in the present application, the drug combination described in the present application, or the drug combination described in the present application to a subject in need thereof. said pharmaceutical composition.
  • the present application provides a method for killing malignant T cells, the method comprising combining the malignant T cells with the modified immune effector cells described in the present application, the drug combination described in the present application, or the combination of the drugs described in the present application. contact with the pharmaceutical composition described above.
  • the present application provides a method for preparing a chimeric antigen receptor T (CAR-T) cell population, wherein the CAR targets CD7, comprising the following steps:
  • CD7 is absent or suppressed in the modified population of T cells compared to the corresponding unmodified cells.
  • said modification comprises administering to said T cell population one or more substances selected from the group consisting of antisense RNA, siRNA, shRNA, transcription activator-like effector nuclease (TALEN) , zinc finger nuclease (ZFN) and CRISPR/Cas system.
  • antisense RNA siRNA, shRNA
  • transcription activator-like effector nuclease TALEN
  • ZFN zinc finger nuclease
  • CRISPR/Cas system CRISPR/Cas system
  • wherein said modification comprises administering a CRISPR/Cas system to said population of T cells.
  • wherein said modification comprises administering a CRISPR/Cas9 system to said population of T cells.
  • said modification comprises administering Cas9 and a gRNA targeting the CD7 gene to said T cell population.
  • the gRNA targeting the CD7 gene comprises the nucleotide sequence shown in any one of SEQ ID NO:211 to SEQ ID NO:218.
  • nucleic acid molecule of the CD7 CAR nucleic acid molecule and the fusion protein is mRNA.
  • nucleic acid molecule of the CD7 CAR nucleic acid molecule and the fusion protein is a circRNA.
  • the cirRNA chimeric antigen receptor comprises elements arranged in the following order: an internal ribosome entry site (IRES) element, a protein coding sequence targeting CD7, and polyadenylic acid (polyA),
  • the circRNA fusion protein contains elements arranged in the following order: internal ribosome entry site (IRES) element, fusion protein coding sequence, and polyadenylic acid (polyA).
  • the transducing the anti-CD7 CAR and the fusion protein into the T cell comprises introducing the nucleic acid molecule encoding the anti-CD7 CAR and the nucleic acid molecule encoding the fusion protein into the T cell.
  • the present application provides a cell population comprising the modified immune effector cells described in the present application or the CAR-T cells described in the present application, wherein the cell population is derived from peripheral blood mononuclear cells (PBMC), Peripheral blood leukocytes (PBL), tumor-infiltrating lymphocytes (TIL), cytokine-induced killer cells (CIK), lymphokine-activated killer cells (LAK), or bone marrow-infiltrating lymphocytes (MIL).
  • PBMC peripheral blood mononuclear cells
  • PBL Peripheral blood leukocytes
  • TIL tumor-infiltrating lymphocytes
  • CIK cytokine-induced killer cells
  • LAK lymphokine-activated killer cells
  • MIL bone marrow-infiltrating lymphocytes
  • the present application provides an antigen-binding protein that specifically binds to CD7 protein.
  • the present application provides a CD7-targeted chimeric antigen receptor and CAR-T cells.
  • the present application provides a CD7-targeting CAR-T cell that avoids self-killing and its use.
  • an isolated antigen-binding protein which specifically binds to a CD7 protein, wherein the isolated antigen-binding protein comprises a heavy chain variable region (VH), and the VH comprises HCDR1, HCDR2 and HCDR3, wherein HCDR1 Comprising the amino acid sequence shown in SEQ ID NO:1.
  • VH heavy chain variable region
  • HCDR1 comprises the amino acid sequence shown in any one of SEQ ID NO:2 to SEQ ID NO:9.
  • HCDR2 comprises the amino acid sequence shown in any one of SEQ ID NO:10 to SEQ ID NO:17.
  • HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 18 or SEQ ID NO: 27.
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18 the amino acid sequence of;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:3
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:11
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:19; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:20; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:4, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:12, and the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:21; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:22; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:6, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:14, and the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:23; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:24; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:25; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:8, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:16, and the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:26; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:27.
  • the isolated antigen-binding protein comprises VH, wherein the VH includes framework regions HFR1, HFR2, HFR3 and HFR4, and the C-terminus of HFR1 is directly or indirectly connected to the N-terminus of HCDR1.
  • Linked, and the HFR1 comprises the amino acid sequence shown in any one of SEQ ID NO:28 to SEQ ID NO:38.
  • the HFR2 is located between the HCDR1 and the HCDR2, and the HFR2 comprises the amino acid sequence shown in any one of SEQ ID NO:39 to SEQ ID NO:47.
  • the HFR3 is located between the HCDR2 and the HCDR3, and the HFR3 comprises the amino acid sequence shown in any one of SEQ ID NO:48 to SEQ ID NO:56.
  • the N-terminal of the HFR4 is directly or indirectly connected to the C-terminal of the HCDR3, and the HFR4 comprises amino acids shown in any one of SEQ ID NO:57 to SEQ ID NO:60 sequence.
  • the isolated antigen-binding protein comprises VH, wherein the VH includes framework regions HFR1, HFR2, HFR3 and HFR4, and the C-terminus of HFR1 is directly or indirectly connected to the N-terminus of HCDR1.
  • the HFR2 is located between the HCDR1 and the HCDR2
  • the HFR3 is located between the HCDR2 and the HCDR3
  • the N-terminal of the HFR4 is directly or indirectly connected to the C-terminal of the HCDR3;
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:28
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:48
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 57; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:29
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:40
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:49
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:30
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:48
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR2 includes the amino acid sequence shown in SEQ ID NO:41
  • the HFR3 includes the amino acid sequence shown in SEQ ID NO:50
  • HFR4 includes the amino acid sequence shown in SEQ ID NO:59 the amino acid sequence shown; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:32
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:42
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:51
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:33
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:48
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:34
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:48
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:35
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:43
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:52
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:28, and the HFR2 comprises SEQ ID NO:39
  • the amino acid sequence shown includes the amino acid sequence shown in SEQ ID NO:48
  • HFR4 includes the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:30
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:44
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:53
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:36
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:45
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:54
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 60; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:37
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:46
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:55
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 60; or
  • the HFR1 comprises the amino acid sequence shown in SEQ ID NO:38
  • the HFR2 comprises the amino acid sequence shown in SEQ ID NO:47
  • the HFR3 comprises the amino acid sequence shown in SEQ ID NO:56
  • HFR4 comprises the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 58.
  • the isolated antigen-binding protein comprises VH, wherein the VH comprises the amino acid sequence shown in SEQ ID NO:61 to SEQ ID NO:73.
  • said isolated antigen-binding protein comprises VL, wherein said VL comprises LCDR1, LCDR2, LCDR3, said LCDR1 comprising any one of SEQ ID NO:74 to SEQ ID NO:86 The amino acid sequence shown.
  • said isolated antigen binding protein comprises VL, wherein said VL comprises LCDR1, LCDR2, LCDR3, said LCDR2 comprising any one of SEQ ID NO:87 to SEQ ID NO:98 The amino acid sequence shown.
  • the isolated antigen-binding protein comprises a VL, wherein the VL comprises LCDR1, LCDR2, LCDR3, and the LCDR3 comprises any one of SEQ ID NO:99 to SEQ ID NO:111 The amino acid sequence shown.
  • the isolated antigen-binding protein comprises a VL, wherein the VL comprises LCDR1, LCDR2 and LCDR3, the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:74, and the LCDR2 comprises SEQ ID NO:74 The amino acid sequence shown in ID NO:87, the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:99; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:75
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:88
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:100;
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:76
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:89
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:101; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:77
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:90
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:102; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:78
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:91
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:103; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:79
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:104; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:80
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:93
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:105; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:81
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:94
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:106; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:82
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:95
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:107; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:84
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:97
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:109; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:85
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:110; or
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:86
  • the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:111.
  • said isolated antigen binding protein comprises VH and VL, wherein said VH comprises HCDR1, HCDR2 and HCDR3, said VL comprises LCDR1, LCDR2 and LCDR3; wherein said HCDR1 comprises SEQ ID NO The amino acid sequence shown in: 2, 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: 18, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO: 74 Amino acid sequence, the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:87, and the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:99; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:3, and the HCDR2 comprises the SEQ ID NO:
  • the amino acid sequence shown in 11 the HCDR3 includes the amino acid sequence shown in SEQ ID NO:19
  • the LCDR1 includes the amino acid sequence shown in SEQ ID NO:75
  • the LCDR2 includes the amino acid sequence shown in SEQ ID NO:88 Sequence
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO: 100; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:20
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:20
  • the amino acid sequence shown in ID NO:76, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:89, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:101; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:4, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:12, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:21, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:21
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:22
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:22
  • the amino acid sequence shown in ID NO:91, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:91
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:103;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:79, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:104; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:80, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:93, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:105; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:6, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:14, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:23, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:23
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprising the amino acid sequence shown in SEQ ID NO:82
  • the LCDR2 comprising the amino acid sequence shown in SEQ ID NO:95
  • the LCDR3 comprising the amino acid sequence shown in SEQ ID NO:107; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:24
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:24
  • the amino acid sequence shown in ID NO:83, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:96
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:108;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:25
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:25
  • the amino acid sequence shown in ID NO:84, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:97
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:109;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:8, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:16, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:26, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:26
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:27
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:27
  • the amino acid sequence shown in ID NO:86, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:111.
  • said isolated antigen binding protein comprises VL, wherein said VL comprises framework regions LFR1, LFR2, LFR3 and LFR4, wherein the C-terminus of said LFR1 is directly or directly to the N-terminus of said LCDR1 Indirectly connected, and the LFR1 comprises the amino acid sequence shown in any one of SEQ ID NO:112 to SEQ ID NO:123.
  • the isolated antigen binding protein comprises a VL, wherein the VL comprises a framework region LFR2, wherein the LFR2 is located between the LCDR1 and the LCDR2, and the LFR2 comprises SEQ ID The amino acid sequence shown in any one of NO:124 to SEQ ID NO:130.
  • the isolated antigen binding protein comprises a VL, wherein the VL comprises a framework region LFR3, the LFR3 is located between the LCDR2 and the LCDR3, and the LFR3 comprises SEQ ID NO : 131 to the amino acid sequence shown in any one of SEQ ID NO: 141.
  • said isolated antigen binding protein comprises a VL, wherein said VL comprises a framework Region LFR4, the N-terminal of the LFR4 is directly or indirectly connected to the C-terminal of the LCDR3, and the LFR4 comprises the amino acid sequence shown in any one of SEQ ID NO:142 to SEQ ID NO:147.
  • said isolated antigen binding protein comprises VL, wherein said VL comprises framework regions LFR1, LFR2, LFR3 and LFR4, wherein the C-terminus of said LFR1 is directly or directly to the N-terminus of said LCDR1 Indirectly connected, the LFR2 is located between the LCDR1 and the LCDR2, the LFR3 is located between the LCDR2 and the LCDR3, and the N-terminal of the LFR4 is directly or indirectly connected to the C-terminal of the LCDR3; wherein
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:112
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:124
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:131
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:131 The amino acid sequence shown in ID NO:142;
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:113
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:125
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:132
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:132.
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:114
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:133
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:133 The amino acid sequence shown in ID NO:142; or
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:115
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:127
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:134
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:134.
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:116
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:125
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:13
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:135.
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:117
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:136
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:136 The amino acid sequence shown in ID NO:143; or
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:118
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:1208
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:137
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:137
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:119
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:125
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:138
  • the LFR4 comprising the amino acid sequence shown in SEQ ID NO: 143; or
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:120
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:129
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:139
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:139.
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:114
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:140
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:140 The amino acid sequence shown in ID NO:144; or
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:121
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:141 The amino acid sequence shown in ID NO:145; or
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:122
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:141
  • the LFR1 comprises the amino acid sequence shown in SEQ ID NO:123
  • the LFR2 comprises the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 comprises the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 comprises the amino acid sequence shown in SEQ ID NO:141 Amino acid sequence shown in ID NO:147.
  • the isolated antigen-binding protein comprises a VL, wherein the VL comprises the amino acid sequence shown in any one of SEQ ID NO: 148 to SEQ ID NO: 160.
  • the isolated antigen-binding protein comprises VH and VL, wherein the VH comprises the amino acid sequence shown in SEQ ID NO:61, and the VL comprises the amino acid sequence shown in SEQ ID NO:148 sequence; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:62, and the VL comprises the amino acid sequence shown in SEQ ID NO:149; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:63
  • the VL comprises the amino acid sequence shown in SEQ ID NO:150; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:64, and the VL comprises the amino acid sequence shown in SEQ ID NO:151; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:65
  • the VL comprises the amino acid sequence shown in SEQ ID NO:152; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:66
  • the VL comprises the amino acid sequence shown in SEQ ID NO:153; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:67
  • the VL comprises the amino acid sequence shown in SEQ ID NO:154; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:68, and the VL comprises the amino acid sequence shown in SEQ ID NO:155; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:69
  • the VL comprises the amino acid sequence shown in SEQ ID NO:156; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:70, and the VL comprises the amino acid sequence shown in SEQ ID NO:157; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:71, and the VL comprises the amino acid sequence shown in SEQ ID NO:158; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:72, and the VL comprises the amino acid sequence shown in SEQ ID NO:159; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:73
  • the VL comprises the amino acid sequence shown in SEQ ID NO:160.
  • the isolated antigen-binding protein comprises an antibody or antigen-binding fragment thereof.
  • the antibodies include monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies, intact antibodies, antibody fragments, human antibodies, humanized antibodies, or chimeric antibodies.
  • said antigen-binding fragment comprises Fab, Fab', Fv fragment, F(ab') 2 , scFv, di-scFv and/or dAb.
  • the antigen binding protein comprises a scFv.
  • VL and VH are linked by a linker.
  • linker comprises a polypeptide linker
  • the polypeptide linker comprises an amino acid sequence represented by (GGGGS)n, wherein n is any integer from 1 to 5.
  • it comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the present application provides an isolated polypeptide comprising the isolated antigen binding protein described herein.
  • the present application provides an immunoconjugate comprising the antigen-binding protein described in the present application.
  • the immunoconjugate comprises:
  • a conjugation moiety selected from the group consisting of detectable markers, drugs, toxins, cytokines, viral coat proteins or VLPs, or combinations thereof.
  • a chimeric antigen receptor comprising at least one extracellular antigen binding domain comprising an antigen binding protein described herein.
  • the extracellular antigen binding domain comprises a scFv.
  • the chimeric antigen receptor comprises a transmembrane domain comprising a transmembrane domain derived from one or more proteins selected from the group consisting of: CD8, CD28, CD3 ⁇ (CD3e), 4-1BB, CD4, CD27, CD7, PD-1, TRAC, TRBC, CD3 ⁇ , CTLA-4, LAG-3, CD5, ICOS, OX40, NKG2D, 2B4(CD244), Fc ⁇ RI ⁇ , BTLA, CD30 , GITR, HVEM, DAP10, CD2, NKG2C, LIGHT, DAP12, CD40L (CD154), TIM1, CD226, DR3, CD45, CD80, CD86, CD9, CD16, CD22, CD33, CD37, CD64, and SLAM.
  • transmembrane domain comprises a CD8-derived transmembrane domain.
  • transmembrane domain comprises the amino acid sequence shown in SEQ ID NO: 177.
  • the chimeric antigen receptor includes an intracellular co-stimulatory signaling domain comprising one or more proteins derived from the group consisting of Intracellular co-stimulatory signaling domains: CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GITR, HVEM, DAP10, DAP12, CD30, CD40, CD40L, TIM1, PD-1, LFA-1, LIGHT, JAML, CD244, CD100, ICOS, CD40, and MyD88.
  • Intracellular co-stimulatory signaling domains CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GI
  • the intracellular costimulatory signaling domain is derived from a costimulatory signaling domain of 4-1BB.
  • the intracellular co-stimulatory signaling domain comprises the amino acid sequence shown in any one of SEQ ID NO:178.
  • the chimeric antigen receptor includes an intracellular signal transduction domain comprising a cellular protein derived from one or more proteins selected from the group consisting of: Internal signaling domain: CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa, bovine leukemia virus gp30, Epstein-Barr virus (EBV) LMP2A, simian immunodeficiency virus PBj14Nef, DAP10, DAP-12 and domains containing at least one ITAM.
  • Internal signaling domain CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa, bovine leukemia virus gp30, Epstein-Barr virus (EBV) LMP2A, simian immunodeficiency virus PBj14Nef, DAP10, DAP-12 and domains containing at least
  • the intracellular signaling domain comprises a signaling domain derived from CD3 ⁇ .
  • said intracellular signal transduction domain comprises the amino acid sequence shown in SEQ ID NO: 179.
  • it includes a hinge region between the extracellular antigen binding domain and the transmembrane domain, said hinge region comprising a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • the hinge region comprises a hinge region derived from CD8.
  • the hinge region comprises the amino acid sequence shown in SEQ ID NO: 176.
  • the non-targeting portion of the chimeric antigen receptor comprises a transmembrane domain, a hinge region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain.
  • the non-targeting portion of the chimeric antigen receptor comprises a CD8 molecular transmembrane domain, a hinge region of CD8, an intracellular co-stimulatory signaling domain of 4-1BB, and a CD3 ⁇ intracellular signaling structure area.
  • it further comprises a signal peptide fragment, the C-terminus of the signal peptide fragment is connected to the N-terminus of the extracellular antigen-binding domain.
  • the signal peptide fragment comprises a CD8 signal peptide fragment.
  • the signal peptide fragment comprises the amino acid sequence shown in SEQ ID NO: 175.
  • it comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • the application provides an isolated nucleic acid molecule or molecules encoding an isolated antigen binding protein described herein, a polypeptide described herein, or a chimeric antigen receptor described herein.
  • the isolated nucleic acid molecule comprises the nucleotide sequence shown in any one of SEQ ID NO:193 to SEQ ID NO:205.
  • the present application provides an expression vector comprising the nucleic acid molecule described in the present application.
  • the vector is selected from DNA vectors, RNA vectors, plasmids, lentiviral vectors, adenoviral vectors, adeno-associated viral vectors and retroviral vectors.
  • the application provides a cell, the cell comprises the nucleic acid molecule of the application or the expression vector described in the application, and/or ii) the cell expresses the antigen binding protein described in the application, the expression vector described in the application.
  • the polypeptide described above or the application Please refer to chimeric antigen receptors.
  • the application provides a method for preparing the isolated antigen-binding protein described in the application, the method comprising culturing the antigen-binding protein described in the application under conditions that allow the expression of the isolated antigen-binding protein described in the application. Cell.
  • the present application provides an engineered cell comprising the nucleic acid molecule or the vector described in the present application, and/or expressing the chimeric antigen receptor described in the present application.
  • the cells include immune effector cells.
  • the immune effector cells include human cells.
  • the immune effector cells include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • NK cells natural killer cells
  • macrophages include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • the immune effector cells comprise autologous or allogeneic immune effector cells.
  • the immune effector cells include allogeneic T cells or autologous T cells.
  • the immune effector cells include modified immune effector cells.
  • the expression of CD7 in the modified immune effector cells is absent or suppressed.
  • the modified immune effector cells have reduced surface expression of CD7 and express an anti-CD7 CAR compared to corresponding immune cells.
  • the immune effector cells include CAR-T cells.
  • the CAR-T cells do not induce cannibalism.
  • the present application provides a method for preparing a chimeric antigen receptor T (CAR-T) cell population, wherein the CAR targets CD7, comprising the following steps:
  • CD7 is absent or suppressed in the modified population of T cells compared to the corresponding unmodified cells.
  • said modification comprises administering to said T cell population one or more substances selected from the group consisting of antisense RNA, siRNA, shRNA, transcription activator-like effector nuclease (TALEN) , zinc finger nucleic acid Enzymes (ZFNs) and CRISPR/Cas systems.
  • one or more substances selected from the group consisting of antisense RNA, siRNA, shRNA, transcription activator-like effector nuclease (TALEN) , zinc finger nucleic acid Enzymes (ZFNs) and CRISPR/Cas systems.
  • wherein said modification comprises administering a CRISPR/Cas system to said population of T cells.
  • wherein said modification comprises administering a CRISPR/Cas9 system to said population of T cells.
  • said modification comprises administering Cas9 and a gRNA targeting the CD7 gene to said T cell population.
  • the gRNA targeting the CD7 gene comprises the nucleotide sequence shown in any one of SEQ ID NO:211 to SEQ ID NO:218.
  • the transducing chimeric antigen receptor comprises transducing a circRNA chimeric antigen receptor.
  • the cirRNA chimeric antigen receptor comprises an internal ribosome entry site (IRES) element, a CD7-targeting protein coding sequence, and polyadenylic acid (polyA) in the following order.
  • IRS internal ribosome entry site
  • CD7-targeting protein coding sequence CD7-targeting protein coding sequence
  • polyA polyadenylic acid
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the immunoconjugate described in the present application, the nucleic acid molecule described in the present application , the expression vector described in the present application, the cell described in the present application, the chimeric antigen receptor described in the present application and/or the engineered cell described in the present application, and optionally a pharmaceutically acceptable carrier.
  • the present application provides a kit comprising the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the immunoconjugate described in the present application, the nucleic acid molecule described in the present application, The expression vector described in the present application, the cell described in the present application, the chimeric antigen receptor described in the present application, the engineered cell described in the present application, or the pharmaceutical composition described in the present application.
  • the isolated antigen binding protein described herein, the polypeptide described herein, the immunoconjugate described herein, the nucleic acid molecule described herein, the expression vector described herein, the cell described herein , the chimeric antigen receptor described in the application, the engineered cell described in the application, or the application of the pharmaceutical composition described in the application in the preparation of medicines, the medicine is used to prevent and/or treat CD7-related disease or condition.
  • said CD7-associated disease or condition comprises a tumor.
  • the tumor comprises a CD7 expressing tumor.
  • the tumor comprises a hematoma.
  • the tumor comprises a CD7-positive hematologic malignancy.
  • the tumor comprises a T cell malignancy.
  • the present application provides a method for treating tumors, the method comprising administering an effective amount of the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the polypeptide described in the present application to a subject in need thereof.
  • the tumor comprises a CD7 expressing tumor.
  • the tumor comprises a hematoma.
  • the tumor comprises a CD7-positive hematologic malignancy.
  • the tumor comprises a T cell malignancy.
  • T-cell malignancy comprises acute T-lymphoblastic leukemia (T-ALL), acute myeloid leukemia, or NK/T-cell lymphoma.
  • T-ALL acute T-lymphoblastic leukemia
  • NK/T-cell lymphoma NK/T-cell lymphoma
  • the present application provides a method for killing malignant T cells, the method comprising contacting the malignant T cells with the engineered cells described in the present application.
  • This application uses CRISPR/Cas9 technology to delete the expression of CD7 in T cells to prevent the occurrence of cannibalism between CAR-T cells; screen out a fully human anti-CD7 scFv from an antibody phage display library, which may help reduce The rejection of the human immune system improves the persistence and therapeutic effect of CAR-T cells.
  • the purpose of this application is to use CRISPR/Cas9 technology to delete the expression of CD7 in T cells to prevent the occurrence of cannibalism among CAR-T cells.
  • the application provides a gRNA targeting CD7 gene, which comprises the nucleotide sequence described in any one of SEQ ID NO:212 to SEQ ID NO:218 or with SEQ ID NO:212 to SEQ ID NO
  • the nucleotide sequence of any one of: 218 has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% , about 99%, about 99.5% amino acid sequence identity.
  • the present application provides an isolated nucleic acid molecule comprising the gRNA described in the present application or a DNA molecule encoding the gRNA.
  • the present application provides an expression vector comprising the gRNA described in the present application or the nucleic acid molecule described in the present application.
  • the present application provides a gene editing system, which comprises the gRNA described in the present application, the nucleic acid molecule described in the present application or the expression vector described in the present application.
  • the gene editing system includes a CRISPR/Cas gene editing system.
  • the gene editing system includes a CRISPR/Cas9 gene editing system.
  • the gene editing system further comprises Cas9-encoding DNA, Cas9-encoding mRNA or Cas9 protein molecule.
  • the gene editing system comprises an expression vector encoding the gRNA targeting the CD7 gene described in the present application and Cas9.
  • the present application provides a cell comprising the gRNA described in the present application, the nucleic acid molecule described in the present application, the expression vector described in the present application, or the gene editing system described in the present application.
  • it includes cells expressing CD7.
  • it includes immune effector cells.
  • the immune effector cells include T cells, B cells, natural killer (NK) cells, mast cells or phagocytes.
  • the immune effector cells comprise engineered immune effector cells.
  • the engineered immune effector cells include CAR-T cells.
  • the engineered immune effector cells include anti-CD7 CAR-T cells.
  • the VH can be Comprising HCDR1, HCDR2 and HCDR3, wherein the HCDR1 can comprise the amino acid sequence shown in SEQ ID NO:2, the HCDR2 can comprise the amino acid sequence shown in SEQ ID NO:10, and the HCDR3 can comprise the amino acid sequence shown in SEQ ID NO: the amino acid sequence shown in 18; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:3, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:11, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:19; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:2
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:10
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:20; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:4, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:12, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:21; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:22; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:6, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:14, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:23; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:2, 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:24; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:25; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:8, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:16, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:26; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:27.
  • VH may comprise the amino acid sequence shown in SEQ ID NO:61 to SEQ ID NO:73.
  • the LCDR1 can comprise the amino acid sequence shown in SEQ ID NO:74
  • the LCDR2 can comprise the amino acid sequence shown in SEQ ID NO:87
  • the LCDR3 can comprise the amino acid sequence shown in SEQ ID NO:99; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:75
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:88
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:100;
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:76
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:89
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:101; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:77
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:90
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:102; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:78
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:91
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:103; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:79
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:104; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:80
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:93
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:105; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:81
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:94
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:106; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:82
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:95
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:107; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:84
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:97
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:109; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:85
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:110; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:86
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:111.
  • said VH comprises HCDR1, HCDR2 and HCDR3
  • said VL comprises LCDR1, LCDR2 and LCDR3
  • said HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • said HCDR2 comprises SEQ ID
  • the amino acid sequence shown in NO:10, the HCDR3 includes the amino acid sequence shown in SEQ ID NO:18, the LCDR1 includes the amino acid sequence shown in SEQ ID NO:74, and the LCDR2 includes the amino acid sequence shown in SEQ ID NO:87
  • the amino acid sequence of said LCDR3 comprises the amino acid shown in SEQ ID NO:99 sequence; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:3
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:11
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:19
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:19
  • the amino acid sequence shown in ID NO:75, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:88, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:100; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:20
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:20
  • the amino acid sequence shown in ID NO:76, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:89, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:101; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:4, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:12, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:21, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:21
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:22
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:22
  • the amino acid sequence shown in ID NO:91, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:91
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:103;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:79, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:104; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:80, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:93, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:105; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:6, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:14, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:23, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:23
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:82, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:95
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:107;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:24
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:24
  • the amino acid sequence shown in ID NO:83, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:96
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:108;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:25
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:25
  • the amino acid sequence shown in ID NO:84, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:97
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:109;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:8, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:16, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:26, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:26
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:27
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:27
  • the amino acid sequence shown in ID NO:86, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:111.
  • VL may comprise the amino acid sequence shown in any one of SEQ ID NO: 148 to SEQ ID NO: 160.
  • VH may comprise the amino acid sequence shown in SEQ ID NO:61
  • said VL may comprise the amino acid sequence shown in SEQ ID NO:148;
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:62, and the VL may comprise the amino acid sequence shown in SEQ ID NO:149; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:63, and the VL may comprise the amino acid sequence shown in SEQ ID NO:150; or
  • the VH can comprise the amino acid sequence shown in SEQ ID NO:64, and the VL can comprise SEQ ID NO: the amino acid sequence shown in 151; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:65, and the VL may comprise the amino acid sequence shown in SEQ ID NO:152; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:66, and the VL may comprise the amino acid sequence shown in SEQ ID NO:153; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:67, and the VL may comprise the amino acid sequence shown in SEQ ID NO:154; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:68, and the VL may comprise the amino acid sequence shown in SEQ ID NO:155; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:69, and the VL may comprise the amino acid sequence shown in SEQ ID NO:156; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:70, and the VL may comprise the amino acid sequence shown in SEQ ID NO:157; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:71, and the VL may comprise the amino acid sequence shown in SEQ ID NO:158; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:72, and the VL may comprise the amino acid sequence shown in SEQ ID NO:159; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:73
  • the VL may comprise the amino acid sequence shown in SEQ ID NO:160.
  • the extracellular antigen binding domain comprises a scFv.
  • the extracellular antigen binding domain can be an anti-CD7 scFv.
  • VL and VH are linked by a linker.
  • linker comprises a polypeptide linker
  • the polypeptide linker comprises an amino acid sequence represented by (GGGGS)n, wherein n is any integer from 1 to 5.
  • the CAR-T cells of the present application comprise the extracellular domain of a chimeric antigen receptor that specifically binds to CD7.
  • CD7 is a T cell surface membrane-associated glycoprotein.
  • CD7 can be overexpressed in T cell malignancies including T cell acute lymphoblastic leukemia (T-ALL) and non-Hodgkin's T cell lymphoma (NHL).
  • T-ALL T cell acute lymphoblastic leukemia
  • NHL non-Hodgkin's T cell lymphoma
  • the CAR-T cells of the present disclosure can be used to target malignant T cells overexpressing CD7.
  • the antigen-specific extracellular domain of the chimeric antigen receptor of the present application can specifically bind to CD7, which comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174 or with SEQ ID
  • the amino acid sequence shown in any one of NO:162 to SEQ ID NO:174 has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97% %, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • the chimeric antigen receptor comprises a transmembrane domain comprising a transmembrane domain derived from one or more proteins selected from the group consisting of: CD8, CD28, CD3 ⁇ (CD3e), 4-1BB, CD4, CD27, CD7, PD-1, TRAC, TRBC, CD3 ⁇ , CTLA-4, LAG-3, CD5, ICOS, OX40, NKG2D, 2B4(CD244), Fc ⁇ RI ⁇ , BTLA, CD30 , GITR, HVEM, DAP10, CD2, NKG2C, LIGHT, DAP12, CD40L (CD154), TIM1, CD226, DR3, CD45, CD80, CD86, CD9, CD16, CD22, CD33, CD37, CD64, and SLAM.
  • transmembrane domain may comprise a transmembrane domain derived from CD8.
  • the transmembrane domain comprises the amino acid sequence shown in SEQ ID NO: 177 or has at least about 90%, about 91%, about 92%, about 93% of the amino acid sequence shown in SEQ ID NO: 177, Amino acid sequences that are about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical.
  • the chimeric antigen receptor includes an intracellular co-stimulatory signaling domain comprising one or more proteins derived from the group consisting of Intracellular co-stimulatory signaling domains: CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GITR, HVEM, DAP10, DAP12, CD30, CD40, CD40L, TIM1, PD-1, LFA-1, LIGHT, JAML, CD244, CD100, ICOS, CD40, and MyD88.
  • Intracellular co-stimulatory signaling domains CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GI
  • the intracellular co-stimulatory signaling domain may comprise a co-stimulatory signaling domain derived from 4-1BB.
  • intracellular co-stimulatory signaling domain may comprise the amino acid sequence shown in any one of SEQ ID NO:178.
  • the chimeric antigen receptor comprises an intracellular signaling domain comprising an intracellular protein derived from one or more proteins selected from the group consisting of Signal transduction domain: CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa, bovine leukemia virus gp30, Epstein-Barr virus (EBV) LMP2A, simian immunodeficiency virus PBj14 Nef, DAP10, DAP-12 and contain at least one Domains of ITAMs.
  • Signal transduction domain CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa
  • bovine leukemia virus gp30 Epstein-Barr virus (EBV) LMP2A
  • PBj14 Nef simian immunodeficiency virus
  • the intracellular signaling domain may comprise a signaling domain derived from CD3 ⁇ .
  • intracellular signal transduction domain may comprise the amino acid sequence shown in SEQ ID NO: 179.
  • it includes a hinge region between the extracellular antigen binding domain and the transmembrane domain, said hinge region comprising a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • the hinge region may comprise a hinge region derived from CD8.
  • the hinge region may comprise the amino acid sequence shown in SEQ ID NO: 176.
  • the non-targeting portion of the chimeric antigen receptor comprises a transmembrane domain, a hinge region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain.
  • the non-targeting portion of the chimeric antigen receptor comprises the CD8 molecular transmembrane domain, the hinge region of CD8, the intracellular co-stimulatory signaling domain of 4-1BB, and the CD3 ⁇ intracellular signaling domain.
  • it further comprises a signal peptide fragment, the C-terminus of the signal peptide fragment is connected to the N-terminus of the extracellular antigen-binding domain.
  • the signal peptide fragment may comprise a CD8 signal peptide fragment.
  • the signal peptide fragment may comprise the amino acid sequence shown in SEQ ID NO:175.
  • the chimeric antigen receptor of the present application may comprise the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192 or any one of SEQ ID NO: 180 to SEQ ID NO: 192
  • the amino acid sequence shown has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • the present application provides the gRNA described in the present application, the nucleic acid molecule described in the present application, the expression vector described in the present application, the gene editing system described in the present application or the cells described in the present application in the preparation of drugs for treating tumors in the application.
  • the tumor comprises a solid tumor or a hematological tumor.
  • the tumor comprises a CD7 positive tumor.
  • the drug comprises CAR-T cells.
  • the drug includes targeting CD7 CAR-T cells.
  • the present application provides a method for gene editing of the CD7 gene in cells, including using this
  • the gRNA described in the application mediates the gene editing of the CD7 gene by Cas9.
  • said gene editing comprises gene knockout.
  • the present application provides a method for regulating T cell function, the method comprising the gRNA described in the application, the nucleic acid molecule described in the application, the expression vector described in the application or the gene described in the application
  • the editing system is introduced into T cells.
  • the Cas enzyme comprises a Cas9 protein.
  • the regulated T cells have down-regulated or knocked-out CD7 gene expression compared to unregulated T cells.
  • the method further comprises modifying the specificity of the T cell by introducing into the T cell a nucleic acid molecule encoding a CAR.
  • the CAR-encoding nucleic acid molecule comprises mRNA.
  • the mRNA encodes an anti-CD7 CAR.
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the anti-CD7 CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • regulated T cells have reduced surface expression of CD7 and express an anti-CD7 CAR compared to corresponding T cells.
  • the present application also provides a circRNA which comprises an internal ribosome entry site (IRES) element, a protein coding sequence targeting CD7, and polyadenylic acid in the following order.
  • IRS internal ribosome entry site
  • the polyA is at least 45 nucleotides in length.
  • the polyA is at least 70 nucleotides in length.
  • the CD7-targeting protein comprises an antibody or antigen-binding fragment thereof, a chimeric antigen receptor (CAR) and/or a T cell receptor (TCR).
  • CAR chimeric antigen receptor
  • TCR T cell receptor
  • the binding domain of the CAR comprises a CD7 scFv.
  • the CD7-targeting protein comprises an antibody comprising a light chain variable region comprising LCDR1, LCDR2, and LCDR3 and a heavy chain variable region comprising HCDR1, HCDR2, and HCDR3, wherein:
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2, 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:18, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the protein targeting CD7 includes an antibody, and the antibody includes VH and VL, wherein the VH includes the amino acid sequence shown in SEQ ID NO:61, and the VL includes SEQ ID NO:148 Place or the VH comprises the amino acid sequence shown in SEQ ID NO:62, and the VL comprises the amino acid sequence shown in SEQ ID NO:149; or the VH comprises the amino acid sequence shown in SEQ ID NO:63 sequence, the VL comprises the amino acid sequence shown in SEQ ID NO:150; or the VH comprises the amino acid sequence shown in SEQ ID NO:64, and the VL comprises the amino acid sequence shown in SEQ ID NO:151; or the VH comprises the amino acid sequence shown in SEQ ID NO:151;
  • the VH comprises the amino acid sequence shown in SEQ ID NO:65, the VL comprises the amino acid sequence shown in SEQ ID NO:152; or the VH comprises the amino acid sequence shown in SEQ ID NO:66, and the VL comprises the amino acid sequence shown in SEQ ID NO:66 The amino acid sequence shown in S
  • the circRNA comprises the nucleotide sequence shown in any one of SEQ ID NO:256-257.
  • the present application also provides a reagent for regulating the expression level and/or activity of CD7, which comprises the circRNA described in the present application.
  • the present application also provides the application of the circRNA described in the present application in regulating the expression level and/or activity of CD7.
  • Figure 1 shows the readouts of the anti-human CD7-Fc monoclonal phage ELISA for three 96-well plates.
  • Figure 2 shows a schematic diagram of the pDA-CAR vector used to generate anti-CD7 CAR mRNA.
  • Figure 3 shows the results of FACS staining of T cells electroporated with different CD7 gRNAs with anti-CD7 antibody.
  • Figure 4 shows the results of FACS staining of different anti-CD7 scFv CAR-T cells with CD7-Fc protein.
  • Figure 5 shows the results of FACS staining of A549 cells electroporated with different amounts of CD7 mRNA with anti-CD7 antibody.
  • Figure 11 shows the CD107a staining results of anti-CD7 CAR-T cells (A40C28+/-) with different mRNAs in co-culture and killing experiments with A549-GFP tumor cells electroporated with 0 or 2ug CD7 mRNA.
  • Figure 12 shows the results of FACS staining of 17 different tumor cell lines with isotype control and anti-CD7 mAb.
  • Figures 13-14 show the CD107a staining results of anti-CD7 CAR-T cells (A40C28+/-) with different mRNAs in the co-culture and killing experiments with different tumor cell lines.
  • Figure 15 shows a schematic representation of the LACO-Stim molecule described in this application.
  • Figure 16 shows the results of examining the expression of CAR/LACO-stim by flow cytometry.
  • FIG 17 shows tumor growth after co-culture with T cells co-expressing CAR and LACO-Stim as shown in Table 3.
  • the E/T ratio is 30:1.
  • E effector cells (T cells); T: target cells (A549-ESO cells).
  • FIG. 18 shows tumor growth after co-culture with T cells co-expressing TCR and LACO-Stim as shown in Table 1.
  • FIG. The E/T ratio is 30:1.
  • E effector cells (T cells); T: target cells (A549-ESO cells).
  • Figure 19 shows the structure of the circRNA described in this application.
  • Figures 20A-20B show the effect of the circRNA described in this application on target cells.
  • the term "antigen binding protein” generally refers to a protein comprising a moiety that binds an antigen, and optionally a scaffold or backbone moiety that allows the moiety that binds the antigen to adopt a conformation that facilitates binding of the antigen binding protein to the antigen.
  • antigen binding proteins include, but are not limited to, antibodies, antigen binding fragments (e.g., Fab, Fab', F(ab) 2 , Fv fragments, F(ab') 2 , scFv, di-scFv, and/or dAb), immune Conjugates, multispecific antibodies (such as bispecific antibodies), antibody fragments, antibody derivatives, antibody analogs, or fusion proteins, etc., as long as they exhibit the desired antigen-binding activity.
  • antigen binding fragments e.g., Fab, Fab', F(ab) 2 , Fv fragments, F(ab') 2 , scFv, di-scFv, and/or dAb
  • immune Conjugates e.g., multispecific antibodies (such as bispecific antibodies), antibody fragments, antibody derivatives, antibody analogs, or fusion proteins, etc., as long as they exhibit the desired antigen-binding activity.
  • an “antibody” is generally used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibody), and antibody fragments, as long as they show the desired biological activity (Miller et al (2003) Jour. of Immunology 170:4854-4861).
  • Antibodies can be murine, human, humanized, chimeric, or derived from other species.
  • an “antibody” may typically comprise a protein of at least two heavy chains (HC) and two light chains (LC) interconnected by disulfide bonds, or an antigen-binding fragment thereof. Each heavy chain comprises a heavy chain variable region (VH) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, CH1, CH2 and CH3.
  • each light chain comprises a light chain variable region (VL) and a light chain constant region.
  • the light chain constant region consists of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, called complementarity determining regions (CDRs), which alternate with more conserved regions called framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each VH and VL comprises three CDRs and four framework regions (FRs), arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • variable domains of native heavy and light chains each comprise four FR regions (HFR1, HFR2, HFR3, HFR4, LFR1, LFR2, LFR3, LFR4), mostly in a ⁇ -sheet configuration, connected by three CDRs, Loop links are formed and in some cases form part of a ⁇ -sheet structure.
  • the CDRs in each chain are in close proximity by the FR regions and, together with the CDRs from the other chain, form the antigen-binding site of the antibody.
  • 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.
  • the term "located between” usually means that the C-terminal of a certain amino acid fragment is directly or indirectly connected to the N-terminal of the first amino acid fragment, and its N-terminal is connected to the second amino acid fragment.
  • the C-termini of the amino acid fragments are linked directly or indirectly.
  • the N-terminal of LFR2 is directly or indirectly linked to the C-terminal of LCDR1
  • the C-terminal of LFR2 is directly or indirectly linked to the N-terminal of LCDR2.
  • the N-terminal of LFR3 is directly or indirectly connected to the C-terminal of LCDR2, and the C-terminal of LFR3 is directly or indirectly connected to the N-terminal of LCDR3.
  • the N-terminal of HFR2 is directly or indirectly linked to the C-terminal of HCDR1
  • the C-terminal of HFR2 is directly or indirectly linked to the N-terminal of HCDR2.
  • the N-terminal of HFR3 is directly or indirectly connected to the C-terminal of HCDR2, and the C-terminal of HFR3 is directly or indirectly connected to the N-terminal of HCDR3.
  • antigen-binding fragment generally refers to a portion of an antibody molecule comprising the amino acids responsible for the specific binding between the antibody and the antigen.
  • Antigen-binding fragments may include: Fab, Fab', F(ab) 2 fragments, Fv fragments, dsFv, F(ab') 2 , scFv, di-scFv, dAb fragments.
  • Fab generally refers to an Fv region comprising the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. Fab' differs from Fab by the addition of several residues at the carboxy-terminus of the CH1 domain of the heavy chain, including one or more cysteines from the antibody hinge region.
  • Fv fragment generally refers to an antibody fragment consisting of the VL and VH domains of a single arm of an antibody, which in some cases may consist of a dimer of one VH and one VL in tight non-covalent association.
  • the term “(Fab) 2 " generally refers to a bivalent fragment comprising the hinge region and the variable and first constant domains of the heavy and light chains.
  • the term “F(ab') 2 " generally refers to an antibody fragment comprising two Fab' fragments linked by a disulfide bond at the hinge region.
  • scFv fragment generally comprises the VH and VL domains of an antibody, wherein these domains are present as a single polypeptide chain.
  • scFv polypeptides further comprise a polypeptide linker between the VH and VL domains, which enables the scFv to form the desired structure for antigen binding.
  • dAb fragment generally refers to an antibody fragment consisting of a VH domain.
  • dsFv generally refers to a disulfide-stabilized Fv fragment in which the linkage between a single light chain variable domain and a single heavy chain variable domain is a disulfide bond.
  • variable generally refers to the fact that certain parts of the sequence of the variable domains of antibodies vary strongly, which contributes to the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments in the light and heavy chain variable regions, called complementarity determining regions (CDRs) or hypervariable regions (HVRs). The more highly conserved portions of variable domains are called the frameworks (FRs).
  • CDRs complementarity determining regions
  • HVRs hypervariable regions
  • the CDRs of antibodies can be defined by various methods, such as the Kabat definition rules based on sequence variability (see, Kabat et al., Protein Sequences in Immunology, 5th edition, National Institutes of Health, Besse Star, MD (1991)), Chothia definition rules based on the location of structural ring regions (cf., A1-Lazikani et al., JMol Biol 273:927-48, 1997) and KABAT definition rules based on the concepts of the IMGT Ontology (IMGT-ONTOLOGY) and the IMGT Scientific diagram rules.
  • the methods used herein may utilize CDRs defined according to any of these systems, in some embodiments using Kabat or Chothia defined CDRs, or may refer to http://abysis.org/ to delineate the CDRs of scFv antibodies.
  • isolated antigen binding protein generally refers to an antigen binding protein that has been identified, separated and/or recovered from a component of the environment in which it was produced (eg, natural or recombinant). Contaminating components of the environment in which it is produced are usually substances that interfere with its research, diagnostic or therapeutic use and can include enzymes, hormones and other proteinaceous or nonproteinaceous solutes. Isolated antigen binding protein or antibody will usually be prepared by at least one purification step.
  • the term "monoclonal antibody” generally refers to an antibody obtained from a population of substantially homogeneous antibodies, ie, the individual antibodies in the population are identical except for minor natural mutations that may be present.
  • Monoclonal antibodies are usually highly specific against a single antigenic site. Furthermore, each monoclonal antibody is directed against a single determinant on the antigen, unlike conventional polyclonal antibody preparations, which typically have different antibodies directed against different determinants.
  • monoclonal antibodies have the advantage that they can be synthesized by hybridoma cultures without contamination from other immunoglobulins.
  • monoclonal denote the characteristics of an antibody obtained from a substantially homogeneous population of antibodies and is not to be construed as requiring that the antibody be produced by any particular method.
  • monoclonal antibodies used herein can be produced in hybridoma cells, or can be produced by recombinant DNA methods.
  • humanized antibody generally refers to an antibody in which some or all of the amino acids other than the CDR region of a non-human antibody (such as a mouse antibody) are replaced with corresponding amino acids derived from human immunoglobulins. In the CDR regions, small additions, deletions, insertions, substitutions or modifications of amino acids may also be permissible so long as they still retain the ability of the antibody to bind a particular antigen.
  • a humanized antibody optionally will comprise at least a portion of a human immunoglobulin constant region.
  • a "humanized antibody” retains antigen specificity similar to the original antibody.
  • “Humanized” forms of non-human (eg, murine) antibodies may contain, at a minimum, chimeric antibodies of sequence derived from non-human immunoglobulin.
  • CDR region residues in a human immunoglobulin can be replaced with a non-human species (donor antibody) (such as mouse, rat) having the desired properties, affinity and/or capabilities. , rabbit or non-human primate) residue substitution in the CDR region.
  • donor antibody such as mouse, rat
  • rabbit or non-human primate residue substitution in the CDR region such as mouse, rat
  • FR region residues of the human immunoglobulin may be replaced with corresponding non-human residues.
  • humanized antibodies can comprise amino acid modifications that are absent in the recipient antibody or in the donor antibody. These modifications may be made to further refine antibody properties, such as binding affinity.
  • Fully human antibody generally refers to the antibody that is expressed by transferring the gene encoding the human antibody into a genetically engineered animal lacking the antibody gene. All parts of the antibody (including variable and constant regions of the antibody Regions) are encoded by genes of human origin. Fully human antibodies can greatly reduce the immune side effects caused by heterologous antibodies on the human body. Methods for obtaining fully human antibodies in this field include phage display technology, transgenic mouse technology, ribosome display technology and RNA-polypeptide technology.
  • the term "scFv” generally refers to a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chains can be The variable regions are contiguous (eg via a synthetic linker such as a short flexible polypeptide linker) and can be expressed as a single chain polypeptide wherein the scFv retains the specificity of the intact antibody from which it was derived.
  • a scFv may have the VL and VH variable regions described in any order (e.g., relative to the N- and C-terminus of the polypeptide), and the scFv may include a VL-linker-VH or VH-Linker-VL can be included.
  • the term “specificity” generally refers to the different types of antigens or antigens to which a particular immunoglobulin sequence, antigen-binding molecule or antigen-binding protein (such as an immunoglobulin single variable domain, or a polypeptide of the invention) can bind. Number of epitopes.
  • the specificity of an antigen binding protein can be determined in terms of affinity and/or avidity. , the terms “specific” and “specifically” are used interchangeably to indicate that biomolecules other than CD7 do not significantly bind the antibody.
  • affinity generally refers to a measure of the binding constant of a single monovalent ligand for its cognate binding partner, for example, the binding of a Fab' for an antigen or epitope.
  • Affinity can be measured in several ways, including measuring association and dissociation rates (kon and koff, respectively) by, for example, plasmon resonance (BiaCore), and expressed as an overall association (Kass) or dissociation constant (KD), where Kass is kon/koff, and KD is koff/kon.
  • KD can also be measured empirically by, for example, measuring the concentration at which binding of the ligand to the binding partner is half-saturated.
  • KD KD
  • a competition assay in which one binder or ligand is labeled or tagged and kept at a constant concentration while the test binder or ligand is added at various concentrations to The labeled substance competes away from its cognate binding partner, and the concentration at which the label is reduced by half is determined.
  • KD KD
  • KD KD
  • dissociation rate constant kdis, also known as “off-rate ) (koff)” or “kd”
  • association rate (kon) also known as “association rate (kon)” or “ka”
  • the binding affinity of an antigen-binding protein (eg, an antibody) for an antigen can be expressed using the on-rate constant (kon), the dissociation rate constant (kdis), and the equilibrium dissociation constant ( KD ).
  • association and dissociation rate constants include, but are not limited to, biofilm interferometry (BLI), radioimmunoassay (RIA), equilibrium dialysis, surface plasmon resonance (SPR), fluorescence resonance energy transfer (FRET) , Co-immunoprecipitation (Co-IP) and protein chip technology. If measured under different conditions (e.g. salt concentration, pH), then The measured affinity for a particular protein-protein interaction can vary.
  • reference antibody generally refers to an antibody with which an antigen-binding protein described herein competes for antigen binding.
  • consortium when used in the context of antigen binding proteins competing for the same epitope generally refers to competition between antigen binding proteins, as prevented or inhibited by the antigen binding proteins (e.g., antibodies or immunologically functional fragments thereof) which are tested As determined by an assay for (eg, reduced) specific binding of a reference antigen binding protein (eg, a ligand or a reference antibody) to a common antigen (eg, CD7 or a fragment thereof).
  • antigen binding proteins e.g., antibodies or immunologically functional fragments thereof
  • solid-phase direct or indirect radioimmunoassay
  • EIA solid-phase direct or indirect enzyme immunoassay
  • sandwich competition assay See, e.g., Stahli et al., 1983, Methods in Enzymology 9:242-253
  • solid phase direct biotin-avidin EIA see, e.g., Kirkland et al., 1986, J. Immunol.
  • solid phase Direct labeling assay solid-phase direct-labeling sandwich assay (see, e.g., Harlow and Lane, 1988, Antibodies, A Laboratory Manual, Cold Spring Harbor Press); solid-phase direct-labeled RIA using I-125 labeling (see, e.g., Morel et al. , 1988, Molec.Immunol.25: 7-15); solid-phase direct biotin-avidin EIA (see, for example, Cheung et al., 1990, Virology 176: 546-552); and direct label RIA (Moldenhauer et al., 1990, Scand. J. Immunol. 32:77-82).
  • such assays involve the use of purified antigen bound to a solid surface or a unit carrying either of these, an unlabeled test antigen binding protein and a labeled reference antigen binding protein.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or unit in the presence of the test antigen binding protein.
  • the test antigen binding protein is present in excess.
  • Antigen-binding proteins identified by competition assays include antigen-binding proteins that bind the same epitope as the reference antigen-binding protein and neighbors that bind the epitope of the reference antigen-binding protein in close enough proximity to be sterically hindered. Epitope binding antigen binding protein.
  • the competing antigen binding protein when it is present in excess, it inhibits (e.g., reduces) specific binding of the reference antigen binding protein to the common antigen by at least about 40-45%, about 45-50%, about 50-55%, About 55-60%, about 60-65%, about 65-70%, about 70-75%, or about 75% or more. In some instances, binding is inhibited by at least about 80-85%, about 85-90%, about 90-95%, about 95-97%, or about 97% or more.
  • Fc generally refers to a polypeptide comprising at least a portion of the CH3, CH2 and hinge regions of an antibody constant domain.
  • the Fc region may comprise a CH4 domain present in some antibody classes.
  • Fc may include: native monomers, native dimers (disulfide-bonded), modified dimers (disulfide-bonded and/or non-covalently linked), and modified monomers (i.e. derivative).
  • Exemplary modifications include addition, deletion or substitution of one or more amino acids in one or more domains. Such changes can be included to optimize effector function, half-life, and the like.
  • isolated antigen binding protein generally refers to an antigen binding protein that has been identified, separated and/or recovered from a component of the environment in which it was produced (eg, natural or recombinant). Contaminating components of the environment in which it is produced are usually substances that interfere with its research, diagnostic or therapeutic use and can include enzymes, hormones and other proteinaceous or nonproteinaceous solutes. Isolated antigen binding protein or antibody will usually be prepared by at least one purification step.
  • percent identity generally refers to the degree to which two or more nucleic acid or polypeptide sequences are identical.
  • percent identity generally refers to the degree to which two or more nucleic acid or polypeptide sequences are identical.
  • identity generally describe the ratio of two or more aligned amino acid sequences compared to the number of amino acid residues that make up the total length of those amino acid sequences The number of matches ("hits") of identical amino acids.
  • sequences compared and aligned for maximum correspondence as measured using sequence comparison algorithms known in the art
  • percentage of amino acid residues that are identical eg, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identity
  • sequences compared to determine sequence identity can be distinguished by one or more amino acid substitutions, additions or deletions. Suitable programs for aligning protein sequences are known to those skilled in the art.
  • the percent sequence identity of protein sequences can be determined, for example, with programs such as CLUSTALW, Clustal Omega, FASTA or BLAST, for example using the NCBI BLAST algorithm (AltschulSF et al. (1997), Nucleic Acids Res. 25:3389-3402) .
  • the term "immunoconjugate” or “antibody conjugate” generally refers to the linkage of an antibody or antibody fragment thereof to other active agents, such as chemotherapeutic agents, cytotoxins (cytotoxic agents), immunotherapeutic agents, Imaging probes, spectroscopic probes, and more.
  • the linkage may be a covalent bond, or a non-covalent interaction such as through electrostatic forces.
  • linkers known in the art can be used to form immunoconjugates.
  • the immunoconjugate can be provided as a fusion protein that can be expressed from a polynucleotide encoding the immunoconjugate.
  • fusion protein generally refers to a protein produced by joining two or more genes or gene fragments that originally encoded separate proteins, including peptides and polypeptides. Translation of the fusion gene produces a single protein with functional properties derived from each original protein.
  • cytotoxin or “cytotoxic agent” may include any agent that is detrimental to (eg, kills) cells.
  • chimeric antigen receptor generally refers to a group of polypeptides, usually two in the simplest embodiment, which, when in immune effector cells, provide cellular (usually cancer cells) and generate intracellular signals.
  • a CAR comprises at least one extracellular antigen-binding domain (such as a VHH, scFv, or portion thereof), a transmembrane domain, and a cytoplasmic signaling domain (also referred to herein as an "intracellular signaling domain”).
  • signaling domain comprising a functional signaling domain derived from a stimulatory molecule and/or a co-stimulatory molecule as defined below.
  • the set of polypeptides are in the same polypeptide chain (for example, comprising chimeric fusion protein). In some embodiments, the set of polypeptides is discontinuous from each other, such as in different polypeptide chains. In some aspects, the set of polypeptides includes a dimerization switch that, in the presence of a dimerization molecule, can The polypeptides are coupled to each other, for example, the antigen binding domain can be coupled to the intracellular signaling domain. In one aspect, the stimulating molecule of the CAR is the zeta chain associated with the T cell receptor complex. In one aspect, the cytoplasmic signaling The structural domain comprises a primary signaling domain (for example, the primary signaling domain of CD3- ⁇ ).
  • the cytoplasmic signaling domain further comprises one or more co-stimulatory molecules derived from at least one co-stimulatory molecule as defined below A functional signaling domain.
  • co-stimulatory molecules can be selected from 4-1BB (ie, CD137), CD27, ICOS and/or CD28.
  • CAR comprises a chimeric fusion protein, which can comprise extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein, which may comprise an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain, the intracellular signaling domain comprises a functional signaling domain derived from a co-stimulatory molecule and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein, which may comprise an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from one or more co-stimulatory molecules and derived from Stimulate the functional signaling domain of molecule.
  • CAR comprises chimeric fusion protein, and it can comprise extracellular antigen recognition domain, transmembrane domain and intracellular signaling domain, and intracellular signaling domain comprises at least Two functional signaling domains derived from one or more co-stimulatory molecules and a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises the amino terminal (N-ter) of the CAR fusion protein. Selected leader sequence.
  • the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen recognition domain, wherein the leader sequence is optionally cleaved from the antigen recognition domain (e.g., VHH) during cellular processing and will CAR localizes to the cell membrane.
  • the antigen recognition domain e.g., VHH
  • isolated nucleic acid molecule or isolated polynucleotide
  • isolated nucleic acid molecule generally refers to DNA or RNA of genomic, mRNA, cDNA or synthetic origin or some combination thereof, which is not identical to polynuclear molecules found in nature. All or a portion of the nucleotides are associated with, or linked to, a polynucleotide to which they are not linked in nature.
  • the term "vector” generally refers to a nucleic acid molecule capable of self-replication in a suitable host, which transfers an inserted nucleic acid molecule into and/or between host cells.
  • the vectors may include vectors mainly used for inserting DNA or RNA into cells, vectors mainly used for replicating DNA or RNA, and vectors mainly used for expression of transcription and/or translation of DNA or RNA.
  • the carrier also includes a carrier having various functions as described above. Said contained An entity may be a polynucleotide capable of being transcribed and translated into a polypeptide when introduced into a suitable host cell.
  • the vector can produce the desired expression product by culturing an appropriate host cell containing the vector.
  • the term "cell” generally refers to a plasmid or vector that can or has contained the nucleic acid molecule described in the application, or can express the antibody or antigen-binding fragment thereof, polypeptide or immunoconjugate described in the application individual cells, cell lines or cell cultures.
  • the cells may include progeny of a single host cell. Due to natural, accidental or deliberate mutations, the progeny cells may not necessarily be completely identical in shape or genome to the original parent cells, but it is sufficient to be able to express the antibodies or antigen-binding fragments thereof described in this application.
  • the cells can be obtained by transfecting cells in vitro with the vectors described in this application.
  • the cells can be prokaryotic cells (such as Escherichia coli) or eukaryotic cells (such as yeast cells, such as COS cells, Chinese hamster ovary (CHO) cells, HeLa cells, HEK293 cells, COS-1 cells, NSO cells or myeloma cells).
  • the cells may be mammalian cells.
  • the mammalian cells may be CHO-K1 cells.
  • the term "recombinant cell” generally refers to a cell into which a recombinant expression vector has been introduced.
  • the recombinant host cells include not only certain specific cells, but also the progeny of these cells.
  • T cell or "T lymphocyte” may be any T cell, such as a cultured T cell, such as a primary T cell, or a T cell from a cultured T cell line, such as Jurkat, SupTI, etc., or T cells obtained from a mammal (preferably a primate, species including monkey, dog or human). If obtained from a mammal, T cells can be obtained from a number of sources including, but not limited to, blood, bone marrow, lymph nodes, thymus or other tissues or fluids. T cells can also be enriched or normalized. T cells can be obtained by maturing hematopoietic stem cells into T cells in vitro or in vivo.
  • a cultured T cell such as a primary T cell
  • a T cell from a cultured T cell line such as Jurkat, SupTI, etc.
  • T cells obtained from a mammal preferably a primate, species including monkey, dog or human.
  • T cells can be obtained from a number of sources including, but not
  • the T cells are human T cells.
  • the T cells are T cells isolated from humans.
  • T cells can be of any type, including NKT cells, and can be of any developmental stage, including but not limited to CD4+/CD8+ double positive T cells; CDA+ helper T cells; e.g. Th1 and Th2 cells, CD8+ T cells (e.g. Cytotoxic T cells); peripheral blood mononuclear cells (PBMC); peripheral blood leukocytes (PBL); tumor infiltrating cells (TIL); memory T cells; untreated T cells, etc.
  • the T cells are CD8+ T cells or CD4+ T cells.
  • the T cells are allogeneic (from a different donor of the same species) to the subject receiving the cells or cells to be received (e.g., the cells are in the form of a therapeutic composition); in some alternatives, the T cells are autologous (donor and recipient are identical); in some alternative approaches, T cells are syngeneic (donor and recipient are different, but identical twins).
  • immune effector cells generally refers to immune cells that participate in the immune response and perform effector functions.
  • the exercising effector functions may include clearing foreign antigens or promoting immune effector responses and the like.
  • immune effect Response cells may include plasma cells, T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells, and myeloid-derived phagocytes.
  • the immune effector cells of the present application may be autologous/autogeneic ("own") or non-autologous ("non-self", eg, allogeneic, syngeneic or allogeneic).
  • autologous generally refers to cells from the same subject.
  • Allogeneic generally refers to cells that are of the same species but are genetically different from those to which they are being compared.
  • Isgeneic generally refers to cells of a different subject that are genetically identical to the cells being compared.
  • Allogeneic generally refers to a cell of a different species than the compared cell.
  • the cells of the present application are autologous or allogeneic.
  • the term "modify” generally refers to altering the state or structure of a cell and/or changing the state or structure of a cell.
  • the change is usually compared with the state or structure of the corresponding unmodified cell, and the change may include a change in the expression level or function of an endogenous gene, such as down-regulating the expression level of an endogenous gene in a cell by means of genetic engineering, Up-regulation or non-expression, the genetic engineering means may include homologous recombination, CRISPR/Cas9 system gene editing, etc.; the change may also include changes in cellular protein expression, structure or function, such as through the endogenous gene expression level or Changes in protein expression, changes in structure or function achieved by changes in function, such as changes in protein expression, changes in structure or function achieved by regulating protein translation, post-translational modification; the changes may also include introducing exogenous Genes, expression of foreign proteins, etc.
  • CRISPR/Cas system generally refers to a group of molecules comprising an RNA-guided nuclease or other effector molecule and a gRNA molecule capable of directing and implementing the RNA-guided nuclease or other effector molecule Nucleic acid is modified at a target sequence, eg, causing degradation of the target sequence.
  • a CRISPR system comprises a gRNA and a Cas protein, e.g., a Cas9 protein.
  • Cas9 systems systems comprising Cas9 or functional mutants thereof are referred to herein as “Cas9 systems” or "CRISPR/Cas9 systems”.
  • the gRNA molecule and the Cas molecule can complex to form a ribonucleoprotein (RNP) complex.
  • RNP ribonucleoprotein
  • gRNA molecule In this application, the terms "gRNA molecule”, “guide RNA”, “guide RNA”, “guide RNA”, “guide RNA molecule” or “gRNA” are used interchangeably and generally refer to Nucleases or other effector molecules (generally complexed with gRNA molecules) to nucleic acid molecules on the target sequence. In certain embodiments, this is accomplished by hybridization of a portion of the gRNA to DNA (e.g., via the gRNA guidance domain) and by binding of a portion of the gRNA molecule to an RNA-guided nuclease or other effector molecule (e.g., at least via the gRNA tracr) the bootstrap.
  • DNA e.g., via the gRNA guidance domain
  • RNA-guided nuclease or other effector molecule e.g., at least via the gRNA tracr
  • a gRNA molecule consists of a single contiguous polynucleotide molecule, referred to herein as a "single guide RNA” or “sgRNA” or the like.
  • the gRNA molecule is itself capable of associating Multiple (eg, two) polynucleotide molecules (generally by hybridization), referred to herein as “dual guide RNA” or “dgRNA” and the like.
  • Cas protein generally refers to the enzyme responsible for cutting DNA in the CRISPR/Cas system. Enzymes from Type I, II, and III CRISPR/Cas systems may be included. For example, Cas3, Cas9, Cas10.
  • Cas9 protein generally refers to the enzyme from the bacterial type II CRISPR/Cas system responsible for cutting DNA. Cas9 can include the wild-type protein and its functional mutants.
  • the term “reduces" the expression level/amount of a gene, gene product such as a protein or a biomarker in a first sample which can be detected by standard methods known in the art (such as those described in this application) About 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70% compared to the expression level/amount of the corresponding gene, gene product such as protein or biomarker in the second sample %, 80%, 85%, 90%, 95%, or 100% overall reduction.
  • the term "decrease” refers to a reduction in the expression level/amount of a gene or biomarker in a first sample, wherein the reduction is the expression level/amount of the corresponding gene or biomarker in a second sample At least about 0.9 times, 0.8 times, 0.7 times, 0.6 times, 0.5 times, 0.4 times, 0.3 times, 0.2 times, 0.1 times, 0.05 times, or 0.01 times.
  • the first sample is a sample obtained from a subject and the second sample is a reference sample.
  • the term "pharmaceutically acceptable carrier” generally refers to one or more non-toxic substances that do not interfere with the effectiveness of the biological activity of the active ingredient.
  • Such formulations will generally contain salts, buffers, preservatives, compatible carriers, adjuvants and, optionally, other therapeutic agents.
  • Such pharmaceutically acceptable formulations may also generally contain compatible solid or liquid fillers, diluents or encapsulating materials suitable for human administration.
  • pharmaceutically acceptable carriers can include liquids such as water, saline, glycerol and ethanol.
  • Auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may also be present in these carriers.
  • adjuvant generally refers to any substance that assists or modulates the action of a drug, including but not limited to an immunological adjuvant, which enhances or diversifies the immune response to an antigen.
  • prevention and/or treatment includes not only preventing and/or treating a disease, but also generally preventing the onset of a disease, slowing or reversing the progression of a disease, preventing or slowing down one or more symptoms associated with a disease onset, reduction and/or alleviation of one or more symptoms associated with the disease, reduction of the severity and/or duration of the disease and/or any symptoms associated therewith and/or prevention of the disease and/or any symptoms associated therewith further increase in the severity of the disease, preventing, reducing or reversing any physiological impairment caused by the disease, and generally beneficial to the patient being treated pharmacological effects.
  • compositions of the present application need not achieve a complete cure or eradicate any symptom or manifestation of a disease to form a viable therapeutic agent.
  • drugs used as therapeutic agents may reduce the severity of a given disease state, but need not eliminate every manifestation of the disease to be considered a useful therapeutic agent.
  • a treatment administered prophylactically need not be fully effective in preventing the onset of the disorder to constitute a viable prophylactic. Simply reducing the effects of disease in a subject (for example, by reducing the number or severity of its symptoms, or by increasing the effectiveness of another treatment, or by producing another beneficial effect), or reducing disease occurrence or The possibility of deterioration is enough.
  • disease or “condition” are used interchangeably and generally refer to any deviation from the normal state of a subject, such as any change in the state of the body or certain organs that prevents or disrupts the performance of function , and/or cause symptoms such as malaise, dysfunction, suffering or even death in those who are sick or come into contact with it.
  • tumor generally refers to all neoplastic cell growth and proliferation, whether malignant or benign, and to all pre-cancerous and cancerous cells and tissues.
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • the term "administration” generally refers to the delivery of proteins, including immunoglobulins, to a human or animal in need thereof by any route known in the art.
  • Pharmaceutically acceptable carriers and formulations or compositions are also well known in the art.
  • Routes of administration may include: intravenous, intramuscular, intradermal, subcutaneous, transdermal, mucosal, intratumoral or mucosal.
  • these terms may refer to the delivery of a vector for expression of a recombinant protein to a cell or to a cell in culture and/or to a cell or organ of a subject. Such administration or introduction can occur in vivo, in vitro or ex vivo.
  • Vectors for recombinant protein or polypeptide expression can be introduced into cells by transfection, which usually means, by physical means (eg, calcium phosphate transfection, electroporation, microinjection, or lipofection) Insertion of heterologous DNA into a cell; infection, which generally refers to the introduction by means of an infectious agent (i.e., a virus); or transduction, which generally refers to the stable infection of a cell by a virus, or the passage of genetic material by a viral agent ( For example, the transfer of bacteriophage) from one microorganism to another.
  • transfection which usually means, by physical means (eg, calcium phosphate transfection, electroporation, microinjection, or lipofection) Insertion of heterologous DNA into a cell
  • infection which generally refers to the introduction by means of an infectious agent (i.e., a virus)
  • transduction which generally refers to the stable infection of a cell by a virus, or the passage of genetic material by a viral agent (
  • the term “contacting” generally means that two or more substances of different types are brought into contact together in any order, in any manner, and for any length of time.
  • the term “contacting” generally refers to the method whereby an antigen binding protein, polypeptide, immunoconjugate, nucleic acid, vector, cell, and/or pharmaceutical composition of the present application is delivered to or placed in direct proximity to a target cell.
  • the delivery may be in vitro or in vivo and may involve the use of recombinant vector systems.
  • "contacting” can include placing a polynucleotide in a beaker, microtiter plate, cell culture flask, or microarray, etc., containing the nucleic acid molecule.
  • contacting can include placing the antibody in a beaker containing the polypeptide, a microtiter plate, a cell In cell culture flasks or microarrays, etc., the contact can occur in vivo, ex vivo or in vitro.
  • the term "effective amount” or “effective dose” generally refers to the amount of active therapeutic agent sufficient to produce the desired therapeutic response without undue adverse side effects such as toxicity, irritation or allergic response.
  • the specific "effective amount” will vary with factors such as the particular condition being treated, the physiological condition of the patient, the type of animal being treated, the duration of the treatment, the presence of concomitant therapies (if any) properties, as well as the specific formulation used and the structure of the compound or its derivatives.
  • an amount will be considered therapeutically effective if it results in, without limitation, one or more of: (a) inhibiting the growth of cancer cells (e.g., AML cells); and (b) killing Dead cancer cells (such as AML cells).
  • the term “subject” generally refers to human or non-human animals, including but not limited to cats, dogs, horses, pigs, cows, sheep, rabbits, mice, rats or monkeys. In certain embodiments, the subject is a human.
  • Subject in need can refer to a patient with or in developing disease or risk that can be treated (e.g., ameliorated, improved, prevented) by inducing T cells to exert specific effects on malignant T cell cytotoxicity subjects (such as patients).
  • circular RNA is generally referred to as circular RNA, which is a new type of noncoding RNA (noncoding RNA, ncRNA) molecule.
  • ncRNA noncoding RNA
  • circular RNA can be divided into three categories: exon circular RNA (ecircRNA), intron circular RNA (circular intronic RNAs, ciRNAs) and exon-intron circRNA (exon-intron circRNA, EIciRNA).
  • the term “about” generally means approximately, in the region of, roughly, or around.
  • a cut-off or a specific value is used to indicate that the stated value may vary by as much as 10% from the recited value. For example, 0.5%, 1%, 1.5%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, 5%, 5.5%, 6%, 6.5%, 7%, 7.5% above or below the specified value %, 8%, 8.5%, 9%, 9.5%, or 10%.
  • the application provides a modified immune effector cell comprising a nucleic acid fraction encoding a fusion protein
  • the fusion protein comprises a first domain that activates an antigen presenting cell (APC) and a second domain that activates an immune effector cell, wherein (i) the first domain includes (a) an activating receptor ligand that binds to the APC or a receptor-binding fragment thereof, or (b) an activating receptor antibody or antigen-binding fragment thereof that binds APC; and (ii) the second domain includes (a) a co-stimulatory ligand for an immune effector cell or a receptor-binding fragment thereof , (b) an antibody or antigen-binding fragment thereof that binds to a costimulatory receptor of an immune effector cell, or (c) a costimulatory receptor or a functional fragment thereof of an immune effector cell.
  • APC antigen presenting cell
  • the APCs are selected from the group consisting of dendritic cells, macrophages, myeloid-derived suppressor cells, monocytes, B cells, T cells, and Langerhans cells.
  • the immune effector cells are selected from the group consisting of T cells, NK cells, NKT cells, macrophages, neutrophils, and granulocytes.
  • the fusion protein is a membrane protein. In some embodiments, the fusion protein is a soluble protein. In some embodiments, the fusion protein is a bispecific antibody. In some embodiments, the C-terminus of the first domain is linked to the N-terminus of the second domain. In some embodiments, the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • first domain and the second domain are linked by a linker.
  • the linker comprises a peptide linker.
  • the linker can be a flexible linker or a rigid linker.
  • the linker is an IgG Fc hinge.
  • the linker may be a trimerization motif selected from the group consisting of T4 fibrin trimerization motif, isoleucine zipper motif, GCN4II motif, Matrilin-1 motif, and type XV A group of collagen trimerization motifs.
  • an APC refers to any cell that displays one or more antigens on its surface, eg, in association with one or more major histocompatibility complex (MHC) proteins.
  • MHC/antigen complexes can be recognized by T cells using their T cell receptors (TCRs) and elicit an immune response.
  • TCRs T cell receptors
  • the APC is selected from the tree group consisting of myelin-derived suppressor cells, myeloid-derived suppressor cells, monocytes, B cells, T cells, and Langerhans cells.
  • APCs activate receptors
  • a molecule can promote APC maturation, proinflammatory state, cytotoxicity, antigen presentation, epitope diffusion, cytokine production, co-stimulation of immune effector cells (such as T cells), or any combination thereof.
  • Activate APCs the first domain of the fusion protein provided by the present invention activates APC by promoting the maturation and activation of APC (eg, DC).
  • the first domain of the fusion protein provided herein activates APC by promoting epitope diffusion between APC and other immune effector cells (eg, T cells).
  • the first domain of the fusion protein provided by the present invention activates APC by promoting antigen presentation by APC.
  • the first domain of the fusion protein provided by the present invention activates APC by promoting the cytotoxicity of APC to foreign bodies (such as cancer cells).
  • the fusion protein provided by the present invention includes a first domain that activates APC, which includes a ligand that binds to an activating receptor of APC or a receptor-binding fragment thereof.
  • Activating receptor refers to a membrane protein expressed on APC, which can trigger a signal to promote the mobilization, differentiation, proliferation and/or activation of APC after binding to a ligand or an antibody.
  • APC activating receptors include, for example, CD40, CD80, CD86, CD91, DEC-205, and DC-SIGN.
  • a “ligand” for a receptor refers to a molecule that selectively binds to the receptor.
  • the ligand is a polypeptide.
  • a "receptor binding fragment” of a ligand refers to a fragment of a ligand that retains its receptor binding ability.
  • Various ligands can stimulate the growth, differentiation, migration and/or activation of dendritic cells or other APCs by binding to activating receptors on APCs. (see Banchereau J et al., Nature (1998) 392:245-52; Young JW et al., Stem Cells (1996) 14:376-387; Cella M et al., Curr Opin Immunol.
  • ligands capable of modulating differentiation, maturation, expansion and/or activation of dendritic cells or other APCs include, for example, CD40 ligand (CD40L), CD80 ligand, CD86 ligand, CD91 ligand (RAP1), DEC- 205 ligand and DC-SIGN ligand.
  • CD40L CD40 ligand
  • CD80 ligand CD80 ligand
  • CD86 ligand CD91 ligand
  • RAP1 CD91 ligand
  • DEC- 205 ligand DEC- 205 ligand
  • DC-SIGN ligand DC-SIGN ligand.
  • the fusion protein provided by the present invention includes a first domain, and the first domain includes the ligand or receptor-binding fragment thereof disclosed in the present invention that binds to an APC-activated receptor.
  • CD40/CD40L is a 48kD transmembrane glycoprotein surface receptor that is a member of the tumor necrosis factor receptor superfamily (TNFRSF).
  • TNFRSF tumor necrosis factor receptor superfamily
  • the typical amino acid sequence of human CD40 is described (see, accession number: ALQ33424.1 GI:957949089), which was originally described as a co-stimulatory receptor expressed on APCs and plays a central role in B- and T-cell activation.
  • the ligand for CD40, CD154 also known as TRAP, T-BAM, CD40 ligand or CD40L
  • CD40L is a type II integral membrane protein. It has been reported that CD40L promotes the induction of dendritic cells and promotes immunogenic response answer happened.
  • the first domain of the fusion protein provided herein includes CD40L or a receptor-binding fragment of CD40L.
  • the receptor binding fragment of CD40L comprises amino acids 119-261 of CD40L (SEQ ID NO: 223).
  • the receptor binding fragment of CD40L comprises the extracellular domain of CD40L.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds to said activated receptor of APC.
  • the first domain of the fusion protein provided by the invention includes an antibody or antigen-binding fragment that binds to CD40, CD80, CD86, CD91, DEC-205 or DC-SIGN.
  • the first knot domain can be an anti-CD40 antibody or an antigen-binding fragment thereof.
  • the fusion protein provided by the invention comprises a first domain that activates antigen-presenting cells (eg, dendritic cells) and a second domain that activates immune effector cells (eg, T cells), wherein
  • the second domain comprises (a) a costimulatory receptor or a functional fragment thereof of the immune effector cell, (b) a costimulatory ligand of the immune effector cell, or a receptor binding fragment thereof, or (c) An antibody, or antigen-binding fragment thereof, that binds to an immune effector cell co-stimulatory receptor.
  • an "immune effector cell” refers to a cell of hematopoietic origin that plays a direct role in the immune response against a target, such as a pathogen, cancer cell, or foreign substance.
  • Immune effector cells include T cells, B cells, natural killer (NK) cells, NKT cells, macrophages, granulocytes, neutrophils, eosinophils, mast cells and basophils.
  • the second domain that activates immune effector cells includes costimulatory receptors for immune effector cells.
  • the immune effector cells are T cells, NK cells, NKT cells, macrophages, neutrophils, or granulocytes.
  • the immune effector cells are T cells.
  • the immune effector cells are NK cells.
  • the immune effector cells are macrophages.
  • stimulation of immune effector cells refers to the induction of primary responses through the binding of stimulatory molecules to their cognate ligands to mediate signaling events in immune effector cells, which can alter the expression of certain genes and/or cytoskeletal structure reorganization, etc.
  • the "stimulatory molecule” of immune effector cells refers to a molecule on immune effector cells that, after binding to cognate ligands usually present on APCs, can mediate signal transduction to promote the maturation, differentiation, Proliferation and/or activation.
  • the T-cell stimulatory molecule, the TCR/CD3 complex triggers T-cell activation.
  • Ligands of stimulatory molecules or "stimulatory ligands” are ligands that are normally present on APCs and are capable of binding to stimulatory molecules on immune effector cells to mediate primary responses of immune effector cells, including but not limited to Maturation, differentiation, activation, initiation of immune response, proliferation, etc.
  • Stimulatory ligands are well known in the art and include MHC class I molecules such as loaded peptides, anti-CD3 antibodies, superagonist anti-CD28 antibodies, and superagonist anti-CD2 antibodies.
  • co-stimulatory signal refers to a signal from a co-stimulatory receptor (such as CD28 or 4-1BB), which in combination with a primary signal (such as TCR/CD3) promotes immune effector cells ( Optimal clonal expansion, differentiation and effector function of T cells).
  • co-stimulatory receptor of immune effector cells refers to molecules on immune effector cells that specifically bind to "costimulatory ligands" to mediate costimulatory responses of immune effector cells, The co-stimulatory response, for example, enhances the activation or proliferation of immune effector cells.
  • Costimulatory receptors for immune effector cells include, but are not limited to, CD28, 4-1BB, ICOS, CD27, OX40, DAP10, CD30, 2B4, CD2, LIGHT, GITR, TLR, DR3, and CD43.
  • a "functional fragment" of a costimulatory receptor is a fragment of a costimulatory receptor that retains the costimulatory receptor's ability to mediate costimulatory signals and stimulate immune effector cells.
  • the functional fragment of a costimulatory receptor retains the costimulatory domain of the costimulatory receptor.
  • the costimulatory domain is the cytoplasmic domain of a costimulatory receptor.
  • a signal from a co-stimulatory receptor of an immune effector cell lowers the activation threshold of the immune effector cell.
  • signals from T cell co-stimulatory receptors result in enhanced TCR signaling events, wherein said TCR signaling is efficient cytokine production (by enhancing transcriptional activity and messenger RNA stability), cell cycle progression, survival, metabolic Necessary for regulatory and T cell responses.
  • costimulatory ligand refers to a molecule that specifically binds to a cognate costimulatory receptor on an immune effector cell, thereby providing a signal in addition to the primary signal provided by the stimulatory molecule , the signal Mediates responses in immune effector cells, including but not limited to proliferation, activation, differentiation, and the like.
  • Costimulatory ligands can be present on APCs (eg, dendritic cells).
  • Costimulatory ligands include but are not limited to CD58, CD70, CD83, CD80, CD86, CD137L (4-1BBL), CD252 (OX40L), CD275 (ICOS-L), CD54 (ICAM-1), CD49a, CD112 (PVRL2) , CD150(SLAM), CD155(PVR), CD265(RANK), CD270(HVEM), TL1A, CD127, IL-4R, GITR-L, TIM-4, CD153(CD30L), CD48, CD160, CD200R(OX2R) and CD44.
  • a "receptor-binding fragment" of a co-stimulatory ligand refers to a fragment of a ligand that retains its ability to bind a receptor.
  • costimulatory receptors and costimulatory ligands are exemplified below. It should be understood that any costimulatory receptor and/or costimulatory ligand provided herein or known in the art can be used as part of the fusion protein provided herein.
  • CD28 Cluster of Differentiation 28
  • CD28 is a protein expressed on T cells that provides co-stimulatory signals for T cell activation and survival.
  • CD28 is the receptor for the CD80 (B7.1) and CD86 (B7.2) proteins.
  • CD28 is a co-stimulatory receptor for optimal clonal expansion, differentiation, and effector function of T cells. CD28 binding lowers the T cell activation threshold and leads to enhanced TCR signaling events that are required for efficient cytokine production (through enhanced transcriptional activity and messenger RNA stability), cell cycle progression, survival, metabolic regulation, and T cell responses. required.
  • CD28 is a key factor in the organization of the immune synapse (IS), where CD28 enhances intimate contacts between T cells and APCs.
  • IS immune synapse
  • the fusion protein provided by the present invention comprises a first domain for activating APC and a second domain for activating immune effector cells, wherein the second domain comprises a CD28 polypeptide or a functional fragment thereof. In some embodiments, the second domain comprises the cytoplasmic domain of CD28. In some embodiments, the fusion protein provided by the present invention comprises a first domain that activates APC and a second domain that activates immune effector cells, wherein the second domain comprises a ligand that binds CD28 or its receptor binding fragment.
  • the fusion protein provided by the present invention comprises a first domain that activates APC and a second domain that activates immune effector cells, wherein the second domain comprises an antibody that binds CD28 or an antigen-binding fragment thereof.
  • the second domain of the fusion protein provided by the invention includes a functional fragment of CD28, and the functional fragment of CD28 includes a part of the intracellular/cytoplasmic domain of CD28, which can serve as a co-stimulatory signal transduction structure domain function.
  • CD28 may have an amino acid sequence corresponding to the sequence of GenBank No. P10747 (P10747.1, GI: 115973) or NP_006130 (NP_006130.1, GI: 5453611) or a functional fragment thereof.
  • the fusion protein disclosed in the present invention may have an amino acid sequence comprising a CD28 cytoplasmic domain or a fragment thereof, and the CD28 cytoplasmic domain corresponds to amino acids 180 to 122 of CD28 (the underlined part of the following sequence, SEQ ID NO: 226).
  • the fusion protein disclosed in the present invention may further comprise an amino acid sequence of a CD28 transmembrane domain or a functional fragment thereof, and the CD28 transmembrane domain corresponds to amino acids 153 to 179. It should be understood that If desired, CD28 sequences shorter or longer than the specifically described domains can be included in the fusion proteins disclosed herein.
  • the second domain is an antibody or antigen-binding fragment thereof that binds a co-stimulatory receptor.
  • co-stimulatory receptor is selected from CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 and CD43.
  • the co-stimulatory receptor is CD28.
  • the second domain can be an anti-CD28 antibody or an antigen-binding fragment thereof.
  • said first domain comprises (a) a ligand that binds an activating receptor of said APC, or a receptor binding fragment thereof, or (b) an antibody that binds an activating receptor of said APC, or an antigen-binding fragment thereof, wherein the APC-activating receptor is selected from the group consisting of CD40, CD80, CD86, CD91, DEC-205 and DC-SIGN.
  • the first domain includes a ligand that binds an activating receptor of APC, or a receptor-binding fragment thereof.
  • the first domain includes a ligand that binds CD40 or a receptor-binding fragment thereof.
  • the first domain includes CD40L.
  • the receptor binding fragment of CD40L comprises amino acids 119-261 of CD40L (SEQ ID NO: 223). In some embodiments, the receptor binding fragment of CD40L comprises the extracellular domain of CD40L. In some embodiments, the first domain of the fusion protein provided herein includes three copies of CD40L or a receptor-binding fragment of CD40L. In some embodiments, the first domain of the fusion protein provided by the invention includes three copies of amino acids 119-261 of CD40L (SEQ ID NO: 223). In some embodiments, the first domain comprises a ligand that binds CD80 or a receptor binding fragment thereof. In some embodiments, the first domain comprises a ligand that binds CD86 or a receptor binding fragment thereof.
  • the first domain comprises a CD28 extracellular domain. In some embodiments, the first domain includes CD28. In some embodiments, the first domain comprises the extracellular domain of CTLA-4. In some embodiments, the first domain includes CTLA-4. In some embodiments, the first domain comprises a ligand that binds CD91 or a receptor binding fragment thereof. In some embodiments, the first domain comprises domain 3 of RAP1. In some embodiments, the first domain comprises RAP1. In some embodiments, the first domain comprises a ligand or receptor-binding fragment thereof that binds DEC-205. In some embodiments, the first domain includes a ligand that binds DC-SIGN or a receptor-binding fragment thereof.
  • the first domain includes ICAM2, ICAM3, CD18 or CEACAM1 or a receptor binding fragment thereof. In some embodiments, the first domain comprises ICAM2 or a receptor binding fragment thereof. In some embodiments, the first domain comprises ICAM3 or a receptor binding fragment thereof. In some embodiments, the first domain comprises CD18, or a receptor binding fragment thereof. In some embodiments, the first domain comprises CEACAM1 or a receptor-binding fragment thereof.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds an activating receptor for APC.
  • the activating receptor of the APC is selected from the group consisting of CD40, CD80, CD86, CD91, DEC-205, and DC-SIGN.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD80.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD86.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD91.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds DEC-205. In some embodiments, the first domain comprises an antibody or antigen-binding fragment thereof that binds DC-SIGN. In some embodiments, the first domain comprises a monoclonal antibody. In some embodiments, the first domain comprises a chimeric antibody. In some embodiments, the first domain comprises a humanized antibody. In some embodiments, the first domain comprises a human antibody. In some embodiments, the first domain comprises Fab, Fab', F(ab')2, Fv, scFv, (scFv)2, single chain antibody, double variable region antibody, bispecific antibody, nanobody antibody or single variable domain antibody. In some embodiments, the first domain comprises a human antibody. In some embodiments, the first construct comprises a scFv.
  • the first domain of the fusion protein provided herein includes an anti-CD40 antibody or an antigen-binding fragment thereof. In some embodiments, the first domain of the fusion protein provided herein includes an anti-CD40 scFv. In some embodiments, the anti-CD40 antibody or antigen-binding fragment thereof comprises an antibody labeled F2.103, F5.157, F5.77, 4D11, A40C or 119, as shown in Table 1 below.
  • the first domain of the fusion protein provided herein includes anti-CD40 scFv.
  • the first structural domain of the fusion protein provided by the invention comprises an anti-CD40scFv, and the anti-CD40scFv has at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97% , at least 98% or at least 99% sequence identity.
  • the first structural domain of the fusion protein provided by the invention comprises having SEQ ID NO:227 Anti-CD40 scFv to the amino acid sequence shown in any one of SEQ ID NO:232.
  • the second domain of the fusion protein provided by the present invention includes costimulatory receptors of immune effector cells or functional fragments thereof, wherein the immune cells are T cells, NK cells, NKT cells, macrophages, Neutrophils or granulocytes.
  • the costimulatory receptors of the immune effector cells are selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, DR3, and CD43.
  • the second domain of the fusion protein provided by the invention includes a functional fragment of a co-stimulatory receptor selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, Group consisting of CD30, CD2, LIGHT, GITR, DR3 and CD43.
  • a co-stimulatory receptor selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, Group consisting of CD30, CD2, LIGHT, GITR, DR3 and CD43.
  • the functional fragment comprises the cytoplasmic domain of a co-stimulatory receptor.
  • the second domain of the fusion protein provided by the invention further includes a transmembrane domain of a co-stimulatory receptor.
  • the second domain of the fusion protein provided by the invention includes functional fragments of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, DR3 or CD43.
  • the second domain of the fusion protein provided by the invention comprises the cytoplasmic domain of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, DR3 or CD43 .
  • the second domain of the fusion protein provided by the invention includes an antibody or an antigen-binding fragment thereof that binds to a co-stimulatory receptor of an immune effector cell.
  • the immune effector cells may be selected from the group consisting of T cells, NK cells, NKT cells, macrophages, neutrophils and granulocytes.
  • the costimulatory receptors of the immune effector cells are selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, DR3, and CD43.
  • the second domain of the fusion protein provided by the invention comprises an antibody or an antibody that binds to CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, DR3 or CD43 Antigen-binding fragments.
  • the second domain comprises a monoclonal antibody. In some embodiments, the second domain comprises a chimeric antibody. In some embodiments, the second domain comprises a humanized antibody. In some embodiments, the second domain comprises a human antibody. In some embodiments, the second domain comprises Fab, Fab', F(ab')2, Fv, scFv, (scFv)2, single chain antibody, dual variable region antibody, bispecific antibody, nano antibody or single variable domain antibody. In some embodiments, the second domain comprises a human antibody. In some embodiments, the second domain comprises a scFv.
  • the second domain of the fusion protein provided herein includes an anti-CD28 antibody or an antigen-binding fragment thereof. In some embodiments, the second domain of the fusion protein provided herein includes an anti-CD28 scFv. In some embodiments, the anti-CD28 antibody or antigen-binding fragment thereof comprises an antibody labeled 1412.
  • the second domain of the fusion protein provided by the invention comprises an anti-CD28scFv
  • the anti-CD28scFv has at least 80%, at least 85%, at least 86%, at least 87% of the sequence shown in SEQ ID NO:233 , at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity sex.
  • the second domain of the fusion protein provided by the invention includes an anti-CD28 scFv having the amino acid sequence shown in SEQ ID NO:233.
  • the fusion protein (i.e., LACO-Stim molecule) described in the present invention can comprise APC activator (ligand or antibody that binds to activating receptor) and immune effector cell activator (co-stimulatory) disclosed in the present invention or other known in the art. receptors or antibodies that bind co-stimulatory receptors).
  • APC activator ligand or antibody that binds to activating receptor
  • immune effector cell activator co-stimulatory
  • receptors or antibodies that bind co-stimulatory receptors include CD40-C28LACO-Stim fusion proteins.
  • APCs e.g., dendritic cells
  • CD40/CD40L signaling e.g., T cells
  • LACO-Stim (1) Ligand of APC activating receptor + co-stimulatory receptor (eg, CD40L-CD28)
  • the fusion protein provided by the invention includes a first domain that activates antigen-presenting cells (APC) and a second domain that activates immune effector cells, wherein the first domain includes an activation receptor that binds to APC.
  • the second domain includes costimulatory receptors of immune effector cells or functional fragments thereof.
  • the second domain comprises the cytoplasmic domain of a costimulatory receptor of an immune effector cell.
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the fusion proteins provided herein are membrane fusion proteins.
  • the first domain and the second domain are linked by a linker.
  • the linker can be a flexible linker or a rigid linker.
  • the linker has Amino acid sequence of GSGGGGSGGGGSGGGGS.
  • the linker has the amino acid sequence of GGGGS.
  • the first domain comprises a ligand or receptor-binding fragment thereof that binds an APC-activating receptor selected from the group consisting of CD40, CD80, CD86, CD91, DEC-205, and DC- A group composed of SIGN.
  • the second domain comprises a co-stimulatory receptor or a functional fragment thereof selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, Group consisting of LIGHT, GITR, TLR, DR3 and CD43.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes CD28 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor-binding fragment, and the second structural domain includes 4-1BB cytoplasmic domain.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes the ICOS cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes CD27 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes the OX40 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes the DAP10 cytoplasmic domain .
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes CD40L or its receptor binding fragment, and the second domain includes a 2B4 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes CD30 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes CD2 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes the LIGHT cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes GITR cytoplasmic domain .
  • the fusion protein provided by the invention has a first domain and a second domain, and the first domain includes CD40L or a receptor binding fragment thereof, wherein the second domain comprises a TLR cytoplasmic domain.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes DR3 cytoplasmic domain .
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes CD43 cytoplasmic domain .
  • the receptor-binding fragment of CD40L may be amino acids 119-261 of CD40L (SEQ ID NO: 223).
  • the first domain comprises full-length CD40L.
  • the fusion protein provided by the invention has an amino acid sequence that is at least 80%, at least 85%, at least 86% identical to the sequence of the fusion protein marked as 40L. %, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or At least 99% identity.
  • CD40L functional fragment or full-length CD40L in the fusion protein exemplified in the present invention can be replaced by different ligands of APC-activating receptors disclosed in the present invention or other known in the art, Including, for example, the extracellular domain of a CD80 ligand (e.g., CD28 or CTLA-4) or its full length, a CD86 ligand (e.g., CD28 or CTLA-4), a CD91 ligand (e.g., RAP1 ), a DEC-205 ligand or DC-SIGN ligands (eg, ICAM2, ICAM3, CD18, or CEACAM1).
  • a CD80 ligand e.g., CD28 or CTLA-4
  • CD91 ligand e.g., RAP1
  • DEC-205 ligand or DC-SIGN ligands eg, ICAM2, ICAM3, CD18, or CEACAM1.
  • the CD28 cytoplasmic domain in the fusion protein exemplified in the present invention can use the cytoplasmic structure of different co-stimulatory factors disclosed in the present invention or other known immune effector cells in the art Domain substitutions, including, for example, the cytoplasmic domain of 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3, or CD43; or 4-1BB, ICOS, CD27, OX40, Different functional fragments of DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 or CD43, the functional fragments retain the function of the full-length protein to activate immune effector cells.
  • the cytoplasmic domain of 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 or CD43 the functional fragments retain the function of the full-length protein to activate immune effector cells
  • LACO-Stim (2) APC activating receptor ligand + co-stimulatory receptor binding antibody (e.g., aCD28- CD40L)
  • the fusion protein provided by the invention includes a first domain that activates APC and a second domain that activates immune effector cells, wherein the first domain includes a ligand that binds to an activating receptor of APC or its A receptor binding fragment, and wherein said second domain comprises an antibody or antigen binding fragment thereof that binds a co-stimulatory receptor of an immune effector cell.
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the fusion proteins provided herein are soluble antibody-based proteins.
  • the two domains of the fusion proteins disclosed herein are linked by a trimerization motif.
  • the linker is a trimerization motif selected from the group consisting of a T4 fibrin trimerization motif, an isoleucine zipper, a GCN4II motif, a Matrilin-1 motif, and a type XV collagen trimerization motif composed of groups.
  • the linker is a T4 fibrin trimerization motif.
  • the first domain comprises a ligand or receptor-binding fragment thereof that binds an APC-activating receptor selected from the group consisting of CD40, CD80, CD86, CD91, DEC-205, and DC-SIGN group.
  • the second domain comprises an antibody or antigen-binding fragment thereof that binds a co-stimulatory receptor of an immune effector cell, wherein the co-stimulatory receptor is selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40 , DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3, and CD43.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-CD28 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes CD40L or its receptor-binding fragment, and the second domain includes an anti-4-1BB antibody or an antigen-binding fragment thereof.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-ICOS antibody or its Antigen-binding fragments.
  • the fusion protein provided by the present invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-CD27 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-OX40 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-DAP10 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes CD40L or its receptor binding fragment, The second domain comprises an anti-2B4 antibody or an antigen-binding fragment thereof.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-CD30 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-CD2 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-LIGHT antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-GITR antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-TLR antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-DR3 antibody or its Antigen-binding fragments.
  • the fusion protein provided by the invention has a first structural domain and a second structural domain, the first structural domain includes CD40L or its receptor binding fragment, and the second structural domain includes an anti-CD43 antibody or its Antigen-binding fragments.
  • the receptor binding fragment of CD40L may have amino acids 119-261 of CD40L (SEQ ID NO: 223).
  • fusion proteins provided herein have a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising an anti-CD28 antibody or an antigen-binding fragment thereof.
  • the anti-CD28 antibody or antigen-binding fragment can be any anti-CD28 antibody or antigen-binding fragment disclosed in the present invention or otherwise known in the art that can activate CD28 signal transmission.
  • the anti-CD28 antibody or antigen-binding fragment is an antibody labeled 1412.
  • the anti-CD28 antibody or antigen-binding fragment thereof comprises an anti-CD28 scFv having the amino acid sequence set forth in SEQ ID NO:233.
  • the fusion protein provided herein has an amino acid sequence that is at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, At least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity.
  • the extracellular domain of CD40L in the fusion protein exemplified by the present invention can use the extracellular domain or receptor of different ligands of APC-activating receptors disclosed in the present invention or other known in the art Replacement of antibody-binding fragments, including, for example, CD80 ligands (e.g., CD28 or CTLA-4), CD86 ligands (e.g., CD28 or CTLA-4), CD91 ligands (e.g., RAP1), DEC-205 ligands, or DC- The extracellular or receptor binding domain of a SIGN ligand (eg, ICAM2, ICAM3, CD18, or CEACAM1).
  • a SIGN ligand eg, ICAM2, ICAM3, CD18, or CEACAM1
  • the anti-CD28 antibody or antigen-binding fragment in the fusion protein exemplified in the present invention can use different co-stimulatory factors disclosed in the present invention or other known in the art to bind and activate immune effector cells Replacement of antibodies or antigen-binding fragments of antibodies or antigen-binding fragments including, for example, antibodies or antigen-binding fragments that bind 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3, or CD43.
  • Exemplary LACO-Stim (3) Antibody to APC-activating receptor + Antibody to co-stimulatory receptor (e.g., aCD40/aCD28 Bispecific Ab)
  • the invention provides bispecific antibodies.
  • bispecific antibody refers to an antibody that has binding specificities for at least two different antigenic epitopes.
  • the epitopes can be from the same antigen or from two different antigens.
  • the fusion protein provided by the invention includes a first domain that activates APC and a second domain that activates immune effector cells, wherein the first domain includes an antibody that binds to an activated receptor of APC or an antigen thereof A binding fragment, and wherein said second domain comprises an antibody or antigen-binding fragment thereof that binds a co-stimulatory receptor of an immune effector cell.
  • the bispecific antibodies disclosed herein have binding specificity for (1) activating receptors of APCs (e.g., dendritic cells) and (2) costimulatory receptors of immune effector cells (e.g., T cells) .
  • APCs e.g., dendritic cells
  • costimulatory receptors of immune effector cells e.g., T cells
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the first domain and the second domain are linked by a linker.
  • the linker can be a flexible linker or a rigid linker.
  • the linker has the amino acid sequence of GSGGGGSGGGGSGGGGS.
  • the linker has the amino acid sequence of GGGGS.
  • the fusion protein provided by the present invention is a bispecific antibody, which includes the first domain of an activating receptor that binds to APC or an antigen-binding fragment thereof, and a costimulatory receptor antibody or an antibody that binds to an immune effector cell.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40, CD80, CD86, CD91, DEC-205, or DC-SIGN.
  • the second domain comprises an antibody or antigen-binding fragment thereof that binds CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3, or CD43.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • CD28 antibody or antigen-binding fragment thereof In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain. 4-1BB antibody or antigen-binding fragment thereof. In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain. DAP10 antibody or antigen-binding fragment thereof. In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain. 2B4 antibody or antigen-binding fragment thereof. In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • the present invention provides A bispecific antibody with a first domain and a second domain, the first domain is an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes an anti-CD2 antibody or an antigen-binding fragment thereof.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain. LIGHT antibody or antigen-binding fragment thereof.
  • the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • GITR antibodies or antigen-binding fragments thereof In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain. TLR antibodies or antigen-binding fragments thereof. In some embodiments, the present invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof, and a second domain comprising an anti-CD40 domain.
  • the invention provides a bispecific antibody comprising a first domain which is an anti-CD40 antibody or an antigen-binding fragment thereof and a second domain comprising an anti-CD43 Antibodies or antigen-binding fragments thereof.
  • bispecific antibodies can be produced recombinantly using the co-expression of two immunoglobulin heavy chain/light chain pairs. See, eg, Milstein et al. (1983) Nature 305:537-39.
  • chemical linkage can be used to prepare bispecific antibodies. See, eg, Brennan et al. (1985) Science 229:81.
  • Bispecific antibodies include bispecific antigen-binding fragments. See, eg, Holliger et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-48; Gruber et al. (1994) J. Immunol. 152:5368.
  • bispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs with different specificities (see Milstein and Cuello, Nature 305:537 (1983), WO 93/08829, and Traunecker et al., EMBO J. 10:3655 (1991 )), and "knob-in-hole” engineering (see eg, US Patent 5,731,168).
  • Multispecific antibodies can also be prepared by: making antibody Fc-engineered electrostatic steering effects on heterodimerized molecules (WO2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., U.S. Pat.
  • bispecific Antibodies engineered to contain three or more functional antigen binding sites are also included herein (see, eg, US2006/0025576A1).
  • bispecific Antibodies can also be constructed by linking two different antibodies or parts thereof.
  • bispecific antibodies can include Fab, F(ab')2, Fab', scFv and sdAb from two different antibodies.
  • the first domain of the fusion protein provided herein includes an anti-CD40 antibody or an antigen-binding fragment thereof.
  • the anti-CD40 antibody or antigen-binding fragment can be any anti-CD40 antibody or antigen-binding fragment disclosed in the present invention or otherwise known in the art that can activate CD40 signal transduction.
  • the anti-CD40 antibody or antigen-binding fragment thereof comprises an antibody labeled F2.103, F5.157, F5.77, 4D11, A40C, or 119 as provided in Table 1 above.
  • the anti-CD40 antibody or antigen-binding fragment thereof comprises an anti-CD40 scFv having the amino acid sequence set forth in any one of SEQ ID NO:227 to SEQ ID NO:232.
  • the anti-CD28 antibody or antigen-binding fragment can be any anti-CD28 antibody or antigen-binding fragment disclosed herein or otherwise known in the art that activates CD28 signaling.
  • the anti-CD28 antibody or antigen-binding fragment is an antibody labeled 1412.
  • the anti-CD28 antibody or antigen-binding fragment thereof comprises an anti-CD28 scFv having the amino acid sequence set forth in SEQ ID NO:233.
  • the fusion protein provided herein has an amino acid sequence that is at least 80% identical to the sequence of a fusion protein labeled 1412-F2.103, 1412-F5.157, 1412-F5.77, or 1412-4D11 , at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity.
  • the anti-CD40 antibody or antigen-binding fragment thereof in the exemplified fusion protein of the present invention can be bound by antibodies or antigens disclosed in the present invention or other known in the art that bind to different APC-activated receptors Fragment substitutions, including for example CD80, CD86, CD91, DEC-205 or DC-SIGN.
  • the anti-CD28 antibodies or antigen-binding fragments in the fusion proteins exemplified in the present invention can be combined with different co-stimulatory factors disclosed in the present invention or other known in the art that bind immune effector cells
  • Antibody or antigen-binding fragments are substituted, including for example antibodies or antigen-binding fragments that bind to 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 or CD43.
  • LACO-Stim (4) antibody to activate receptor + co-stimulatory receptor (eg, aCD40-CD28; aCD40-4- 1BB)
  • the fusion protein provided by the present invention includes a first domain that activates APC and a second domain that activates immune effector cells, wherein the first domain includes an antibody that binds to an activated receptor of APC or its An antigen-binding fragment, and wherein said second domain comprises a co-stimulatory receptor of an immune effector cell or a functional fragment thereof.
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the present invention provides antibody-based membrane fusion proteins.
  • the first and second domains are connected by a CD8 hinge, a CD28 hinge, or an IgG Fc region.
  • the fusion protein provided by the present invention includes a first domain and a second domain, wherein the first domain includes an antibody or an antigen-binding fragment thereof that binds to an activated receptor of APC, and the second Domains include antibodies or antigen-binding fragments thereof that bind costimulatory receptors of immune effector cells.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40, CD80, CD86, CD91, DEC-205, or DC-SIGN.
  • the first domain of the fusion protein provided herein includes an anti-CD40 antibody or an antigen-binding fragment thereof.
  • the anti-CD40 antibody or antigen-binding fragment may be any anti-CD40 antibody or antigen-binding fragment disclosed herein or otherwise known in the art that activates CD40 signaling.
  • the anti-CD40 antibody or antigen-binding fragment thereof comprises an antibody labeled F2.103, F5.157, F5.77, 4D11, A40C, or 119 as provided in Table 1 above.
  • the second domain comprises a co-stimulatory receptor or a functional fragment thereof selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, Group consisting of GITR, TLR, DR3 and CD43.
  • the second domain comprises the cytoplasmic domain of a co-stimulatory receptor selected from the group consisting of CD28, 4-1BB, ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2 , LIGHT, GITR, TLR, DR3 and CD43 group.
  • the second domain of the fusion protein provided herein includes CD28 cytoplasmic domain. In some embodiments, the second domain of the fusion protein provided by the invention can have at least 85%, at least 88%, at least 90%, at least 95%, at least 98%, or 100% of the sequence shown in the CD28 cytoplasmic domain. % identity amino acid sequence. In some embodiments, the second domain of the fusion protein provided by the invention further includes a CD28 transmembrane domain.
  • the second domain of the fusion protein provided herein includes a 4-1BB cytoplasmic domain.
  • the second domain of a fusion protein provided herein can have a cytoplasmic domain that is at least 85%, at least 88%, at least 90%, at least 95%, at least 98%, or 100% identical to a 4-1BB cytoplasmic domain.
  • Sexual amino acid sequence In some embodiments, the second domain of the fusion protein provided by the invention further includes a 4-1BB transmembrane domain.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a CD28 cytoplasmic structure area.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a 4-1BB cell quality domain.
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes an ICOS cytoplasmic structure area.
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a CD27 cytoplasmic structure area. In some embodiments, the fusion protein provided by the invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes an OX40 cytoplasmic structure area. In some embodiments, the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a DAP10 cytoplasmic structure area.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a 2B4 cytoplasmic structure area. In some embodiments, the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a CD30 cytoplasmic structure area. In some embodiments, the fusion protein provided by the invention has a first domain and a second domain, and the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, wherein the second domain comprises a CD2 cytoplasmic domain.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a LIGHT cytoplasmic structure area. In some embodiments, the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a GITR cytoplasmic structure area. In some embodiments, the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a TLR cytoplasmic structure area.
  • the fusion protein provided by the invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a DR3 cytoplasmic structure area.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a CD43 cytoplasmic structure area.
  • the first domain comprises full-length CD40L.
  • the fusion protein provided by the present invention further includes a transmembrane region.
  • the transmembrane regions are derived from the same costimulatory receptor.
  • the transmembrane regions are derived from different costimulatory receptors.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a CD28 transmembrane region and CD28 cytoplasmic domain.
  • the fusion protein provided by the present invention has a first domain and a second domain, the first domain includes an anti-CD40 antibody or an antigen-binding fragment thereof, and the second domain includes a 4-1BB spanning Membrane region and 4-1BB cytoplasmic domain.
  • the amino acid sequence of the fusion protein provided by the invention is the same as that marked as F2.103.CD28, F5.157.CD28, F5.77.CD28, F5.157.BB, F5.77.BB,
  • the sequence of the fusion protein of 4D11.CD28, A40C.CD28 or 119.CD28 has at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity.
  • the anti-CD40 antibody or antigen-binding fragment thereof in the exemplified fusion protein of the present invention can be disclosed by the present invention or other antibodies known in the art that bind and activate different activating receptors of APC or antigen-binding fragments, including for example CD80, CD86, CD91, DEC-205 or DC-SIGN.
  • the CD28 cytoplasmic domain or 4-1BB cytoplasmic domain in the fusion protein exemplified in the present invention can be different from the immune effector cells disclosed in the present invention or other known in the art.
  • Cytoplasmic domain substitutions of co-stimulatory factors including for example the cytoplasmic domains of ICOS, CD27, OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 or CD43; or 4-1BB, ICOS, CD27, Different functional fragments of OX40, DAP10, 2B4, CD30, CD2, LIGHT, GITR, TLR, DR3 or CD43, the functional fragments retain the function of the full-length protein to activate immune effector cells.
  • Exemplary LACO-Stim (5) antibody to APC activating receptor + ligand to co-stimulatory receptor (e.g., aCD40-CD80; aCD40-CD86)
  • the fusion protein provided by the present invention includes a first domain that activates APC and a second domain that activates immune effector cells, wherein the first domain includes an activation receptor that binds to an antigen-presenting cell An antibody or antigen-binding fragment thereof, and wherein said second domain comprises a co-stimulatory ligand for an immune effector cell or a receptor-binding fragment thereof.
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the present invention provides antibody-based soluble fusion proteins.
  • the present invention provides antibody-based soluble fusion proteins.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40, CD80, CD86, CD91, DEC-205, and DC-SIGN.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD80.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD86.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD91.
  • the first domain comprises an antibody or an antigen binding thereof that binds DEC-205 Combine fragments.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds DC-SIGN.
  • the antibodies and antigen-binding fragments may be any antibodies or antigen-binding fragments disclosed in the present invention or otherwise known in the art.
  • the fusion protein provided by the present invention includes a first domain and a second domain, wherein the first domain includes an antibody or an antigen-binding fragment thereof that binds to an APC-activated receptor, and the second domain Domains include costimulatory ligands or receptor binding fragments thereof for immune effector cells.
  • the first domain comprises an antibody or antigen-binding fragment thereof that binds CD40, CD80, CD86, CD91, DEC-205, or DC-SIGN.
  • the second domain comprises a protein selected from the group consisting of CD58, CD70, CD83, CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL-4R , GITR-L, TIM-4, CD153, CD48, CD160, CD200R and CD44 group of ligands, or receptor-binding fragments thereof.
  • the first domain of the fusion protein provided herein includes an anti-CD40 antibody or an antigen-binding fragment thereof.
  • the anti-CD40 antibody or antigen-binding fragment may be any anti-CD40 antibody or antigen-binding fragment disclosed herein or otherwise known in the art that activates CD40 signaling.
  • the anti-CD40 antibody or antigen-binding fragment thereof comprises an antibody labeled as F2.103, F5.157, F5.77, 4D11, A40C or 119 as provided in Table 1 above.
  • the anti-CD40 antibody or antigen-binding fragment thereof comprises an anti-CD40 scFv having the amino acid sequence set forth in any one of SEQ ID NO:227 to SEQ ID NO:232.
  • the fusion protein includes a first domain and a second domain, the first domain includes an antibody or antigen-binding fragment that binds CD40, and the second domain includes a ligand and its receptor A binding fragment, the ligand being selected from the group consisting of CD58, CD70, CD83, CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL-4R, GITR-L , TIM-4, CD153, CD48, CD160, CD200R, CD44 group.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD58 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD70 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD83 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD80 or a receptor-binding fragment thereof.
  • the fusion protein includes a first domain and a second domain, the first domain includes an antibody or antigen-binding fragment that binds CD40, and the second domain includes CD86 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD137L or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD252 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD275 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD54 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD49a or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD112 or a receptor-binding fragment thereof. In some embodiments, the fusion protein includes a first domain including an antibody that binds CD40 or an antigen-binding fragment thereof, and a second domain that includes CD150 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD155 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD265 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD270 or a receptor-binding fragment thereof.
  • the fusion protein includes a first domain that includes an antibody or antigen-binding fragment that binds CD40, and a second domain that includes TL1A or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD127 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising IL-4R or a receptor-binding fragment thereof.
  • the fusion protein includes a first domain that includes an antibody or antigen-binding fragment that binds CD40, and a second domain that includes GITR-L or a receptor-binding fragment thereof. In some embodiments, the fusion protein includes a first domain and a second domain, the first domain includes an antibody or antigen-binding fragment that binds CD40, and the second domain includes TIM-4 or a receptor-binding fragment thereof. In some embodiments, the fusion protein includes a first domain and a second domains, the first domain includes an antibody or antigen-binding fragment that binds to CD40, and the second domain includes CD153 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD48 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD160 or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising an antibody or antigen-binding fragment that binds CD40 and a second domain comprising CD200R or a receptor-binding fragment thereof.
  • the fusion protein includes a first domain including an antibody that binds CD40 or an antigen-binding fragment thereof, and a second domain that includes CD44 or a receptor-binding fragment thereof.
  • a suitable receptor-binding fragment of a ligand that retains its binding affinity for the receptor and functions to activate the receptor.
  • the anti-CD40 antibody or antigen-binding fragment thereof in the exemplified fusion protein of the present invention can be disclosed in the present invention or other antibodies known in the art that bind to different activation receptors of APC or Antigen binding fragments are substituted, including for example CD80, CD86, CD91, DEC-205 or DC-SIGN.
  • LACO-Stim (6) APC-activating receptor ligand + co-stimulatory ligand (e.g., CD40L-CD86; CD40L-CD80)
  • the fusion protein provided by the invention includes a first domain that activates APC and a second domain that activates immune effector cells, wherein the first domain includes a ligand that binds to an activating receptor of APC or its A receptor binding fragment, and wherein said second domain comprises a co-stimulatory ligand for an immune effector cell or a receptor binding fragment thereof.
  • the C-terminus of the first domain is linked to the N-terminus of the second domain.
  • the N-terminus of the first domain is linked to the C-terminus of the second domain.
  • the fusion protein provided by the invention includes a first domain and a second domain
  • the first domain includes a ligand or a functional fragment thereof that binds to an activating receptor selected from the group consisting of CD40 , CD80, CD86, CD91, DEC-205, and DC-SIGN
  • the second domain includes a co-stimulatory ligand or a receptor-binding fragment thereof
  • the co-stimulatory ligand is selected from the group consisting of CD58, CD70, CD83 , CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL-4R, GITR-L, TIM-4, CD153, CD48, CD160, CD200R, CD44 composed of groups.
  • the first domain of the fusion protein provided herein includes the extracellular domain of CD40L. In some embodiments, the first domain of the fusion protein provided by the invention may have at least 85%, at least 88%, at least 90%, at least 95%, at least 98%, or 100% identity with the extracellular domain of CD40L. Sexual amino acid sequence List. In some embodiments, the first domain of the fusion protein provided by the invention includes full-length CD40L. In some embodiments, the first domain of the fusion protein provided by the invention may have amino acids 119-261 of CD40L (SEQ ID NO: 223). In some embodiments, the first domain of the fusion protein provided by the present invention includes three copies of CD40L or a functional fragment thereof.
  • the first domain of the fusion protein provided herein includes three copies of the extracellular domain of CD40L. In some embodiments, the first domain of the fusion protein provided by the invention includes three copies of amino acids 119-261 of CD40L (SEQ ID NO: 223).
  • the fusion protein comprises a first domain comprising CD40L or its receptor binding fragment and a second domain comprising a ligand or its receptor binding Fragments, the ligand being selected from the group consisting of CD58, CD70, CD83, CD80, CD86, CD137L, CD252, CD275, CD54, CD49a, CD112, CD150, CD155, CD265, CD270, TL1A, CD127, IL-4R, GITR-L, Group consisting of TIM-4, CD153, CD48, CD160, CD200R, CD44.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD58 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD70 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD83 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD80 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD86 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD137L or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD252 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD275 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising CD54 or a receptor-binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising CD49a or a receptor-binding fragment thereof.
  • the fusion protein includes a first domain and a second domain, the first domain includes CD40L or a receptor binding fragment thereof, and the second domain Domains include CD112 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising binding CD40L or a receptor binding fragment thereof and a second domain comprising CD150 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD155 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD265 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD270 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising TL1A or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD127 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising IL-4R or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor-binding fragment thereof and a second domain comprising GITR-L or a receptor-binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising TIM-4 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD153 or a receptor binding fragment thereof.
  • the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD48 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD160 or a receptor binding fragment thereof. In some embodiments, the fusion protein comprises a first domain comprising CD40L or a receptor binding fragment thereof and a second domain comprising CD200R or a receptor binding fragment thereof. In some embodiments, the fusion protein includes a first domain that binds CD40L or a receptor-binding fragment thereof and a second domain that includes CD44 or a receptor-binding fragment thereof.
  • One of ordinary skill in the art can readily determine a suitable receptor-binding fragment of a ligand that retains its binding affinity for the receptor and functions to activate the receptor.
  • CD40L or its receptor-binding fragment in the fusion protein exemplified in the present invention can be replaced by different ligands of APC-activating receptors disclosed in the present invention or other known in the art, including example Such as CD80 ligand (for example, CD28 or CTLA-4), CD86 ligand (for example, CD28 or CTLA-4), CD91 ligand (for example, RAP1), DEC-205 ligand or DC-SIGN ligand (for example, Extracellular domain or full-length ligand of ICAM2, ICAM3, CD18 or CEACAM1).
  • CD80 ligand for example, CD28 or CTLA-4
  • CD86 ligand for example, CD28 or CTLA-4
  • CD91 ligand for example, RAP1
  • DEC-205 ligand DEC-205 ligand
  • DC-SIGN ligand for example, Extracellular domain or full-length ligand of ICAM2, ICAM3, CD18 or CEACAM1.
  • the antigen-specific extracellular domain of a chimeric antigen receptor recognizes and specifically binds an antigen, usually an antigen expressed on the surface of a malignant tumor (eg, CD7).
  • an antigen usually an antigen expressed on the surface of a malignant tumor (eg, CD7).
  • an antigen-specific extracellular domain binds the antigen with an affinity constant or interaction affinity (KD) of about 0.1 pM to about 10 ⁇ M, or about 0.1 pM to about 1 ⁇ M, or about 0.1 pM to about 100 nM
  • KD affinity constant or interaction affinity
  • Methods for determining interaction affinity are known in the art.
  • An antigen-specific extracellular domain suitable for use in a CAR of the present disclosure can be any antigen-binding polypeptide, a wide variety of which are known in the art.
  • the antigen binding domain is a single chain Fv (scFv).
  • scFv single chain Fv
  • Other antibody-based recognition domains cAb VHH (camel antibody variable domain) and its humanized form
  • T cell receptor (TCR) based recognition domains such as single chain TCR (scTv) are also suitable for use.
  • Suitable antigens may include T cell-specific antigens and/or antigens that are not specific for T cells.
  • the antigen specifically bound by the CAR-T cell chimeric antigen receptor and the CAR-T cell deficient antigen is an antigen expressed on malignant T cells, more preferably in combination with non-malignant T cells. cells compared to antigens overexpressed on malignant T cells.
  • a "malignant T cell” is a T cell derived from a T cell malignancy.
  • T cell malignancies refers to a broad and highly heterogeneous group of malignancies derived from T cell precursors, mature T cells or natural killer cells.
  • T-cell malignancies include T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1 positive (HTLV-1 +) Adult T-cell leukemia/lymphoma (ATL), T-cell prolymphocytic leukemia (T-PLL), and various peripheral T-cell lymphomas (PTCL), including but not limited to angioimmunoblastic T-cell lymphoma ( AITL), ALK-positive anaplastic large cell lymphoma, and ALK-negative anaplastic large cell lymphoma.
  • T-ALL T-cell acute lymphoblastic leukemia/lymphoma
  • LGL lymphocyte
  • HTLV-1 + human T-cell leukemia virus type 1 positive
  • ATL adult T-cell leukemia/lymphoma
  • T-PLL T-cell prolymphocytic leukemia
  • PTCL peripheral T-cell lymphomas
  • the CAR-T cells of the present application comprise the extracellular domain of a chimeric antigen receptor that specifically binds to CD7.
  • CD7 is a T cell surface membrane-associated glycoprotein.
  • CD7 can be overexpressed in T cell malignancies including T cell acute lymphoblastic leukemia (T-ALL) and non-Hodgkin's T cell lymphoma (NHL).
  • T-ALL T cell acute lymphoblastic leukemia
  • NHL non-Hodgkin's T cell lymphoma
  • the CAR-T cells of the present disclosure can be used to target malignant T cells overexpressing CD7.
  • the extracellular antigen binding domain comprises a heavy chain variable region (VH) and a light chain variable region (VL), wherein the VH comprises the amino acid sequence shown in SEQ ID NO:61 to SEQ ID NO:73.
  • VL comprises the amino acid sequence shown in any one of SEQ ID NO: 148 to SEQ ID NO: 160.
  • VH comprises the amino acid sequence shown in SEQ ID NO:61
  • said VL comprises the amino acid sequence shown in SEQ ID NO:148;
  • the VH comprises the amino acid sequence shown in SEQ ID NO:62, and the VL comprises the amino acid sequence shown in SEQ ID NO:149; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:63
  • the VL comprises the amino acid sequence shown in SEQ ID NO:150; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:64, and the VL comprises the amino acid sequence shown in SEQ ID NO:151; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:65
  • the VL comprises the amino acid sequence shown in SEQ ID NO:152; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:66
  • the VL comprises the amino acid sequence shown in SEQ ID NO:153; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:67
  • the VL comprises the amino acid sequence shown in SEQ ID NO:154; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:68, and the VL comprises the amino acid sequence shown in SEQ ID NO:155; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:69
  • the VL comprises the amino acid sequence shown in SEQ ID NO:156; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:70, and the VL comprises the amino acid sequence shown in SEQ ID NO:157; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:71, and the VL comprises the amino acid sequence shown in SEQ ID NO:158; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:72, and the VL comprises the amino acid sequence shown in SEQ ID NO:159; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:73
  • the VL comprises the amino acid sequence shown in SEQ ID NO:160 The amino acid sequence shown.
  • the extracellular antigen binding domain comprises a scFv.
  • the extracellular antigen binding domain can be an anti-CD7 scFv.
  • VL and VH are linked by a linker.
  • linker comprises a polypeptide linker
  • the polypeptide linker comprises an amino acid sequence represented by (GGGGS)n, wherein n is any integer from 1 to 5.
  • the antigen-specific extracellular domain of the chimeric antigen receptor of the present application can specifically bind to CD7, which comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174 or with SEQ ID
  • the amino acid sequence shown in any one of NO:162 to SEQ ID NO:174 has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97% %, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • the chimeric antigen receptors of the present application also comprise an intracellular domain that provides an intracellular signal to the T cell following antigen binding to the antigen-specific extracellular domain.
  • suitable intracellular domains include the zeta chain of a T cell receptor or any of its homologues (e.g., ⁇ , ⁇ , ⁇ , or ⁇ ), MB 1 chain, B29, Fc RIII, Fc Combinations of RI and signaling molecules such as CD3. ⁇ . and CD28, CD27, 4-1BB, DAP-10, OX40 and combinations thereof and other similar molecules and fragments.
  • the intracellular signaling portions of other members of the activin family such as Fc. ⁇ .RIII and Fc. ⁇ .RI, can be used.
  • the intracellular domain of the present application is intended to include any truncated portion of the intracellular domain sufficient to transduce an effector function signal.
  • the antigen-specific extracellular domain is linked to the intracellular domain of the chimeric antigen receptor through a transmembrane domain.
  • the transmembrane domain traverses the cell membrane, anchors the CAR to the T cell surface, and connects the extracellular domain to the intracellular signaling domain, thereby affecting the expression of the CAR on the T cell surface.
  • a chimeric antigen receptor may further comprise one or more co-stimulatory domains and/or one or more spacers.
  • Costimulatory domains are derived from intracellular signaling domains of costimulatory proteins that enhance cytokine production, proliferation, cytotoxicity and/or persistence in vivo.
  • the spacer (i) connects the antigen-specific extracellular domain to the transmembrane domain, (ii) connects the transmembrane domain to the costimulatory domain, (iii) connects the costimulatory domain to the intracellular domain, And/or (iv) linking the transmembrane domain to the intracellular domain.
  • the inclusion of a spacer domain between the antigen-specific extracellular domain and the transmembrane domain can affect the flexibility of the antigen-binding domain and Thus affecting the CAR function.
  • Suitable transmembrane domains, co-stimulatory domains and spacers are known in the art.
  • the chimeric antigen receptor comprises a transmembrane domain comprising a transmembrane domain derived from one or more proteins selected from the group consisting of: CD8, CD28, CD3 ⁇ (CD3e), 4-1BB, CD4, CD27, CD7, PD-1, TRAC, TRBC, CD3 ⁇ , CTLA-4, LAG-3, CD5, ICOS, OX40, NKG2D, 2B4(CD244), Fc ⁇ RI ⁇ , BTLA, CD30 , GITR, HVEM, DAP10, CD2, NKG2C, LIGHT, DAP12, CD40L (CD154), TIM1, CD226, DR3, CD45, CD80, CD86, CD9, CD16, CD22, CD33, CD37, CD64, and SLAM.
  • transmembrane domain may comprise a transmembrane domain derived from CD8.
  • the transmembrane domain comprises the amino acid sequence shown in SEQ ID NO: 177 or has at least about 90%, about 91%, about 92%, about 93% of the amino acid sequence shown in SEQ ID NO: 177, Amino acid sequences that are about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical.
  • the chimeric antigen receptor includes an intracellular co-stimulatory signaling domain comprising one or more proteins derived from the group consisting of Intracellular co-stimulatory signaling domains: CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GITR, HVEM, DAP10, DAP12, CD30, CD40, CD40L, TIM1, PD-1, LFA-1, LIGHT, JAML, CD244, CD100, ICOS, CD40, and MyD88.
  • Intracellular co-stimulatory signaling domains CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GI
  • the intracellular co-stimulatory signaling domain may comprise a co-stimulatory signaling domain derived from 4-1BB.
  • the intracellular co-stimulatory signaling domain may comprise the amino acid sequence shown in SEQ ID NO: 178.
  • the chimeric antigen receptor comprises an intracellular signaling domain comprising an intracellular protein derived from one or more proteins selected from the group consisting of Signal transduction domain: CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa, bovine leukemia virus gp30, Epstein-Barr virus (EBV) LMP2A, simian immunodeficiency virus PBj14 Nef, DAP10, DAP-12 and domains containing at least one ITAM.
  • Signal transduction domain CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa
  • bovine leukemia virus gp30 Epstein-Barr virus (EBV) LMP2A
  • simian immunodeficiency virus PBj14 Nef simian immunodeficiency virus
  • the intracellular signaling domain may comprise a signaling domain derived from CD3 ⁇ .
  • intracellular signal transduction domain may comprise the amino acid sequence shown in SEQ ID NO: 179.
  • it comprises a hinge region between the extracellular antigen binding domain and the transmembrane domain, said hinge The chain region comprises a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D , NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D , NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • the hinge region may comprise a hinge region derived from CD8.
  • the hinge region may comprise the amino acid sequence shown in SEQ ID NO: 176.
  • the non-targeting portion of the chimeric antigen receptor comprises a transmembrane domain, a hinge region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain.
  • the non-targeting portion of the chimeric antigen receptor comprises the CD8 molecular transmembrane domain, the hinge region of CD8, the intracellular co-stimulatory signaling domain of 4-1BB, and the CD3 ⁇ intracellular signaling domain.
  • it further comprises a signal peptide fragment, the C-terminus of the signal peptide fragment is connected to the N-terminus of the extracellular antigen-binding domain.
  • the signal peptide fragment may comprise a CD8 signal peptide fragment.
  • the signal peptide fragment may comprise the amino acid sequence shown in SEQ ID NO:175.
  • the chimeric antigen receptor of the present application may comprise the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192 or any one of SEQ ID NO: 180 to SEQ ID NO: 192
  • the amino acid sequence shown has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • immune effector cells include engineered immune effector cells.
  • engineered immune cell generally refers to an immune cell that has been genetically modified by adding additional genetic material in the form of DNA or RNA to the total genetic material of the cell, also known as an immune effector cell.
  • engineered immune cells have been genetically modified to express a CD7-targeting CAR according to the invention.
  • the immune effector cells include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells cell.
  • the immune effector cells comprise autologous or allogeneic immune effector cells.
  • the immune effector cells may include allogeneic T cells or autologous T cells.
  • the engineered immune effector cells include CAR-T cells, CAR-NK or TCR-T cells.
  • the engineered immune effector cells include anti-CD7 CAR-T cells.
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • the expression of CD7 in the modified immune effector cells is absent or suppressed.
  • the CAR-T cells lack an antigen to which the chimeric antigen receptor specifically binds and thus do not induce cannibalism.
  • the antigen of the T cell is modified such that the chimeric antigen receptor no longer specifically binds the modified antigen (eg, CD7).
  • the chimeric antigen receptor no longer specifically binds the modified antigen (eg, CD7).
  • an epitope of an antigen recognized by a chimeric antigen receptor can be modified by one or more amino acid changes (eg, substitution or deletion), or the epitope can be deleted from the antigen.
  • expression of the antigen is reduced in T cells by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or more.
  • Methods for reducing expression of a protein are known in the art and include, but are not limited to, modifying or replacing a promoter operably linked to a nucleic acid sequence encoding a protein.
  • T cells are modified such that the antigen is not expressed, eg, by deletion or disruption of the gene encoding the antigen.
  • Methods of genetically modifying T cells to lack antigens are well known in the art, and non-limiting examples are provided above.
  • CRISPR/cas9 gene editing can be used to modify T cells to lack antigen.
  • an engineered CAR can be introduced into T cells using a retrovirus, which efficiently and stably integrates a nucleic acid sequence encoding a chimeric antigen receptor into the target cell genome.
  • CAR-encoding mRNA is used to transfer into T cells.
  • Other methods known in the art include, but are not limited to, lentiviral transduction, transposon-based systems, direct RNA transfection, and CRISPR/Cas systems.
  • CAR-T cells can be generated from any suitable source of T cells known in the art, including but not limited to T cells collected from a subject.
  • the subject may be a patient with a T-cell malignancy in need of CAR-T cell therapy or the same class of subject as a subject with a T-cell malignancy in need of CAR-T cell therapy.
  • Harvested T cells can be expanded ex vivo using methods generally known in the art and then transduced with CAR to generate CAR-T cells.
  • the present application provides a cell population comprising the modified immune effector cells described in the present application, which
  • the cell populations described in are derived from peripheral blood mononuclear cells (PBMC), peripheral blood leukocytes (PBL), tumor infiltrating lymphocytes (TIL), cytokine-induced killer cells (CIK), lymphokine-activated killer cells (LAK ) or bone marrow infiltrating lymphocytes (MIL).
  • PBMC peripheral blood mononuclear cells
  • PBL peripheral blood leukocytes
  • TIL tumor infiltrating lymphocytes
  • CIK cytokine-induced killer cells
  • LAK lymphokine-activated killer cells
  • MIL bone marrow infiltrating lymphocytes
  • the present application provides a pharmaceutical combination comprising immune effector cells and a fusion protein
  • the fusion protein comprises a first domain for activating antigen-presenting cells (APC) and a second domain for activating immune effector cells
  • the first domain comprises (a) an activating receptor ligand or receptor binding fragment thereof that binds APC, or (b) an activating receptor antibody or an antigen binding fragment thereof that binds APC
  • a second Domains include (a) costimulatory ligands or receptor-binding fragments thereof for immune effector cells, or (b) antibodies or antigen-binding fragments thereof that bind costimulatory receptors for immune effector cells.
  • the fusion protein is a soluble protein. In some embodiments, the fusion protein is a bispecific antibody.
  • first domain and the second domain of the fusion protein can be selected from the following combinations:
  • APC-activating receptor ligand + antibody that binds to a co-stimulatory receptor eg, aCD28-CD40L
  • a co-stimulatory receptor eg, aCD28-CD40L
  • Antibodies to APC-activating receptors + antibodies to co-stimulatory receptors eg, aCD40/aCD28 bispecific Ab
  • Antibodies to APC-activating receptors + costimulatory ligands e.g., aCD40-CD80; aCD40-CD86; and
  • Ligands of APC activating receptors + co-stimulatory ligands eg, CD40L-CD86; CD40L-CD80.
  • the present application provides the use of the modified immune effector cells described in the present application, the pharmaceutical combination described in the present application or the pharmaceutical composition described in the present application in the preparation of medicines, and the medicines are used to treat tumors.
  • the tumor includes hematological tumors and solid tumors.
  • the tumor comprises a CD7 expressing tumor.
  • the tumor comprises a CD7-positive hematologic malignancy.
  • the tumor comprises a T cell malignancy.
  • T-cell malignancy comprises acute T-lymphoblastic leukemia (T-ALL), acute myeloid leukemia, or NK/T-cell lymphoma.
  • T-ALL acute T-lymphoblastic leukemia
  • NK/T-cell lymphoma NK/T-cell lymphoma
  • the present application provides a method for treating tumors, the method comprising administering the modified immune effector cells described in the present application, the drug combination described in the present application, or the drug combination described in the present application to a subject in need thereof. said pharmaceutical composition.
  • CAR-T cells can be administered to a subject by intravenous route (eg, by intravenous infusion).
  • CAR-T cells Administration can be in single dose or in multiple doses.
  • CAR-T cells can be injected in a pharmaceutical composition suitable for intravenous administration. Suitable pharmaceutical compositions for IV administration are known in the art.
  • the pharmaceutical compositions of the present disclosure may also comprise additional components. For example, such components can be used to maintain the viability and/or activity of injected CAR-T cells.
  • CAR-T cells can be administered in effective doses.
  • An effective dose can be one dose or multiple doses sufficient to produce the desired therapeutic effect.
  • Typical doses of CAR-T cells may range from about 1 x 105 to 5 x 107 cells/Kg body weight of the subject receiving the therapy.
  • Effective doses can be calculated based on the stage of the malignancy, the health of the subject, and the type of malignancy. Where multiple doses are administered, the dose and the interval between doses can be determined based on the subject's response to the therapy.
  • the present application provides a method for killing malignant T cells, the method comprising combining the malignant T cells with the modified immune effector cells described in the present application, the drug combination described in the present application, or the combination of the drugs described in the present application. contact with the pharmaceutical composition described above.
  • the present application provides a method for preparing a chimeric antigen receptor T (CAR-T) cell population, wherein the CAR targets CD7, comprising the following steps:
  • an engineered CAR can be introduced into T cells using a retrovirus, which efficiently and stably integrates a nucleic acid sequence encoding a chimeric antigen receptor into the target cell genome.
  • CAR-encoding mRNA is used to transfer into T cells.
  • Other methods known in the art include, but are not limited to, lentiviral transduction, transposon-based systems, direct RNA transfection, and CRISPR/Cas systems.
  • CAR-T cells can be generated from any suitable source of T cells known in the art, including but not limited to T cells collected from a subject.
  • the subject may be a patient with a T-cell malignancy in need of CAR-T cell therapy or the same class of subject as a subject with a T-cell malignancy in need of CAR-T cell therapy.
  • Harvested T cells can be expanded ex vivo using methods generally known in the art and then transduced with CAR to generate CAR-T cells.
  • the present disclosure provides a T cell comprising a chimeric antigen receptor that specifically binds CD7, wherein the T cell lacks CD7 (eg, a CD7 KO CAR-T cell).
  • the absence of CD7 results from (a) modification of CD7 expressed by T cells such that the chimeric antigen receptor no longer specifically binds to the modified CD7, (b) modification of T cells such that expression of the antigen on T cells at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more, or (c) modify T cells so that CD7 is not expressed (for example, by deletion or disruption of the gene encoding CD7 ).
  • said modification comprises administering to said T cell population one or more substances selected from the group consisting of antisense RNA, siRNA, shRNA, transcription activator-like effector nuclease (TALEN) , zinc finger nuclease (ZFN) and CRISPR/Cas system.
  • antisense RNA siRNA, shRNA
  • transcription activator-like effector nuclease TALEN
  • ZFN zinc finger nuclease
  • CRISPR/Cas system CRISPR/Cas system
  • wherein said modification comprises administering a CRISPR/Cas system to said population of T cells.
  • wherein said modification comprises administering a CRISPR/Cas9 system to said population of T cells.
  • said modification comprises administering Cas9 and a gRNA targeting the CD7 gene to said T cell population.
  • the gRNA targeting the CD7 gene comprises the nucleotide sequence shown in any one of SEQ ID NO:211 to SEQ ID NO:218.
  • the transducing the anti-CD7 CAR and the fusion protein into the T cell comprises introducing the nucleic acid molecule encoding the anti-CD7 CAR and the nucleic acid molecule encoding the fusion protein into the T cell.
  • an isolated antigen binding protein that competes for binding to CD7 with a reference antibody, wherein the reference antibody comprises a light chain variable region VL and a heavy chain variable region VH;
  • VH comprises the amino acid sequence shown in SEQ ID NO: 61
  • VL comprises the amino acid sequence shown in SEQ ID NO: 148
  • the reference antibody is named anti-CD7 scFv-H1 antibody
  • the VH comprises the amino acid sequence shown in SEQ ID NO:62
  • the VL comprises the amino acid sequence shown in SEQ ID NO:149
  • the reference antibody is named anti-CD7 scFv-H4 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:63
  • the VL comprises the amino acid sequence shown in SEQ ID NO:150
  • the reference antibody is named anti-CD7 scFv-H6 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:64
  • the VL comprises the amino acid sequence shown in SEQ ID NO:151
  • the reference antibody is named anti-CD7 scFv-H8 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:65
  • the VL comprises the amino acid sequence shown in SEQ ID NO:152
  • anti-CD7 scFv-H9 antibody or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:66
  • the VL comprises the amino acid sequence shown in SEQ ID NO:153
  • the reference antibody is named anti-CD7 scFv-H10 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:67
  • the VL comprises the amino acid sequence shown in SEQ ID NO:154
  • the reference antibody is named anti-CD7 scFv-H12 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:68
  • the VL comprises the amino acid sequence shown in SEQ ID NO:155
  • the reference antibody is named anti-CD7 scFv-H13 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:69
  • the VL comprises the amino acid sequence shown in SEQ ID NO:156
  • the reference antibody is named anti-CD7 scFv-H17 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:70
  • the VL comprises the amino acid sequence shown in SEQ ID NO:157
  • the reference antibody is named anti-CD7 scFv-H18 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:71
  • the VL comprises the amino acid sequence shown in SEQ ID NO:158
  • the reference antibody is named anti-CD7 scFv-H5 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:72
  • the VL comprises the amino acid sequence shown in SEQ ID NO:159
  • the reference antibody is named anti-CD7 scFv-H7 antibody; or
  • the VH comprises the amino acid sequence shown in SEQ ID NO:73
  • the VL comprises the amino acid sequence shown in SEQ ID NO:160
  • the reference antibody is named anti-CD7 scFv-H15 antibody.
  • the antigen binding protein of described isolation comprises heavy chain variable region (VH), and described VH comprises HCDR1, HCDR2 and HCDR3, wherein HCDR1 comprises the aminoacid sequence shown in SEQ ID NO:1, SEQ ID
  • the general sequence formula of NO:1 is shown as G[FGY][ST][FILV][ST][EGST][LNY], wherein the amino acid in [] is an optional amino acid.
  • HCDR1 may comprise the amino acid sequence shown in any one of SEQ ID NO:2 to SEQ ID NO:9.
  • HCDR2 comprises the amino acid sequence shown in any one of SEQ ID NO:10 to SEQ ID NO:17.
  • HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 18 or SEQ ID NO: 27.
  • the isolated antigen-binding protein comprises a heavy chain variable region (VH), and the VH may comprise HCDR1, HCDR2 and HCDR3, wherein the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:2, Place 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:18; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:3, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:11, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:19; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:2
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:10
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:20; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:4, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:12, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:21; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:22; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:6, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:14, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:23; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:2, 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:24; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:25; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:8, the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:16, and the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:26; or
  • the HCDR1 may comprise the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 may comprise the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 may comprise the amino acid sequence shown in SEQ ID NO:27.
  • said isolated antigen binding protein comprises a VH, wherein said VH comprises a framework Regions HFR1, HFR2, HFR3 and HFR4, the C-terminus of HFR1 is directly or indirectly connected to the N-terminus of HCDR1, and the HFR1 comprises any one of SEQ ID NO:28 to SEQ ID NO:38 amino acid sequence.
  • the HFR2 is located between the HCDR1 and the HCDR2, and the HFR2 comprises the amino acid sequence shown in any one of SEQ ID NO:39 to SEQ ID NO:47.
  • the HFR3 is located between the HCDR2 and the HCDR3, and the HFR3 comprises the amino acid sequence shown in any one of SEQ ID NO:48 to SEQ ID NO:56.
  • the N-terminal of the HFR4 is directly or indirectly connected to the C-terminal of the HCDR3, and the HFR4 comprises amino acids shown in any one of SEQ ID NO:57 to SEQ ID NO:60 sequence.
  • the isolated antigen-binding protein comprises VH, wherein the VH may include framework regions HFR1, HFR2, HFR3 and HFR4, the C-terminus of HFR1 is directly or indirectly linked to the N-terminus of HCDR1, the HFR2 is located between the HCDR1 and the HCDR2, the HFR3 is located between the HCDR2 and the HCDR3, and the N-terminal of the HFR4 is directly or indirectly connected to the C-terminal of the HCDR3;
  • the HFR1 can be Comprising the amino acid sequence shown in SEQ ID NO: 28, the HFR2 can include the amino acid sequence shown in SEQ ID NO: 39, the HFR3 can include the amino acid sequence shown in SEQ ID NO: 48, and HFR4 can include the amino acid sequence shown in SEQ ID NO : the amino acid sequence shown in 57; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:29
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:40
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:49
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:30
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:48
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:31
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:41
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:50
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:59; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:32
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:42
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:51
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:33, and the HFR2 can comprise SEQ ID NO:33
  • the amino acid sequence shown in ID NO:39, the HFR3 can include the amino acid sequence shown in SEQ ID NO:48, and the HFR4 can include the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:34
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:48
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:35
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:43
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:52
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:28
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:39
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:48
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:30
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:44
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:53
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:58; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:36
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:45
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:54
  • HFR4 can comprise comprising the amino acid sequence shown in SEQ ID NO:60; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:37
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:46
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:55
  • HFR4 can comprise the amino acid sequence shown in SEQ ID NO:55 the amino acid sequence shown in SEQ ID NO:60; or
  • the HFR1 can comprise the amino acid sequence shown in SEQ ID NO:38
  • the HFR2 can comprise the amino acid sequence shown in SEQ ID NO:47
  • the HFR3 can comprise the amino acid sequence shown in SEQ ID NO:56
  • HFR4 can comprise Comprising the amino acid sequence shown in SEQ ID NO:58.
  • the isolated antigen-binding protein comprises VH, wherein the VH may comprise the amino acid sequence shown in SEQ ID NO:61 to SEQ ID NO:73.
  • said isolated antigen-binding protein comprises VL, wherein said VL comprises LCDR1, LCDR2, LCDR3, said LCDR1 comprising any one of SEQ ID NO:74 to SEQ ID NO:86 The amino acid sequence shown.
  • said isolated antigen binding protein comprises VL, wherein said VL comprises LCDR1, LCDR2, LCDR3, the LCDR2 comprises the amino acid sequence shown in any one of SEQ ID NO:87 to SEQ ID NO:98.
  • said isolated antigen-binding protein comprises VL, wherein said VL comprises LCDR1, LCDR2, LCDR3, said LCDR3 comprising any one of SEQ ID NO:99 to SEQ ID NO:101 The amino acid sequence shown.
  • the isolated antigen-binding protein comprises VL, wherein the VL may comprise LCDR1, LCDR2 and LCDR3, the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:74, and the LCDR2 may comprise SEQ ID NO The amino acid sequence shown in: 87, described LCDR3 can comprise the amino acid sequence shown in SEQ ID NO: 99; Or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:75
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:88
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:100;
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:76
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:89
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:101; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:77
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:90
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:102; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:78
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:91
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:103; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:79
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:104; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:80
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:93
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:105; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:81
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:94
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:106 acid sequence
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:82
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:95
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:107; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:84
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:97
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:109; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:85
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:110; or
  • the LCDR1 may comprise the amino acid sequence shown in SEQ ID NO:86
  • the LCDR2 may comprise the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 may comprise the amino acid sequence shown in SEQ ID NO:111.
  • said isolated antigen binding protein comprises VH and VL, wherein said VH comprises HCDR1, HCDR2 and HCDR3, said VL comprises LCDR1, LCDR2 and LCDR3; wherein said HCDR1 comprises SEQ ID NO The amino acid sequence shown in: 2, 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: 18, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO: 74 Amino acid sequence, the LCDR2 comprises the amino acid sequence shown in SEQ ID NO:87, and the LCDR3 comprises the amino acid sequence shown in SEQ ID NO:99; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:3
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:11
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:19
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:19
  • the amino acid sequence shown in ID NO:75, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:88, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:100; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:20
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:20
  • the amino acid sequence shown in ID NO:76, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:89, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:101; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:4, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:12, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:21, and the LCDR1 Comprising the amino acid sequence shown in SEQ ID NO:77, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:90, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:102; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:22
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:22
  • the amino acid sequence shown in ID NO:91, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:91
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:103;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:79, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:92
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:104; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:80, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:93, and the LCDR3 includes the amino acid sequence shown in SEQ ID NO:105; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:6, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:14, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:23, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:23
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:18
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:18
  • the amino acid sequence shown in ID NO:82, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:95
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:107;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:24
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:24
  • the amino acid sequence shown in ID NO:83, said LCDR2 comprises the amino acid sequence shown in SEQ ID NO:96
  • said LCDR3 comprises the amino acid sequence shown in SEQ ID NO:108;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:7
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:15
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:25
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:25
  • said LCDR2 comprises the amino acid shown in SEQ ID NO:97 Sequence
  • the LCDR3 comprises the amino acid sequence shown in SEQ ID NO: 109; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:8, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:16, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:26, and the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:26
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:9
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:17
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:27
  • the LCDR1 comprises the amino acid sequence shown in SEQ ID NO:27
  • the amino acid sequence shown in ID NO:86, the LCDR2 includes the amino acid sequence shown in SEQ ID NO:98
  • the LCDR3 includes the amino acid sequence shown in SEQ ID NO:111.
  • said isolated antigen binding protein comprises VL, wherein said VL comprises framework regions LFR1, LFR2, LFR3 and LFR4, wherein the C-terminus of said LFR1 is directly or directly to the N-terminus of said LCDR1 Indirectly connected, and the LFR1 comprises the amino acid sequence shown in any one of SEQ ID NO:112 to SEQ ID NO:123.
  • the isolated antigen binding protein comprises a VL, wherein the VL comprises a framework region LFR2, wherein the LFR2 is located between the LCDR1 and the LCDR2, and the LFR2 comprises SEQ ID The amino acid sequence shown in any one of NO:124 to SEQ ID NO:130.
  • the isolated antigen binding protein comprises a VL, wherein the VL comprises a framework region LFR3, the LFR3 is located between the LCDR2 and the LCDR3, and the LFR3 comprises SEQ ID NO : 131 to the amino acid sequence shown in any one of SEQ ID NO: 141.
  • the isolated antigen-binding protein comprises a VL, wherein the VL comprises a framework region LFR4, the N-terminus of the LFR4 is directly or indirectly connected to the C-terminus of the LCDR3, and the LFR4 Comprising the amino acid sequence shown in any one of SEQ ID NO:142 to SEQ ID NO:147.
  • said isolated antigen binding protein comprises VL, wherein said VL may comprise framework regions LFR1, LFR2, LFR3 and LFR4, wherein the C-terminus of said LFR1 is directly or indirectly linked to the N-terminus of said LCDR1, said The LFR2 is located between the LCDR1 and the LCDR2, the LFR3 is located between the LCDR2 and the LCDR3, and the N-terminal of the LFR4 is directly or indirectly connected to the C-terminal of the LCDR3; wherein the LFR1 can be Comprising the amino acid sequence shown in SEQ ID NO:112, the LFR2 can include the amino acid sequence shown in SEQ ID NO:124, the LFR3 can include the amino acid sequence shown in SEQ ID NO:131, and the LFR4 can include the amino acid sequence shown in SEQ ID NO:131, and the LFR4 can include the amino acid sequence shown in SEQ ID NO:131 The amino acid sequence shown in ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:113
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:125
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:132
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:114
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:133
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:115
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:127
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:134
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:116
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:125
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:13
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 143; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:117
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:136
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 143; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO: 118
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO: 1208
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO: 137
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO: 119
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO: 125
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO: 138
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 143; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:120
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:129
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:139
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 142; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:114
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:126
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:140
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 144; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:121
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 145; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:122
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 146; or
  • the LFR1 may comprise the amino acid sequence shown in SEQ ID NO:123
  • the LFR2 may comprise the amino acid sequence shown in SEQ ID NO:130
  • the LFR3 may comprise the amino acid sequence shown in SEQ ID NO:141
  • the LFR4 may comprise the amino acid sequence shown in SEQ ID NO: 147.
  • the isolated antigen-binding protein comprises VL, wherein the VL may comprise the amino acid sequence shown in any one of SEQ ID NO:148 to SEQ ID NO:160.
  • the isolated antigen-binding protein comprises VH and VL, wherein the VH may comprise the amino acid sequence shown in SEQ ID NO:61, and the VL may comprise the amino acid sequence shown in SEQ ID NO:148; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:62, and the VL may comprise the amino acid sequence shown in SEQ ID NO:149; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:63, and the VL may comprise the amino acid sequence shown in SEQ ID NO:150; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:64, and the VL may comprise the amino acid sequence shown in SEQ ID NO:151; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:65, and the VL may comprise the amino acid sequence shown in SEQ ID NO:152; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:66, and the VL may comprise the amino acid sequence shown in SEQ ID NO:153; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:67, and the VL may comprise the amino acid sequence shown in SEQ ID NO:154; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:68, and the VL may comprise the amino acid sequence shown in SEQ ID NO:155; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:69, and the VL may comprise the amino acid sequence shown in SEQ ID NO:156; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:70, and the VL may comprise SEQ ID NO: the amino acid sequence shown at 157; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:71, and the VL may comprise the amino acid sequence shown in SEQ ID NO:158; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:72, and the VL may comprise the amino acid sequence shown in SEQ ID NO:159; or
  • the VH may comprise the amino acid sequence shown in SEQ ID NO:73
  • the VL may comprise the amino acid sequence shown in SEQ ID NO:160.
  • the isolated antigen-binding protein comprises an antibody or antigen-binding fragment thereof.
  • the antibodies include monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies, intact antibodies, antibody fragments, human antibodies, humanized antibodies, or chimeric antibodies.
  • said antigen-binding fragment comprises Fab, Fab', Fv fragment, F(ab') 2 , scFv, di-scFv and/or dAb.
  • the antigen binding protein comprises a scFv.
  • the antigen binding protein can be an anti-CD7 scFv antibody.
  • VL and VH are linked by a linker.
  • linker comprises a polypeptide linker
  • the polypeptide linker comprises an amino acid sequence represented by (GGGGS)n, wherein n is any integer from 1 to 5.
  • the antigen binding protein comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the present application provides an immunoconjugate comprising the antigen-binding protein described in the present application.
  • the term "immunoconjugate” or “antibody conjugate” generally refers to the linkage of an antibody or antibody fragment thereof to other active agents, such as chemotherapeutic agents, toxins, immunotherapeutic agents, imaging probes, spectroscopic probes, Needle, wait.
  • the linkage may be a covalent bond, or a non-covalent interaction such as through electrostatic forces.
  • linkers known in the art can be used to form immunoconjugates.
  • the immunoconjugate can be provided as a fusion protein that can be expressed from a polynucleotide encoding the immunoconjugate.
  • Fusion protein refers to a protein produced by linking two or more genes or gene fragments that originally encoded separate proteins, including peptides and polypeptides. Translation of the fusion gene produces a single protein with functional properties derived from each original protein.
  • the immunoconjugate comprises:
  • a conjugation moiety selected from the group consisting of detectable markers, drugs, toxins, cytokines, viral coat proteins or VLPs, or combinations thereof.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to (eg, kills) cells.
  • agents include paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin Bixin, daunorubicin, dihydroxyanthracene, mitoxantrone, plicamycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine Caine, propranolol, and puromycin and their analogs or congeners.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil diazepam), alkylating agents (e.g., nitrogen mustard, thiotepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, silk split mycin C and cis-dichlorodiaminoplatinum(II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly known as erythromycin) and doxorubicin), antibiotics ( For example, actinomycin (proactinomycin)), bleomycin, plicamycin, and anthranimycin (AMC)), and antimitotic agents (eg, vincristine
  • the antigen-specific extracellular domain of a chimeric antigen receptor recognizes and specifically binds an antigen, usually an antigen expressed on the surface of a malignant tumor (eg, CD7).
  • an antigen usually an antigen expressed on the surface of a malignant tumor (eg, CD7).
  • an antigen-specific extracellular domain binds the antigen with an affinity constant or interaction affinity (KD) of about 0.1 pM to about 10 ⁇ M, or about 0.1 pM to about 1 ⁇ M, or about 0.1 pM to about 100 nM
  • KD affinity constant or interaction affinity
  • Methods for determining interaction affinity are known in the art.
  • An antigen-specific extracellular domain suitable for use in a CAR of the present disclosure can be any antigen-binding polypeptide, a wide variety of which are known in the art.
  • the antigen binding domain is a single chain Fv (scFv).
  • scFv single chain Fv
  • Other antibody-based recognition domains cAb VHH (camel antibody variable domain) and its humanized form
  • T cell receptor (TCR) based recognition domains such as single chain TCR (scTv) are also suitable for use.
  • Suitable antigens may include T cell-specific antigens and/or antigens that are not specific for T cells.
  • the antigen specifically bound by the CAR-T cell chimeric antigen receptor and the CAR-T cell deficient antigen is an antigen expressed on malignant T cells, more preferably in combination with non-malignant T cells. cells compared to malignant T cells expressed antigen.
  • a "malignant T cell” is a T cell derived from a T cell malignancy.
  • T cell malignancies refers to a broad and highly heterogeneous group of malignancies derived from T cell precursors, mature T cells or natural killer cells.
  • T-cell malignancies include T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), T-cell large granular lymphocyte (LGL) leukemia, human T-cell leukemia virus type 1 positive (HTLV-1 +) Adult T-cell leukemia/lymphoma (ATL), T-cell prolymphocytic leukemia (T-PLL), and various peripheral T-cell lymphomas (PTCL), including but not limited to angioimmunoblastic T-cell lymphoma ( AITL), ALK-positive anaplastic large cell lymphoma, and ALK-negative anaplastic large cell lymphoma.
  • T-ALL T-cell acute lymphoblastic leukemia/lymphoma
  • LGL lymphocyte
  • HTLV-1 + human T-cell leukemia virus type 1 positive
  • ATL adult T-cell leukemia/lymphoma
  • T-PLL T-cell prolymphocytic leukemia
  • PTCL peripheral T-cell lymphomas
  • the CAR-T cells of the present application comprise the extracellular domain of a chimeric antigen receptor that specifically binds to CD7.
  • CD7 is a T cell surface membrane-associated glycoprotein.
  • CD7 can be overexpressed in T cell malignancies including T cell acute lymphoblastic leukemia (T-ALL) and non-Hodgkin's T cell lymphoma (NHL).
  • T-ALL T cell acute lymphoblastic leukemia
  • NHL non-Hodgkin's T cell lymphoma
  • the CAR-T cells of the present disclosure can be used to target malignant T cells overexpressing CD7.
  • the antigen-specific extracellular domain of the chimeric antigen receptor of the present application can specifically bind to CD7, which comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174 or with SEQ ID
  • the amino acid sequence shown in any one of NO:162 to SEQ ID NO:174 has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97% %, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • the chimeric antigen receptors of the present application also comprise an intracellular domain that provides an intracellular signal to the T cell following antigen binding to the antigen-specific extracellular domain.
  • suitable intracellular domains include the zeta chain of a T cell receptor or any of its homologues (e.g., ⁇ , ⁇ , ⁇ , or ⁇ ), MB 1 chain, B29, Fc RIII, Fc Combinations of RI and signaling molecules such as CD3. ⁇ . and CD28, CD27, 4-1BB, DAP-10, OX40 and combinations thereof and other similar molecules and fragments.
  • the intracellular signaling portions of other members of the activin family such as Fc. ⁇ .RIII and Fc. ⁇ .RI, can be used.
  • the intracellular domain of the present application is intended to include any truncated portion of the intracellular domain sufficient to transduce an effector function signal.
  • the antigen-specific extracellular domain is linked to the intracellular domain of the chimeric antigen receptor through a transmembrane domain.
  • the transmembrane domain traverses the cell membrane, anchors the CAR to the T cell surface, and connects the extracellular domain to the intracellular signaling domain, thereby affecting the expression of the CAR on the T cell surface.
  • a chimeric antigen receptor may further comprise one or more co-stimulatory domains and/or one or more spacers. Costimulatory domains derived from enhancers of costimulatory proteins Intracellular signaling domains for cytokine production, proliferation, cytotoxicity and/or persistence.
  • the spacer (i) connects the antigen-specific extracellular domain to the transmembrane domain, (ii) connects the transmembrane domain to the costimulatory domain, (iii) connects the costimulatory domain to the intracellular domain, And/or (iv) linking the transmembrane domain to the intracellular domain.
  • inclusion of a spacer domain between the antigen-specific extracellular domain and the transmembrane domain can affect the flexibility of the antigen-binding domain and thus CAR function.
  • Suitable transmembrane domains, co-stimulatory domains and spacers are known in the art.
  • the chimeric antigen receptor comprises a transmembrane domain comprising a transmembrane domain derived from one or more proteins selected from the group consisting of: CD8, CD28, CD3 ⁇ (CD3e), 4-1BB, CD4, CD27, CD7, PD-1, TRAC, TRBC, CD3 ⁇ , CTLA-4, LAG-3, CD5, ICOS, OX40, NKG2D, 2B4(CD244), Fc ⁇ RI ⁇ , BTLA, CD30 , GITR, HVEM, DAP10, CD2, NKG2C, LIGHT, DAP12, CD40L (CD154), TIM1, CD226, DR3, CD45, CD80, CD86, CD9, CD16, CD22, CD33, CD37, CD64, and SLAM.
  • transmembrane domain may comprise a transmembrane domain derived from CD8.
  • the transmembrane domain comprises the amino acid sequence shown in SEQ ID NO: 177 or has at least about 90%, about 91%, about 92%, about 93% of the amino acid sequence shown in SEQ ID NO: 177, Amino acid sequences that are about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical.
  • the chimeric antigen receptor includes an intracellular co-stimulatory signaling domain comprising one or more proteins derived from the group consisting of Intracellular co-stimulatory signaling domains: CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GITR, HVEM, DAP10, DAP12, CD30, CD40, CD40L, TIM1, PD-1, LFA-1, LIGHT, JAML, CD244, CD100, ICOS, CD40, and MyD88.
  • Intracellular co-stimulatory signaling domains CD28, CD137, CD27, CD2, CD7, CD8, OX40, CD226, DR3, SLAM, CDS, ICAM-1, NKG2D, NKG2C, B7-H3, 2B4, Fc ⁇ RI ⁇ , BTLA, GI
  • the intracellular co-stimulatory signaling domain may comprise a co-stimulatory signaling domain derived from 4-1BB.
  • intracellular co-stimulatory signaling domain may comprise the amino acid sequence shown in any one of SEQ ID NO:178.
  • the chimeric antigen receptor comprises an intracellular signaling domain comprising an intracellular protein derived from one or more proteins selected from the group consisting of Signal transduction domain: CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa, bovine leukemia virus gp30, Epstein-Barr virus (EBV) LMP2A, simian immunodeficiency virus PBj14 Nef, DAP10, DAP-12 and contain at least one Domains of ITAMs.
  • Signal transduction domain CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, FceRI ⁇ , FceRI ⁇ , Fc ⁇ RIIa
  • bovine leukemia virus gp30 Epstein-Barr virus (EBV) LMP2A
  • PBj14 Nef simian immunodeficiency virus
  • the intracellular signaling domain may comprise a signaling domain derived from CD3 ⁇ .
  • intracellular signal transduction domain may comprise the amino acid sequence shown in SEQ ID NO: 179.
  • it includes a hinge region between the extracellular antigen binding domain and the transmembrane domain, said hinge region comprising a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • a hinge region derived from one or more proteins selected from the group consisting of: CD28, IgG1, IgG4, IgD, 4-1BB, CD4, CD27, CD7, CD8, PD-1, ICOS, OX40, NKG2D, NKG2C, Fc ⁇ RI ⁇ , BTLA, GITR, DAP10, TIM1, SLAM, CD30, and LIGHT.
  • the hinge region may comprise a hinge region derived from CD8.
  • the hinge region may comprise the amino acid sequence shown in SEQ ID NO: 176.
  • the non-targeting portion of the chimeric antigen receptor comprises a transmembrane domain, a hinge region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain.
  • the non-targeting portion of the chimeric antigen receptor comprises the CD8 molecular transmembrane domain, the hinge region of CD8, the intracellular co-stimulatory signaling domain of 4-1BB, and the CD3 ⁇ intracellular signaling domain.
  • it further comprises a signal peptide fragment, the C-terminus of the signal peptide fragment is connected to the N-terminus of the extracellular antigen-binding domain.
  • the signal peptide fragment may comprise a CD8 signal peptide fragment.
  • the signal peptide fragment may comprise the amino acid sequence shown in SEQ ID NO:175.
  • the chimeric antigen receptor of the present application may comprise the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192 or any one of SEQ ID NO: 180 to SEQ ID NO: 192
  • the amino acid sequence shown has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identical amino acid sequences.
  • the present application provides an engineered cell comprising the nucleic acid molecule or the vector described in the present application, and/or expressing the chimeric antigen receptor described in the present application.
  • the chimeric antigen receptor can comprise the amino acid sequence shown in any one of SEQ ID NO:180 to SEQ ID NO:192 or the amino acid sequence shown in any one of SEQ ID NO:180 to SEQ ID NO:192
  • the amino acid sequence has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.5% identity sequence.
  • the cells include immune effector cells.
  • the immune effector cells include human cells.
  • the immune effector cells include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • NK cells natural killer cells
  • macrophages include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • the immune effector cells comprise autologous or allogeneic immune effector cells.
  • the immune effector cells include allogeneic T cells or autologous T cells.
  • the immune effector cells include modified immune effector cells.
  • the expression of CD7 in the modified immune effector cells is absent or suppressed.
  • the modified immune effector cells have reduced surface expression of CD7 and express an anti-CD7 CAR compared to corresponding immune cells.
  • the immune effector cells include CAR-T cells.
  • the CAR-T cells lack an antigen to which the chimeric antigen receptor specifically binds and thus do not induce cannibalism.
  • the antigen of the T cell is modified such that the chimeric antigen receptor no longer specifically binds the modified antigen (eg, CD7).
  • the chimeric antigen receptor no longer specifically binds the modified antigen (eg, CD7).
  • an epitope of an antigen recognized by a chimeric antigen receptor can be modified by one or more amino acid changes (eg, substitution or deletion), or the epitope can be deleted from the antigen.
  • expression of the antigen is reduced in T cells by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or more.
  • Methods for reducing expression of a protein are known in the art and include, but are not limited to, modifying or replacing a promoter operably linked to a nucleic acid sequence encoding a protein.
  • T cells are modified such that the antigen is not expressed, eg, by deletion or disruption of the gene encoding the antigen.
  • Methods of genetically modifying T cells to lack antigens are well known in the art, and non-limiting examples are provided above.
  • CRISPR/cas9 gene editing can be used to modify T cells to lack antigen.
  • CAR-T cells encompassed by the present disclosure may also lack endogenous T cell receptor (TCR) signaling.
  • TCR T cell receptor
  • reducing or eliminating endogenous TCR signaling in CAR-T cells can prevent or reduce graft-versus-host disease (GvHD) when allogeneic T cells are used to generate CAR-T cells.
  • GvHD graft-versus-host disease
  • Methods for reducing or eliminating endogenous TCR signaling are known in the art and include, but are not limited to, modifying a portion of a TCR receptor (eg, TCR receptor alpha chain (TRAC), etc.).
  • TRAC modification blocks TCR-mediated signaling. Therefore, TRAC modification may allow the safe use of allogeneic T cells as a source of CAR-T cells without inducing life-threatening GvHD.
  • CAR-T cells encompassed by the present disclosure may also comprise one or more suicide genes.
  • suicide gene refers to a nucleic acid sequence introduced into a CAR-T cell by standard methods known in the art, which when activated, causes the death of the CAR-T cell.
  • Suicide genes facilitate efficient tracking and elimination of CAR-T cells in vivo, if desired. Promotion of killing by activation of suicide genes can occur by methods known in the art.
  • the present disclosure provides a T cell comprising a chimeric antigen receptor that specifically binds CD7, wherein the T cell lacks CD7 (eg, a CD7 ⁇ CART7 cell).
  • the lack of CD7 results from (a) modifying CD7 expressed by T cells such that the chimeric antigen receptor no longer specifically binds the modified CD7, (b) modifying the T cells such that the antigen Expression is reduced in T cells by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or more, or (c) T cells are modified such that CD7 is not expressed (e.g., by the gene encoding CD7 missing or damaged).
  • an engineered CAR can be introduced into T cells using a retrovirus, which efficiently and stably integrates a nucleic acid sequence encoding a chimeric antigen receptor into the target cell genome.
  • CAR-encoding mRNA is used to transfer into T cells.
  • Other methods known in the art include, but are not limited to, lentiviral transduction, transposon-based systems, direct RNA transfection, and CRISPR/Cas systems.
  • CAR-T cells can be generated from any suitable source of T cells known in the art, including but not limited to T cells collected from a subject.
  • the subject may be a patient with a T-cell malignancy in need of CAR-T cell therapy or the same class of subject as a subject with a T-cell malignancy in need of CAR-T cell therapy.
  • Harvested T cells can be expanded ex vivo using methods generally known in the art and then transduced with CAR to generate CAR-T cells.
  • the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the immunoconjugate described in the present application, the nucleic acid molecule described in the present application , the expression vector described in the present application, the cell described in the present application, the chimeric antigen receptor described in the present application and/or the engineered cell described in the present application, and optionally a pharmaceutically acceptable carrier.
  • the present application provides a kit comprising the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the immunoconjugate described in the present application, the nucleic acid molecule described in the present application, The table described in this application The expression carrier, the cell described in the present application, the chimeric antigen receptor described in the present application, the engineered cell described in the present application, or the pharmaceutical composition described in the present application.
  • the isolated antigen binding protein described herein, the polypeptide described herein, the immunoconjugate described herein, the nucleic acid molecule described herein, the expression vector described herein, the cell described herein , the chimeric antigen receptor described in the application, the engineered cell described in the application, or the application of the pharmaceutical composition described in the application in the preparation of medicines, the medicine is used to prevent and/or treat CD7-related disease or condition.
  • said CD7-associated disease or condition comprises a tumor.
  • the tumor comprises a CD7 expressing tumor.
  • the tumor comprises a hematoma.
  • the tumor comprises a CD7-positive hematologic malignancy.
  • the tumor comprises a T cell malignancy.
  • the present application provides a method for treating tumors, the method comprising administering an effective amount of the isolated antigen-binding protein described in the present application, the polypeptide described in the present application, the polypeptide described in the present application to a subject in need thereof.
  • the T-cell malignancy comprises a CD7-positive hematologic malignancy.
  • T-cell malignancy comprises acute T-lymphoblastic leukemia (T-ALL), acute myeloid leukemia, or NK/T-cell lymphoma.
  • T-ALL acute T-lymphoblastic leukemia
  • NK/T-cell lymphoma NK/T-cell lymphoma
  • the present application provides a method for treating tumors, the method comprising administering an effective amount of the CAR-T cells described in the present application to a subject in need thereof.
  • the CAR-T cells target CD7, and CD7 is deleted or inhibited.
  • CAR-T cell therapy can also be accompanied by other therapies, including but not limited to immunotherapy, chemotherapy or radiation therapy.
  • CAR-T cells can be administered to a subject by intravenous route (eg, by intravenous infusion).
  • CAR-T cells can be administered in a single dose or in multiple doses.
  • CAR-T cells can be injected in a pharmaceutical composition suitable for intravenous administration. Suitable pharmaceutical compositions for IV administration are known in the art.
  • the pharmaceutical compositions of the present disclosure may also comprise additional components. For example, such components can be used to maintain the viability and/or activity of injected CAR-T cells.
  • CAR-T cells can be administered in effective doses.
  • An effective dose can be one dose or multiple doses sufficient to produce the desired therapeutic effect.
  • Typical doses of CAR-T cells may range from about 1 x 105 to 5 x 107 cells/Kg body weight of the subject receiving the therapy.
  • Effective doses can be based on the stage of the malignancy, the health of the subject, and the malignancy. Tumor type is calculated. Where multiple doses are administered, the dose and the interval between doses can be determined based on the subject's response to the therapy.
  • the present application provides a method for killing malignant T cells, the method comprising contacting the malignant T cells with the engineered cells described in the present application.
  • the application provides a gRNA targeting CD7 gene, which comprises the nucleotide sequence described in any one of SEQ ID NO:212 to SEQ ID NO:218 or with SEQ ID NO:212 to SEQ ID NO
  • the nucleotide sequence of any one of: 218 has at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% %, about 99%, about 99.5% identical amino acid sequences.
  • the present application provides an isolated nucleic acid molecule comprising the gRNA described in the present application or a DNA molecule encoding the gRNA.
  • the present application provides a vector comprising the nucleic acid molecule described in the present application.
  • the "vector” generally refers to a nucleic acid molecule capable of self-replication in a suitable host, so as to transfer the inserted nucleic acid molecule into and/or between host cells.
  • the vectors may include vectors mainly used for inserting DNA or RNA into cells, vectors mainly used for replicating DNA or RNA, and vectors mainly used for expression of transcription and/or translation of DNA or RNA.
  • the carrier also includes a carrier having various functions as described above.
  • the vector may be a polynucleotide capable of being transcribed and translated into a polypeptide when introduced into a suitable host cell. Generally, the vector can produce the desired expression product by culturing an appropriate host cell containing the vector.
  • Vectors can contain additional features beyond the transgene insert and backbone: promoters, genetic markers, antibiotic resistance, reporter genes, targeting sequences, protein purification tags.
  • Vectors known as expression vectors are used in particular for the expression of transgenes in target cells and usually have control sequences.
  • the carrier described in the present application can be an expression vector, which can include viral vectors (lentiviral vectors and/or retroviral vectors), phage vectors, phagemids, cosmids, cosmids, artificial chromosomes such as yeast artificial chromosomes (YAC), Bacterial Artificial Chromosome (BAC) or P1-derived Artificial Chromosome (PAC) and/or Plasmid.
  • viral vectors lentiviral vectors and/or retroviral vectors
  • phage vectors phagemids
  • cosmids cosmids
  • cosmids cosmids
  • artificial chromosomes such as yeast artificial chromosomes (YAC), Bacterial
  • the vector includes a virus vector
  • the nucleic acid molecule of the present application can be introduced into cells by using the virus vector.
  • viral vectors include, for example, recombinant retroviruses, adenoviruses, adeno-associated viruses, and herpes simplex virus-1.
  • Retroviral vectors and adeno-associated viral vectors are usually understood as recombinant gene delivery systems for the transfer of exogenous genes in vivo, especially into the human body. Alternatively, they can be used to introduce exogenous genes into T cells ex vivo. These vectors efficiently deliver genes into T cells, and the transferred nucleic acid is stably integrated into the host cell chromosome DNA.
  • the present application provides a cell comprising the nucleic acid molecule described in the present application or the vector described in the present application.
  • it includes immune effector cells.
  • the immune effector cells include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • NK cells natural killer cells
  • macrophages include T cells, B cells, natural killer cells (NK cells), macrophages, NKT cells, monocytes, dendritic cells, granulocytes, lymphocytes, leukocytes and/or peripheral blood mononuclear cells.
  • the immune effector cells comprise autologous or allogeneic immune effector cells.
  • the immune effector cells include allogeneic T cells or autologous T cells.
  • the immune effector cells comprise engineered immune effector cells.
  • the engineered immune effector cells include CAR-T cells.
  • expression of the antigen CD7 is reduced in T cells by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or more.
  • CRISPR/cas9 gene editing can be used to modify T cells to lack the antigen CD7.
  • the gRNA of the present invention can be introduced into T cells by transfection methods well known in the art. These methods include sonication, electrical pulses, electroporation, osmotic shock, calcium phosphate precipitation and DEAE-dextran transfection, lipid-mediated delivery, passive delivery, and the like.
  • transfection includes a variety of techniques that can be used to introduce nucleic acid into mammalian cells, including electroporation, calcium phosphate precipitation, DEAE-dextran treatment, lipofection, microinjection, and viral infection. Suitable methods for transfecting mammalian cells can be found in Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press (1989)) and other experiments. room textbook.
  • the invention comprises a method of delivering a CRISPR enzyme comprising delivering a nucleic acid molecule, such as a plasmid or RNA or mRNA, encoding the CRISPR enzyme to a cell.
  • the CRISPR enzyme is Cas9. In other embodiments, the CRISPR enzyme can be delivered directly into the cell.
  • chimeric antigen receptor generally refers to a group of polypeptides, usually two in the simplest embodiment, which, when in immune effector cells, provide cellular (usually cancer cells) and generate intracellular signals.
  • the CAR comprises at least one extracellular antigen-binding domain, a transmembrane domain, and a cytoplasmic signaling domain (also referred to herein as an "intracellular signaling domain”) comprising a derivative A functional signaling domain derived from a stimulatory and/or co-stimulatory molecule as defined below.
  • the set of polypeptides are in the same polypeptide chain (eg, comprising chimeric fusion proteins).
  • the set of polypeptides is discontinuous from each other, eg, in different polypeptide chains.
  • the set of polypeptides includes a dimerization switch that, in the presence of a dimerization molecule, can couple the polypeptides to each other, eg, can couple an antigen binding domain to an intracellular signaling domain.
  • the stimulatory molecule of the CAR is the zeta chain associated with the T cell receptor complex.
  • the cytoplasmic signaling domain comprises a primary signaling domain (eg, the primary signaling domain of CD3-zeta).
  • the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one co-stimulatory molecule as defined below.
  • the co-stimulatory molecule is selected from 4-1BB (ie CD137), CD27, ICOS and/or CD28.
  • a CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a co-stimulatory molecule and a functional signaling domain derived from a stimulatory molecule.
  • a CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising a functional domain derived from one or more co-stimulatory molecules. Signaling domains and functional signaling domains derived from stimulatory molecules.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen recognition domain, a transmembrane domain and an intracellular signaling domain comprising at least two co-stimulatory molecules derived from and functional signaling domains derived from stimulatory molecules.
  • the CAR comprises an optional leader sequence at the amino terminus (N-ter) of the CAR fusion protein.
  • the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen recognition domain, wherein the leader sequence is optionally cleaved from the antigen recognition domain (e.g. scFv) during cellular processing and localizes the CAR to the cell membrane.
  • the present application provides a CAR-T cell, and the CAR-T cell is a CAR-T cell modified by the nucleic acid molecule described in the present application.
  • CAR-T or “CAR-T cell” generally refers to a T cell capable of expressing a CAR (also known as a "chimeric antigen receptor”).
  • the CAR generally refers to a fusion protein comprising an extracellular domain capable of binding an antigen and at least one intracellular domain.
  • CAR is a core component of chimeric antigen receptor T cells (CAR-T), which can include a targeting moiety (for example, a moiety that binds tumor-associated antigen (TAA)), a hinge region, a transmembrane region and intracellular domain.
  • TAA tumor-associated antigen
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the anti-CD7 CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • composition comprising:
  • nucleic acid molecule described in the present application a. the nucleic acid molecule described in the present application, the vector described in the present application, the cell described in the present application, or the CAR-T cell described in the present application;
  • a pharmaceutically acceptable carrier b.
  • a pharmaceutically acceptable carrier generally refers to a substance suitable for administration to a subject, wherein the carrier is not biologically harmful, or does not cause other adverse effects.
  • Such carriers are usually inert ingredients of the drug.
  • the carrier is administered to a subject with the active ingredient without causing any undesired biological effects or interacting in a deleterious manner with any other components of the pharmaceutical composition contained therein.
  • Suitable pharmaceutical carriers are described in Martin, Remington's Pharmaceutical Sciences, 18th ed., Mark Press., Easton, Pa., (1990), the contents of which are incorporated herein by reference.
  • a more specific form of the present application provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a nucleic acid molecule, vector or cell and a pharmaceutically acceptable diluent, preservative, solubilizer, emulsifier, adjuvant and/or carrier.
  • Such compositions include various buffer contents (e.g., phosphate, Tris-HCl, acetate), pH and ionic strength agents, and additives, such as detergents and solubilizers (e.g., Tween 80, polysorbate 80), antioxidants (eg, ascorbic acid, sodium metabisulfite), preservatives (eg, thimerosal, benzyl alcohol), and fillers (eg, lactose, mannitol).
  • buffer contents e.g., phosphate, Tris-HCl, acetate
  • pH and ionic strength agents e.g., phosphate, Tris-HCl, acetate
  • additives
  • compositions can affect the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the disclosed compositions.
  • the composition may be prepared in liquid form, or may be a dry powder, for example in lyophilized form. It should be understood that the pharmaceutical compositions provided in this disclosure may be administered by any means known in the art. For example, pharmaceutical compositions for administration may be administered by injection, orally, or by pulmonary or nasal routes.
  • the present application provides a method for regulating T cell function, the method comprising the gRNA described in the application, the nucleic acid molecule described in the application, the expression vector described in the application or the gene described in the application
  • the editing system is introduced into T cells.
  • Cas enzyme and CRISPR enzyme are generally used herein interchangeably unless otherwise stated.
  • the Cas enzyme comprises a Cas9 protein.
  • the present invention includes more Cas9s from other microbial species, such as SpCas9, SaCa9, StlCas9 and the like.
  • the regulated T cells have down-regulated or knocked-out CD7 gene expression compared to unregulated T cells.
  • the method further comprises modifying the specificity of the T cell by introducing into the T cell a nucleic acid molecule encoding a CAR.
  • the CAR-encoding nucleic acid molecule comprises mRNA.
  • the mRNA encodes an anti-CD7 CAR.
  • the extracellular antigen binding domain comprises the amino acid sequence shown in any one of SEQ ID NO: 162 to SEQ ID NO: 174.
  • the anti-CD7 CAR comprises the amino acid sequence shown in any one of SEQ ID NO: 180 to SEQ ID NO: 192.
  • regulated T cells have reduced surface expression of CD7 and express an anti-CD7 CAR compared to corresponding T cells.
  • the present application provides a method for treating tumors, the method comprising administering an effective amount of the cell described in the present application or the pharmaceutical composition described in the present application to a subject in need thereof.
  • the T-cell malignancy comprises a CD7-positive hematologic malignancy.
  • T-cell malignancy comprises acute T-lymphoblastic leukemia (T-ALL), acute myeloid leukemia, or NK/T-cell lymphoma.
  • T-ALL acute T-lymphoblastic leukemia
  • NK/T-cell lymphoma NK/T-cell lymphoma
  • the present application provides a method for treating tumors, the method comprising administering an effective amount of the CAR-T cells described in the present application to a subject in need thereof.
  • the CAR-T cells target CD7, and CD7 is deleted or inhibited.
  • CAR-T cell therapy can also be accompanied by other therapies, including but not limited to immunotherapy, chemotherapy or radiation therapy.
  • CAR-T cells can be administered to a subject by intravenous route (eg, by intravenous infusion).
  • CAR-T cells can be administered in a single dose or in multiple doses.
  • CAR-T cells can be injected in a pharmaceutical composition suitable for intravenous administration. Suitable pharmaceutical compositions for IV administration are known in the art.
  • the pharmaceutical compositions of the present disclosure may also comprise additional components. For example, such components can be used to maintain the viability and/or activity of injected CAR-T cells.
  • CAR-T cells can be administered in effective doses.
  • An effective dose can be one dose or multiple doses sufficient to produce the desired therapeutic effect.
  • Typical doses of CAR-T cells may range from about 1 x 105 to 5 x 107 cells/Kg body weight of the subject receiving the therapy.
  • Effective doses can be calculated based on the stage of the malignancy, the health of the subject, and the type of malignancy. Where multiple doses are administered, the dose and the interval between doses can be determined based on the subject's response to the therapy.
  • the present application provides a method for killing malignant T cells, the method comprising contacting the malignant T cells with the CAR-T cells described in the present application.
  • antigen-binding protein of separation its specific binding CD7 albumen, wherein said antigen-binding protein of separation comprises heavy chain variable region (VH), and described VH comprises HCDR1, HCDR2 and HCDR3, and wherein HCDR1 comprises SEQ ID NO: The amino acid sequence shown in 1.
  • VH heavy chain variable region
  • HCDR1 comprises the amino acid sequence shown in any one of SEQ ID NO:2 to SEQ ID NO:9.
  • HCDR2 comprises the amino acid sequence shown in any one of SEQ ID NO: 10 to SEQ ID NO: 17.
  • HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 18 or SEQ ID NO: 27.
  • HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • said HCDR2 comprises the amino acid sequence shown in SEQ ID NO:10
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 18;
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:3
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:11
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:19; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2, and the HCDR2 comprises the SEQ ID NO: The amino acid sequence shown in 10, the HCDR3 comprises the amino acid sequence shown in SEQ ID NO: 20; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:4, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:12, and the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:21; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:5
  • the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:13
  • the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:22; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:6, the HCDR2 comprises the amino acid sequence shown in SEQ ID NO:14, and the HCDR3 comprises the amino acid sequence shown in SEQ ID NO:23; or
  • the HCDR1 comprises the amino acid sequence shown in SEQ ID NO:2
  • 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:24; or

Abstract

本申请涉及一种经修饰的免疫效应细胞,其包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,还包含一种分离的核酸分子,其用于在细胞内抑制CD7基因的表达。本申请还涉及所述核酸分子在制备抗CD7CAR-T细胞中的用途。引入本申请所述的核酸分子的抗CD7CAR-T细胞表达的CD7被下调,不影响CAR的表达且避免CAR-T细胞相互残杀。

Description

经修饰的免疫效应细胞及其用途 技术领域
本申请涉及生物医药领域,具体的涉及一种经修饰的免疫效应细胞及其用途。
背景技术
T淋巴细胞可以被设计成表达识别肿瘤抗原的T细胞受体(TCRs)[1-3]或嵌合抗原受体(CAR)[4,5],以杀死肿瘤,用于治疗癌症和其他疾病。尽管用CD19等B细胞标志物特异性的CAR设计的T淋巴细胞在血液学恶性肿瘤中显示出巨大的临床反应,但在实体瘤中有效的免疫治疗被证明是具有挑战性的,这主要是由于复杂、动态的肿瘤微环境(TME)引起的免疫逃逸,诱发T细胞功能低下和衰竭,限制了抗肿瘤免疫反应[6]。设计T细胞以规避TME的抑制作用的策略提供了规避抑制途径的机会,而不会引起全身性的毒性,例如,在使用PD-1和CTLA-4检查点抑制剂的联合治疗中出现的高不良反应率[7]
T细胞激活阈值的提高和T细胞激活状态的降低是大多数肿瘤相关的对T细胞的抑制作用的主要机制,这可以通过提供足够的成本刺激来抵消[8]。CD28被认为是最佳T细胞克隆扩展、分化和效应功能的最突出的共刺激分子。CD28的参与降低了T细胞的激活阈值,并导致TCR信号事件的增强,这对有效的细胞因子的产生(通过增强的转录活性和信使RNA的稳定)、细胞周期的进展、生存、代谢的调节和T细胞的反应是必要的。这是因为CD28是免疫学突触(IS)组织的一个关键角色,它加强了T细胞和APCs之间的密切接触。
研究表明,可以通过遗传方式将代价刺激受体或其配体引入T细胞,以增强其效应功能、持久性和抗肿瘤活性[8-10]。然而,由于受体和配体的表达贫乏,以及抑制性配体(如PD-L1)在TME中的过度表达,预计表达共刺激受体或配体的T细胞在肿瘤性病变中的功能将不足[11]。在以前的研究中,我们和其他人已经发现,PD1-CD28转换受体是由PD1的细胞外部分和CD28的细胞内部分组成的融合蛋白,在体外和肿瘤动物模型中都能增强T细胞功能[12-16]。研究表明,与在CART细胞中引入额外的成本刺激配体[8-10]或使CART细胞分泌检查点抑制剂的策略相比,引入PD1-CD28嵌合开关受体可能是拯救T细胞免受肿瘤免疫抑制的更高效和有效的方法[17,18]
对复发或难治的T细胞急性淋巴细胞白血病(T-ALL)的有效系统治疗是有限的。最近嵌合抗原受体(CAR)免疫疗法的临床应用表明,CAR-T细胞成功地控制了B细胞恶性肿瘤;然而,为T-ALL设计CAR仍然是一个挑战。
T细胞恶性肿瘤中CD7的过度表达可能是T-ALL免疫疗法的一个有吸引力的目标。研究发现30%的AML表达CD7。但是,它在正常T细胞上的存在意味着CD7 CAR在这些细胞上的表达是不可行的,因为它将证明是自相残杀。因此,开发安全有效的自体CD7-CAR T细胞用于治疗T-ALL和AML是非常迫切的。
对复发或难治的T细胞急性淋巴细胞白血病(T-ALL)的有效系统治疗是有限的。最近嵌合抗原受体(CAR)免疫疗法的临床应用表明,CAR-T细胞成功地控制了B细胞恶性肿瘤;然而,为T-ALL设计CAR仍然是一个挑战。
T细胞恶性肿瘤中CD7的过度表达可能是T-ALL免疫疗法的一个有吸引力的目标。研究发现30%的AML表达CD7。但是,它在正常T细胞上的存在意味着CD7 CAR在这些细胞上的表达是不可行的,因为它将证明是自相残杀。能否设计、制备出精确性和特异性靶向CD7基因的SgRNA成为CRISPR/cas9特异性敲除CD7基因的关键技术。因此,开发安全有效的自体CD7-CAR T细胞用于治疗T-ALL和AML是非常迫切的。
CRISPR(Clustered Regularly Interspaced ShortPalindromic Repeats)由高度保守的重复序列(repeats)和多个不同的间隔序列(spacer)顺序排列组成,重复序列长度通常21~48bp,重复序列之间被26~72bp的间隔序列隔开。CRISPR特异性编辑靶序列是通过crRNA和tracrRNA和靶序列互补识别实现的。现已将tracrRNA与crRNA表达为一条嵌合的向导RNA(single guide RNA,sgRNA),将CRISPR-Cas9系统简化为Cas9蛋白和sgRNA两个组分,使得CRISPR-Cas9系统具有构建简单、效率高、成本低廉等优点,是基因组编辑技术最合适的选择。
环状RNA(circRNA)是一类由特殊的选择性剪切产生的首尾相接的非编码RNA。circRNA呈共价闭合的环形结构,没有5'和3'极性,不能被核糖核酸酶降解。研究已证实circRNA在疾病的发生和发展过程中具有重要的作用。有的circRNA可作为翻译模板,有的circRNA可以通过顺式或反式作用影响亲本基因的表达.
发明内容
本申请提供了一种经修饰的免疫效应细胞,其表达一组嵌合蛋白分子,称为淋巴细 胞-APCs共刺激分子(Lymphocytes-APCs Co-stimulators,LACO-Stim),包括:a.由细胞外CD40配体(CD40L)和细胞内CD28组成的膜融合蛋白,或与细胞内CD28融合的针对CD40的scFv;b.由细胞外CD40L和针对CD28的scFv组成的可溶融合蛋白;c.针对CD40和CD28的双特异性抗体。
当与CARs或TCRs共同引入人类T淋巴细胞时,LACO-stim分子对增加T细胞抗肿瘤功能,刺激和成熟APCs、巨噬细胞和髓系衍生细胞表现出强烈的作用。表达LACO-刺激分子的T细胞有能力克服TEM的抑制作用,如PD1/PD-L1、Treg和TGF-β的抑制作用。LACO-stim可以协调T淋巴细胞和APCs之间的相互作用,促进APCs的表位扩散能力,进一步提高抗肿瘤活性。
一方面,本申请提供一种经修饰的免疫效应细胞,其包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
在某些实施方式中,其中所述第一结构域的N端与第二结构域的C端连接。
在某些实施方式中,其中所述第一结构域的C端与第二结构域的N端连接。
在某些实施方式中,其中所述第一结构域与第二结构域直接连接或间接连接。
在某些实施方式中,其中所述第一结构域与第二结构域通过连接子连接。
在某些实施方式中,其中所述连接子包括肽连接子。
在某些实施方式中,其中所述抗体或其抗原结合片段为scFv。
在某些实施方式中,其中所述APC选自由树突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
在某些实施方式中,其中所述APC的激活受体选自CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
在某些实施方式中,其中所述第一结构域包含结合CD40、CD80、CD86、CD91、DEC-205、DC-SIGN的配体或它们的受体结合片段。
在某些实施方式中,其中所述第一结构域包含CD40配体(CD40L)的受体结合片段。
在某些实施方式中,其中所述第一结构域包含结合所述APC的激活受体的抗体或其 抗原结合片段。
在某些实施方式中,其中所述第一结结构域为抗CD40抗体或其抗原结合片段。
在某些实施方式中,其中所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、嗜中性粒细胞和粒细胞组成的组。
在某些实施方式中,其中第二结构域包含共刺激受体的胞内结构域。
在某些实施方式中,其中所述共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
在某些实施方式中,其中所述共刺激受体为CD28或4-1BB。
在某些实施方式中,其中所述第二结构域是免疫效应细胞的共刺激配体或其受体结合片段。
在某些实施方式中,其中共刺激配体选自CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44。
在某些实施方式中,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
在某些实施方式中,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
在某些实施方式中,其中共刺激受体为CD28或4-1BB。
在某些实施方式中,其中所述第一结构域与第二结构域选自以下组合:
i)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28或4-1BB胞内结构域;
ii)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
iii)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28胞内结构域;
iv)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
v)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;和
vi)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28 共刺激配体或其受体结合片段。
在某些实施方式中,其中所述融合蛋白包含SEQ ID NO:234至SEQ ID NO:250中任一项所示的氨基酸序列。
在某些实施方式中,其中所述免疫效应细胞包括工程化的免疫效应细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
在某些实施方式中,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中所述CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
在某些实施方式中,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
在某些实施方式中,所述免疫效应细胞不诱导自相残杀。
嵌合抗原受体T(CAR-T)细胞,其表达抗CD7 CAR,并且包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
在某些实施方式中,其中所述CAR-T细胞的CD7的表达缺失或受抑制。
在某些实施方式中,所述CAR-T细胞不诱导自相残杀。
另一方面,本申请提供一种药物组合,其包含免疫效应细胞和融合蛋白,其中所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,或(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。
在某些实施方式中,其中所述第一结构域的N端与第二结构域的C端连接。
在某些实施方式中,其中所述第一结构域的C端与第二结构域的N端连接。
在某些实施方式中,其中所述第一结构域与第二结构域直接连接或间接连接。
在某些实施方式中,其中所述第一结构域与第二结构域通过连接子连接。
在某些实施方式中,其中所述连接子包括肽连接子。
在某些实施方式中,其中所述抗体或其抗原结合片段为scFv。
在某些实施方式中,其中所述APC选自由树突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
在某些实施方式中,其中所述APC的激活受体选自CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
在某些实施方式中,其中所述第一结构域包含结合CD40、CD80、CD86、CD91、DEC-205、DC-SIGN的配体或它们的受体结合片段。
在某些实施方式中,其中所述第一结构域包含CD40配体(CD40L)的受体结合片段。
在某些实施方式中,其中所述第一结构域包含结合所述APC的激活受体的抗体或其抗原结合片段。
在某些实施方式中,其中所述为抗CD40抗体或其抗原结合片段。
在某些实施方式中,其中所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、嗜中性粒细胞和粒细胞组成的组。
在某些实施方式中,其中第二结构域包含共刺激受体的胞内结构域。
在某些实施方式中,其中所述共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
在某些实施方式中,其中所述共刺激受体为CD28。
在某些实施方式中,其中所述第二结构域是免疫效应细胞的共刺激配体或其受体结合片段。
在某些实施方式中,其中共刺激配体选自CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44。
在某些实施方式中,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
在某些实施方式中,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
在某些实施方式中,其中共刺激受体为CD28或4-1BB。
在某些实施方式中,其中所述第一结构域与第二结构域选自以下组合:
i)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28或4-1BB共刺激配体或其受体结合片段;
ii)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
iii)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;和
iv)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段。
在某些实施方式中,其中所述融合蛋白包含SEQ ID NO:237至SEQ ID NO:241中任一项所示的氨基酸序列。
在某些实施方式中,其中所述免疫效应细胞包括工程化的免疫效应细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
在某些实施方式中,与相应的免疫细胞相比,其中所述药物组合CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中所述CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
在某些实施方式中,所述免疫效应细胞不诱导自相残杀。
另一方面,本申请提供一种药物组合物,其包含本申请所述的经修饰的免疫效应细胞或本申请所述的药物组合,以及药学上可接受的载体。
另一方,本申请提供本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物在制备药物中的用途,所述药物用于治疗肿瘤。
在某些实施方式中,其中所述肿瘤包括血液瘤和实体瘤。
在某些实施方式中,其中所述肿瘤包括表达CD7的肿瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述肿瘤包括T细胞恶性肿瘤。
在某些实施方式中,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物。
另一方面,本申请提供一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物接触。
另一方面,本申请提供一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
a.对T细胞群中的CD7基因进行修饰;
b.激活所述T细胞群;
c.将抗CD7 CAR和融合蛋白转导所述T细胞群;以及
d.扩增所述嵌合抗原受体T细胞群。
在某些实施方式中,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
在某些实施方式中,其中所述靶向所述CD7基因的gRNA包含SEQ ID NO:211至SEQ ID NO:218中任一项所示的核苷酸序列。
在某些实施方式中,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
在某些实施方式中,其中所述gRNA与所述Cas9同时递送。
在某些实施方式中,其中所述递送是通过电穿孔进行的。
在某些实施方式中,其中所述CD7 CAR核酸分子和所述融合蛋白的核酸分子为mRNA。
在某些实施方式中,其中所述CD7 CAR核酸分子和所述融合蛋白的核酸分子为circRNA。
在某些实施方式中,所述cirRNA嵌合抗原受体包含按以下顺序排列的元件:内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA),circRNA融合蛋白包含按以下顺序排列的元件:内部核糖体进入位点(IRES)元件、融合蛋白编码序列和聚腺苷酸(polyA)。
在某些实施方式中,其中所述将抗CD7 CAR和融合蛋白转导所述T细胞包括将编码所述抗CD7 CAR的核酸分子和编码所述融合蛋白的核酸分子引入T细胞。
另一方面,本申请提供一种包含本申请所述的经修饰的免疫效应细胞或本申请所述CAR-T细胞的细胞群,其中所述细胞群来源于外周血单核细胞(PBMC)、外周血血液白细胞(PBL)、肿瘤浸润淋巴细胞(TIL)、细胞因子诱导的杀伤细胞(CIK)、淋巴因子激活的杀伤细胞(LAK)或骨髓浸润淋巴细胞(MIL)。
一方面,本申请提供了一种特异性结合CD7蛋白的抗原结合蛋白。
另一方面,本申提供了一种靶向CD7的嵌合抗原受体及CAR-T细胞。
另一方面,本申请提供了一种避免自我杀伤的靶向CD7的CAR-T细胞及其用途。
本申请公开了以下技术方案:
一方面,提供了一种分离的抗原结合蛋白,其特异性结合CD7蛋白,其中所述分离的抗原结合蛋白包含重链可变区(VH),所述VH包含HCDR1、HCDR2和HCDR3,其中HCDR1包SEQ ID NO:1所示的氨基酸序列。
在某些实施方式中,其中所述HCDR1包含SEQ ID NO:2至SEQ ID NO:9中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HCDR2包含SEQ ID NO:10至SEQ ID NO:17中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HCDR3包含SEQ ID NO:18或SEQ ID NO:27所示的氨基酸序列。
在某些实施方式中,其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示 的氨基酸序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH,其中所述VH包括框架区HFR1,HFR2、HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,且所述HFR1包含SEQ ID NO:28至SEQ ID NO:38中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR2位于所述HCDR1与所述HCDR2之间,且所述HFR2包含SEQ ID NO:39至SEQ ID NO:47中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR3位于所述HCDR2与所述HCDR3之间,且所述HFR3包含SEQ ID NO:48至SEQ ID NO:56中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR4的N末端与所述HCDR3的C末端直接或间接相连,且所述HFR4包含SEQ ID NO:57至SEQ ID NO:60中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH,其中所述VH包括框架区HFR1,HFR2,HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,所述HFR2位于所述HCDR1与所述HCDR2之间,所述HFR3位于所述HCDR2与所述HCDR3之间,所述HFR4的N末端与所述HCDR3的C末端直接或间接相连;其中,所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:57所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:29所示的氨基酸序列,所述HFR2包含SEQ ID NO:40所示的氨基酸序列,所述HFR3包含SEQ ID NO:49所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58示的氨基酸序列;或
所述SEQ ID NO:31所示的氨基酸序列,所述HFR2包含SEQ ID NO:41所示的氨基酸序列,所述HFR3包含SEQ ID NO:50所示的氨基酸序列,HFR4包含SEQ ID NO:59所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:32所示的氨基酸序列,所述HFR2包含SEQ ID NO:42所示的氨基酸序列,所述HFR3包含SEQ ID NO:51所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:33所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:34所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:35所示的氨基酸序列,所述HFR2包含SEQ ID NO:43所示的氨基酸序列,所述HFR3包含SEQ ID NO:52所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39 所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:44所示的氨基酸序列,所述HFR3包含SEQ ID NO:53所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:36所示的氨基酸序列,所述HFR2包含SEQ ID NO:45所示的氨基酸序列,所述HFR3包含SEQ ID NO:54所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:37所示的氨基酸序列,所述HFR2包含SEQ ID NO:46所示的氨基酸序列,所述HFR3包含SEQ ID NO:55所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:38所示的氨基酸序列,所述HFR2包含SEQ ID NO:47所示的氨基酸序列,所述HFR3包含SEQ ID NO:56所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH,其中所述VH包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR1包含SEQ ID NO:74至SEQ ID NO:86中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR2包含SEQ ID NO:87至SEQ ID NO:98中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR3包含SEQ ID NO:99至SEQ ID NO:111中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1,LCDR2和LCDR3,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:78所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH和VL,其中所述VH包含HCDR1,HCDR2和HCDR3,所述VL包含LCDR1,LCDR2和LCDR3;其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO: 11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1 包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,且所述LFR1包含SEQ ID NO:112至SEQ ID NO:123中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR2,其中所述LFR2位于所述LCDR1与所述LCDR2之间,且所述LFR2包含SEQ ID NO:124至SEQ ID NO:130中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR3,所述LFR3位于所述LCDR2与所述LCDR3之间,且所述LFR3包含SEQ ID NO:131至SEQ ID NO:141中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架 区LFR4,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连,且所述LFR4包含SEQ ID NO:142至SEQ ID NO:147中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,所述LFR2位于所述LCDR1与所述LCDR2之间,所述LFR3位于所述LCDR2与所述LCDR3之间,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连;其中所述LFR1包含SEQ ID NO:112所示的氨基酸序列,所述LFR2包含SEQ ID NO:124所示的氨基酸序列,所述LFR3包含SEQ ID NO:131所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:113所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:132所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:133所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:115所示的氨基酸序列,所述LFR2包含SEQ ID NO:127所示的氨基酸序列,所述LFR3包含SEQ ID NO:134所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:116所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:135所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:117所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:136所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:118所示的氨基酸序列,所述LFR2包含SEQ ID NO:128所示的氨基酸序列,所述LFR3包含SEQ ID NO:137所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:119所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:138所示的氨基酸序列,所述LFR4 包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:120所示的氨基酸序列,所述LFR2包含SEQ ID NO:129所示的氨基酸序列,所述LFR3包含SEQ ID NO:139所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:140所示的氨基酸序列,所述LFR4包含SEQ ID NO:144所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:121所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:145所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:122所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:146所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:123所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:147所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH和VL,其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所示的氨基酸序列;或
所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或
所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或
所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或
所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或
所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列;或
所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或
所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或
所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或
所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或
所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或
所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或
所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白包括抗体或其抗原结合片段。
在某些实施方式中,所述抗体包括单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体、完整抗体、抗体片段、人抗体、人源化抗体或嵌合抗体。
在某些实施方式中,其中所述抗原结合片段包括Fab,Fab’,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb。
在某些实施方式中,所述的抗原结合蛋白包括scFv。
在某些实施方式中,其中所述VL和VH通过接头连接。
在某些实施方式中,其中所述接头包括多肽接头。
在某些实施方式中,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
在某些实施方式中,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
另一方面,本申请提供一种分离的多肽,其包含本申请所述的分离的抗原结合蛋白。
另一方面,本申请提供一种免疫缀合物,其包含本申请所述的抗原结合蛋白。
在某些实施方式中,所述免疫缀合物包含:
(a)本申请所述的抗原结合蛋白;
(b)选自下组的缀合部分:可检测标记物、药物、毒素、细胞因子、病毒外壳蛋白或VLP、或其组合。
嵌合抗原受体,其包含至少一个包含本申请所述的抗原结合蛋白的细胞外抗原结合结构域。
在某些实施方式中,其中所述细胞外抗原结合结构域包括scFv。
在某些实施方式中,所述的嵌合抗原受体包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
在某些实施方式中,其中所述跨膜域包含源自CD8的跨膜域。
在某些实施方式中,其中所述跨膜域包含SEQ ID NO:177所示的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
在某些实施方式中,其中所述胞内共刺激信号传导结构域源自4-1BB的共刺激信号传导结构域。
在某些实施方式中,其中所述胞内共刺激信号传导结构域包含SEQ ID NO:178中任一项所示的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14Nef、DAP10、DAP-12 和至少包含一个ITAM的结构域。
在某些实施方式中,其中所述胞内信号转导结构域包含源自CD3ζ的信号传导结构域。
在某些实施方式中,其中所述胞内信号转导结构域包含SEQ ID NO:179所示的氨基酸序列。
在某些实施方式中,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
在某些实施方式中,所述铰链区包含源自CD8的铰链区。
在某些实施方式中,所述铰链区包含SEQ ID NO:176所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
在某些实施方式中,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
在某些实施方式中,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
在某些实施方式中,所述信号肽片段包括CD8信号肽片段。
在某些实施方式中,所述信号肽片段包含如SEQ ID NO:175所示的氨基酸序列。
在某些实施方式中,其包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
另一方面,本申请提供一种分离的一种或多种核酸分子,其编码本申请所述的分离的抗原结合蛋白、本申请所述的多肽或本申请所述的嵌合抗原受体。
在某些实施方式中,所述的分离的核酸分子包含SEQ ID NO:193至SEQ ID NO:205中任一项所示的核苷酸序列。
另一方面,本申请提供一种表达载体,其包含本申请所述的核酸分子。
在某些实施方式中,其中所述载体选自DNA载体、RNA载体、质粒、慢病毒载体、腺病毒载体、腺相关病毒载体和逆转录病毒载体。
另一方面,本申请提供一种细胞,所述细胞包含本申请的核酸分子或本申请所述的表达载体,和/或ii)所述细胞表达本申请所述的抗原结合蛋白、本申请所述的多肽或本申 请所述的嵌合抗原受体。
另一方面,本申请提供一种制备本申请所述的分离的抗原结合蛋白的方法,所述方法包括在使得本申请所述的分离的抗原结合蛋白表达的条件下,培养根据本申请所述的细胞。
另一方面,本申请提供一种工程化的细胞,其包含本申请所述的核酸分子或本申请所述的载体,和/或表达本申请所述的嵌合抗原受体。
在某些实施方式中,所述细胞包括免疫效应细胞。
在某些实施方式中,所述的免疫效应细胞包括人细胞。
在某些实施方式中,所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
在某些实施方式中,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。
在某些实施方式中,其中所述免疫效应细胞包括同种异体T细胞或自体T细胞。
在某些实施方式中,所述的免疫效应细胞包括经修饰的免疫效应细胞。
在某些实施方式中,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
在某些实施方式中,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,所述免疫效应细胞包括CAR-T细胞。
在某些实施方式中,所述CAR-T细胞不诱导自相残杀。
另一方面,本申请提供一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
a.对T细胞群中的CD7基因进行修饰;
b.激活所述T细胞群;
c.用本申请所述的嵌合抗原受体转导所述T细胞;以及
d.扩增所述嵌合抗原受体T细胞。
在某些实施方式中,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸 酶(ZFN)和CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
在某些实施方式中,其中所述靶向CD7基因的gRNA包含SEQ ID NO:211至SEQ ID NO:218中任一项所示的核苷酸序列。
在某些实施方式中,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
在某些实施方式中,其中所述gRNA与所述Cas9同时递送。
在某些实施方式中,其中所述递送是通过电穿孔进行的。
在某些实施方式中,所述转导嵌合抗原受体包括向转导circRNA嵌合抗原受体。
在某些实施方式中,所述cirRNA嵌合抗原受体按以下顺序包含内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA)。
另一方面,本申请提供一种药物组合物,其包含本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体和/或本申请所述的工程化细胞,以及任选地药学上可接受的载体。
另一方面,本申请提供一种试剂盒,其包含本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物。
本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物在制备药物中的用途,所述药物用于预防和/或治疗CD7相关的疾病或病症。
在某些实施方式中,其中所述CD7相关的疾病或病症包括肿瘤。
在某些实施方式中,其中所述肿瘤包括表达CD7的肿瘤。
在某些实施方式中,其中所述肿瘤包括血液瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述肿瘤包括T细胞恶性肿瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物。
在某些实施方式中,其中所述肿瘤包括表达CD7的肿瘤。
在某些实施方式中,其中所述肿瘤包括血液瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述肿瘤包括T细胞恶性肿瘤。
在某些实施方式中,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
另一方面,本申请提供一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与本申请所述的工程化的细胞接触。
本申请利用CRISPR/Cas9技术删除了T细胞中CD7的表达,以防止CAR-T细胞之间自相残杀的发生;从抗体噬菌体展示库中筛选出全人抗CD7 scFv,这可能有助于减少人类免疫系统的排斥反应,提高CAR-T细胞的持久性和治疗效果。
本申请目的在于利用CRISPR/Cas9技术删除T细胞中CD7的表达,以防止CAR-T细胞之间自相残杀的发生。
一方面,本申请提供一种靶向CD7基因的gRNA,其包含SEQ ID NO:212至SEQ ID NO:218中任一项所述的核苷酸序列或与SEQ ID NO:212至SEQ ID NO:218中任一项所述的核苷酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
另一方面,本申请提供一种分离的核酸分子,其包含本申请所述的gRNA或编码所述gRNA的DNA分子。
另一方面,本申请提供一种表达载体,其包含本申请所述的gRNA或本申请所述的核酸分子。
另一方面,本申请提供一种基因编辑系统,其包含本申请所述的gRNA、本申请所述的核酸分子或本申请所述的表达载体。
在某些实施方式中,所述的基因编辑系统包括CRISPR/Cas基因编辑系统。
在某些实施方式中,所述的基因编辑系统包括CRISPR/Cas9基因编辑系统。
在某些实施方式中,所述的基因编辑系统还包含编码Cas9的DNA、编码Cas9的mRNA或Cas9蛋白分子。
在某些实施方式中,所述的基因编辑系统包含编码本申请所述的靶向CD7基因的gRNA和Cas9的表达载体。
另一方面,本申请提供一种细胞,其包含本申请所述的gRNA、本申请所述的核酸分子、本申请所述的表达载体、或本申请所述的基因编辑系统。
在某些实施方式中,其包括表达CD7的细胞。
在某些实施方式中,其包括免疫效应细胞。
在某些实施方式中,其中所述免疫效应细胞的包括T细胞、B细胞、自然杀伤(NK)细胞、肥大细胞或吞噬细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞靶向CD7。
在某些实施方式中,其中所述免疫效应细胞包括工程化的免疫效应细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括CAR-T细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含重链可变区(VH)和轻链可变区(VL),所述VH可以包含HCDR1、HCDR2和HCDR3,,其中所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:18所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:19所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:20所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:21所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:22所示的氨基酸序列; 或
所述HCDR1可以包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:23所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:24所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:25所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:26所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:27所示的氨基酸序列。
在某些实施方式中,其中所述VH可以包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
在某些实施方式中,其中所述VL可以包含LCDR1,LCDR2和LCDR3,所述LCDR1可以包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:99所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:100所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:101所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:102所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:78所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:103所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:104所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:105所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:106所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:107所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:109所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:110所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,其中所述VH包含HCDR1,HCDR2和HCDR3,所述VL包含LCDR1,LCDR2和LCDR3;其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸 序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,其中所述VL可以包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
在某些实施方式中,其中所述VH可以包含SEQ ID NO:61所示的氨基酸序列,所述VL可以包含SEQ ID NO:148所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:62所示的氨基酸序列,所述VL可以包含SEQ ID NO:149所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:63所示的氨基酸序列,所述VL可以包含SEQ ID NO:150所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:64所示的氨基酸序列,所述VL可以包含SEQ ID NO: 151所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:65所示的氨基酸序列,所述VL可以包含SEQ ID NO:152所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:66所示的氨基酸序列,所述VL可以包含SEQ ID NO:153所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:67所示的氨基酸序列,所述VL可以包含SEQ ID NO:154所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:68所示的氨基酸序列,所述VL可以包含SEQ ID NO:155所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:69所示的氨基酸序列,所述VL可以包含SEQ ID NO:156所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:70所示的氨基酸序列,所述VL可以包含SEQ ID NO:157所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:71所示的氨基酸序列,所述VL可以包含SEQ ID NO:158所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:72所示的氨基酸序列,所述VL可以包含SEQ ID NO:159所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:73所示的氨基酸序列,所述VL可以包含SEQ ID NO:160所示的氨基酸序列。
在某些实施方式中,所述的细胞外抗原结合结构域包括scFv。
例如,所述细胞外抗原结合结构域可以是抗CD7 scFv。
在某些实施方式中,其中所述VL和VH通过接头连接。
在某些实施方式中,其中所述接头包括多肽接头。
在某些实施方式中,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
在一些实施方案中,本申请的CAR-T细胞包含特异性地结合到CD7的嵌合抗原受体的胞外结构域。CD7是T细胞表面膜相关糖蛋白。CD7可在T细胞恶性肿瘤包括T细胞急性成淋巴细胞性白血病(T-ALL)和非霍奇金氏T细胞淋巴瘤(NHL)中过表达。本公开的CAR-T细胞可用于靶向过表达CD7的恶性T细胞。
例如,本申请的嵌合抗原受体的抗原特异性胞外结构域可以特异性结合CD7,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列或与SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
例如,其中所述跨膜域可以包含源自CD8的跨膜域。
又例如,其中所述跨膜域包含SEQ ID NO:177所示的氨基酸序列或与SEQ ID NO:177所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
例如,其中所述胞内共刺激信号传导结构域可以包含源自4-1BB的共刺激信号传导结构域。
又例如,其中所述胞内共刺激信号传导结构域可以包含SEQ ID NO:178中任一项所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14 Nef、DAP10、DAP-12和至少包含一个 ITAM的结构域。
例如,其中所述胞内信号转导结构域可以包含源自CD3ζ的信号传导结构域。
又例如,其中所述胞内信号转导结构域可以包含SEQ ID NO:179所示的氨基酸序列。
在某些实施方式中,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
例如,所述铰链区可以包含源自CD8的铰链区。
例如,所述铰链区可以包含SEQ ID NO:176所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
例如,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
在某些实施方式中,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
例如,所述信号肽片段可以包含CD8信号肽片段。
例如,所述信号肽片段可以包含SEQ ID NO:175所示的氨基酸序列。
又例如,本申请的嵌合抗原受体可以包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列或与SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
另一方面,本申请提供本申请所述的gRNA,本申请所述的核酸分子,本申请所述的表达载体,本申请所述的基因编辑系统或本申请所述的细胞在制备治疗肿瘤药物中的应用。
在某些实施方式中,其中所述肿瘤包括实体瘤或血液瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性肿瘤。
在某些实施方式中,其中所述药物包括CAR-T细胞。
在某些实施方式中,其中所述药物包括靶向CD7 CAR-T细胞。
另一方面,本申请提供一种对细胞中CD7基因进行基因编辑的方法,包括利用本 申请所述的gRNA介导Cas9对CD7基因进行基因编辑。
在某些实施方式中,其中所述基因编辑包括基因敲除。
另一方面,本申请提供一种调节T细胞功能的方法,所述方法包括将本申请所述的gRNA、本申请所述的核酸分子、本申请所述的表达载体或本申请所述的基因编辑系统引入T细胞。
在某些实施方式中,还包括向所述细胞施用Cas酶。
在某些实施方式中,其中Cas酶包括Cas9蛋白。
在某些实施方式中,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
在某些实施方式中,其中所述gRNA与所述Cas9同时递送。
在某些实施方式中,其中所述递送是通过电穿孔进行的。
在某些实施方式中,与未经调节的T细胞相比,经调节的所述T细胞的CD7基因表达下调或被敲除。
在某些实施方式中,所述方法还包括通过向所述T细胞引入编码CAR的核酸分子来修饰所述T细胞的特异性。
在某些实施方式中,其中所述编码CAR的核酸分子包括mRNA。
在某些实施方式中,其中mRNA编码anti-CD7 CAR。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中anti-CD7 CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
在某些实施方式中,与相应的T细胞相比,其中所述经调节的T细胞CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,其通过选自以下的任一方式将本申请所述的gRNA、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的基因编辑系统和/或所述编码CAR的核酸分子引入T细胞:超声处理、电脉冲、电穿孔、渗透压冲击、磷酸钙沉淀、DEAE葡聚糖转染、脂质介导的递送和被动递送。
另一方面,本申请还提供了一种circRNA其按以下顺序包含内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸。
在某些实施方式中,所述polyA的长度至少为45个核苷酸。
在某些实施方式中,所述polyA的长度至少为70个核苷酸。
在某些实施方式中,所述靶向CD7的蛋白质包括抗体或其抗原结合片段、嵌合抗原受体(CAR)和/或T细胞受体(TCR)。
在某些实施方式中,所述CAR的结合结构域包括CD7 scFv。
在某些实施方式中,所述靶向CD7的蛋白质包括抗体,所述抗体包括包含LCDR1、LCDR2和LCDR3的轻链可变区和包含HCDR1、HCDR2和HCDR3的重链可变区,其中:
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO: 104所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,所述靶向CD7的蛋白质包括抗体,所述抗体包括VH和VL,其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所 示的氨基酸序列;或所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列;或所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列。
在某些实施方式中,所述的circRNA包含SEQ ID NO:256-257中任一项所示的核苷酸序列。
另一方面,本申请还提供一种调节CD7表达水平和/或活性的试剂,其包含本申请所述的circRNA。
另一方面,本申请还提供本申请所述的circRNA在调节CD7表达水平和/或活性中的应用。
本领域技术人员能够从下文的详细描述中容易地洞察到本申请的其它方面和优势。下文的详细描述中仅显示和描述了本申请的示例性实施方式。如本领域技术人员将认识到的,本申请的内容使得本领域技术人员能够对所公开的具体实施方式进行改动而不脱离本申请所涉及发明的精神和范围。相应地,本申请的附图和说明书中的描述仅仅是示例性的,而非为限制性的。
附图说明
本申请所涉及的发明的具体特征如所附权利要求书所显示。通过参考下文中详细描 述的示例性实施方式和附图能够更好地理解本申请所涉及发明的特点和优势。对附图简要说明如下:
图1显示的是三个96孔板的抗人CD7-Fc单克隆噬菌体ELISA的读数。
图2显示的是用于生成抗CD7 CAR mRNA的pDA-CAR载体的示意图。
图3显示的是用抗CD7抗体对电穿孔不同CD7 gRNA的T细胞进行FACS染色的结果。
图4显示的是用CD7-Fc蛋白对不同抗CD7 scFv CAR-T细胞进行FACS染色的结果。
图5显示的是用抗CD7抗体对电穿孔不同数量的CD7 mRNA的A549细胞进行FACS染色的结果。
图6-图8显示的是基于mRNA的抗CD7 CAR-T细胞(H1-H18)在E/T比例=3:1时对A549-GFP肿瘤细胞的杀伤曲线。
图9显示的是五种抗CD7 CAR-T细胞(H1,H7,H9,H10,H17)在E/T比=3:1时对电穿孔了0或2ug CD7 mRNA的A549-GFP肿瘤细胞的杀伤曲线。
图10显示的是基于mRNA的抗CD7 CAR(H1,H7,H9,H10,H17)+A40C28 T细胞在E/T比例=3:1时对电穿孔了0或2ug CD7 mRNA的A549-GFP肿瘤细胞的杀伤曲线。
图11显示的是在与电穿孔0或2ug CD7 mRNA的A549-GFP肿瘤细胞的共培养和杀伤试验中,不同mRNA的抗CD7 CAR-T细胞(A40C28+/-)的CD107a染色结果。
图12显示的是用同型对照和抗CD7 mAb对17个不同种类的肿瘤细胞系进行FACS染色的结果。
图13-14显示的是在与不同肿瘤细胞系的共培养和杀伤试验中,不同mRNA的抗CD7 CAR-T细胞(A40C28+/-)的CD107a染色结果。
图15显示的是本申请所述LACO-Stim分子的示意图。
图16显示的是通过流式细胞仪检查CAR/LACO-stim的表达的结果。
图17显示的是与共表达CAR和如表3所示的LACO-Stim的T细胞共培养后的肿瘤生长。E/T比值为30:1。E:效应细胞(T细胞);T:靶细胞(A549-ESO细胞)。
图18显示的是与如表1所示的共表达TCR和LACO-Stim的T细胞共培养后的肿瘤生长。E/T比值为30:1。E:效应细胞(T细胞);T:靶细胞(A549-ESO细胞)。
图19显示的是本申请所述circRNA的结构。
图20A-20B显示的是本申请所述circRNA对靶细胞的作用效果。
具体实施方式
以下由特定的具体实施例说明本申请发明的实施方式,熟悉此技术的人士可由本说明书所公开的内容容易地了解本申请发明的其他优点及效果。
术语定义
在本申请中,术语“抗原结合蛋白”通常是指包含结合抗原的部分的蛋白质,以及任选地允许结合抗原的部分采用促进抗原结合蛋白与抗原结合的构象的支架或骨架部分。抗原结合蛋白的实例包括但不限于抗体、抗原结合片段(例如,Fab,Fab’,F(ab)2,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb)、免疫缀合物、多特异性抗体(例如双特异性抗体)、抗体片段、抗体衍生物、抗体类似物或融合蛋白等,只要它们显示出所需的抗原结合活性即可。
在本申请中,术语“抗体”通常是在最广泛的意义上加以使用,并且具体地涵盖单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体(例如,双特异性抗体)、和抗体片段,只要它们显示所期望的生物活性(Milleretal(2003)Jour.ofImmunology170:4854-4861)。抗体可以是鼠、人、人源化、嵌合抗体,或源于其它物种。非限制性的,“抗体”典型地可以包含通过二硫键互相连接的至少两条重链(HC)和两条轻链(LC)的蛋白,或其抗原结合片段。每条重链包含重链可变区(VH)和重链恒定区。在某些天然存在的IgG、IgD和IgA抗体中,重链恒定区包含三个结构域,CH1、CH2和CH3。在某些天然存在的抗体中,各轻链包含轻链可变区(VL)和轻链恒定区。轻链恒定区包含一个结构域,CL。VH和VL区可进一步细分为超变性的区域,称为互补决定区(CDR),其与称为框架区(FR)的较保守的区域交替。各VH和VL包含三个CDR和四个框架区(FR),从氨基端至羧基端按以下顺序排列:FR1,CDR1,FR2,CDR2,FR3,CDR3和FR4。天然重链和轻链的可变结构域各自包含四个FR区(HFR1,HFR2,HFR3,HFR4,LFR1,LFR2,LFR3,LFR4),大部分采用β-折叠构型,通过三个CDRs连接,形成环连接,并且在一些情况下形成β-折叠结构的一部分。每条链中的CDRs通过FR区紧密靠近在一起,并与来自另一条链的CDR一起形成抗体的抗原结合位点。抗体的恒定区可介导免疫球蛋白与宿主组织或因子,包括免疫系统的各种细胞(例如,效应细胞)和经典补体系统的第一组分(Clq)结合。
在描述抗体序列中氨基酸片段的位置关系时,术语“位于……之间”通常是指某种氨基酸片段的C端与第一氨基酸片段的N端直接或间接连接,并且其N端与第二氨基酸片段的C端直接或间接连接。在轻链中,例如,LFR2的N末端与LCDR1的C末端直接或间接相连,且LFR2的C末端与LCDR2的N末端直接或间接相连。又例如,LFR3的N末端与LCDR2的C末端直接或间接相连,且LFR3的C末端与LCDR3的N末端直接或间接相连。在重链中,例如,HFR2的N末端与HCDR1的C末端直接或间接相连,且HFR2的C末端与HCDR2的N末端直接或间接相连。又例如,HFR3的N末端与HCDR2的C末端直接或间接相连,且HFR3的C末端与HCDR3的N末端直接或间接相连。
在本申请中,术语“抗原结合片段”通常是指抗体分子的一部分,其包含负责抗体与抗原之间的特异性结合的氨基酸。抗原结合片段可以包括:Fab,Fab’,F(ab)2片段,Fv片段,dsFv,F(ab’)2,scFv,di-scFv,dAb片段。术语“Fab”通常指含有Fv区、轻链的恒定结构域及重链的第一恒定结构域(CH1)。Fab’与Fab的不同处在于在重链CH1结构域的羧基末端添加几个残基,包括一或多个来自抗体铰链区的半胱胺酸。术语“Fv片段”通常指由抗体的单臂的VL和VH结构域组成的抗体片段,在某些情形中,该片段可以由一个VH和一个VL以紧密非共价结合的二聚体组成。术语“(Fab)2”通常是指含有铰链区和重链、轻链的可变区和第一个恒定区的二价片段。术语“F(ab’)2”通常指包含通过铰链区上的二硫键连接的两个Fab’片段的抗体片段。术语“scFv片段”通常包含抗体的VH和VL结构域,其中这些结构域是以单一多肽链存在。通常,scFv多肽进一步包含VH和VL结构域之间的多肽连接体,其使得scFv能够形成用于抗原结合的期望结构。术语“dAb片段”通常指由VH结构域组成的抗体片段。术语“dsFv”通常是指二硫键稳定的Fv片段,其单个轻链可变区与单个重链可变区之间的键是二硫键。
在本申请中,术语“可变”通常是指这样的事实,即抗体的可变结构域的序列的某些部分变化强烈,它形成各种特定抗体对其特定抗原的结合和特异性。然而,变异性并非均匀地分布在抗体的整个可变区中。它集中在轻链和重链可变区中的三个区段,被称为互补决定区(CDR)或高变区(HVR)。可变域中更高度保守的部分被称为框架(FR)。在本领域中,可以通过多种方法来定义抗体的CDR,例如基于序列可变性的Kabat定义规则(参见,Kabat等人,免疫学的蛋白质序列,第五版,美国国立卫生研究院,贝塞斯达,马里兰州(1991))、基于结构环区域位置的Chothia定义规则(参见,A1-Lazikani 等人,JMol Biol 273:927-48,1997)和基于IMGT本体论(IMGT-ONTOLOGY)的概念和IMGT Scientific图表规则的KABAT定义规则。本文使用的方法可以利用根据这些系统中的任何一种限定的CDRs,在一些实施方案使用Kabat或Chothia限定的CDRs,或者可以参考http://abysis.org/对scFv抗体的CDR进行划分。
在本申请中,术语“分离的”抗原结合蛋白通常是指已经从其产生环境(例如,天然的或重组的)的组分中识别,分离和/或回收的抗原结合蛋白。其产生环境的污染组分通常是干扰其研究、诊断或治疗用途的物质,可以包括酶、激素和其他蛋白质或非蛋白质溶质。分离的抗原结合蛋白或抗体通常将通过至少一个纯化步骤来制备。
在本申请中,术语“单克隆抗体”通常是指从一群基本上同质的抗体获得的抗体,即集群中的个别抗体是相同的,除了可能存在的少量的自然突变。单克隆抗体通常针对单个抗原位点具有高度特异性。而且,与常规多克隆抗体制剂(通常具有针对不同决定簇的不同抗体)不同,各单克隆抗体是针对抗原上的单个决定簇。除了它们的特异性之外,单克隆抗体的优点在于它们可以通过杂交瘤培养合成,不受其他免疫球蛋白污染。修饰语“单克隆”表示从基本上同质的抗体群体获得的抗体的特征,并且不被解释为需要通过任何特定方法产生抗体。例如,本申请使用的单克隆抗体可以在杂交瘤细胞中制备,或者可以通过重组DNA方法制备。
在本申请中,术语“人源化抗体”通常是指非人抗体(例如小鼠抗体)的CDR区以外的部分或全部有的氨基酸被源自人免疫球蛋白的相应的氨基酸置换的抗体。在CDR区中,氨基酸的小的添加、缺失、插入、置换或修饰也可以是允许的,只要它们仍保留抗体结合特定抗原的能力。人源化抗体可任选地包含人类免疫球蛋白恒定区的至少一部分。“人源化抗体”保留类似于原始抗体的抗原特异性。非人(例如鼠)抗体的“人源化”形式可以最低限度地包含衍生自非人免疫球蛋白的序列的嵌合抗体。在某些情形中,可以将人免疫球蛋白(受体抗体)中的CDR区残基用具有所期望性质、亲和力和/或能力的非人物种(供体抗体)(诸如小鼠,大鼠,家兔或非人灵长类动物)的CDR区残基替换。在某些情形中,可以将人免疫球蛋白的FR区残基用相应的非人残基替换。此外,人源化抗体可包含在受体抗体中或在供体抗体中没有的氨基酸修饰。进行这些修饰可以是为了进一步改进抗体的性能,诸如结合亲和力。
在本申请中,术语“全人源抗体”通常是指将人类编码抗体的基因转移至基因工程改造的抗体基因缺失动物中,使动物表达的抗体。抗体所有部分(包括抗体的可变区和恒 定区)均由人类来源的基因所编码。全人源抗体可以大大减少异源抗体对人体造成的免疫副反应。本领域获得全人源抗体的方法可以有噬菌体展示技术、转基因小鼠技术、核糖体展示技术和RNA-多肽技术等。
在本申请中,术语“scFv”通常是指包含至少一个包括轻链的可变区抗体片段和至少一个包括重链的可变区的抗体片段的融合蛋白,其中所述轻链和重链可变区是邻接的(例如经由合成接头例如短的柔性多肽接头),并且能够以单链多肽形式表达,且其中所述scFv保留其所来源的完整抗体的特异性。除非特别说明,否则如本文中使用的那样,scFv可以以任何顺序(例如相对于多肽的N-末端和C末端)具有所述的VL和VH可变区,scFv可以包括VL-接头-VH或可以包括VH-接头-VL。
在本申请中,术语“特异性”通常是指特定免疫球蛋白序列、抗原结合分子或抗原结合蛋白(例如免疫球蛋白单可变结构域、或本发明多肽)可以结合的不同类型的抗原或抗原决定簇的数量。抗原结合蛋白的特异性可以根据亲和力(affinity)和/或抗体亲抗原性(avidity)来确定。,术语“特异性的”和“特异性地”可被可互换地用于指示除CD7以外的生物分子不显著结合所述抗体。
在本申请中,术语“亲和力”通常是指单个单价配体对于其关联结合配偶体的结合常数的量度,例如,Fab’对于抗原或表位的结合。亲和力可以以几种方法进行测量,包括通过例如等离振子共振(BiaCore)测量结合和解离速率(分别为kon和koff),并且表示为总体结合(Kass)或解离常数(KD),其中Kass是kon/koff,和KD是koff/kon。KD也可以通过例如测量在其下配体与结合配偶体的结合是半饱和的浓度凭经验进行测量。测量KD的另一种方法是通过竞争测定,其中一种粘合剂或配体进行标记或加上标签,并且保持在恒定浓度下,而测试粘合剂或配体以各种浓度加入,以从其关联结合配偶体中竞争走标记的物质,且测定在其下标记减少一半的浓度。
在本申请中,术语“KD”、“KD”可互换地使用,通常是指平衡解离常数,“KD”是解离速率常数(kdis,也称为“解离率(off-rate)(koff)”或“kd”)与结合速率常数(kon,也称为“结合率(kon)”或“ka”)的比值。可使用结合速率常数(kon)、解离速率常数(kdis)和平衡解离常数(KD)表示抗原结合蛋白(例如抗体)对抗原的结合亲和力。确定结合和解离速率常数的方法为本领域熟知,包括但不限于生物膜干涉技术(BLI)、放射免疫法(RIA)、平衡透析法、表面等离子共振(SPR)、荧光共振能量迁移(FRET)、免疫共沉淀(Co-IP)以及蛋白质芯片技术。如果在不同的条件(例如盐浓度、pH)下测量,则 所测得的某种特定蛋白-蛋白相互作用的亲和力可不同。
在本申请中,术语“参比抗体”通常是指本申请所述抗原结合蛋白与之竞争结合抗原的抗体。
当在竞争相同表位的抗原结合蛋白上下文中使用的术语“竞争”通常表示抗原结合蛋白之间的竞争,如通过其中被测试的抗原结合蛋白(例如,抗体或其免疫功能片段)预防或抑制(例如,减少)参比抗原结合蛋白(例如,配体或参比抗体)与共同抗原(例如,CD7或其片段)特异性结合的测定所确定的。许多类型的竞争性结合测定可用于确定一个抗原结合蛋白是否与另一个竞争,例如:固相直接或间接放射性免疫测定(RIA)、固相直接或间接酶免疫测定(EIA)、夹心竞争测定(参见例如,Stahli等人,1983,Methods in Enzymology9:242-253);固相直接生物素-亲和素EIA(参见例如,Kirkland等人,1986,J.Immunol.137:3614-3619)固相直接标记测定、固相直接标记夹心测定(参见例如,Harlow和Lane,1988,Antibodies,A Laboratory Manual,Cold Spring Harbor Press);使用I-125标记的固相直接标记RIA(参见例如,Morel等人,1988,Molec.Immunol.25:7-15);固相直接生物素-亲和素EIA(参见例如,Cheung等人,1990,Virology176:546-552);和直接标记RIA(Moldenhauer等人,1990,Scand.J.Immunol.32:77-82)。通常,这种测定涉及与固体表面结合的纯化抗原或者携带这些任何一个的单元、未标记的测试抗原结合蛋白和标记的参比抗原结合蛋白的使用。竞争性抑制通过测定在测试抗原结合蛋白存在下于固体表面或单元结合的标记的量来测量。通常,测试抗原结合蛋白过量存在。通过竞争测定鉴定的抗原结合蛋白(竞争抗原结合蛋白)包括与参比抗原结合蛋白结合相同表位的抗原结合蛋白和与参比抗原结合蛋白结合的表位足够靠近而发生空间位阻的相邻表位结合的抗原结合蛋白。通常,当竞争抗原结合蛋白过量存在时,它使参比抗原结合蛋白与共同抗原的特异性结合抑制(例如,减少)至少约40-45%、约45-50%、约50-55%、约55-60%、约60-65%、约65-70%、约70-75%或约75%或更多。在一些情况下,结合被抑制至少约80-85%、约85-90%、约90-95%、约95-97%或约97%或更多。
术语“Fc”通常是指包含抗体恒定域的CH3、CH2和铰链区的至少一部分的多肽。任选地,Fc区可以包含存在于一些抗体类别中的CH4结构域。“Fc”可以包括:天然单体、天然二聚体(二硫键连接的)、修饰二聚体(二硫键和/或非共价连接的)和修饰单体(即 衍生物)。示例性修饰包括添加、缺失或置换一个或多个结构域中的一个或多个氨基酸。可以包括这类变化以优化效应子功能、半衰期等。
在本申请中,术语“分离的”抗原结合蛋白通常是指已经从其产生环境(例如,天然的或重组的)的组分中识别,分离和/或回收的抗原结合蛋白。其产生环境的污染组分通常是干扰其研究、诊断或治疗用途的物质,可以包括酶、激素和其他蛋白质或非蛋白质溶质。分离的抗原结合蛋白或抗体通常将通过至少一个纯化步骤来制备。
在本申请中,术语“百分比同一性”或“同一性”通常指两条或多条核酸或多肽序列相同的程度。在氨基酸序列的上下文中,术语“百分比同一性”或“同一性”通常描述了两个或更多个经比对的氨基酸序列与组成这些氨基酸序列的总长度的氨基酸残基数相比而言一致的氨基酸的匹配(“命中”)数。换言之,使用比对,对于两个或更多个序列,当将这些序列针对最大对应(如使用本领域已知的序列比较算法测量的)进行比较和比对时,或者当手动比对和视觉检查时,可以确定相同的氨基酸残基的百分比(例如65%、70%、75%、80%、85%、90%、95%或99%同一性)。因此,进行比较以确定序列一致性的序列可以通过一个或多个氨基酸取代、添加或缺失来区分。用于比对蛋白序列的适合程序是本领域技术人员已知的。蛋白序列的百分比序列一致性可以例如用程序如CLUSTALW、Clustal Omega、FASTA或BLAST来确定,例如使用NCBI BLAST算法(AltschulSF等人(1997),Nucleic Acids Res.[核酸研究]25:3389-3402)。
在本申请中,术语“免疫缀合物”或“抗体缀合物”通常是指抗体或其抗体片段与其它活性剂的连接,诸如化疗剂,细胞毒素(细胞毒性剂),免疫治疗剂,成像探针,光谱探针,等等。所述连接可以是共价键,或例如通过静电力的非共价相互作用。可以使用本领域中已知的多种接头以形成免疫缀合物。此外,该免疫缀合物可以以融合蛋白的形式提供,所述融合蛋白可以从编码该免疫缀合物的多核苷酸表达。术语“融合蛋白”通常指的是通过连接两个或多个最初编码独立的蛋白(包括肽和多肽)的基因或基因片段产生的蛋白。融合基因的翻译产生具有来自各原始蛋白的功能特性的单一蛋白。术语“细胞毒素”或“细胞毒性剂”可包括对细胞有害(例如,杀死)的任何药剂。
在本申请中,术语“嵌合抗原受体”或“CAR”通常是指一组多肽,在最简单的实施方案中通常有两种,其当在免疫效应细胞中时,提供细胞对靶细胞(通常为癌细胞)的特异性,并产生细胞内信号。在一些实施方案中,CAR包含至少一个细胞外抗原结合结构域(如VHH、scFv或其部分),跨膜结构域和胞质信号传导结构域(本文中也称为“胞内信 号传导结构域”),其包含衍生自如下所定义的刺激分子和/或共刺激分子的功能性信号传导结构域。在一些实施方案中,该组多肽在相同的多肽链中(例如,包含嵌合融合蛋白)。在一些实施方案中,该组多肽彼此不连续,例如在不同的多肽链中。在一些方面,该组多肽包括二聚化开关,其在二聚化分子的存在下可将多肽彼此偶联,例如可将抗原结合结构域偶联至胞内信号传导结构域。一方面,CAR的刺激分子是与T细胞受体复合物相关的ζ链。在一个方面,细胞质信号传导结构域包含一级信号传导结构域(例如,CD3-ζ的一级信号传导结构域)。在一个方面,细胞质信号传导结构域还包含一个或多个衍生自如下定义的至少一种共刺激分子的功能性信号传导结构域。一方面,共刺激分子可以选自4-1BB(即CD137),CD27,ICOS和/或CD28。一方面,CAR包含嵌合融合蛋白,其可以包含细胞外抗原识别结构域,跨膜结构域和包含衍生自刺激分子的功能性信号传导结构域的细胞内信号传导结构域。一方面,CAR包含嵌合融合蛋白,其可以包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含衍生自共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。一方面,CAR包含嵌合融合蛋白,其可以包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含衍生自一个或多个共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。一方面,CAR包括嵌合融合蛋白,其可以包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含至少两个衍生自一个或多个共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。在一个方面,CAR包含CAR融合蛋白的氨基末端(N-ter)上任选的前导序列。在一个方面,CAR进一步包含在细胞外抗原识别结构域的N末端的前导序列,其中前导序列任选地在细胞加工过程中从抗原识别结构域(例如VHH)切除,并将CAR定位于细胞膜。
在本申请中,术语“分离的核酸分子”或“分离的多核苷酸”通产是指基因组、mRNA、cDNA或合成来源的DNA或RNA或其一定组合,其不与在自然界中发现的多核苷酸的全部或一部分缔合,或连接至其在自然界中不连接的多核苷酸。
在本申请中,术语“载体”通常是指能够在合适的宿主中自我复制的核酸分子,其将插入的核酸分子转移到宿主细胞中和/或宿主细胞之间。所述载体可包括主要用于将DNA或RNA插入细胞中的载体、主要用于复制DNA或RNA的载体,以及主要用于DNA或RNA的转录和/或翻译的表达的载体。所述载体还包括具有多种上述功能的载体。所述载 体可以是当引入合适的宿主细胞时能够转录并翻译成多肽的多核苷酸。通常,通过培养包含所述载体的合适的宿主细胞,所述载体可以产生期望的表达产物。
在本申请中,术语“细胞”通常是指可以或已经含有包括本申请所述的核酸分子的质粒或载体,或者能够表达本申请所述的抗体或其抗原结合片段、多肽或免疫缀合物的个体细胞、细胞系或细胞培养物。所述细胞可以包括单个宿主细胞的子代。由于天然的、意外的或故意的突变,子代细胞与原始亲本细胞在形态上或在基因组上可能不一定完全相同,但能够表达本申请所述的抗体或其抗原结合片段即可。所述细胞可以通过使用本申请所述的载体体外转染细胞而得到。所述细胞可以是原核细胞(例如大肠杆菌),也可以是真核细胞(例如酵母细胞,例如COS细胞,中国仓鼠卵巢(CHO)细胞,HeLa细胞,HEK293细胞,COS-1细胞,NS0细胞或骨髓瘤细胞)。在某些情形中,所述细胞可以是哺乳动物细胞。例如,所述哺乳动物细胞可以是CHO-K1细胞。在本申请中,术语“重组细胞”通常是指在其中引入了重组表达载体的细胞。所述重组宿主细胞不仅包括某种特定的细胞,还包括这些细胞的后代。
在本申请中,术语“T细胞”或“T淋巴细胞”可以是任何T细胞,如培养T细胞,例如原代T细胞,或来自培养T细胞系的T细胞,例如Jurkat、SupTI等,或获自哺乳动物的T细胞(优选灵长类动物,物种,包括猴、狗或人)。如果获自哺乳动物,那么T细胞可以获自诸多来源,包括但不限于血液、骨髓、淋巴结、胸腺或其它组织或流体。T细胞还可以被富集或被化。T细胞可以通过在体外或体内将造血干细胞成熟化成T细胞而获得。在示范性方面中,T细胞是人类T细胞。在示范性方面中,T细胞是从人类中分离的T细胞。T细胞可以是任何类型的T细胞,包括NKT细胞,并且可以具有任何发育阶段,包括但不限于CD4+/CD8+双阳性T细胞;CDA+辅助T细胞;例如Th1和Th2细胞,CD8+T细胞(例如细胞毒性T细胞);外周血液单核细胞(PBMC);外周血液白细胞(PBL);肿瘤浸润细胞(TIL);记忆T细胞;未处理T细胞等等。优选地,T细胞是CD8+T细胞或CD4+T细胞。在一些替代方式中,T细胞与接受细胞或待接受细胞(例如所述细胞处于治疗组合物的形式)的接受受试者是同种异体的(来自相同物种的不同供体);在一些替代方式中,T细胞是自体的(供体和接受者相同);在一些替代方式中,T细胞是同基因的(syngeneic)(供体和接受者不同,但为同卵双胞胎)。
在本申请中,术语“免疫效应细胞”通常是指参与免疫应答,行使效应功能的免疫细胞。例如所述行使效应功能可以包括清除异物抗原或促进免疫效应子应答等。免疫效 应细胞可以包括浆细胞、T细胞、B细胞、自然杀伤(NK)细胞、自然杀伤T(NKT)细胞、肥大细胞和骨髓源性吞噬细胞。
本申请的免疫效应细胞可以是自体/自身的(autologous/autogeneic)(“自己的”)或非自体的(“非自己的”,例如同种异体的、同基因的或异基因的)。在本申请中,术语“自体的”通常是指来自相同受试者的细胞。“同种异体的”通常是指与相比较的为相同物种但在遗传上不同的细胞。“同基因的”通常是指在遗传上与相比较的细胞相同的不同受试者的细胞。“异基因的”通常是指物种与相比较的细胞不同的细胞。在一些实施方式中,本申请的细胞是自体的或同种异体的。
在本申请中,术语“修饰”通常是指改变细胞的状态或结构和/或细胞的状态或结构的改变。所述改变通常是与相应未经所述修饰的细胞的状态或结构相比,所述改变可以包括内源基因表达水平或功能的变化,例如通过基因工程手段使得细胞内源基因表达水平下调、上调或不表达,所述基因工程手段可以包括同源重组、CRISPR/Cas9系统基因编辑等;所述改变还可以包括细胞蛋白质表达、结构或功能的变化,例如通过所述内源基因表达水平或功能的变化而实现的相应蛋白质表达的变化、结构或功能的变化,例如通过调节蛋白质翻译、翻译后修饰而实现的蛋白质表达的变化、结构或功能的变化;所述改变还可以包括引入外源基因、表达外源蛋白质等。
在本申请中,术语“CRISPR/Cas系统”通常是指包含RNA引导的核酸酶或其他效应分子和gRNA分子的一组分子,所述分子能够指引和实现由RNA引导的核酸酶或其他效应分子在靶序列处修饰核酸,例如引起靶序列降解。在某些实施方式中,CRISPR系统包含gRNA和Cas蛋白,例如,Cas9蛋白。包含Cas9或其功能性突变体的系统在本申请中称作“Cas9系统”或“CRISPR/Cas9系统”。在某些实施方式中,gRNA分子和Cas分子可以复合,以形成核糖核蛋白(RNP)复合体。
在本申请中,术语“gRNA分子”、“向导RNA”、“指导RNA”、“指引RNA”、“向导RNA分子”或“gRNA”可互换使用,通常是指能够促进特异性指引RNA引导的核酸酶或其他效应分子(一般与gRNA分子复合)至靶序列上的核酸分子。在某些实施方案中,通过gRNA的一部分与DNA(例如,通过gRNA导引结构域)杂交并且通过gRNA分子的一部分与RNA指导的核酸酶或其他效应分子结合(例如,至少通过gRNA tracr)实现所述引导。在某些实施方案中,gRNA分子由单一的连续多核苷酸分子组成,在本文中称作“单一向导RNA”或“sgRNA”等。在其他实施方案中,gRNA分子由本身能够缔合 (一般通过杂交)的多个(例如二个)多核苷酸分子组成,在本文中称作“双重向导RNA”或“dgRNA”等。
在本申请中,术语“Cas蛋白”通常是指CRISPR/Cas系统中负责剪切DNA的酶。可以包括来自Ⅰ、Ⅱ、Ⅲ型CRISPR/Cas系统的酶。例如,Cas3、Cas9、Cas10。
在本申请中,术语“Cas9蛋白”通常是指负责剪切DNA的来自细菌II型CRISPR/Cas系统的酶。Cas9可以包括野生型蛋白及其有功能性突变体。
在本申请中,术语“减少”和“降低”可互换使用并且通常表示小于原来的任何变化。“减少”和“降低”是相对的术语,需要在测量前和测量后间进行比较。“减少”和“降低”包括完全耗竭。
在本申请中,术语“降低”可以通过本领域已知标准方法(诸如本申请中描述的那些)检测的,基因、基因产物例如蛋白质或生物标志物在第一样品中的表达水平/量与相应基因、基因产品例如蛋白质或生物标志物在第二样品中的表达水平/量相比约5%、10%、20%、25%、30%、40%、50%、60%、70%、80%、85%、90%、95%、或100%的总体降低。在某些实施方式中,术语“降低”指第一样品中基因或生物标志物的表达水平/量的降低,其中该降低是第二样品中相应基因或生物标志物的表达水平/量的至少约0.9倍、0.8倍、0.7倍、0.6倍、0.5倍、0.4倍、0.3倍、0.2倍、0.1倍、0.05倍、或0.01倍。在某些实施方式中,第一样品是自受试者获得的样品,而第二样品是参照样品。
在本申请中,术语“药学上可接受的载体”通常是指不干扰活性成分生物学活性的有效性的一种或多种无毒物质。这类制剂通常可含有盐、缓冲剂、防腐剂、相容性载体、佐剂和任选的其它治疗剂。这类药学上可接受的制剂通常也可包含适合给予人的相容性固体或液体填料、稀释剂或包囊材料。非限制性的,药学上可接受的载体可包括液体,例如水、盐水、甘油和乙醇。这些载体中也可存在辅助物质,例如润湿剂或乳化剂、pH缓冲物质等。术语“佐剂”通常是指辅助或调节药物作用的任何物质,包括但不仅限于免疫学佐剂,它使对抗原的免疫反应增强或免疫反应多样化。
在本申请中,术语“预防和/或治疗”不仅包括预防和/或治疗疾病,还通常包括预防疾病的发作,减缓或逆转疾病的进展,预防或减缓与疾病相关的一种或多种症状的发作,减少和/或减轻与疾病相关的一种或多种症状,降低疾病和/或与其相关的任何症状的严重程度和/或持续时间和/或预防疾病和/或与其相关的任何症状的严重程度的进一步增加,预防、减少或逆转由疾病引起的任何生理损伤,以及通常对正在治疗的患者有益的任何 药理学作用。本申请的组合物形成可行的治疗剂不需要实现完全治愈或根除疾病的任何症状或表现。如在相关领域中所认识到的,用作治疗剂的药物可降低给定疾病状态的严重程度,但不需要消除疾病的每种表现才能被认为是有用治疗剂。类似地,预防性施用的治疗构成可行的预防剂不需要完全有效地预防病症的发作。简单地在受试者中减少疾病的影响(例如,通过减少其症状的数量或严重程度,或通过提高另一种治疗的有效性,或通过产生另一种有益效果),或减少疾病发生或恶化的可能性就足够了。
在本申请中,术语“疾病”或“病症”可以互换使用,通常是指受试者与正常状态的任意偏离,例如身体或某些器官的状态的任何变化,妨碍或扰乱了功能的履行,和/或在患病或与其接触的人中引起症状例如不适、机能障碍、痛苦或甚至死亡。
在本申请中,术语“肿瘤”通常是指所有赘生性(neoplastic)细胞生长和增殖,无论是恶性的还是良性的,及所有癌前(pre-cancerous)和癌性细胞和组织。术语“癌症”,“癌性”,“细胞增殖性病症”,“增殖性病症”和“肿瘤”在本文中提到时并不互相排斥。在本申请中,所述肿瘤可以包括实体瘤和/或血液瘤。
在本申请中,术语“施用”通常是指通过本领域已知的任意途径,将蛋白(包括免疫球蛋白)递送给有此需要的人或动物。药用载体和制剂或组合物也是本领域众所周知的。给药途径可以包括:静脉内的、肌肉内的、真皮内的、皮下的、透皮的、粘膜的、瘤内的或粘膜的。或者,这些术语可以表示,将用于重组蛋白表达的载体递送给细胞或培养的细胞和/或受试者的细胞或器官。这样的施用或导入可以在体内、在体外或先体外后体内地发生。通过下述方式,可以将用于重组蛋白或多肽表达的载体导入细胞中:转染,其通常是指,通过物理方式(例如,磷酸钙转染、电穿孔、显微注射或脂转染)将异源DNA插入细胞中;感染,其通常是指,借助于传染性病原体(即病毒)的导入;或转导,其通常是指,病毒对细胞的稳定感染,或遗传物质通过病毒剂(例如,噬菌体)从一种微生物向另一种微生物的转移。
在本申请中,术语“接触”通常是指两种两个或更多个不同类型的物质以任何顺序、任何方式以及任何时长接触在一起。当应用于细胞时,术语“接触”通常指借以将本申请的抗原结合蛋白、多肽、免疫缀合物、核酸、载体、细胞和/或药物组合物递送到靶细胞或与靶细胞直接靠近放置的方法,该递送可以是体外的或体内的并且可以涉及使用重组载体系统。例如,“接触”可以包括将多核苷酸置于含有核酸分子的烧杯、微量滴定板、细胞培养瓶或微阵列等中。又例如,接触可以包括将抗体置于含有多肽的烧杯、微量滴定板、细 胞培养瓶或微阵列等中,接触可以发生在体内(in vivo)、间接体内(ex vivo)或体外(in vitro)。
在本申请中,术语“有效量”或“有效剂量”通常是指足以产生所需的治疗反应而没有过度的不利副作用如毒性、刺激性或变态反应的活性治疗剂的量。显然,具体的“有效量”将随各种因素而变化,所述因素如治疗的特定病症,患者的生理状况,治疗的动物的类型,治疗的持续时间,并存疗法(如果有的话)的性质,以及所用的具体制剂和化合物或其衍生物的结构。在此情况中,如果某个量导致(不限于)以下中的一种或多种则其将被认为是治疗有效的:(a)抑制癌细胞生长(例如AML细胞);以及(b)杀死癌细胞(例如AML细胞)。
在本申请中,术语“受试者”通常是指人类或非人类动物,包括但不限于猫、狗、马、猪、奶牛、羊、兔、小鼠、大鼠或猴等。在某些实施方式中,所述受试者是人。“有需要的受试者”可以指患有或处于发展中的疾病或风险中的可通过诱导T细胞对恶性T细胞的细胞毒性发挥特异性作用而被治疗(例如,改善、改进、预防)的受试者(例如患者)。
在本申请中,术语“circRNA”通常称环形RNA,是一种新型的非编码RNA(noncoding RNA,ncRNA)分子。根据RNA构成的不同,环状RNA可分为三类:外显子环状RNA(exon circular RNA,ecircRNA)、内含子环状RNA(circular intronic RNAs,ciRNAs)和外显子-内含子circRNA(exon-intron circRNA,EIciRNA)。
在本申请中,术语“包括”、“包含”、“具有”、“可以”、“含有”及其变体通常旨在是开放式过渡性短语、术语或词语,其不排除额外行为或结构的可能性。术语“由……组成”通常表示不能存在别的组分(或同样地,特征、整数、步骤、等)。除非上下文另有明确规定,单数形式如英文的“a”,“an”,“the”,中文的“一个”、“一种”和“所述/该”一般包括所指代事物的复数形式。
在本申请中,术语“约”通常意指大约(approximately)、在......的附近(in the region of)、粗略地(roughly)、或左右(around)。当术语“约”当用于指涉数值范围时,截值或特定数值用于指示所载明的数值可与该列举数值有多达10%的差异。例如在指定数值以上或以下0.5%、1%、1.5%、2%、2.5%、3%、3.5%、4%、4.5%、5%、5.5%、6%、6.5%、7%、7.5%、8%、8.5%、9%、9.5%、或10%的范围内变动。
发明详述
经修饰的免疫效应细胞
一方面,本申请提供一种经修饰的免疫效应细胞,其包含编码融合蛋白的核酸分 子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
在一些实施方案中,所述APC选自由树突状细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的群组。
在一些实施方案中,所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、中性粒细胞和粒细胞组成的群组。
在一些实施方案中,所述融合蛋白为膜蛋白。在一些实施方案中,所述融合蛋白为可溶性蛋白。在一些实施方案中,融合蛋白为双特异性抗体。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中,第一结构域的N-末端连接到第二结构域的C-末端。
在某些实施方式中,其中所述第一结构域与第二结构域直接连接或间接连接。
在某些实施方式中,其中所述第一结构域与第二结构域通过连接子连接。
在某些实施方式中,其中所述连接子包括肽连接子。所述连接子可以为柔性连接子或刚性连接子。
在一些实施方案中,所述连接子具有(GGGGS)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(EAAAK)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(PA)nP,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有GSGGGGSGGGGSGGGGS(SEQ ID NO:252)的氨基酸序列。在一些实施方案中,所述连接子具有GGGGS(SEQ ID NO:253)的氨基酸序列。在一些实施方案中,所述连接子为CD8铰链。在一些实施例中,所述连接子为CD28铰链。在一些实施例中,所述连接子为IgG Fc铰链。在一些实施例中,所述连接子可以为一种三聚基序,其选自由T4纤维蛋白三聚基序、异亮氨酸拉链基序、GCN4II基序、Matrilin-1基序和XV型胶原三聚基序组成的群组。
在本申请中,APC是指在其表面显示一种或多种抗原的任何细胞,例如,与一种或多种主要组织相容性复合体(MHC)蛋白结合。所述MHC/抗原复合物可被T细胞利用其T细胞受体(TCRs)识别并引起免疫应答。在某些实施方式中,其中所述APC选自由树 突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
APCs激活受体
如本领域所理解,一种分子可以通过促进APC的成熟、促炎状态、细胞毒性、抗原呈递、表位扩散、细胞因子产生、免疫效应细胞(例如T细胞)的共刺激或其中任何组合来激活APC。在一些实施方案中,本发明所提供融合蛋白的第一结构域通过促进APC(例如DC)的成熟和活化来激活APC。在一些实施方案中,本发明所提供融合蛋白的第一结构域通过促进APC和其他免疫效应细胞(例如T细胞)之间的表位扩散来激活APC。在一些实施方案中,本发明所提供融合蛋白的第一结构域通过促进APC的抗原呈递来激活APC。在一些实施方案中,本发明所提供融合蛋白的第一结构域通过促进APC对异物(例如癌细胞)的细胞毒性来激活APC。
在一些实施方案中,本发明提供的融合蛋白包括激活APC的第一结构域,其包括结合APC的激活受体的配体或其受体结合片段。“激活受体”是指APC上表达的一种膜蛋白,它与配体或抗体结合后,能引发信号以促进APC的流动、分化、增殖和/或激活。APC激活受体包括,例如,CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
受体的“配体”是指能选择性结合受体的分子。在一些实施方案中,所述配体为一种多肽。配体的“受体结合片段”是指配体保留其受体结合能力的片段。各种配体可通过与APC上的激活受体结合,刺激树突状细胞或其他APCs的生长、分化、迁移和/或激活。(见Banchereau J等,Nature(1998)392:245-52;Young JW等,Stem Cells(1996)14:376-387;Cella M等,Curr Opin Immunol.(1997)9:10-16;Curti A等,J.Biol.Regul.Homeost.Agents(2001)15:49-52)。能够调节树突状细胞或其他APCs分化、成熟、扩增和/或激活的配体实例包括,例如CD40配体(CD40L)、CD80配体、CD86配体、CD91配体(RAP1)、DEC-205配体和DC-SIGN配体。在一些实施方案中,本发明提供的融合蛋白包括第一结构域,所述第一结构域包括本发明公开的结合APC激活受体的配体或其受体结合片段。
CD40/CD40L:CD40是一种48kD的跨膜糖蛋白表面受体,其为肿瘤坏死因子受体超家族(TNFRSF)的成员。描述了人CD40的典型氨基酸序列(参见,登录号:ALQ33424.1 GI:957949089),CD40最初被描述为APC上表达的共刺激受体,在B细胞和T细胞激活中发挥核心作用。CD40的配体CD154(也称为TRAP、T-BAM、CD40配体或CD40L)是一种II型整合膜蛋白。据报道,CD40L可促进树突状细胞的诱导,并促进免疫原性应 答的发生。参见,例如,Elgueta R等,Immunol Rev.(2009)229(1):10.1111;Ma D&Clark EA,Semin Immunol.2009 21(5):265–272;Borges L等J Immunol.(1999)163:1289-1297;Grewal I,Immunol Res.(1997)16:59-70。描述了编码CD40配体和等同物的示例性多核苷酸(参见,例如,Genbank登录号:X65453和L07414),以及制剂、组合物和使用方法(美国专利No.6,290,972)。以下提供了人CD40L的示例性氨基酸序列。胞外结构域(SEQ ID NO:224)以下划线标出。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括CD40L或CD40L的受体结合片段。在一些实施方案中,CD40L的受体结合片段包括CD40L(SEQ ID NO:223)的119-261位氨基酸。在一些实施方案中,CD40L的受体结合片段包括CD40L的胞外结构域。
在某些实施方式中,其中所述第一结构域包含结合所述APC的激活受体的抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括结合CD40、CD80、CD86、CD91、DEC-205或DC-SIGN的抗体或抗原结合片段。例如,其中所述第一结结构域可以为抗CD40抗体或其抗原结合片段。
免疫效应细胞的共刺激受体
在一些实施方案中,本发明提供的融合蛋白包含激活抗原提呈细胞(例如,树突状细胞)的第一结构域的和激活免疫效应细胞(例如,T细胞)的第二结构域,其中所述第二结构域包括(a)所述免疫效应细胞的共刺激受体或其功能片段,(b)所述免疫效应细胞的共刺激配体,或其受体结合片段,或(c)结合免疫效应细胞共刺激受体的抗体,或其抗原结合片段。
如本发明所用且如本领域所理解,“免疫效应细胞”是指具有造血起源并在针对靶标(如病原体、癌细胞或外来物质)的免疫应答中发挥直接作用的细胞。免疫效应细胞包括T细胞、B细胞、自然杀伤(NK)细胞、NKT细胞、巨噬细胞、粒细胞、中性粒细胞、嗜酸性粒细胞、肥大细胞和嗜碱性粒细胞。在一些实施方案中,本发明所提供融合蛋白的 激活免疫效应细胞的第二结构域包括免疫效应细胞的共刺激受体。在一些实施方案中,所述免疫效应细胞为T细胞、NK细胞、NKT细胞、巨噬细胞、中性粒细胞或粒细胞。在一些实施方案中,所述免疫效应细胞为T细胞。在一些实施方案中,所述免疫效应细胞为NK细胞。在一些实施方案中,所述免疫效应细胞为巨噬细胞。
免疫效应细胞的“刺激”是指通过刺激分子与其同源配体结合从而在免疫效应细胞中介导信号转导事件而诱导的初级应答,其可以改变某些基因的表达和/或细胞骨架结构的重组等。免疫效应细胞的“刺激分子”是指免疫效应细胞上的一种分子,它与通常存在于APC上的同源配体结合后,可以介导信号转导以促进免疫效应细胞的成熟、分化、增殖和/或激活。例如,T细胞的刺激分子,TCR/CD3复合物触发T细胞的激活。刺激分子的配体或“刺激配体”是指通常存在于APC上并能与免疫效应细胞上的刺激分子结合以介导免疫效应细胞的初级应答的配体,所述初级应答包括但不限于成熟、分化、激活、启动免疫应答、增殖等。刺激配体在本领域中是众所周知的,并且包括例如负载肽、抗CD3抗体、超激动剂抗CD28抗体和超激动剂抗CD2抗体的MHC I类分子。
如本发明所用且如本领域所理解,“共刺激信号”是指来自共刺激受体(例如CD28或4-1BB)的信号,其与初级信号(例如TCR/CD3)结合促进免疫效应细胞(例如T细胞)的最佳克隆扩增、分化和效应功能。如本发明所用且如本领域所理解,免疫效应细胞的“共刺激受体”是指免疫效应细胞上与“共刺激配体”特异性结合以介导免疫效应细胞的共刺激反应的分子,所述共刺激反应例如增强免疫效应细胞的激活或增殖。免疫效应细胞的共刺激受体包括但不限于CD28、4-1BB、ICOS、CD27、OX40、DAP10、CD30、2B4、CD2、LIGHT、GITR、TLR、DR3和CD43。共刺激受体的“功能片段”是共刺激受体的片段,其保留共刺激受体介导共刺激信号和刺激免疫效应细胞的功能。在一些实施方案中,共刺激受体的功能片段保留了共刺激受体的共刺激结构域。在一些实施方案中,共刺激结构域为共刺激受体的胞质结构域。在一些实施方案中,来自免疫效应细胞(例如T细胞)的共刺激受体的信号降低了免疫效应细胞的激活阈值。在一些实施方案中,来自T细胞共刺激受体的信号导致TCR信号事件增强,其中所述TCR信号为高效细胞因子产生(通过增强转录活性和信使RNA稳定性)、细胞周期进程、存活、代谢调节和T细胞应答所必需。
如本发明所用且如本领域所理解,“共刺激配体”是指特异地结合免疫效应细胞上的同源共刺激受体的分子,从而提供除了由刺激分子提供的初级信号之外的信号,该信号 介导免疫效应细胞中的应答,包括但不限于增殖、激活、分化等。共刺激配体可以存在于APC上(例如,树突状细胞)。共刺激配体包括但不限于CD58、CD70、CD83、CD80、CD86、CD137L(4-1BBL)、CD252(OX40L)、CD275(ICOS-L)、CD54(ICAM-1)、CD49a、CD112(PVRL2)、CD150(SLAM)、CD155(PVR)、CD265(RANK)、CD270(HVEM)、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153(CD30L)、CD48、CD160、CD200R(OX2R)和CD44。共刺激配体的“受体结合片段”是指配体保留其结合受体能力的片段。
下面举例说明了一些共刺激受体和共刺激配体。应理解,本发明提供的或本领域已知的任何共刺激受体和/或共刺激配体可用作本发明提供的融合蛋白的一部分。
CD28:分化簇28(CD28)是一种在T细胞上表达的蛋白,其为T细胞激活和存活提供共刺激信号。CD28是CD80(B7.1)和CD86(B7.2)蛋白的受体。CD28是T细胞最佳克隆扩增、分化和效应功能的共刺激受体。CD28结合降低了T细胞激活阈值并导致TCR信号事件增强,其中所述TCR信号为高效细胞因子产生(通过增强转录活性和信使RNA稳定性)、细胞周期进程、存活、代谢调节和T细胞应答所必需。CD28是免疫突触(IS)组织的关键因素,在其中CD28增强T细胞和APC之间的密切接触。
在一些实施方案中,本发明所提供的融合蛋白包含激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第二结构域包含CD28多肽或其功能片段。在一些实施方案中,所述第二结构域包括CD28的胞质结构域。在一些实施方案中,本发明所提供的融合蛋白包含激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第二结构域包含结合CD28的配体或其受体结合片段。在一些实施方案中,本发明所提供的融合蛋白包含激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第二结构域包含结合CD28的抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括CD28的功能片段,所述CD28的功能片段包括CD28的胞内/胞质结构域的一部分,其可作为共刺激信号传导结构域发挥作用。CD28可以具有如下氨基酸序列或其功能片段:所述氨基酸序列对应于GenBank No.P10747(P10747.1,GI:115973)或NP_006130(NP_006130.1,GI:5453611)的序列。在一个实施方案中,本发明公开的融合蛋白可具有包含CD28胞质结构域的氨基酸序列或其片段,所述CD28胞质结构域对应于CD28的180至122位氨基酸(如下序列的下划线部分,SEQ ID NO:226)。在另一实施方案中,本发明公开的融合蛋白可以具有进一步包含CD28跨膜结构域的氨基酸序列或其功能片段,所述CD28跨膜结构域对应于153至179位氨基酸。应理解, 如果需要,短于或长于特定描述结构域的CD28序列可包含在本发明公开的融合蛋白中。
在某些实施方式中,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
在某些实施方式中,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
在某些实施方式中,其中共刺激受体为CD28。例如,其中第二结构域可以是抗CD28抗体或其抗原结合片段。
在一些实施方案中,所述第一结构域包括(a)结合所述APC的激活受体的配体,或其受体结合片段,或(b)结合所述APC的激活受体的抗体,或其抗原结合片段,其中所述APC的激活受体选自由CD40、CD80、CD86、CD91、DEC-205和DC-SIGN组成的群组。在一些实施方案中,所述第一结构域包括结合APC的激活受体的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括结合CD40的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括CD40L。在一些实施方案中,CD40L的受体结合片段包括CD40L(SEQ ID NO:223)的119-261位氨基酸。在一些实施方案中,CD40L的受体结合片段包括CD40L的胞外结构域。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括三个拷贝的CD40L或CD40L的受体结合片段。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括三个拷贝的CD40L(SEQ ID NO:223)的119-261位氨基酸。在一些实施方案中,所述第一结构域包括结合CD80的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括结合CD86的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括CD28胞外结构域。在一些实施方案中,所述第一结构域包括CD28。在一些实施方案中,所述第一结构域包括CTLA-4的胞外结构域。在一些实施方案中,所述第一结构域包括CTLA-4。在一些实施方案中,所述第一结构域包括结合CD91的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括RAP1的结构域3。在一些实施方案中,所述第一结构域包括RAP1。在一些实施方案中,所述第一结构域包括结合DEC-205的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括结合DC-SIGN的配体或其受体结合片段。在一些实施方案中,所述第一结构域包括 ICAM2、ICAM3、CD18或CEACAM1或其受体结合片段。在一些实施方案中,所述第一结构域包括ICAM2或其受体结合片段。在一些实施方案中,所述第一结构域包括ICAM3或其受体结合片段。在一些实施方案中,所述第一结构域包括CD18,或其受体结合片段。在一些实施方案中,所述第一结构域包括CEACAM1或其受体结合片段。
在一些实施方案中,所述第一结构域包括结合APC的激活受体的抗体或其抗原结合片段。在一些实施方案中,所述APC的激活受体选自由CD40、CD80、CD86、CD91、DEC-205和DC-SIGN组成的群组。在一些实施方案中,所述第一结构域包括结合CD40的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD80的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD86的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD91的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合DEC-205的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合DC-SIGN的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括单克隆抗体。在一些实施方案中,所述第一结构域包括嵌合抗体。在一些实施方案中,所述第一结构域包括人源化抗体。在一些实施方案中,所述第一结构域包括人抗体。在一些实施方案中,所述第一结构域包括Fab、Fab'、F(ab')2、Fv、scFv、(scFv)2、单链抗体、双可变区抗体、双特异性抗体、纳米抗体或单可变区抗体。在一些实施方案中,所述第一结构域包括人抗体。在一些实施方案中,所述第一结构包括scFv。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40 scFv。在一些实施方案中,所述抗CD40抗体或其抗原结合片段包括标记为F2.103、F5.157、F5.77、4D11、A40C或119的抗体,如下表1所示。
表1:抗CD40抗体

在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40scFv。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40scFv,所述抗CD40scFv与SEQ ID NO:227至SEQ ID NO:232中任一项所示序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%序列同一性。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括具有SEQ ID NO:227 至SEQ ID NO:232中任一项所示氨基酸序列的抗CD40scFv。
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括免疫效应细胞的共刺激受体或其功能片段,其中所述免疫细胞为T细胞、NK细胞、NKT细胞、巨噬细胞、中性粒细胞或粒细胞。在一些实施方案中,所述免疫效应细胞的共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3和CD43组成的群组。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括共刺激受体的功能片段,所述共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3和CD43组成的群组。
在一些实施方案中,所述功能片段包括共刺激受体的胞质结构域。在一些实施方案中,本发明提供的融合蛋白的第二结构域进一步包括共刺激受体的跨膜结构域。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3或CD43的功能片段。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3或CD43的胞质结构域。
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。所述免疫效应细胞可选自由T细胞、NK细胞、NKT细胞、巨噬细胞、中性粒细胞和粒细胞组成的群组。在一些实施方案中,所述免疫效应细胞的共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3和CD43组成的群组。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括结合CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、DR3或CD43的抗体或其抗原结合片段。
在一些实施方案中,所述第二结构域包括单克隆抗体。在一些实施方案中,所述第二结构域包括嵌合抗体。在一些实施方案中,所述第二结构域包括人源化抗体。在一些实施方案中,所述第二结构域包括人抗体。在一些实施方案中,所述第二结构域包括Fab、Fab'、F(ab')2、Fv、scFv、(scFv)2、单链抗体、双可变区抗体、双特异性抗体、纳米抗体或单可变区抗体。在一些实施方案中,所述第二结构域包括人抗体。在一些实施例中,所述第二结构域包括scFv。
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括抗CD28抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括抗CD28 scFv。 在一些实施方案中,所述抗CD28抗体或其抗原结合片段包括标记为1412的抗体。
表2:示例性抗CD28抗体
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括抗CD28scFv,所述抗CD28scFv与SEQ ID NO:233所示序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%序列同一性。在一些实施方案中,本发明提供的融合蛋白的第二结构域包括具有SEQ ID NO:233所示氨基酸序列的抗CD28 scFv。
本发明描述的融合蛋白(即,LACO-Stim分子)可以包括本发明公开的或本领域其他已知的APC激活剂(结合激活受体的配体或抗体)和免疫效应细胞激活剂(共刺激受体或结合共刺激受体的抗体)的任何组合。为便于说明,下文提供了各种形式的CD40-C28LACO-Stim融合蛋白,其通过CD40/CD40L信号转导激活APCs(例如,树突状细胞),并通过CD28信号转导激活免疫效应细胞(例如,T细胞)。
示例性LACO-Stim(1):APC激活受体的配体+共刺激受体(例如,CD40L-CD28)
在一些实施方案中,本发明提供的融合蛋白包括激活抗原提呈细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合APC的激活受体的配体或其受体结合片段,所述第二结构域包括免疫效应细胞的共刺激受体或其功能片段。在一些实施方案中,所述第二结构域包括免疫效应细胞的共刺激受体的胞质结构域。在一些实施方案中,所述第一结构域的C-末端连接到所述第二结构域的N-末端。在一些实施方案中,所述第一结构域的N-末端连接到所述第二结构域的C-末端。在一些实施方案中,本发明所提供融合蛋白为膜融合蛋白。在一些实施方案中,第一结构域和第二结构域通过连接子连接。所述连接子可以为柔性连接子或刚性连接子。在一些实施方案中,所述连接子具有(GGGGS)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(EAAAK)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(PA)nP,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有 GSGGGGSGGGGSGGGGS的氨基酸序列。在一些实施方案中,所述连接子具有GGGGS的氨基酸序列。
在一些实施方案中,所述第一结构域包括结合APC激活受体的配体或其受体结合片段,所述APC激活受体选自由CD40、CD80、CD86、CD91、DEC-205和DC-SIGN组成的群组。在一些实施方案中,所述第二结构域包括共刺激受体或其功能片段,所述共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43组成的群组。
在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括CD28胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括4-1BB胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括ICOS胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括CD27胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括OX40胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括DAP10胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括2B4胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括CD30胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括CD2胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括LIGHT胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括GITR胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L 或其受体结合片段,所述第二结构域包括TLR胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括DR3胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括CD43胞质结构域。所述CD40L的受体结合片段可以为CD40L(SEQ ID NO:223)的119-261位氨基酸。在一些实施方案中,所述第一结构域包括全长CD40L。

在一些实施方案中,本发明提供的融合蛋白具有的氨基酸序列与标记为40L.28.40L.40L,TriCD40L_8-28,或TriCD40L_28-28的融合蛋白的序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%同一性。
如本领域普通技术人员所理解的,本发明所示例的融合蛋白中的CD40L功能片段或全长CD40L可以用本发明所公开的或本领域其他已知的APC激活受体的不同配体替换,包括例如CD80配体(例如,CD28或CTLA-4)的胞外结构域或其全长、CD86配体(例如,CD28或CTLA-4)、CD91配体(例如,RAP1)、DEC-205配体或DC-SIGN配体(例如,ICAM2、ICAM3、CD18或CEACAM1)。如本领域普通技术人员所理解的,本发明所例示的融合蛋白中的CD28胞质结构域可以用本发明所公开的或本领域其他已知的免疫效应细胞的不同共刺激因子的胞质结构域替换,包括例如,4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的胞质结构域;或4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的不同功能片段,所述功能片段保留全长蛋白的激活免疫效应细胞的功能。
示例性LACO-Stim(2):APC激活受体配体+结合共刺激受体的抗体(例如,aCD28- CD40L)
在一些实施方案中,本发明提供的融合蛋白包括激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合APC的激活受体的配体或其受体结合片段,并且其中所述第二结构域包括结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中第一结构域的N-末端连接到第二结构域的C-末端。在一些实施方案中,本发明提供的融合蛋白是基于抗体的可溶性蛋白。
在一些实施方案中,本发明公开的融合蛋白的两个结构域通过三聚基序连接。在一些实施方案中,连接子为一种三聚基序,其选自由T4纤维蛋白三聚基序、异亮氨酸拉链、GCN4II基序、Matrilin-1基序和XV型胶原三聚基序组成的群组。在一些实施方案中,连接子为T4纤维蛋白三聚基序。
在一些实施方案中,第一结构域包括结合APC激活受体的配体或其受体结合片段,所述APC激活受体选自由CD40、CD80、CD86、CD91、DEC-205和DC-SIGN组成的群组。在一些实施方案中,所述第二结构域包括结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,其中所述共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43组成的群组。
在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗CD28抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗4-1BB抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗ICOS抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗CD27抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗OX40抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗DAP10抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段, 所述第二结构域包括抗2B4抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗CD30抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗CD2抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗LIGHT抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗GITR抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗TLR抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗DR3抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括抗CD43抗体或其抗原结合片段。在一些实施方案中,CD40L的受体结合片段可以具有CD40L(SEQ ID NO:223)的119-261位氨基酸。
在一些实施方案中,本发明提供的融合蛋白具有包括CD40L或其受体结合片段的第一结构域和包括抗CD28抗体或其抗原结合片段的第二结构域。所述抗CD28抗体或抗原结合片段可以是本发明公开的或本领域其他已知的可激活CD28信号传递的任何抗CD28抗体或抗原结合片段。在一些实施方案中,所述抗CD28抗体或抗原结合片段为标记为1412的抗体。在一些实施方案中,所述抗CD28抗体或其抗原结合片段包括具有SEQ ID NO:233所示氨基酸序列的抗CD28 scFv。

在一些实施方案中,本发明提供的融合蛋白具有的氨基酸序列与标记为1412-T4-CD40L的融合蛋白的序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%同一性。
如本领域普通技术人员所理解的,本发明示例的融合蛋白中CD40L胞外结构域可以用本发明公开的或本领域其他已知的APC激活受体的不同配体的胞外结构域或受体结合片段替换,包括例如CD80配体(例如,CD28或CTLA-4)、CD86配体(例如,CD28或CTLA-4)、CD91配体(例如,RAP1)、DEC-205配体或DC-SIGN配体(例如,ICAM2、ICAM3、CD18或CEACAM1)的胞外结构域或受体结合结构域。如本领域普通技术人员所理解的,本发明所示例的融合蛋白中的抗CD28抗体或抗原结合片段可以用本发明所公开的或本领域其他已知的结合和激活免疫效应细胞不同共刺激因子的抗体或抗原结合片段替换,包括例如结合4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的抗体或抗原结合片段。
示例性LACO-Stim(3):APC激活受体的抗体+共刺激受体的抗体(例如,aCD40/aCD28 双特异性Ab)
在一些实施方案中,本发明提供了双特异性抗体。如本发明所用和如本领域所理解的,“双特异性抗体”是指对至少两个不同的抗原表位具有结合特异性的抗体。所述表位可以来自同一抗原,也可以来自两个不同的抗原。在一些实施方案中,本发明提供的融合蛋白包括激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合APC的激活受体的抗体或其抗原结合片段,并且其中所述第二结构域包括结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。因此,本发明公开的双特异性抗体具有对(1)APC(例如,树突状细胞)的激活受体和(2)免疫效应细胞(例如,T细胞)的共刺激受体的结合特异性。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中,第一结构域的N-末端连接到第二结构域的C-末端。
在一些实施方案中,第一结构域和第二结构域通过连接子连接。所述连接子可以为柔性连接子或刚性连接子。在一些实施方案中,所述连接子具有(GGGGS)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(EAAAK)n,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有(PA)nP,n=1、2、3、4或5的氨基酸序列。在一些实施方案中,所述连接子具有GSGGGGSGGGGSGGGGS的氨基酸序列。在一些实施方案中,所述连接子具有GGGGS的氨基酸序列。
在一些实施方案中,本发明提供的融合蛋白为双特异性抗体,其包括结合APC的激活受体的第一结构域或其抗原结合片段,和结合免疫效应细胞的共刺激受体抗体或其抗原结合片段的第二结构域。在一些实施方案中,第一结构域包括结合CD40、CD80、CD86、CD91、DEC-205或DC-SIGN的抗体或其抗原结合片段。在一些实施方案中,第二结构域包括结合CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的抗体或其抗原结合片段。
在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗CD28抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗4-1BB抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗ICOS抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗CD27抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗OX40抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗DAP10抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗2B4抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗CD30抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括 第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗CD2抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗LIGHT抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗GITR抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗TLR抗体或其抗原结合片段。在一些实施方案中,本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗DR3抗体或其抗原结合片段。在一些实施方案中本发明提供了包括第一结构域和第二结构域的双特异性抗体,所述第一结构域为抗CD40抗体或其抗原结合片段,所述第二结构域包括抗CD43抗体或其抗原结合片段。
制备双特异性抗体的方法为本领域已知。例如,可以利用两个免疫球蛋白重链/轻链对的共表达重组生产双特异性抗体。参见,例如Milstein等,(1983)Nature 305:537-39。或者,可以使用化学连接来制备双特异性抗体。参见,例如Brennan等,(1985)Science 229:81。双特异性抗体包括双特异性抗原结合片段。参见,例如Holliger等,(1993)Proc.Natl.Acad.Sci.U.S.A.90:6444-48;Gruber et al.(1994)J.Immunol.152:5368。制备双特异性抗体的技术包括但不限于重组共表达两个具有不同特异性的免疫球蛋白重链-轻链对(参见Milstein和Cuello,Nature 305:537(1983),WO 93/08829,和Traunecker等,EMBO J.10:3655(1991)),和“knob-in-hole”工程(参见例如,美国专利5,731,168)。多特异性抗体也可以通过以下方式制备:制备抗体Fc-异源二聚化分子的工程化静电转向效应(WO2009/089004A1);交联两个或多个抗体或片段(参见,例如,美国专利4,676,980,和Brennan等,Science 229:81(1985));用亮氨酸拉链产生双特异性抗体(参见,例如Kostelny等,J.Immunol.148(5):1547-1553(1992));使用“diabody”技术制备双特异性抗体片段(参见,例如Hollinger等,Proc.Natl.Acad.Sci.USA,90:6444-6448(1993));和使用单链Fv(scFv)二聚体(参见例如,Gruber等,J.Immunol.,152:5368(1994));以及如,例如在Tutt et al.J.Immunol.147:60(1991)中所述制备双特异性抗体。含有三个或更多个功能抗原结合位点的工程抗体,包括“章鱼抗体”(Octopus antibodies),也包括于此(参见,例如,US2006/0025576A1)。双特异性 抗体也可以通过连接两个不同的抗体或其部分来构建。例如,双特异性抗体可以包括来自两种不同的抗体的Fab、F(ab')2、Fab'、scFv和sdAb。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40抗体或其抗原结合片段。所述抗CD40抗体或抗原结合片段可以是本发明公开的或本领域其他已知的,可以激活CD40信号转导的任何抗CD40抗体或抗原结合片段。在一些实施方案中,所述抗CD40抗体或其抗原结合片段包括如上表1中所提供的标记为F2.103、F5.157、F5.77、4D11、A40C或119的抗体。在一些实施方案中,抗CD40抗体或其抗原结合片段包括具有SEQ ID NO:227至SEQ ID NO:232中任一项所示氨基酸序列的抗CD40 scFv。
在一些实施方案中,抗CD28抗体或抗原结合片段可以是本发明公开的或本领域其他已知的激活CD28信号传导的任何抗CD28抗体或抗原结合片段。在一些实施方案中,抗CD28抗体或抗原结合片段是标记为1412的抗体。在一些实施方案中,抗CD28抗体或其抗原结合片段包括具有SEQ ID NO:233所示氨基酸序列的抗CD28 scFv。

在一些实施方案中,本发明提供的融合蛋白具有的氨基酸序列与标记为1412-F2.103,1412-F5.157,1412-F5.77,或1412-4D11的融合蛋白的序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少 92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%同一性。
如本领域普通技术人员所理解,本发明所示例的融合蛋白中的抗CD40抗体或其抗原结合片段可被本发明公开的或本领域其他已知的结合不同APC激活受体的抗体或抗原结合片段替换,包括例如CD80、CD86、CD91、DEC-205或DC-SIGN。如本领域普通技术人员所理解,本发明所示例的融合蛋白中的抗CD28抗体或抗原结合片段可被与本发明所公开的或本领域其他已知的结合免疫效应细胞的不同共刺激因子的抗体或抗原结合片段所取代,包括例如与4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43结合的抗体或抗原结合片段。
示例性LACO-Stim(4):激活受体的抗体+共刺激受体(例如,aCD40-CD28;aCD40-4- 1BB)
在一些实施方案中,本发明所提供的融合蛋白包括激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合APC的激活受体的抗体或其抗原结合片段,并且其中所述第二结构域包括免疫效应细胞的共刺激受体或其功能片段。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中,第一结构域的N-末端连接到第二结构域的C-末端。在一些实施方案中,本发明提供了基于抗体的膜融合蛋白。
在一些实施方案中,第一和第二结构域通过CD8铰链、CD28铰链或IgG Fc区连接。
在一些实施方案中,本发明所提供的融合蛋白包括第一结构域和第二结构域,其中所述第一结构域包括结合APC的激活受体的抗体或其抗原结合片段,所述第二结构域包括结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD40、CD80、CD86、CD91、DEC-205或DC-SIGN的抗体或其抗原结合片段。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40抗体或其抗原结合片段。所述抗CD40抗体或抗原结合片段可以是本发明公开的或本领域其他已知的激活CD40信号传导的任何抗CD40抗体或抗原结合片段。在一些实施方案中,所述抗CD40抗体或其抗原结合片段包括如上表1中提供的标记为F2.103、F5.157、F5.77、4D11、A40C或119的抗体。
在一些实施方案中,所述第二结构域包括共刺激受体或其功能片段,所述共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、 GITR、TLR、DR3和CD43组成的群组。在一些实施方案中,所述第二结构域包括共刺激受体的胞质结构域,所述共刺激受体选自由CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43组成的群组。
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括CD28胞质结构域。在一些实施方案中,本发明提供的融合蛋白的第二结构域可以具有与CD28胞质结构域所示序列有至少85%、至少88%、至少90%、至少95%、至少98%或100%同一性的氨基酸序列。在一些实施方案中,本发明提供的融合蛋白的第二结构域进一步包括CD28跨膜结构域。
在一些实施方案中,本发明提供的融合蛋白的第二结构域包括4-1BB胞质结构域。在一些实施方案中,文提供的融合蛋白的第二结构域可以具有与4-1BB胞质结构域有至少85%、至少88%、至少90%、至少95%、至少98%或100%同一性的氨基酸序列。在一些实施方案中,本发明提供的融合蛋白的第二结构域进一步包括4-1BB跨膜结构域。
在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括CD28胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括4-1BB胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括ICOS胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括CD27胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括OX40胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括DAP10胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括2B4胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括CD30胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体 或其抗原结合片段,所述第二结构域包括CD2胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括LIGHT胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括GITR胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括TLR胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括DR3胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括CD43胞质结构域。在一些实施方案中,所述第一结构域包括全长CD40L。
在一些实施方案中,本发明提供的融合蛋白进一步包括跨膜区。在一些实施方案中,所述跨膜区来源于相同的共刺激受体。在一些实施方案中,所述跨膜区来源于不同的共刺激受体。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括CD28跨膜区和CD28胞质结构域。在一些实施方案中,本发明提供的融合蛋白具有第一结构域和第二结构域,所述第一结构域包括抗CD40抗体或其抗原结合片段,所述第二结构域包括4-1BB跨膜区和4-1BB胞质结构域。


在一些实施方案中,本发明提供的融合蛋白具有的氨基酸序列与标记为F2.103.CD28,F5.157.CD28,F5.77.CD28,,F5.157.BB,F5.77.BB,4D11.CD28,A40C.CD28或119.CD28的融合蛋白的序列具有至少80%、至少85%、至少86%、至少87%、至少88%、至少89%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%或至少99%同一性。
如本领域普通技术人员所理解的,本发明所示例的融合蛋白中的抗CD40抗体或其抗原结合片段可以被本发明公开的或本领域其他已知的结合和激活APC不同激活受体的抗体或抗原结合片段所替代,包括例如CD80、CD86、CD91、DEC-205或DC-SIGN。如本领域普通技术人员所理解的,本发明所示例的融合蛋白中的CD28胞质结构域或4-1BB胞质结构域可以用本发明公开的或本领域其他已知的免疫效应细胞的不同共刺激因子的胞质结构域替换,包括例如ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的胞质结构域;或4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3或CD43的不同功能片段,所述功能片段保留全长蛋白的激活免疫效应细胞的功能。
示例性LACO-Stim(5):APC激活受体的抗体+共刺激受体的配体(例如,aCD40-CD80; aCD40-CD86)
在一些实施方案中,本发明所提供的融合蛋白包括激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合抗原提呈细胞的激活受体的抗体或其抗原结合片段,并且其中所述第二结构域包括免疫效应细胞的共刺激配体或其受体结合片段。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中,第一结构域的N-末端连接到第二结构域的C-末端。在一些实施方案中,本发明提供了基于抗体的可溶性融合蛋白。在一些实施方案中,本发明提供了基于抗体的可溶性融合蛋白。
在一些实施方案中,所述第一结构域包括结合CD40、CD80、CD86、CD91、DEC-205和DC-SIGN的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD40的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD80的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD86的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合CD91的抗体或其抗原结合片段。在一些实施方案中,所述第一结构域包括结合DEC-205的抗体或其抗原结 合片段。在一些实施方案中,所述第一结构域包括结合DC-SIGN的抗体或其抗原结合片段。所述抗体和抗原结合片段可以为本发明公开的或本领域其他已知的任何抗体或抗原结合片段。
在一些实施方案中,本发明所提供得融合蛋白包括第一结构域和第二结构域,其中所述第一结构域包括结合APC激活受体的抗体或其抗原结合片段,所述第二结构域包括免疫效应细胞的共刺激配体或其受体结合片段。在一些实施方案中,第一结构域包括结合CD40、CD80、CD86、CD91、DEC-205或DC-SIGN的抗体或其抗原结合片段。在一些实施方案中,第二结构域包括选自由CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44组成得群组的配体,或其受体结合片段。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括抗CD40抗体或其抗原结合片段。所述抗CD40抗体或抗原结合片段可以是本发明公开的或本领域其他已知的激活CD40信号传导的任何抗CD40抗体或抗原结合片段。在一些实施方案中,抗CD40抗体或其抗原结合片段包括如上表1中提供的标记为为F2.103、F5.157、F5.77、4D11、A40C或119的抗体。在一些实施方案中,所述抗CD40抗体或其抗原结合片段包括具有SEQ ID NO:227至SEQ ID NO:232中任一项所示氨基酸序列的抗CD40scFv。
在一些实施方案中,所述融合蛋白包括第一结构域和第二结构域,所述第一结构域包括结合CD40的抗体或抗原结合片段,所述第二结构域包括配体及其受体结合片段,所述配体选自由CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R、CD44组成的群组。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD58或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD70或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD83或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD80或其受体结合片段。在一些 实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD86或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD137L或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD252或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD275或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD54或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD49a或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD112或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或其抗原结合片段,第二结构域包括CD150或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD155或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD265或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD270或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域,其包括结合CD40的抗体或抗原结合片段,第二结构域包括TL1A或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD127或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括IL-4R或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域,其包括结合CD40的抗体或抗原结合片段,第二结构域包括GITR-L或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括TIM-4或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结 构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD153或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD48或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD160或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或抗原结合片段,第二结构域包括CD200R或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40的抗体或其抗原结合片段,第二结构域包括CD44或其受体结合片段。本领域普通技术人员可以容易地确定配体合适的受体结合片段,所述受体结合片段保持其对受体的结合亲和力并具有激活受体的功能。
如本领域普通技术人员所理解的,本发明所示例的融合蛋白中的抗CD40抗体或其抗原结合片段可被本发明公开的或本领域其他已知的结合APC的不同激活受体的抗体或抗原结合片段所替换,包括例如CD80、CD86、CD91、DEC-205或DC-SIGN。
示例性LACO-Stim(6):APC激活受体的配体+共刺激配体(例如,CD40L-CD86; CD40L-CD80)
在一些实施方案中,本发明提供的融合蛋白包括激活APC的第一结构域和激活免疫效应细胞的第二结构域,其中所述第一结构域包括结合APC的激活受体的配体或其受体结合片段,并且其中所述第二结构域包括免疫效应细胞的共刺激配体或其受体结合片段。在一些实施方案中,第一结构域的C-末端连接到第二结构域的N-末端。在一些实施方案中,第一结构域的N-末端连接到第二结构域的C-末端。
在一些实施方案中,本发明提供的融合蛋白包括第一结构域和第二结构域,所述第一结构域包括结合激活受体的配体或其功能片段,所述激活受体选自由CD40、CD80、CD86、CD91、DEC-205和DC-SIGN组成的群组,所述第二结构域包括共刺激配体或其受体结合片段,所述共刺激配体选自由CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R、CD44组成的群组。
在一些实施方案中,本发明提供的融合蛋白的第一结构域包括CD40L的胞外结构域。在一些实施方案中,本发明提供的融合蛋白的第一结构域可以具有与CD40L的胞外结构域有至少85%、至少88%、至少90%、至少95%、至少98%或100%同一性的氨基酸序 列。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括全长CD40L。在一些实施方案中,本发明提供的融合蛋白的第一结构域可以具有CD40L(SEQ ID NO:223)的119-261位氨基酸。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括三个拷贝的CD40L或其功能片段。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括三个拷贝的CD40L胞外结构域。在一些实施方案中,本发明提供的融合蛋白的第一结构域包括三个拷贝的CD40L(SEQ ID NO:223)的119-261位氨基酸。
在一些实施方案中,所述融合蛋白包括第一结构域和第二结构域,所述第一结构域包括CD40L或其受体结合片段,所述第二结构域包括配体或其受体结合片段,所述配体选自由CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R、CD44组成的群组。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD58或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD70或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD83或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD80或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD86或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD137L或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD252或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD275或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD54或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD49a或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结 构域包括CD112或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40L或其受体结合片段,第二结构域包括CD150或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD155或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD265或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD270或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括TL1A或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD127或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括IL-4R或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括GITR-L或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括TIM-4或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD153或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD48或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD160或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括CD40L或其受体结合片段,第二结构域包括CD200R或其受体结合片段。在一些实施方案中,融合蛋白包括第一结构域和第二结构域,第一结构域包括结合CD40L或其受体结合片段,第二结构域包括CD44或其受体结合片段。本领域普通技术人员可以容易地确定配体合适的受体结合片段,所述受体结合片段保持其对受体的结合亲和力并具有激活受体的功能。
如本领域普通技术人员所理解,本发明所示例的融合蛋白中的CD40L或其受体结合片段可以用本发明公开的或本领域已其他已知的APC激活受体的不同配体替换,包括例 如CD80配体(例如,CD28或CTLA-4)、CD86配体(例如,CD28或CTLA-4)、CD91配体(例如,RAP1)、DEC-205配体或DC-SIGN配体(例如,ICAM2、ICAM3、CD18或CEACAM1)的胞外结构域或全长配体。
抗CD7 CAR
嵌合抗原受体的抗原特异性胞外结构域识别并特异性地结合抗原,通常是恶性肿瘤的表面表达的抗原(如CD7)。例如,当抗原特异性胞外结构域以约0.1pM至约10μM、或约0.1pM至约1μM、或约0.1pM至约100nM的亲和力常数或相互作用亲和力(KD)结合抗原时,所述抗原特异性胞外结构域特异性地结合抗原。用于确定相互作用亲和力的方法在本领域中是已知的。适用于本公开的CAR的抗原特异性胞外结构域可以是任何抗原结合多肽,各种各样的抗原结合多肽在本领域中是已知的。在一些情况下,所述抗原结合结构域是单链Fv(scFv)。其他基于抗体的识别结构域(cAb VHH(骆驼抗体可变结构域)及其人源化形式、IgNAR VH(鲨鱼抗体可变结构域)及其人源化形式、sdAb VH(单结构域抗体可变结构域)和“骆驼化”抗体可变结构域适合使用。在一些情况下,基于T细胞受体(TCR)的识别结构域诸如单链TCR(scTv)也适合使用。
合适的抗原可包括T细胞特异性抗原和/或对于T细胞不是特异性的抗原。在一些的实施方案中,由CAR-T细胞的嵌合抗原受体特异性地结合的抗原和CAR-T细胞缺乏的抗原是在恶性T细胞上表达的抗原,更优选地是与非恶性T细胞相比在恶性T细胞上过表达的抗原。“恶性T细胞”是源自T细胞恶性肿瘤的T细胞。术语“T细胞恶性肿瘤”是指广泛的高度异质分组的源自T细胞前体、成熟T细胞或自然杀伤细胞的恶性肿瘤。T细胞恶性肿瘤的非限制性实例包括T细胞急性成淋巴细胞性白血病/淋巴瘤(T-ALL)、T细胞大颗粒淋巴细胞(LGL)白血病、人T细胞白血病病毒1型阳性(HTLV-1+)成人T细胞白血病/淋巴瘤(ATL)、T细胞幼淋巴细胞白血病(T-PLL)以及各种外周T细胞淋巴瘤(PTCL),包括但不限于血管免疫母细胞性T细胞淋巴瘤(AITL)、ALK阳性间变性大细胞淋巴瘤和ALK阴性间变性大细胞淋巴瘤。
在一些实施方案中,本申请的CAR-T细胞包含特异性地结合到CD7的嵌合抗原受体的胞外结构域。CD7是T细胞表面膜相关糖蛋白。CD7可在T细胞恶性肿瘤包括T细胞急性成淋巴细胞性白血病(T-ALL)和非霍奇金氏T细胞淋巴瘤(NHL)中过表达。本公开的CAR-T细胞可用于靶向过表达CD7的恶性T细胞。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结 合结构域包含重链可变区(VH)和轻链可变区(VL),其中所述VH包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
在某些实施方式中,其中所述VL包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
在某些实施方式中,其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所示的氨基酸序列;或
所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或
所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或
所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或
所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或
所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列;或
所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或
所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或
所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或
所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或
所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或
所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或
所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所 示的氨基酸序列。
在某些实施方式中,所述的细胞外抗原结合结构域包括scFv。
例如,所述细胞外抗原结合结构域可以是抗CD7 scFv。
在某些实施方式中,其中所述VL和VH通过接头连接。
在某些实施方式中,其中所述接头包括多肽接头。
在某些实施方式中,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
例如,本申请的嵌合抗原受体的抗原特异性胞外结构域可以特异性结合CD7,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列或与SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
本申请的嵌合抗原受体还包含胞内结构域,在抗原结合到抗原特异性胞外结构域之后,所述胞内结构域向T细胞提供胞内信号。合适的胞内结构域的非限制性实例包括T细胞受体的ζ链或其同系物中的任一种(例如,η、δ、γ或ε)、MB 1链、B29、Fc RIII、Fc RI以及信号传导分子(诸如CD3.ζ.和CD28、CD27、4-1BB、DAP-10、OX40和它们的组合以及其他相似的分子和片段)的组合。可使用激活蛋白家族的其他成员的胞内信号传导部分,诸如Fc.γ.RIII和Fc.ε.RI。虽然通常采用整个胞内结构域,但是在许多情况下,不需要使用整个胞内多肽。在胞内信号传导结构域的截短部分可使用的程度下,可使用此截短部分代替完整的链,只要所述截短部分仍然转导效应子功能信号即可。因此,本申请的胞内结构域旨在包括胞内结构域的足以转导效应子功能信号的任何截短部分。
通常,抗原特异性胞外结构域通过跨膜结构域连接到嵌合抗原受体的胞内结构域。跨膜结构域横贯细胞膜,将CAR锚固到T细胞表面,并且将胞外结构域连接到胞内信号传导结构域,从而影响CAR在T细胞表面上的表达。嵌合抗原受体还可进一步包含一个或多个共刺激结构域和/或一个或多个间隔子。共刺激结构域来源于共刺激蛋白的增强体内细胞因子产生、增殖、细胞毒性和/或持续性的胞内信号传导结构域。间隔子(i)将抗原特异性胞外结构域连接到跨膜结构域,(ii)将跨膜结构域连接到共刺激结构域,(iii)将共刺激结构域连接到胞内结构域,和/或(iv)将跨膜结构域连接到胞内结构域。例如,在抗原特异性胞外结构域与跨膜结构域之间包含间隔子结构域可影响抗原结合结构域的柔性并且 因此影响CAR功能。合适的跨膜结构域、共刺激结构域和间隔子在本领域中是已知的。
在某些实施方式中,所述的嵌合抗原受体包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
例如,其中所述跨膜域可以包含源自CD8的跨膜域。
又例如,其中所述跨膜域包含SEQ ID NO:177所示的氨基酸序列或与SEQ ID NO:177所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
例如,其中所述胞内共刺激信号传导结构域可以包含源自4-1BB的共刺激信号传导结构域。
又例如,其中所述胞内共刺激信号传导结构域可以包含SEQ ID NO:178所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14 Nef、DAP10、DAP-12和至少包含一个ITAM的结构域。
例如,其中所述胞内信号转导结构域可以包含源自CD3ζ的信号传导结构域。
又例如,其中所述胞内信号转导结构域可以包含SEQ ID NO:179所示的氨基酸序列。
在某些实施方式中,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰 链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
例如,所述铰链区可以包含源自CD8的铰链区。
例如,所述铰链区可以包含SEQ ID NO:176所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
例如,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
在某些实施方式中,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
例如,所述信号肽片段可以包含CD8信号肽片段。
例如,所述信号肽片段可以包含SEQ ID NO:175所示的氨基酸序列。
又例如,本申请的嵌合抗原受体可以包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列或与SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
工程化的免疫效应细胞
在本申请中,免疫效应细胞包括工程化的免疫效应细胞。术语“工程化免疫细胞”通常是指通过将DNA或RNA形式的额外遗传物质加入细胞的总遗传物质而被基因修饰的免疫细胞,也称为免疫效应细胞。例如,工程化免疫细胞已经过基因修饰以表达根据本发明的靶向CD7的CAR。所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
在某些实施方式中,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。例如,其中所述免疫效应细胞可以包括同种异体T细胞或自体T细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
在某些实施方式中,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7 表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中所述CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
在某些实施方式中,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
在某些实施方式中,所述CAR-T细胞缺乏嵌合抗原受体特异性地结合的抗原并且因此不诱导自相残杀。
在一些实施方案中,T细胞的抗原被修饰,使得嵌合抗原受体不再特异性地结合修饰的抗原(如CD7)。例如,被嵌合抗原受体识别的抗原的表位可通过一个或多个氨基酸变化(例如,取代或缺失)来修饰,或者所述表位可从抗原缺失。在其他实施方案中,抗原的表达在T细胞中减少至少50%、至少60%、至少70%、至少80%、至少90%或更多。用于降低蛋白质的表达的方法在本领域中是已知的,并且包括但不限于修饰或替换可操作地连接到编码蛋白质的核酸序列的启动子。在其他的实施方案中,T细胞被修饰,使得例如通过编码抗原的基因的缺失或破坏而不表达抗原。将T细胞遗传修饰成缺乏抗原的方法在本领域中是熟知的,并且非限制性实例在以上提供。在一个示例性实施方案中,CRISPR/cas9基因编辑可用于将T细胞修饰成缺乏抗原。
用于T细胞中的CAR设计、递送和表达的方法以及临床级CAR-T细胞群体的制造在本领域中是已知的。参见例如,Lee等人,Clin.Cancer Res.,2012,18(10):2780-90。例如,工程化的CAR可使用逆转录酶病毒引入到T细胞中,这将编码嵌合抗原受体的核酸序列有效并稳定地整合到靶细胞基因组中。又例如,利用编码CAR的mRNA转入T细胞中。本领域中已知的其他方法包括但不限于慢病毒转导、基于转座子的系统、直接RNA转染和CRISPR/Cas系统。
CAR-T细胞可由本领域已知的任何合适的T细胞来源生成,所述T细胞包括但不限于从受试者采集的T细胞。受试者可以是需要CAR-T细胞疗法的患有T细胞恶性肿瘤的患者或与需要CAR-T细胞疗法的患有T细胞恶性肿瘤的受试者相同种类的受试者。采集的T细胞可使用本领域中通常已知的方法进行离体扩展,之后用CAR转导以生成CAR-T细胞。
另一方面,本申请提供一种包含本申请所述的经修饰的免疫效应细胞的细胞群,其 中所述细胞群来源于外周血单核细胞(PBMC)、外周血血液白细胞(PBL)、肿瘤浸润淋巴细胞(TIL)、细胞因子诱导的杀伤细胞(CIK)、淋巴因子激活的杀伤细胞(LAK)或骨髓浸润淋巴细胞(MIL)。
药物组合
另一方面,本申请提供一种药物组合,其包含免疫效应细胞和融合蛋白,其中所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,或(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。
在一些实施方案中,所述融合蛋白为可溶性蛋白。在一些实施方案中,融合蛋白为双特异性抗体。
例如,融合蛋白的第一结构域和第二结构域可以选自以下组合:
APC激活受体配体+结合共刺激受体的抗体(例如,aCD28-CD40L);
APC激活受体的抗体+共刺激受体的抗体(例如,aCD40/aCD28双特异性Ab);
APC激活受体的抗体+共刺激配体(例如,aCD40-CD80;aCD40-CD86);和
APC激活受体的配体+共刺激配体(例如,CD40L-CD86;CD40L-CD80)。
方法和用途
另一方,本申请提供本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物在制备药物中的用途,所述药物用于治疗肿瘤。
在某些实施方式中,其中所述肿瘤包括血液瘤和实体瘤。
在某些实施方式中,其中所述肿瘤包括表达CD7的肿瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述肿瘤包括T细胞恶性肿瘤。
在某些实施方式中,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物。
CAR-T细胞可通过静脉内途径(例如,通过静脉内输注)施用给受试者。CAR-T细胞 可以单剂量或以多剂量施用。CAR-T细胞可在适用于静脉内施用的药物组合物中注射。用于IV施用的合适的药物组合物在本领域中是已知的。本公开的药物组合物还可包含另外的组分。例如,此类组分可用于维持注射的CAR-T细胞的活力和/或活性。
CAR-T细胞可以有效剂量施用。有效剂量可以是一个剂量或多个剂量,并且足以产生所需的治疗效果。CAR-T细胞的典型剂量的范围可以是约1×105至5×107个细胞/Kg接受疗法的受试者的体重。有效剂量可基于恶性肿瘤的阶段、受试者的健康状况和恶性肿瘤的类型来计算。在施用多个剂量的情况下,此剂量和剂量之间的间隔可基于受试者对于疗法的反应来确定。
另一方面,本申请提供一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与本申请所述的经修饰的免疫效应细胞、本申请所述的药物组合或本申请所述的药物组合物接触。
另一方面,本申请提供一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
a.对T细胞群中的CD7基因进行修饰;
b.激活所述T细胞群;
c.将抗CD7 CAR和融合蛋白转导所述T细胞群;以及
d.扩增所述嵌合抗原受体T细胞群。
用于T细胞中的CAR设计、递送和表达的方法以及临床级CAR-T细胞群体的制造在本领域中是已知的。参见例如,Lee等人,Clin.Cancer Res.,2012,18(10):2780-90。例如,工程化的CAR可使用逆转录酶病毒引入到T细胞中,这将编码嵌合抗原受体的核酸序列有效并稳定地整合到靶细胞基因组中。又例如,利用编码CAR的mRNA转入T细胞中。本领域中已知的其他方法包括但不限于慢病毒转导、基于转座子的系统、直接RNA转染和CRISPR/Cas系统。
CAR-T细胞可由本领域已知的任何合适的T细胞来源生成,所述T细胞包括但不限于从受试者采集的T细胞。受试者可以是需要CAR-T细胞疗法的患有T细胞恶性肿瘤的患者或与需要CAR-T细胞疗法的患有T细胞恶性肿瘤的受试者相同种类的受试者。采集的T细胞可使用本领域中通常已知的方法进行离体扩展,之后用CAR转导以生成CAR-T细胞。
在一个示例性实施方案中,本公开提供一种T细胞,其包含特异性地结合CD7的嵌合抗原受体,其中所述T细胞缺乏CD7(例如,CD7 KO CAR-T细胞)。在非限制性实 例中,缺乏CD7由以下产生:(a)修饰由T细胞表达的CD7,使得嵌合抗原受体不再特异性地结合修饰的CD7,(b)修饰T细胞,使得抗原的表达在T细胞中减少至少50%、至少60%、至少70%、至少80%、至少90%或更多,或者(c)修饰T细胞,使得CD7不被表达(例如,通过编码CD7的基因的缺失或破坏)。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
在某些实施方式中,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
在某些实施方式中,其中所述靶向所述CD7基因的gRNA包含SEQ ID NO:211至SEQ ID NO:218中任一项所示的核苷酸序列。
在某些实施方式中,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
在某些实施方式中,其中所述gRNA与所述Cas9同时递送。
在某些实施方式中,其中所述递送是通过电穿孔进行的。
在某些实施方式中,其中所述将抗CD7 CAR和融合蛋白转导所述T细胞包括将编码所述抗CD7 CAR的核酸分子和编码所述融合蛋白的核酸分子引入T细胞。
抗原结合蛋白
一方面,提供了一种分离的抗原结合蛋白,所述分离的抗原结合蛋白与参比抗体竞争结合CD7,其中所述参比抗体包含轻链可变区VL和重链可变区VH;
其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H1抗体;或
所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H4抗体;或
所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H6抗体;或
所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H8抗体;或
所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所 示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H9抗体;或
所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H10抗体;或
所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H12抗体;或
所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H13抗体;或
所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H17抗体;或
所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H18抗体;或
所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H5抗体;或
所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H7抗体;或
所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列,该参比抗体命名为抗CD7 scFv-H15抗体。
在某些实施方式中,所述分离的抗原结合蛋白包含重链可变区(VH),所述VH包含HCDR1、HCDR2和HCDR3,其中HCDR1包SEQ ID NO:1所示的氨基酸序列,SEQ ID NO:1序列通式如G[FGY][ST][FILV][ST][EGST][LNY]所示,其中[]中的氨基酸为任选的一个氨基酸。
例如,其中所述HCDR1可以包含SEQ ID NO:2至SEQ ID NO:9中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HCDR2包含SEQ ID NO:10至SEQ ID NO:17中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HCDR3包含SEQ ID NO:18或SEQ ID NO:27所示的氨基酸序列。
又例如,,所述分离的抗原结合蛋白包含重链可变区(VH),所述VH可以包含HCDR1、HCDR2和HCDR3,,其中所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所 述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:18所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:19所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:20所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:21所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:22所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:23所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:24所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:25所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:26所示的氨基酸序列;或
所述HCDR1可以包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2可以包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3可以包含SEQ ID NO:27所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH,其中所述VH包括框架 区HFR1,HFR2、HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,且所述HFR1包含SEQ ID NO:28至SEQ ID NO:38中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR2位于所述HCDR1与所述HCDR2之间,且所述HFR2包含SEQ ID NO:39至SEQ ID NO:47中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR3位于所述HCDR2与所述HCDR3之间,且所述HFR3包含SEQ ID NO:48至SEQ ID NO:56中任一项所示的氨基酸序列。
在某些实施方式中,其中所述HFR4的N末端与所述HCDR3的C末端直接或间接相连,且所述HFR4包含SEQ ID NO:57至SEQ ID NO:60中任一项所示的氨基酸序列。
例如,所述的分离的抗原结合蛋白其包含VH,其中所述VH可以包括框架区HFR1,HFR2,HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,所述HFR2位于所述HCDR1与所述HCDR2之间,所述HFR3位于所述HCDR2与所述HCDR3之间,所述HFR4的N末端与所述HCDR3的C末端直接或间接相连;其中,所述HFR1可以包含SEQ ID NO:28所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:39所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:48所示的氨基酸序列,HFR4可以包含SEQ ID NO:57所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:29所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:40所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:49所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:30所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:39所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:48所示的氨基酸序列,HFR4可以包含SEQ ID NO:58示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:31所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:41所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:50所示的氨基酸序列,HFR4可以包含SEQ ID NO:59所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:32所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:42所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:51所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:33所示的氨基酸序列,所述HFR2可以包含SEQ  ID NO:39所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:48所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:34所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:39所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:48所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:35所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:43所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:52所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:28所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:39所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:48所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:30所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:44所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:53所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:36所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:45所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:54所示的氨基酸序列,HFR4可以包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:37所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:46所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:55所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1可以包含SEQ ID NO:38所示的氨基酸序列,所述HFR2可以包含SEQ ID NO:47所示的氨基酸序列,所述HFR3可以包含SEQ ID NO:56所示的氨基酸序列,HFR4可以包含SEQ ID NO:58所示的氨基酸序列。
又例如,所述的分离的抗原结合蛋白其包含VH,其中所述VH可以包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR1包含SEQ ID NO:74至SEQ ID NO:86中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含 LCDR1、LCDR2、LCDR3,所述LCDR2包含SEQ ID NO:87至SEQ ID NO:98中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR3包含SEQ ID NO:99至SEQ ID NO:101中任一项所示的氨基酸序列。
例如,所述的分离的抗原结合蛋白其包含VL,其中所述VL可以包含LCDR1,LCDR2和LCDR3,所述LCDR1可以包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:99所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:100所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:101所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:102所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:78所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:103所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:104所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:105所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:106所示的氨基 酸序列;或
所述LCDR1可以包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:107所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:109所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:110所示的氨基酸序列;或
所述LCDR1可以包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2可以包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3可以包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VH和VL,其中所述VH包含HCDR1,HCDR2和HCDR3,所述VL包含LCDR1,LCDR2和LCDR3;其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1 包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸 序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,且所述LFR1包含SEQ ID NO:112至SEQ ID NO:123中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR2,其中所述LFR2位于所述LCDR1与所述LCDR2之间,且所述LFR2包含SEQ ID NO:124至SEQ ID NO:130中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR3,所述LFR3位于所述LCDR2与所述LCDR3之间,且所述LFR3包含SEQ ID NO:131至SEQ ID NO:141中任一项所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白其包含VL,其中所述VL包括框架区LFR4,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连,且所述LFR4包含SEQ ID NO:142至SEQ ID NO:147中任一项所示的氨基酸序列。
例如,所述的分离的抗原结合蛋白其包含VL,其中所述VL可以包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,所述LFR2位于所述LCDR1与所述LCDR2之间,所述LFR3位于所述LCDR2与所述LCDR3之间,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连;其中所述LFR1可以包含SEQ ID NO:112所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:124所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:131所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:113所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:125所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:132所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:114所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:126所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:133所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:115所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:127所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:134所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:116所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:125所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:135所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:117所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:126所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:136所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:118所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:128所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:137所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:119所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:125所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:138所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:120所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:129所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:139所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:114所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:126所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:140所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:144所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:121所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:130所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:141所示的氨基酸序列, 所述LFR4可以包含SEQ ID NO:145所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:122所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:130所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:141所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:146所示的氨基酸序列;或
所述LFR1可以包含SEQ ID NO:123所示的氨基酸序列,所述LFR2可以包含SEQ ID NO:130所示的氨基酸序列,所述LFR3可以包含SEQ ID NO:141所示的氨基酸序列,所述LFR4可以包含SEQ ID NO:147所示的氨基酸序列。
例如,所述的分离的抗原结合蛋白其包含VL,其中所述VL可以包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
又例如,所述的分离的抗原结合蛋白其包含VH和VL,其中所述VH可以包含SEQ ID NO:61所示的氨基酸序列,所述VL可以包含SEQ ID NO:148所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:62所示的氨基酸序列,所述VL可以包含SEQ ID NO:149所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:63所示的氨基酸序列,所述VL可以包含SEQ ID NO:150所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:64所示的氨基酸序列,所述VL可以包含SEQ ID NO:151所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:65所示的氨基酸序列,所述VL可以包含SEQ ID NO:152所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:66所示的氨基酸序列,所述VL可以包含SEQ ID NO:153所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:67所示的氨基酸序列,所述VL可以包含SEQ ID NO:154所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:68所示的氨基酸序列,所述VL可以包含SEQ ID NO:155所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:69所示的氨基酸序列,所述VL可以包含SEQ ID NO:156所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:70所示的氨基酸序列,所述VL可以包含SEQ ID NO: 157所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:71所示的氨基酸序列,所述VL可以包含SEQ ID NO:158所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:72所示的氨基酸序列,所述VL可以包含SEQ ID NO:159所示的氨基酸序列;或
所述VH可以包含SEQ ID NO:73所示的氨基酸序列,所述VL可以包含SEQ ID NO:160所示的氨基酸序列。
在某些实施方式中,所述的分离的抗原结合蛋白包括抗体或其抗原结合片段。
在某些实施方式中,所述抗体包括单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体、完整抗体、抗体片段、人抗体、人源化抗体或嵌合抗体。
在某些实施方式中,其中所述抗原结合片段包括Fab,Fab’,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb。
在某些实施方式中,所述的抗原结合蛋白包括scFv。
例如,所述抗原结合蛋白可以是抗CD7 scFv抗体。
在某些实施方式中,其中所述VL和VH通过接头连接。
在某些实施方式中,其中所述接头包括多肽接头。
在某些实施方式中,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
例如,所述抗原结合蛋白包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
免疫缀合物
另一方面,本申请提供一种免疫缀合物,其包含本申请所述的抗原结合蛋白。
在本申请中,术语“免疫缀合物”或“抗体缀合物”通常是指抗体或其抗体片段与其它活性剂的连接,诸如化疗剂,毒素,免疫治疗剂,成像探针,光谱探针,等等。所述连接可以是共价键,或例如通过静电力的非共价相互作用。可以使用本领域中已知的多种接头以形成免疫缀合物。此外,该免疫缀合物可以以融合蛋白的形式提供,所述融合蛋白可以从编码该免疫缀合物的多核苷酸表达。如本文所用的“融合蛋白”指的是通过连接两个或多个最初编码独立的蛋白(包括肽和多肽)的基因或基因片段产生的蛋白。融合基因的翻译产生具有来自各原始蛋白的功能特性的单一蛋白。
在某些实施方式中,所述免疫缀合物包含:
(a)本申请所述的抗原结合蛋白;
(b)选自下组的缀合部分:可检测标记物、药物、毒素、细胞因子、病毒外壳蛋白或VLP、或其组合。
细胞毒素或细胞毒性剂包括对细胞有害(例如,杀死)的任何药剂。实例包括紫杉醇、细胞松弛素B、短杆菌肽D、溴化乙锭、依米丁、丝裂霉素、依托泊苷、替尼泊苷、长春新碱、长春碱、秋水仙碱、多柔比星、柔红霉素、二羟基蒽二酮、米托蒽醌、普卡霉素、放线菌素D、1-脱氢睾酮、糖皮质激素、普鲁卡因、丁卡因、利多卡因、普萘洛尔和嘌呤霉素及其类似物或同系物。治疗剂包括但不限于,抗代谢物(例如,甲氨蝶呤、6-巯嘌呤、6-硫鸟嘌呤、阿糖胞苷、5-氟尿嘧啶氨烯咪胺)、烷化剂(例如,氮芥、噻替派苯丁酸氮芥、美法仑、卡莫司汀(BSNU)和洛莫司汀(CCNU)、环磷酰胺、白消安、二溴甘露醇、链脲菌素、丝裂霉素C和顺-二氯二胺铂(II)(DDP)顺铂)、蒽环霉素(例如,柔红霉素(以前称为红比霉素)和多柔比星)、抗生素(例如,放线菌素(原放线菌素))、博来霉素、普卡霉素和氨茴霉素(AMC)),和抗有丝分裂剂(例如,长春新碱和长春碱)。用于本申请的抗体可以与放射性同位素(例如,放射性碘)缀合,以生成用于治疗癌症的细胞毒性放射性药物。
嵌合抗原受体
嵌合抗原受体的抗原特异性胞外结构域识别并特异性地结合抗原,通常是恶性肿瘤的表面表达的抗原(如CD7)。例如,当抗原特异性胞外结构域以约0.1pM至约10μM、或约0.1pM至约1μM、或约0.1pM至约100nM的亲和力常数或相互作用亲和力(KD)结合抗原时,所述抗原特异性胞外结构域特异性地结合抗原。用于确定相互作用亲和力的方法在本领域中是已知的。适用于本公开的CAR的抗原特异性胞外结构域可以是任何抗原结合多肽,各种各样的抗原结合多肽在本领域中是已知的。在一些情况下,所述抗原结合结构域是单链Fv(scFv)。其他基于抗体的识别结构域(cAb VHH(骆驼抗体可变结构域)及其人源化形式、IgNAR VH(鲨鱼抗体可变结构域)及其人源化形式、sdAb VH(单结构域抗体可变结构域)和“骆驼化”抗体可变结构域适合使用。在一些情况下,基于T细胞受体(TCR)的识别结构域诸如单链TCR(scTv)也适合使用。
合适的抗原可包括T细胞特异性抗原和/或对于T细胞不是特异性的抗原。在一些的实施方案中,由CAR-T细胞的嵌合抗原受体特异性地结合的抗原和CAR-T细胞缺乏的抗原是在恶性T细胞上表达的抗原,更优选地是与非恶性T细胞相比在恶性T细胞上过 表达的抗原。“恶性T细胞”是源自T细胞恶性肿瘤的T细胞。术语“T细胞恶性肿瘤”是指广泛的高度异质分组的源自T细胞前体、成熟T细胞或自然杀伤细胞的恶性肿瘤。T细胞恶性肿瘤的非限制性实例包括T细胞急性成淋巴细胞性白血病/淋巴瘤(T-ALL)、T细胞大颗粒淋巴细胞(LGL)白血病、人T细胞白血病病毒1型阳性(HTLV-1+)成人T细胞白血病/淋巴瘤(ATL)、T细胞幼淋巴细胞白血病(T-PLL)以及各种外周T细胞淋巴瘤(PTCL),包括但不限于血管免疫母细胞性T细胞淋巴瘤(AITL)、ALK阳性间变性大细胞淋巴瘤和ALK阴性间变性大细胞淋巴瘤。
在一些实施方案中,本申请的CAR-T细胞包含特异性地结合到CD7的嵌合抗原受体的胞外结构域。CD7是T细胞表面膜相关糖蛋白。CD7可在T细胞恶性肿瘤包括T细胞急性成淋巴细胞性白血病(T-ALL)和非霍奇金氏T细胞淋巴瘤(NHL)中过表达。本公开的CAR-T细胞可用于靶向过表达CD7的恶性T细胞。
例如,本申请的嵌合抗原受体的抗原特异性胞外结构域可以特异性结合CD7,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列或与SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
本申请的嵌合抗原受体还包含胞内结构域,在抗原结合到抗原特异性胞外结构域之后,所述胞内结构域向T细胞提供胞内信号。合适的胞内结构域的非限制性实例包括T细胞受体的ζ链或其同系物中的任一种(例如,η、δ、γ或ε)、MB 1链、B29、Fc RIII、Fc RI以及信号传导分子(诸如CD3.ζ.和CD28、CD27、4-1BB、DAP-10、OX40和它们的组合以及其他相似的分子和片段)的组合。可使用激活蛋白家族的其他成员的胞内信号传导部分,诸如Fc.γ.RIII和Fc.ε.RI。虽然通常采用整个胞内结构域,但是在许多情况下,不需要使用整个胞内多肽。在胞内信号传导结构域的截短部分可使用的程度下,可使用此截短部分代替完整的链,只要所述截短部分仍然转导效应子功能信号即可。因此,本申请的胞内结构域旨在包括胞内结构域的足以转导效应子功能信号的任何截短部分。
通常,抗原特异性胞外结构域通过跨膜结构域连接到嵌合抗原受体的胞内结构域。跨膜结构域横贯细胞膜,将CAR锚固到T细胞表面,并且将胞外结构域连接到胞内信号传导结构域,从而影响CAR在T细胞表面上的表达。嵌合抗原受体还可进一步包含一个或多个共刺激结构域和/或一个或多个间隔子。共刺激结构域来源于共刺激蛋白的增强体 内细胞因子产生、增殖、细胞毒性和/或持续性的胞内信号传导结构域。间隔子(i)将抗原特异性胞外结构域连接到跨膜结构域,(ii)将跨膜结构域连接到共刺激结构域,(iii)将共刺激结构域连接到胞内结构域,和/或(iv)将跨膜结构域连接到胞内结构域。例如,在抗原特异性胞外结构域与跨膜结构域之间包含间隔子结构域可影响抗原结合结构域的柔性并且因此影响CAR功能。合适的跨膜结构域、共刺激结构域和间隔子在本领域中是已知的。
在某些实施方式中,所述的嵌合抗原受体包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
例如,其中所述跨膜域可以包含源自CD8的跨膜域。
又例如,其中所述跨膜域包含SEQ ID NO:177所示的氨基酸序列或与SEQ ID NO:177所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
在某些实施方式中,所述的嵌合抗原受体包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
例如,其中所述胞内共刺激信号传导结构域可以包含源自4-1BB的共刺激信号传导结构域。
又例如,其中所述胞内共刺激信号传导结构域可以包含SEQ ID NO:178中任一项所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14 Nef、DAP10、DAP-12和至少包含一个 ITAM的结构域。
例如,其中所述胞内信号转导结构域可以包含源自CD3ζ的信号传导结构域。
又例如,其中所述胞内信号转导结构域可以包含SEQ ID NO:179所示的氨基酸序列。
在某些实施方式中,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
例如,所述铰链区可以包含源自CD8的铰链区。
例如,所述铰链区可以包含SEQ ID NO:176所示的氨基酸序列。
在某些实施方式中,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
例如,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
在某些实施方式中,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
例如,所述信号肽片段可以包含CD8信号肽片段。
例如,所述信号肽片段可以包含SEQ ID NO:175所示的氨基酸序列。
又例如,本申请的嵌合抗原受体可以包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列或与SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
嵌合抗原受体细胞
另一方面,本申请提供一种工程化的细胞,其包含本申请所述的核酸分子或本申请所述的载体,和/或表达本申请所述的嵌合抗原受体。
例如,嵌合抗原受体可以包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列或与SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
在某些实施方式中,所述细胞包括免疫效应细胞。
在某些实施方式中,所述的免疫效应细胞包括人细胞。
在某些实施方式中,所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
在某些实施方式中,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。
在某些实施方式中,其中所述免疫效应细胞包括同种异体T细胞或自体T细胞。
在某些实施方式中,所述的免疫效应细胞包括经修饰的免疫效应细胞。
在某些实施方式中,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
在某些实施方式中,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,所述免疫效应细胞包括CAR-T细胞。
在某些实施方式中,所述CAR-T细胞缺乏嵌合抗原受体特异性地结合的抗原并且因此不诱导自相残杀。
在一些实施方案中,T细胞的抗原被修饰,使得嵌合抗原受体不再特异性地结合修饰的抗原(如CD7)。例如,被嵌合抗原受体识别的抗原的表位可通过一个或多个氨基酸变化(例如,取代或缺失)来修饰,或者所述表位可从抗原缺失。在其他实施方案中,抗原的表达在T细胞中减少至少50%、至少60%、至少70%、至少80%、至少90%或更多。用于降低蛋白质的表达的方法在本领域中是已知的,并且包括但不限于修饰或替换可操作地连接到编码蛋白质的核酸序列的启动子。在其他的实施方案中,T细胞被修饰,使得例如通过编码抗原的基因的缺失或破坏而不表达抗原。将T细胞遗传修饰成缺乏抗原的方法在本领域中是熟知的,并且非限制性实例在以上提供。在一个示例性实施方案中,CRISPR/cas9基因编辑可用于将T细胞修饰成缺乏抗原。
本公开涵盖的CAR-T细胞还可缺乏内源性T细胞受体(TCR)信号传导。在各种实施方案中,可能希望减少或消除本文公开的CAR-T细胞中的内源性TCR信号传导。例如,当同种异体T细胞用于产生CAR-T细胞时,减少或消除CAR-T细胞中的内源性TCR信号传导可预防或减少移植物抗宿主病(GvHD)。用于减少或消除内源性TCR信号传导的方法在本领域中是已知的,并且包括但不限于修饰TCR受体的一部分(例如,TCR受体α链(TRAC)等)。TRAC修饰可阻断TCR介导的信号传导。因此,TRAC修饰可允许安全地使用同种异体T细胞作为CAR-T细胞的来源而不诱导危及生命的GvHD。
可替代地或另外,本公开涵盖的CAR-T细胞还可包含一种或多种自杀基因。如本文所用,“自杀基因”是指通过本领域中已知的标准方法引入到CAR-T细胞的核酸序列,当激活时,所述核酸序列引起CAR-T细胞的死亡。如果需要的话,自杀基因可促进在体内对CAR-T细胞的有效追踪和消除。通过激活自杀基因促进杀死可通过本领域中已知的方法发生。
在一个示例性实施方案中,本公开提供一种T细胞,其包含特异性地结合CD7的嵌合抗原受体,其中所述T细胞缺乏CD7(例如,CD7ΔCART7细胞)。在非限制性实例中,缺乏CD7由以下产生:(a)修饰由T细胞表达的CD7,使得嵌合抗原受体不再特异性地结合修饰的CD7,(b)修饰T细胞,使得抗原的表达在T细胞中减少至少50%、至少60%、至少70%、至少80%、至少90%或更多,或者(c)修饰T细胞,使得CD7不被表达(例如,通过编码CD7的基因的缺失或破坏)。
用于T细胞中的CAR设计、递送和表达的方法以及临床级CAR-T细胞群体的制造在本领域中是已知的。参见例如,Lee等人,Clin.Cancer Res.,2012,18(10):2780-90。例如,工程化的CAR可使用逆转录酶病毒引入到T细胞中,这将编码嵌合抗原受体的核酸序列有效并稳定地整合到靶细胞基因组中。又例如,利用编码CAR的mRNA转入T细胞中。本领域中已知的其他方法包括但不限于慢病毒转导、基于转座子的系统、直接RNA转染和CRISPR/Cas系统。
CAR-T细胞可由本领域已知的任何合适的T细胞来源生成,所述T细胞包括但不限于从受试者采集的T细胞。受试者可以是需要CAR-T细胞疗法的患有T细胞恶性肿瘤的患者或与需要CAR-T细胞疗法的患有T细胞恶性肿瘤的受试者相同种类的受试者。采集的T细胞可使用本领域中通常已知的方法进行离体扩展,之后用CAR转导以生成CAR-T细胞。
用途
另一方面,本申请提供一种药物组合物,其包含本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体和/或本申请所述的工程化细胞,以及任选地药学上可接受的载体。
另一方面,本申请提供一种试剂盒,其包含本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表 达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物。
本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物在制备药物中的用途,所述药物用于预防和/或治疗CD7相关的疾病或病症。
在某些实施方式中,其中所述CD7相关的疾病或病症包括肿瘤。
在某些实施方式中,其中所述肿瘤包括表达CD7的肿瘤。
在某些实施方式中,其中所述肿瘤包括血液瘤。
在某些实施方式中,其中所述肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述肿瘤包括T细胞恶性肿瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的本申请所述的分离的抗原结合蛋白、本申请所述的多肽、本申请所述的免疫缀合物、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的细胞、本申请所述的嵌合抗原受体、本申请所述的工程化细胞、或本申请所述的药物组合物。
在某些实施方式中,其中所述T细胞恶性肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的本申请所述的CAR-T细胞。其中所述CAR-T细胞靶向CD7,且CD7缺失或受抑制。CAR-T细胞疗法还可伴随其他疗法,包括但不限于免疫疗法、化学疗法或放射疗法。
CAR-T细胞可通过静脉内途径(例如,通过静脉内输注)施用给受试者。CAR-T细胞可以单剂量或以多剂量施用。CAR-T细胞可在适用于静脉内施用的药物组合物中注射。用于IV施用的合适的药物组合物在本领域中是已知的。本公开的药物组合物还可包含另外的组分。例如,此类组分可用于维持注射的CAR-T细胞的活力和/或活性。
CAR-T细胞可以有效剂量施用。有效剂量可以是一个剂量或多个剂量,并且足以产生所需的治疗效果。CAR-T细胞的典型剂量的范围可以是约1×105至5×107个细胞/Kg接受疗法的受试者的体重。有效剂量可基于恶性肿瘤的阶段、受试者的健康状况和恶性 肿瘤的类型来计算。在施用多个剂量的情况下,此剂量和剂量之间的间隔可基于受试者对于疗法的反应来确定。
另一方面,本申请提供一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与本申请所述的工程化的细胞接触。
分离的核酸分子和载体
一方面,本申请提供一种靶向CD7基因的gRNA,其包含SEQ ID NO:212至SEQ ID NO:218中任一项所述的核苷酸序列或与SEQ ID NO:212至SEQ ID NO:218中任一项所述的核苷酸序列具有具有至少约90%,约91%,约92%,约93%,约94%,约95%,约96%,约97%,约98%,约99%,约99.5%同一性的氨基酸序列。
另一方面,本申请提供一种分离的核酸分子,其包含本申请所述的gRNA或编码所述gRNA的DNA分子。
另一方面,本申请提供一种载体,其包含本申请所述的核酸分子。
在本申请中,所述“载体”通常是指能够在合适的宿主中自我复制的核酸分子,用以将插入的核酸分子转移到宿主细胞中和/或宿主细胞之间。所述载体可包括主要用于将DNA或RNA插入细胞中的载体、主要用于复制DNA或RNA的载体,以及主要用于DNA或RNA的转录和/或翻译的表达的载体。所述载体还包括具有多种上述功能的载体。所述载体可以是当引入合适的宿主细胞时能够转录并翻译成多肽的多核苷酸。通常,通过培养包含所述载体的合适的宿主细胞,所述载体可以产生期望的表达产物。载体可涵盖除转基因插入序列和主链以外的额外特征:启动子、遗传标记、抗生素抗性、报告基因、靶向序列、蛋白质纯化标签。称为表达载体(表达构建体)的载体具体地讲用于在靶细胞中表达转基因,且通常具有控制序列。本申请所述的载体可以是表达载体,可包括病毒载体(慢病毒载体和/或逆转录病毒载体)、噬菌体载体、噬菌粒、粘粒、cosmid、人工染色体如酵母人工染色体(YAC)、细菌人工染色体(BAC)或P1来源的人工染色体(PAC)和/或质粒。
在某些实施方式中,所述载体包括病毒载体,可以利用病毒载体将本申请的核酸分子引入细胞。此类病毒载体包括,例如重组逆转录病毒、腺病毒、腺伴随病毒和单纯疱疹病毒-1。通常将逆转录病毒载体和腺伴随病毒载体理解为体内转移外源性基因,特别是进入人体而选用的重组基因递送系统。或者,它们可以用于将外源基因离体引入T细胞。这些载体将基因有效递送入T细胞,并且转移的核酸被稳定整合到宿主细胞的染色体 DNA中。
细胞
另一方面,本申请提供了一种细胞,其包括本申请所述的核酸分子或本申请所述的载体。
在某些实施方式中,其包括免疫效应细胞。
在某些实施方式中,所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
在某些实施方式中,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。
在某些实施方式中,其中所述免疫效应细胞包括同种异体T细胞或自体T细胞。
在某些实施方式中,其中所述免疫效应细胞包括工程化的免疫效应细胞。
在某些实施方式中,其中所述工程化的免疫效应细胞包括CAR-T细胞。
在一些实施方案中,抗原CD7的表达在T细胞中减少至少50%、至少60%、至少70%、至少80%、至少90%或更多。在一个示例性实施方案中,CRISPR/cas9基因编辑可用于将T细胞修饰成缺乏抗原CD7。
可通过本领域熟知的转染方法将本发明的gRNA引入T细胞。这些方法包括超声处理、电脉冲、电穿孔、渗透压冲击、磷酸钙沉淀和DEAE葡聚糖转染、脂质介导的递送、被动递送等。术语“转染”包括可用于将核酸引入哺乳动物细胞的多种技术,包括电穿孔、磷酸钙沉淀、DEAE-葡聚糖处理、脂质转染、显微注射和病毒感染。用于转染哺乳动物细胞的合适方法可以参见Sambrook等.(《分子克隆:实验室手册》(Molecular Cloning:A Laboratory Manual),第2版,冷泉港实验室出版社(1989))和其它实验室教科书。
在一些实施方式中,本发明包含一种递送CRISPR酶的方法,该方法包括向细胞递送编码该CRISPR酶的核酸分子,例如质粒或RNA或mRNA。在本发明的一些方法中,该CRISPR酶是Cas9。在另一些实施方式中,可以将CRISPR酶直接递送到所述细胞中。
嵌合抗原受体
在本申请中,术语“嵌合抗原受体”或“CAR”通常是指一组多肽,在最简单的实施方案中通常有两种,其当在免疫效应细胞中时,提供细胞对靶细胞(通常为癌细胞)的特异性,并产生细胞内信号。在一些实施方案中,CAR包含至少一个细胞外抗原结合结构域,跨膜结构域和胞质信号传导结构域(本文中也称为“细胞内信号传导结构域”),其包含衍 生自如下所定义的刺激分子和/或共刺激分子的功能性信号传导结构域。在一些实施方案中,该组多肽在相同的多肽链中(例如,包含嵌合融合蛋白)。在一些实施方案中,该组多肽彼此不连续,例如在不同的多肽链中。在一些方面,该组多肽包括二聚化开关,其在二聚化分子的存在下可将多肽彼此偶联,例如可将抗原结合结构域偶联至胞内信号传导结构域。一方面,CAR的刺激分子是与T细胞受体复合物相关的ζ链。在一个方面,细胞质信号传导结构域包含一级信号传导结构域(例如,CD3-ζ的一级信号传导结构域)。在一个方面,细胞质信号传导结构域还包含一个或多个衍生自如下定义的至少一种共刺激分子的功能性信号传导结构域。一方面,共刺激分子选自4-1BB(即CD137),CD27,ICOS和/或CD28。一方面,CAR包含嵌合融合蛋白,其包含细胞外抗原识别结构域,跨膜结构域和包含衍生自刺激分子的功能性信号传导结构域的细胞内信号传导结构域。一方面,CAR包含嵌合融合蛋白,其包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含衍生自共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。一方面,CAR包含嵌合融合蛋白,其包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含衍生自一个或多个共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。一方面,CAR包括嵌合融合蛋白,其包含细胞外抗原识别结构域,跨膜结构域和细胞内信号传导结构域,细胞内信号传导结构域包含至少两个衍生自一个或多个共刺激分子的功能性信号传导结构域和衍生自刺激分子的功能性信号传导结构域。在一个方面,CAR包含CAR融合蛋白的氨基末端(N-ter)上任选的前导序列。在一个方面,CAR进一步包含在细胞外抗原识别结构域的N末端的前导序列,其中前导序列任选地在细胞加工过程中从抗原识别结构域(例如scFv)切除,并将CAR定位于细胞膜。
另一方面,本申请提供一种CAR-T细胞,所述的CAR-T细胞是由本申请所述的核酸分子修饰的CAR-T细胞。
术语“CAR-T”或“CAR-T细胞”通常是指能够表达CAR(又称“嵌合抗原受体”)的T细胞。所述CAR通常是指包含能够结合抗原的胞外结构域和至少一个胞内结构域的融合蛋白。CAR是嵌合抗原受体T细胞(CAR-T)的核心部件,其可包括靶向部分(例如,结合肿瘤相关抗原(tumor-associated antigen,TAA)的部分)、铰链区、跨膜区和细胞内结构域。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结 合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中anti-CD7 CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
组合物
另一方,本申请提供一种组合物,其包含:
a.本申请所述的核酸分子,本申请所述的载体,本申请所述的细胞,或本申请所述的CAR-T细胞;和
b.药物上可以接受的载体。
药学上可接受的载体通常是指适合给予对象的物质,其中该载体在生物学上无害,或不引起其它不良影响。此类载体通常是药物的惰性成分。通常,将载体与活性成分一起给予对象,而不会引起任何不希望的生物学效应或以有害的方式与其中所包含的药物组合物的任何其它组分相互作用。合适的药物载体描述于Martin,《雷明顿药物科学》(Remington's Pharmaceutical Sciences),第18版.,马克出版社.,伊斯顿,宾夕法尼亚州.,(1990),其内容通过引用纳入本文。
本申请的更具体形式提供了药物组合物,其包含治疗有效量的核酸分子、载体或细胞以及药学上可接受的稀释剂、防腐剂、增溶剂、乳化剂、佐剂和/或载体。此类组合物包括各种缓冲剂内含物(例如磷酸盐、Tris-HCl、乙酸盐)、pH和离子强度剂以及添加剂,例如去污剂和增溶剂(例如吐温80、聚山梨酯80)、抗氧化剂(例如抗坏血酸、偏亚硫酸氢钠)、防腐剂(如硫柳汞、苯甲醇)和填充剂(如乳糖、甘露醇)。可以将这些物质掺入聚合化合物的颗粒制剂中,例如但不限于聚乳酸或聚乙醇酸,或掺入脂质体中。也可以使用透明质酸。此类组合物可影响所公开的组合物的物理状态、稳定性、体内释放率和体内清除率。所述组合物可以制备成液体形式,或者可以为干燥粉末,例如冻干形式。应当理解,本公开提供的药物组合物可以通过本领域已知的任何方式给予。例如,用于给药的药物组合物可以通过注射、口服或通过肺或鼻途径给药。
方法与用途
另一方面,本申请提供一种调节T细胞功能的方法,所述方法包括将本申请所述的gRNA、本申请所述的核酸分子、本申请所述的表达载体或本申请所述的基因编辑系统引入T细胞。
在某些实施方式中,还包括向所述细胞施用Cas酶。
应当理解的是,术语Cas酶和CRISPR酶在本文中通常可互换地使用,除非另外说明。在某些实施方式中,其中Cas酶包括Cas9蛋白。应当理解的是,本发明包括更多的来自其他微生物物种的Cas9,如SpCas9、SaCa9、StlCas9等等。
在某些实施方式中,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
在某些实施方式中,其中所述gRNA与所述Cas9同时递送。
在某些实施方式中,其中所述递送是通过电穿孔进行的。
在某些实施方式中,与未经调节的T细胞相比,经调节的所述T细胞的CD7基因表达下调或被敲除。
在某些实施方式中,所述方法还包括通过向所述T细胞引入编码CAR的核酸分子来修饰所述T细胞的特异性。
在某些实施方式中,其中所述编码CAR的核酸分子包括mRNA。
在某些实施方式中,其中mRNA编码anti-CD7 CAR。
在某些实施方式中,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
在某些实施方式中,其中anti-CD7 CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
在某些实施方式中,与相应的T细胞相比,其中所述经调节的T细胞CD7表面表达降低,并表达抗CD7 CAR。
在某些实施方式中,其通过选自以下的任一方式将本申请所述的gRNA、本申请所述的核酸分子、本申请所述的表达载体、本申请所述的基因编辑系统和/或所述编码CAR的核酸分子引入T细胞:超声处理、电脉冲、电穿孔、渗透压冲击、磷酸钙沉淀、DEAE葡聚糖转染、脂质介导的递送和被动递送。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的本申请所述的细胞或本申请所述的药物组合物。
在某些实施方式中,其中所述T细胞恶性肿瘤包括CD7阳性的恶性血液瘤。
在某些实施方式中,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
另一方面,本申请提供一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的本申请所述的CAR-T细胞。其中所述CAR-T细胞靶向CD7,且CD7缺失或 受抑制。CAR-T细胞疗法还可伴随其他疗法,包括但不限于免疫疗法、化学疗法或放射疗法。
CAR-T细胞可通过静脉内途径(例如,通过静脉内输注)施用给受试者。CAR-T细胞可以单剂量或以多剂量施用。CAR-T细胞可在适用于静脉内施用的药物组合物中注射。用于IV施用的合适的药物组合物在本领域中是已知的。本公开的药物组合物还可包含另外的组分。例如,此类组分可用于维持注射的CAR-T细胞的活力和/或活性。
CAR-T细胞可以有效剂量施用。有效剂量可以是一个剂量或多个剂量,并且足以产生所需的治疗效果。CAR-T细胞的典型剂量的范围可以是约1×105至5×107个细胞/Kg接受疗法的受试者的体重。有效剂量可基于恶性肿瘤的阶段、受试者的健康状况和恶性肿瘤的类型来计算。在施用多个剂量的情况下,此剂量和剂量之间的间隔可基于受试者对于疗法的反应来确定。
另一方面,本申请提供一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与本申请所述的CAR-T细胞接触。
本申请包括以下的实施方式:
1.分离的抗原结合蛋白,其特异性结合CD7蛋白,其中所述分离的抗原结合蛋白包含重链可变区(VH),所述VH包含HCDR1、HCDR2和HCDR3,其中HCDR1包SEQ ID NO:1所示的氨基酸序列。
2.根据实施方式1所述的分离的抗原结合蛋白,其中所述HCDR1包含SEQ ID NO:2至SEQ ID NO:9中任一项所示的氨基酸序列。
3.根据实施方式1-2中任一项所述的分离的抗原结合蛋白,其中所述HCDR2包含SEQ ID NO:10至SEQ ID NO:17中任一项所示的氨基酸序列。
4.根据实施方式1-3中任一项所述的分离的抗原结合蛋白,其中所述HCDR3包含SEQ ID NO:18或SEQ ID NO:27所示的氨基酸序列。
5.根据实施方式1-4中任一项所述的分离的抗原结合蛋白,其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO: 10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列。
6.根据实施方式1-5中任一项所述的分离的抗原结合蛋白,其中所述VH包括框架区HFR1,HFR2、HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,且所述HFR1包含SEQ ID NO:28至SEQ ID NO:38中任一项所示的氨基酸序列。
7.根据实施方式1-6中任一项所述的分离的抗原结合蛋白,其中所述HFR2位于所述HCDR1与所述HCDR2之间,且所述HFR2包含SEQ ID NO:39至SEQ ID NO:47中任一项所示的氨基酸序列。
8.根据实施方式1-7中任一项所述的分离的抗原结合蛋白,其中所述HFR3位于所述HCDR2与所述HCDR3之间,且所述HFR3包含SEQ ID NO:48至SEQ ID NO:56中任一项所示的氨基酸序列。
9.根据实施方式1-8中任一项所述的分离的抗原结合蛋白,其中所述HFR4的N末端与所述HCDR3的C末端直接或间接相连,且所述HFR4包含SEQ ID NO:57至SEQ ID NO:60中任一项所示的氨基酸序列。
10.根据实施方式1-9所述的分离的抗原结合蛋白,其包含VH,其中所述VH包括 框架区HFR1,HFR2,HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,所述HFR2位于所述HCDR1与所述HCDR2之间,所述HFR3位于所述HCDR2与所述HCDR3之间,所述HFR4的N末端与所述HCDR3的C末端直接或间接相连;其中,所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:57所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:29所示的氨基酸序列,所述HFR2包含SEQ ID NO:40所示的氨基酸序列,所述HFR3包含SEQ ID NO:49所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58示的氨基酸序列;或
所述SEQ ID NO:31所示的氨基酸序列,所述HFR2包含SEQ ID NO:41所示的氨基酸序列,所述HFR3包含SEQ ID NO:50所示的氨基酸序列,HFR4包含SEQ ID NO:59所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:32所示的氨基酸序列,所述HFR2包含SEQ ID NO:42所示的氨基酸序列,所述HFR3包含SEQ ID NO:51所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:33所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:34所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:35所示的氨基酸序列,所述HFR2包含SEQ ID NO:43所示的氨基酸序列,所述HFR3包含SEQ ID NO:52所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ  ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:44所示的氨基酸序列,所述HFR3包含SEQ ID NO:53所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:36所示的氨基酸序列,所述HFR2包含SEQ ID NO:45所示的氨基酸序列,所述HFR3包含SEQ ID NO:54所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:37所示的氨基酸序列,所述HFR2包含SEQ ID NO:46所示的氨基酸序列,所述HFR3包含SEQ ID NO:55所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
所述HFR1包含SEQ ID NO:38所示的氨基酸序列,所述HFR2包含SEQ ID NO:47所示的氨基酸序列,所述HFR3包含SEQ ID NO:56所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列。
11.根据实施方式1-10中任一项所述的分离的抗原结合蛋白,其包含VH,其中所述VH包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
12.根据实施方式1-11中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR1包含SEQ ID NO:74至SEQ ID NO:86中任一项所示的氨基酸序列。
13.根据实施方式1-12中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR2包含SEQ ID NO:87至SEQ ID NO:98中任一项所示的氨基酸序列。
14.根据实施方式1-13中任一项所述的分离的抗原结合蛋白,其包括VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR3包含SEQ ID NO:99至SEQ ID NO:111中任一项所示的氨基酸序列。
15.根据实施方式1-14中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1,LCDR2和LCDR3,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO: 88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:78所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
16.根据实施方式1-15中任一项所述的分离的抗原结合蛋白,其包含VH和VL,其中所述VH包含HCDR1,HCDR2和HCDR3,所述VL包含LCDR1,LCDR2和LCDR3;其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1 包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸 序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
17.根据实施方式1-16中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,且所述LFR1包含SEQ ID NO:112至SEQ ID NO:123中任一项所示的氨基酸序列。
18.根据实施方式1-17中任一项所述的分离的抗原结合蛋白,其中所述LFR2位于所述LCDR1与所述LCDR2之间,且所述LFR2包含SEQ ID NO:124至SEQ ID NO:130中任一项所示的氨基酸序列。
19.根据实施方式1-18中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR3,所述LFR3位于所述LCDR2与所述LCDR3之间,且所述LFR3包含SEQ ID NO:131至SEQ ID NO:141中任一项所示的氨基酸序列。
20.根据实施方式1-19中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR4,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连, 且所述LFR4包含SEQ ID NO:142至SEQ ID NO:147中任一项所示的氨基酸序列。
21.根据实施方式1-20中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,所述LFR2位于所述LCDR1与所述LCDR2之间,所述LFR3位于所述LCDR2与所述LCDR3之间,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连;其中所述LFR1包含SEQ ID NO:112所示的氨基酸序列,所述LFR2包含SEQ ID NO:124所示的氨基酸序列,所述LFR3包含SEQ ID NO:131所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:113所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:132所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:133所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:115所示的氨基酸序列,所述LFR2包含SEQ ID NO:127所示的氨基酸序列,所述LFR3包含SEQ ID NO:134所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:116所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:135所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:117所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:136所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:118所示的氨基酸序列,所述LFR2包含SEQ ID NO:128所示的氨基酸序列,所述LFR3包含SEQ ID NO:137所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:119所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:138所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:120所示的氨基酸序列,所述LFR2包含SEQ ID NO:129所示的氨基酸序列,所述LFR3包含SEQ ID NO:139所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:140所示的氨基酸序列,所述LFR4包含SEQ ID NO:144所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:121所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:145所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:122所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:146所示的氨基酸序列;或
所述LFR1包含SEQ ID NO:123所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:147所示的氨基酸序列。
22.根据实施方式1-21中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
23.根据实施方式1-22中任一项所述的分离的抗原结合蛋白,其包含VH和VL,其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所示的氨基酸序列;或
所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或
所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或
所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或
所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或
所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所 示的氨基酸序列;或
所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或
所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或
所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或
所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或
所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或
所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或
所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列。
24.根据实施方式1-23中任一项所述的分离的抗原结合蛋白,其包括抗体或其抗原结合片段。
25.根据实施方式1-24中任一项所述的分离的抗原结合蛋白,所述抗体包括单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体、完整抗体、抗体片段、人抗体、人源化抗体或嵌合抗体。
26.根据实施方式1-25中任一项所述的分离的抗原结合蛋白,其中所述抗原结合片段包括Fab,Fab’,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb。
27.根据实施方式1-26所述的抗原结合蛋白,其包括scFv。
28.根据实施方式1-27所述的抗原结合蛋白,其中所述VL和VH通过接头连接。
29.根据实施方式28所述的抗原结合蛋白,其中所述接头包括多肽接头。
30.根据实施方式29所述的抗原结合蛋白,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
31.根据实施方式1-30中任一项所述的分离的抗原结合蛋白,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
32.分离的多肽,其包含实施方式1-31中任一项所述的分离的抗原结合蛋白。
33.免疫缀合物,其包含实施方式1-31中任一项所述的抗原结合蛋白。
34.嵌合抗原受体,其包含至少一个包含实施方式1-31中任一项的抗原结合蛋白的细胞外抗原结合结构域。
35.根据实施方式34所述的嵌合抗原受体,其中所述细胞外抗原结合结构域包括scFv。
36.根据实施方式1-35中任一项所述的嵌合抗原受体,其包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
37.根据实施方式36所述的嵌合抗原受体,其中所述跨膜域包含源自CD8的跨膜域。
38.根据实施方式34-37中任一项所述的嵌合抗原受体,其中所述跨膜域包含SEQ ID NO:177所示的氨基酸序列。
39.根据实施方式34-38中任一项所述的嵌合抗原受体,其包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
40.根据实施方式39所述的嵌合抗原受体,其中所述胞内共刺激信号传导结构域源自4-1BB的共刺激信号传导结构域。
41.根据实施方式39-40中任一项所述的嵌合抗原受体,其中所述胞内共刺激信号传导结构域包含SEQ ID NO:178中任一项所示的氨基酸序列。
42.根据实施方式34-41中任一项所述的嵌合抗原受体,其包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14 Nef、DAP10、 DAP-12和至少包含一个ITAM的结构域。
43.根据实施方式42所述的嵌合抗原受体,其中所述胞内信号转导结构域包含源自CD3ζ的信号传导结构域。
44.根据实施方式42-43中任一项所述的嵌合抗原受体,其中所述胞内信号转导结构域包含SEQ ID NO:179所示的氨基酸序列。
45.根据实施方式34-44中任一项所述的嵌合抗原受体,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
46.根据实施方式45所述的嵌合抗原受体,所述铰链区包含源自CD8的铰链区。
47.根据实施方式45-46中任一项所述的嵌合抗原受体,所述铰链区包含SEQ ID NO:176所示的氨基酸序列。
48.根据实施方式34-47中任一项所述的嵌合抗原受体,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
49.根据实施方式34-48中任一项所述的嵌合抗原受体,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
50.根据实施方式34-49中任一项所述的嵌合抗原受体,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
51.根据实施方式50所述的嵌合抗原受体,所述信号肽片段包括CD8信号肽片段。
52.根据实施方式中50-51中任一项所述的嵌合抗原受体,所述信号肽片段包含如SEQ ID NO:175所示的氨基酸序列。
53.根据实施方式34-52中任一项所述的嵌合抗原受体,其包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
54.分离的核酸分子,其编码实施方式1-31中任一项所述的分离的抗原结合蛋白、实施方式32所述的多肽或实施方式34-53中任一项所述的嵌合抗原受体。
55.表达载体,其包含根据实施方式54所述的核酸分子。
56.细胞,所述细胞包含根据实施方式54所述的核酸分子或根据实施方式55所述的 表达载体,和/或ii)所述细胞表达根据实施方式1-31中任一项所述的抗原结合蛋白、实施方式32所述的多肽或实施方式34-53中任一项所述的嵌合抗原受体。
57.制备实施方式1-31中任一项所述的分离的抗原结合蛋白的方法,所述方法包括在使得实施方式1-31中任一项所述的分离的抗原结合蛋白表达的条件下,培养根据实施方式56所述的细胞。
58.工程化的细胞,其包含实施方式1-31中任一项所述的核酸分子或实施方式55中任一项所述的载体,和/或表达实施方式34-53中任一项所述的嵌合抗原受体。
59.根据实施方式58所述的工程化的细胞,所述细胞包括免疫效应细胞。
60.根据实施方式59所述的工程化的细胞,所述的免疫效应细胞包括人细胞。
61.根据实施方式59-60中任一项所述的工程化的细胞,所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
62.根据实施方式59-61中任一项所述的工程化的细胞,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。
63.根据实施方式59-62中任一项所述的工程化的细胞,其中所述免疫效应细胞包括同种异体T细胞或自体T细胞。
64.根据实施方式59-63中任一项所述的工程化的细胞,所述的免疫效应细胞包括经修饰的免疫效应细胞。
65.根据实施方式64所述的工程化的细胞,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
66.根据实施方式64-65中任一项所述的工程化的细胞,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
67.根据实施方式69-66中任一项所述的工程化的细胞,所述免疫效应细胞包括CAR-T细胞。
68.根据实施方式67所述的工程化的细胞,所述CAR-T细胞不诱导自相残杀。
69.一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
a.对T细胞群中的CD7基因进行修饰;
b.激活所述T细胞群;
c.用实施方式34-53中任一项所述的嵌合抗原受体转导所述T细胞;以及
d.扩增所述嵌合抗原受体T细胞。
70.根据实施方式69所述的方法,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
71.根据实施方式69-70中任一项所述的方法,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
72.根据实施方式69-71中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
73.根据实施方式68-72中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
74.根据实施方式68-73中任一项所述的方法,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
75.根据实施方式74所述的方法,其中所述靶向CD7基因的gRNA包含SEQ ID NO:211至SEQ ID NO:218中任一项所示的核苷酸序列。
76.根据实施方式74所述的方法,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
77.根据实施方式74所述的方法,其中所述gRNA与所述Cas9同时递送。
78.根据实施方式77所述的方法,其中所述递送是通过电穿孔进行的。
79.药物组合物,其包含实施方式1-31中任一项所述的分离的抗原结合蛋白、实施方式31所述的多肽、实施方式33所述的免疫缀合物、实施方式54所述的核酸分子、实施方式55所述的表达载体、实施方式56所述的细胞、实施方式34-53中任一项所述的嵌合抗原受体和/或实施方式58-68中任一项所述的工程化细胞,以及任选地药学上可接受的载体。
80.试剂盒,其包含实施方式1-31中任一项所述的分离的抗原结合蛋白、实施方式31所述的多肽、实施方式33所述的免疫缀合物、实施方式54所述的核酸分子、实施方式55所述的表达载体、实施方式56所述的细胞、实施方式34-53中任一项所述的嵌合抗原受体、实施方式58-68中任一项所述的工程化细胞、或实施方式79所述的药物组合物。
81.实施方式1-31中任一项所述的分离的抗原结合蛋白、实施方式31所述的多肽、实施方式33所述的免疫缀合物、实施方式54所述的核酸分子、实施方式55所述 的表达载体、实施方式56所述的细胞、实施方式34-53中任一项所述的嵌合抗原受体、实施方式58-68中任一项所述的工程化细胞、或实施方式79所述的药物组合物在制备药物中的用途,所述药物用于预防和/或治疗CD7相关的疾病或病症。
82.根据实施方式81所述的用途,其中所述CD7相关的疾病或病症包括肿瘤。
83.根据实施方式82所述的用途,其中所述肿瘤包括表达CD7的肿瘤。
84.根据实施方式82-83中任一项所述的用途,其中所述肿瘤包括血液瘤。
85.根据实施方式82-84中任一项所述的用途,其中所述肿瘤包括CD7阳性的恶性血液瘤。
86.根据实施方式82-85中任一项所述的用途,其中所述肿瘤包括T细胞恶性肿瘤。
87.一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量实施方式1-31中任一项所述的分离的抗原结合蛋白、实施方式31所述的多肽、实施方式33所述的免疫缀合物、实施方式54所述的核酸分子、实施方式55所述的表达载体、实施方式56所述的细胞、实施方式34-53中任一项所述的嵌合抗原受体、实施方式58-68中任一项所述的工程化细胞、或实施方式79所述的药物组合物。
88.根据实施方式87所述的方法,其中所述肿瘤包括表达CD7的肿瘤。
89.根据实施方式87所述的方法,其中所述肿瘤包括血液瘤。
90.根据实施方式87所述的方法,其中所述肿瘤包括CD7阳性的恶性血液瘤。
91.根据实施方式87所述的方法,其中所述肿瘤包括T细胞恶性肿瘤。
92.根据实施方式91所述的方法,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
93.一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与实施方式58-68中任一项所述的工程化的细胞接触。
本申请还提供以下的实施方式:
1.靶向CD7基因的gRNA,其包含SEQ ID NO:212至SEQ ID NO:218中任一项所述的核苷酸序列。
2.分离的核酸分子,其包含实施方式1所述的gRNA或编码所述gRNA的DNA分子。
3.表达载体,其包含实施方式1所述的gRNA或实施方式2所述的核酸分子。
4.基因编辑系统,其包含实施方式1所述的gRNA、实施方式2所述的核酸分子 或实施方式3所述的表达载体。
5.根据实施方式4所述的基因编辑系统,其包括CRISPR/Cas基因编辑系统。
6.根据实施方式4所述的基因编辑系统,其包括CRISPR/Cas9基因编辑系统。
7.根据实施方式4所述的基因编辑系统,其还包含编码Cas9的DNA、编码Cas9的mRNA或Cas9蛋白分子。
8.根据实施方式4所述的基因编辑系统,其包含编码实施方式1所述的靶向CD7基因的gRNA和Cas9的表达载体。
9.细胞,其包含实施方式1所述的gRNA、实施方式2所述的核酸分子、实施方式3所述的表达载体、或实施方式4中任一项所述的基因编辑系统。
10.根据实施方式9所述的细胞,其包括表达CD7的细胞。
11.根据实施方式9所述的细胞,其包括免疫效应细胞。
12.根据实施方式11所述的细胞,其中所述免疫效应细胞的包括T细胞、B细胞、自然杀伤(NK)细胞、肥大细胞或吞噬细胞。
13.根据实施方式11-12中任一项所述的细胞,其中所述免疫效应细胞包括工程化的免疫效应细胞。
14.根据实施方式13所述的细胞,其中所述工程化的免疫效应细胞靶向CD7。
15.根据实施方式13-14中任一项所述的细胞,其中所述工程化的免疫效应细胞包括CAR-T细胞。
16.根据实施方式13-15中任一项所述的细胞,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
17.根据实施方式15-16中任一项所述的细胞,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
18.根据实施方式15-17中任一项所述的细胞,其中所述CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
19.实施方式1所述的gRNA,实施方式2所述的核酸分子,实施方式3所述的表达载体,实施方式4-8所述的基因编辑系统或实施方式9-18中任一项所述的细胞在制备用于治疗肿瘤药物中的应用。
20.根据实施方式19所述的应用,其中所述肿瘤包括实体瘤或血液瘤。
21.根据实施方式19-20中任一项所述的应用,其中所述肿瘤包括CD7阳性肿瘤。
22.根据实施方式19-21中任一项所述的应用,其中所述药物包括CAR-T细胞。
23.根据实施方式19-22中任一项所述的应用,其中所述药物包括靶向CD7 CAR-T细胞。
24.对细胞中CD7基因进行基因编辑的方法,包括利用实施方式1所述的gRNA介导Cas9对CD7基因进行基因编辑。
25.根据实施方式24所述的方法,其中所述基因编辑包括基因敲除。
26.一种调节T细胞功能的方法,所述方法包括将实施方式1所述的gRNA、实施方式2所述的核酸分子、实施方式3所述的表达载体或实施方式4-8中任一项所述的基因编辑系统引入T细胞。
27.根据实施方式26所述的方法,还包括向所述细胞施用Cas酶。
28.根据实施方式27所述的方法,其中Cas酶包括Cas9蛋白。
29.根据实施方式28所述的方法,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
30.根据实施方式29所述的方法,其中所述gRNA与所述Cas9同时递送。
31.根据实施方式30所述的方法,其中所述递送是通过电穿孔进行的。
32.根据实施方式26-31中任一项所述的方法,与未经调节的T细胞相比,经调节的所述T细胞的CD7基因表达下调或被敲除。
33.根据实施方式26-32中任一项所述的方法,所述方法还包括通过向所述T细胞引入编码CAR的核酸分子来修饰所述T细胞的特异性。
34.根据实施方式33所述的方法,其中所述编码CAR的核酸分子包括mRNA。
35.根据实施方式34所述的方法,其中mRNA编码抗CD7 CAR。
36.根据实施方式35所述的方法,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
37.根据实施方式36所述的方法,其中anti-CD7 CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
38.根据实施方式26-37中任一项所述的方法,与相应的T细胞相比,其中所述经调节的T细胞CD7表面表达降低,并表达抗CD7 CAR。
39.根据实施方式26-38中任一项所述的方法,其通过选自以下的任一方式将实施方式1所述的gRNA、实施方式2所述的核酸分子、实施方式3所述的表达载体、实施方 式4-8中任一项所述的基因编辑系统和/或所述编码CAR的核酸分子引入T细胞:超声处理、电脉冲、电穿孔、渗透压冲击、磷酸钙沉淀、DEAE葡聚糖转染、脂质介导的递送和被动递送。
本申请还提供以下的实施方式:
1.经修饰的免疫效应细胞,其包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
2.根据实施方式1所述的经修饰的免疫效应细胞,其中所述第一结构域的N端与第二结构域的C端连接。
3.根据实施方式1所述的经修饰的免疫效应细胞,其中所述第一结构域的C端与第二结构域的N端连接。
4.根据实施方式1-3中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域与第二结构域直接连接或间接连接。
5.根据实施方式1-4中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域与第二结构域通过连接子连接。
6.根据实施方式5所述的经修饰的免疫效应细胞,其中所述连接子包括肽连接子。
7.根据实施方式1-6中任一项所述的经修饰的免疫效应细胞,其中所述抗体或其抗原结合片段为scFv。
8.根据实施方式1-7中任一项所述的经修饰的免疫效应细胞,其中所述APC选自由树突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
9.根据实施方式1-8中任一项所述的经修饰的免疫效应细胞,其中所述APC的激活受体选自CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
10.根据实施方式1-9中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含结合CD40、CD80、CD86、CD91、DEC-205、DC-SIGN的配体或它们的受体结合片段。
11.根据实施方式1-10中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含CD40配体(CD40L)的受体结合片段。
12.根据实施方式1-11中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含结合所述APC的激活受体的抗体或其抗原结合片段。
13.根据实施方式1-12中任一项所述的经修饰的免疫效应细胞,其中所述第一结结构域为抗CD40抗体或其抗原结合片段。
14.根据实施方式1-13中任一项所述的经修饰的免疫效应细胞,其中所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、嗜中性粒细胞和粒细胞组成的组。
15.根据实施方式1-14中任一项所述的经修饰的免疫效应细胞,其中第二结构域包含共刺激受体的胞内结构域。
16.根据实施方式1-15中任一项所述的经修饰的免疫效应细胞,其中所述共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
17.根据实施方式1-16中任一项所述的经修饰的免疫效应细胞,其中所述共刺激受体为CD28。
18.根据实施方式1-17中任一项所述的经修饰的免疫效应细胞,其中所述第二结构域是免疫效应细胞的共刺激配体或其受体结合片段。
19.根据实施方式1-18中任一项所述的经修饰的免疫效应细胞,其中共刺激配体选自CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44。
20.根据实施方式1-19中任一项所述的经修饰的免疫效应细胞,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
21.根据实施方式1-21中任一项所述的经修饰的免疫效应细胞,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
22.根据实施方式22所述的经修饰的免疫效应细胞,其中共刺激受体为CD28或4-1BB。
23.根据实施方式1-22中任一项所述的经修饰的免疫效应细胞,其中所述第一结构 域与第二结构域选自以下组合:
i)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28或4-1BB胞内结构域;
ii)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
iii)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28胞内结构域;
iv)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
v)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;和
vi)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段。
24.根据实施方式1-23中任一项所述的经修饰的免疫效应细胞,其中所述融合蛋白包含SEQ ID NO:83至SEQ ID NO:99中任一项所示的氨基酸序列。
25.根据实施方式1-24中任一项所述的经修饰的免疫效应细胞,其中所述免疫效应细胞包括工程化的免疫效应细胞。
26.根据实施方式25所述的经修饰的免疫效应细胞,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
27.根据实施方式1-26中任一项所述的经修饰的免疫效应细胞,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
28.根据实施方式27所述的经修饰的免疫效应细胞,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
29.根据实施方式28所述的经修饰的免疫效应细胞,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:28至SEQ ID NO:40中任一项所示的氨基酸序列。
30.根据实施方式29所述的经修饰的免疫效应细胞,其中所述CAR包含SEQ ID NO:46至SEQ ID NO:58中任一项所示的氨基酸序列。
31.根据实施方式1-30中任一项所述的经修饰的免疫效应细胞,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
32.根据实施方式1-31中任一项所述的经修饰的免疫效应细胞,所述经修饰的免疫效应细胞不诱导自相残杀。
33.嵌合抗原受体T(CAR-T)细胞,其表达抗CD7 CAR,并且包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
34.根据实施方式33所述的CAR-T细胞,其中所述CAR-T细胞的CD7的表达缺失或受抑制。
35.根据实施方式33-34中任一项所述的CAR-T细胞,所述CAR-T细胞不诱导自相残杀。
36.药物组合,其包含免疫效应细胞和融合蛋白,其中所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,或(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段。
37.根据实施方式36所述的药物组合,其中所述第一结构域的N端与第二结构域的C端连接。
38.根据实施方式36所述的药物组合,其中所述第一结构域的C端与第二结构域的N端连接。
39.根据实施方式36-38中任一项所述的药物组合,其中所述第一结构域与第二结构域直接连接或间接连接。
40.根据实施方式36-39中任一项所述的药物组合,其中所述第一结构域与第二结构域通过连接子连接。
41.根据实施方式40所述的药物组合,其中所述连接子包括肽连接子。
42.根据实施方式36-41中任一项所述的药物组合,其中所述抗体或其抗原结合片段为scFv。
43.根据实施方式36-42中任一项的药物组合,其中所述APC选自由树突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
44.根据实施方式36-43中任一项所述的药物组合,其中所述APC的激活受体选自CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
45.根据实施方式36-44中任一项所述的药物组合,其中所述第一结构域包含结合CD40、CD80、CD86、CD91、DEC-205、DC-SIGN的配体或它们的受体结合片段。
46.根据实施方式36-45中任一项所述的药物组合,其中所述第一结构域包含CD40配体(CD40L)的受体结合片段。
47.根据实施方式36-46中任一项所述的药物组合,其中所述第一结构域包含结合所述APC的激活受体的抗体或其抗原结合片段。
48.根据实施方式36-47中任一项所述的药物组合,其中所述第一结构域为抗CD40抗体或其抗原结合片段。
49.根据实施方式36-48中任一项所述的药物组合,其中所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、嗜中性粒细胞和粒细胞组成的组。
50.根据实施方式36-49中任一项所述的药物组合,其中第二结构域包含共刺激受体的胞内结构域。
51.根据实施方式36-50中任一项所述的药物组合,其中所述共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
52.根据实施方式36-51中任一项所述的药物组合,其中所述共刺激受体为CD28或4-1BB。
53.根据实施方式36-52中任一项所述的药物组合,其中所述第二结构域是免疫效应细胞的共刺激配体或其受体结合片段。
54.根据实施方式36-53中任一项所述的药物组合,其中共刺激配体选自CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44。
55.根据实施方式36-54中任一项所述的药物组合,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
56.根据实施方式36-55中任一项所述的药物组合,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
57.根据实施方式36-56中任一项所述的药物组合,其中共刺激受体为CD28或4-1BB。
58.根据实施方式36-57中任一项所述的药物组合,其中所述第一结构域与第二结构域选自以下组合:
i)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;
ii)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
iii)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;和
iv)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段。
59.根据实施方式36-58中任一项所述的药物组合,其中所述融合蛋白包含中SEQ ID NO:86至SEQ ID NO:90中任一项所示的氨基酸序列。
60.根据实施方式36-59中任一项所述的药物组合,其中所述免疫效应细胞包括工程化的免疫效应细胞。
61.根据实施方式36-60中任一项所述的药物组合,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
62.根据实施方式36-61中任一项中任一项所述的药物组合,与相应的免疫细胞相比,其中所述药物组合CD7表面表达降低,并表达抗CD7 CAR。
63.根据实施方式36-62中任一项所述的药物组合,其中所述工程化的免疫效应细胞包括抗CD7 CAR-T细胞。
64.根据实施方式36-63中任一项所述的药物组合,其中所述CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:28至SEQ ID NO:40中任一项所示的氨基酸序列。
65.根据实施方式36-64中任一项所述的药物组合,其中所述CAR包含SEQ ID NO:46至SEQ ID NO:58中任一项所示的氨基酸序列。
66.根据实施方式36-65中任一项中任一项所述的药物组合,所述免疫效应细胞不诱导自相残杀。
67.药物组合物,其包含实施方式1-32中任一项所述的经修饰的免疫效应细胞,实 施方式33-35中任一项所述的CAR-T细胞或实施方式36-66中任一项所述的药物组合,以及药学上可接受的载体。
68.实施方式1-32中任一项所述的经修饰的免疫效应细胞、实施方式33-35中任一项所述的CAR-T细胞、实施方式36-66中任一项所述的药物组合或实施方式67所述的药物组合物在制备药物中的用途,所述药物用于治疗肿瘤。
69.根据实施方式68所述的用途,其中所述肿瘤包括血液瘤和实体瘤。
70.根据实施方式68-69中任一项所述的用途,其中所述肿瘤包括表达CD7的肿瘤。
71.根据实施方式68-70中任一项所述的用途,其中所述肿瘤包括CD7阳性的恶性血液瘤。
72.根据实施方式68-71中任一项所述的用途,其中所述肿瘤包括T细胞恶性肿瘤。
73.根据实施方式68-72中任一项所述的用途,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
74.一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的实施方式1-32中任一项所述的经修饰的免疫效应细胞、实施方式33-35中任一项所述的CAR-T细胞、实施方式36-66中任一项所述的药物组合或实施方式67所述的药物组合物。
75.一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与实施方式1-32中任一项所述的经修饰的免疫效应细胞、实施方式33-35中任一项所述的CAR-T细胞、实施方式36-66中任一项所述的药物组合或实施方式67所述的药物组合物接触。
76.一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
a.对T细胞群中的CD7基因进行修饰;
b.激活所述T细胞群;
c.将抗CD7 CAR和融合蛋白转导所述T细胞群;以及
d.扩增所述嵌合抗原受体T细胞群。
77.根据实施方式76所述的方法,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
78.根据实施方式76-77中任一项所述的方法,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物 核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
79.根据实施方式76-78中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
80.根据实施方式76-79中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
81.根据实施方式80所述的方法,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
82.根据实施方式81所述的经修饰的方法,其中所述靶向所述CD7基因的gRNA包含SEQ ID NO:64至SEQ ID NO:71中任一项所示的核苷酸序列。
83.根据实施方式82所述的方法,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
84.根据实施方式83所述的方法,其中所述gRNA与所述Cas9同时递送。
85.根据实施方式84所述的方法,其中所述递送是通过电穿孔进行的。
86.根据实施方式76-85中任一项所述的方法,其中所述将抗CD7 CAR和融合蛋白转导所述T细胞包括将编码所述抗CD7 CAR的核酸分子和编码所述融合蛋白的核酸分子引入T细胞。
87.包含实施方式1-32中任一项所述的经修饰的免疫效应细胞或实施方式33-35中任一项所述CAR-T细胞的细胞群,其中所述细胞群来源于外周血单核细胞(PBMC)、外周血血液白细胞(PBL)、肿瘤浸润淋巴细胞(TIL)、细胞因子诱导的杀伤细胞(CIK)、淋巴因子激活的杀伤细胞(LAK)或骨髓浸润淋巴细胞(MIL)。
circRNA
本申请提供了一种circRNA,其按以下顺序包含内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA)。
在本申请中,所述polyA的长度可以至少为45个核苷酸。例如,所述polyA的长度可以为至少50个核苷酸、至少55个核苷酸、至少60个核苷酸、至少65个核苷酸、至少70个核苷酸、至少75个核苷酸、至少80个核苷酸或更多。
在本申请中,所述靶向CD7的蛋白质可以包括靶向CD7的抗体或其抗原结合片段、嵌合抗原受体(CAR)和/或T细胞受体(TCR)。
在本申请中,所述抗体可以包括单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体、完整抗体、抗体片段、人抗体、人源化抗体或嵌合抗体。
在本申请中,所述抗原结合片段可以包括Fab,Fab’,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb。
在本申请中,所述靶向CD7的蛋白质可以为靶向CD7的嵌合抗原受体(CAR)。所述CAR可以包含特异性地结合到CD7的嵌合抗原受体的胞外结构域。所述胞外结构域也可以称为结合结构域。例如,所述结合结构域可以包括CD7 scFv。
在本申请中,所述抗体(例如,所述抗体或所述CAR的结合结构域,例如所述CD7scFV)可以包括包含LCDR1、LCDR2和LCDR3的轻链可变区和包含HCDR1、HCDR2和HCDR3的重链可变区,其中:所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序 列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
在本申请中,所述抗体(例如,所述抗体或所述CAR的结合结构域,例如所述CD7 scFV)可以包括包括VH和VL,其中所述VH包含SEQ ID NO:61所示的氨基酸序列, 所述VL包含SEQ ID NO:148所示的氨基酸序列;或所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列;或所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列。
在本申请中,所述的circRNA可以包含SEQ ID NO:256-257中任一项所示的核苷酸序列。
本申请还提供了一种调节CD7表达水平和/或活性的试剂,其可以包含本申请所述的circRNA。本申请还提供了本申请所述的circRNA在调节CD7表达水平和/或活性中的应用。
本申请还提供了一种调节CD7表达水平和/或活性的方法,其可以包括以下步骤:施用本申请所述的circRNA。
在本申请中,所述试剂可以以试剂盒的形式提供。所述试剂盒中还可以包括其他可以调节CD7表达水平和/或活性的试剂和/或仪器。
在本申请中,所述表达水平可以包括检测样本的编码CD7的核酸(DNA和/或mRNA)的表达水平;所述表达水平可以包括检测样本的CD7蛋白质的表达水平。所述调节可以在体内、体外和/或离体条件下进行。
所述调节可以包括上调和/或下调。例如,所述下调可以为与不施用所述circRNA相比,CD7表达水平和/或活性下调至少10%、至少20%、至少30%、至少40%、至少50%、 至少60%、至少70%、至少80%、至少90%、至少100%或更多。
所述施用可以包括递送,例如可以通过电穿孔的方法递送本申请所述的circRNA。
在本申请中,所述的circRNA,其还可以包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段。
在本申请中,所述第一结构域可以与第二结构域的C端或者N端连接。
在本申请中,所述连接子可以包括肽连接子。
在本申请中,所述第一结构域可以包含CD40L或者其受体结合片段,抗CD40抗体或其抗原结合片段,第二结构域包含CD28胞内结构域或抗CD28抗体或其抗原结合片段。
在本申请中,所述第一结构域可以包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28胞内结构域。
在本申请中,所述融合蛋白可以包含SEQ ID NO:234至SEQ ID NO:250中任一项所示的氨基酸序列。
在本申请中,所述circRNA可以包含前体RNA。所述前体RNA可以包含环化元件(circularizing element)、内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA)。
所述环化元件可以包含位于内部核糖体进入位点(IRES)元5’的第一内含子序列和位于多聚腺苷酸3’的第二内含子序列。所述第一内含子序列和所述第二内含子序列可以源自I组或II组内含子自剪接序列。
所述第一内含子序列可以包含含有3’剪接位点核苷酸的I组内含子片段,并且所述第二内含子序列可以包含含有5’剪接的I组内含子片段。
所述前体RNA还可以包含在第一内含子序列和IRES之间的5’间隔序列,以及在第一内含子序列和IRES之间的3’间隔序列。
所述前体RNA还可以包含在第一内含子序列外部5'同源臂和在第二内含子序列外部的3'同源臂。
本申请还提供了包含本申请所述前体RNA的载体。所述载体可以包含编码所述前体RNA的DNA。
本申请还提供了生产本申请所述circRNA的方法。例如,其可以包含以下的步骤:转录本申请所述包含前体RNA的载体。所述转录可以在细胞内和/或无细胞条件下进行。在本申请中,所述方法还可以包括纯化本申请所述circRNA的步骤。例如,通过基于oligo dT的捕获来进行纯化。
本申请还提供了包含本申请所述circRNA的细胞和/或细胞群。
本申请还提供了本申请所述circRNA、上述前体RNA、载体、细胞和/或细胞群在制备治疗疾病的药物中的用途,所述药物用于治疗肿瘤。所述肿瘤可以包括血液瘤和实体瘤。
所述肿瘤可以包括表达CD7的肿瘤。所述肿瘤可以包括CD7阳性的恶性血液瘤。所述肿瘤可以包括T细胞恶性肿瘤。所述T细胞恶性肿瘤可以包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
不欲被任何理论所限,下文中的实施例仅仅是为了阐释本申请的细胞、制备方法和用途等,而不用于限制本申请发明的范围。
实施例
实施例1从全人源抗体噬菌体展示库中筛选抗CD7抗体
1.通过加入新鲜解冻的M13K07辅助噬菌体感染对数期TG1库培养物,感染倍数为20:1(噬菌体-细胞比),用IPTG过夜诱导噬菌体展示库的表达。
2.用PEG/NaCl沉淀法纯化噬菌体库,并确定噬菌体滴度。将噬菌体储存在4℃,并尽快进行scFv选择。
3.CD7特异性scFv-噬菌体的选择。对于第一轮选择,用20μg/ml CD7-6His蛋白溶解在1×PBS中涂在Maxisorp平板上,在4℃下孵育过夜。(对于随后的轮次,降低蛋白质浓度以进行更严格的选择,第2轮生物淘选中为2μg/ml,第3轮生物淘选中为0.5ug/ml)。
4.用PBS洗三次,在每个孔中加入封闭缓冲液(5%牛奶+1%BSA,在1×PBS中),在RT孵育2小时。
5.弃去封闭缓冲液,加入噬菌体溶液,用封口膜密封,轻轻摇晃孵育2小时。
6.在第一轮筛选中,用PBST洗10次。在接下来的几轮中,通过增加洗涤周期来 提高洗涤的严格程度(第二轮20次,第三轮30次)。
7.洗脱抗原结合的scFv噬菌体。加入1毫升酸性洗脱缓冲液(pH2.2),孵育约8分钟,然后用吸管将洗脱的噬菌体吸到约100μl中和缓冲液中,放在一个新型PP管中。
8.将15mL对数期的TG1培养物(OD600=0.5)与洗脱的噬菌体溶液接种在新的50mL试管中。在37℃下孵化,静置30分钟,摇晃30分钟。然后在含有2xYT-GA琼脂的15cm平板上进行培养。
9.在30℃下培养平板过夜,并收获细菌以进行后续的筛选工作。
10.三轮选择后,选择数百个阳性菌落进行单克隆噬菌体ELISA(mpELISA)筛选。
11.mpELISA筛选。将单个细菌产生的噬菌体上清液与涂有2μg/ml CD7-6His蛋白的预阻断Maxisorp平板进行孵化。洗涤三次后,加入100μl/孔的HRP结合的抗M13抗体,在阻断缓冲液(5%牛奶+1%BSA在1×PBS中)中稀释为1:5000,在RT下孵育60分钟。用PBST洗板5次后,加入100μl/孔的TMB底物溶液,孵育10-30分钟直到出现蓝色。加入50μl/孔的终止液(2N H2SO4)终止反应。在酶标仪读取在450nm处的吸光度。
12.根据ELISA结果选择阳性克隆,并作为模板进行scFv序列的PCR克隆(正向引物序列:tgcagctggcacgacaggtttc(SEQ ID NO:219),反向引物序列:cgtcagactgtagcacgtt(SEQ ID NO:220))。然后用桑格测序法对PCR产物进行测序(正向引物序列:aacaattgaattcaggagga(SEQ ID NO:221),反向引物序列:cctcctaagaagcgtagtc(SEQ ID NO:222))。
13.通过abysis网站(http://abysis.org/)对scFv的CDR区域进行了分析。
结果如图1所示,经过3轮生物淘洗,选择288个菌落进行培养并产生噬菌体上清液,测试其与CD7-Fc蛋白的结合,阳性克隆为灰色高亮部分。这些数字表明,在噬菌体展示筛选实验中,许多抗CD7抗体被成功筛选出来。在scFv筛选部分,从噬菌体展示库中成功筛选出18个抗CD7抗体:抗CD7 scFv-H1至抗CD7 scFv-H18。
实施例2构建用于生成mRNA的抗CD7 CAR载体
1.用Xba1和Sal1酶消化pDA载体,并通过凝胶纯化法纯化。
2.用PCR扩增scFv序列和CAR片段(从铰链结构域到CD3-zeta结构域),并用凝胶纯化法纯化。
3.用Gibson组装法将scFv片段、CAR片段(从铰链结构域到CD3-zeta结构域)和 pDA载体连接起来(如图2所示),并转化感受态细胞。
4.通过sanger测序确认合适的菌落,并选择进一步实验。
实施例3 CAR mRNA的体外转录(IVT)
1.用Spe1酶消化pDA-CAR质粒,使其线性化。
2.用PCR净化试剂盒纯化线性化载体,用无RNase水洗脱。
3.用纳米滴管测量DNA的浓度,并通过琼脂糖DNA凝胶进行检测。
4.按照制造商(Thermofisher,Cat No:AMB13455)的协议进行IVT。具体地,将1ug模板DNA、NTP/ARCA缓冲液、T7缓冲液、GTP、T7酶和无RNase H2O以20ul体积加入到0.2ml PCR管中,并在37℃下孵育3小时。
5. 3小时后,在每个反应中加入2ul的DNase,并在37℃下孵育15分钟。
6.然后根据制造商的建议进行尾随程序。
7.使用RNasy试剂盒(Qiagen)纯化IVT mRNA。
8.用纳米滴管测量RNA的浓度,并通过运行PAGE凝胶进行检测。
实施例4 CD7 gRNA的体外转录(IVT)
1.用EcoR1酶消化pUC57-CD7-gRNA质粒,使其线性化。
2.用PCR净化试剂盒纯化线性化载体,用无RNase水洗脱。
3.用纳米滴管测量DNA的浓度,并通过琼脂糖DNA凝胶进行检测。
4.按照制造商(Thermofisher,Cat No:AMB13345)的协议进行IVT。简单地说,将1ug模板DNA、NTP缓冲液、T7缓冲液、T7酶和无RNase H2O以20ul的体积加入到0.2ml PCR管中,并在37℃下孵育3-4小时。
5. 3-4小时后,在每个反应中加入1ul的DNase,并在37℃下孵育30分钟。
6.使用RNasy试剂盒(Qiagen)纯化IVT gRNA。
7.用纳米滴管测量gRNA的浓度,并通过运行PAGE凝胶进行检测。
实施例5肿瘤细胞系和原代人淋巴细胞
所有肿瘤细胞系,包括H226、OVCAR3、SKOV3、Caski、U87、U251、ASPC1、A549、Raji、Nalm6、MOLM14、SupT1、DOHH2、RPMI8226、Jurkat和CEM,均在补充有10%FCS的RPMI-1640培养基中培养。用抗CD3/CD28 Dynabeads(Life Technologies)刺激正常供体的初级淋巴细胞,并在R10培养基(RPMI-1640补充有10%FCS,青霉素-链霉素(100倍),HEPES(100倍),丙酮酸钠(100倍),Glutamax(100倍),NEAA(100 倍))中培养。刺激后第12天将T细胞冷冻保存在90%FCS和10%DMSO的溶液中,每瓶1×10e8个细胞。
实施例6将mRNA电穿孔至A549和T细胞
1.收集A549肿瘤细胞和T细胞,用Opti-MEM培养基清洗3次。
2.用Opti-MEM培养基重悬细胞颗粒,细胞浓度应是1×10e7/ml。
3.将10ug RNA分装到1.5ml EP管中,然后加入100ul T细胞或A549细胞,混合均匀。
4.在BTX机上设置参数:
a)用于T细胞:500V,0.7ms;
b)对于A549肿瘤细胞:300V,0.5ms。
5.在BTX电穿孔杯中加入100ul与RNA混合的细胞,轻拍以避免气泡。
6.进行电穿孔,然后将细胞转移到预热的培养基中,在37℃下培养。
用同型和抗CD7抗体对电穿孔的不同数量的CD7 mRNA的A549细胞进行FACS染色。图5显示,CD7的异位表达水平与电穿孔到A549细胞的CD7 mRNA的数量有关。
实施例7 CD7 KO CAR-T细胞和CD7 KO CAR+LACO T细胞的制备
1.第0天,用CD3/CD28 Dynabeads(CTS Dynabeads CD3/CD28:Gibco;Cat#402031;4E8 beads/mL)激活CD3+T细胞,CD3/CD28 beads:T细胞的比例=3:1。
2.第4天,从T细胞中去除CD3/CD28 Dynabeads。将T细胞转移到15ml法氏管中,并放入磁力管中,放置3-5分钟,并将上清液转移到新的15ml法氏管中。
3.计算T细胞的细胞数,用OPTI-MEM洗涤三次,并将T细胞重悬至50E6/mL。
4.用Cas9重组蛋白和CD7 gRNA电穿孔T细胞。用OPTI-MEM洗涤T细胞三次后,将15ug Cas9蛋白和7.5ug CD7 gRNA在室温下混合10分钟。然后将Cas9蛋白/gRNA混合物加入到T细胞中,轻轻地混合均匀。用BTX ECM 830进行电穿孔,条件参数为360V,1ms。继续培养电穿孔的T细胞。
5.第9天,用抗CD7抗体染色测试CD7基因敲除的效率,并用流式细胞仪进行分析。
FACS染色结果(图3)显示通过CRISPR-Cas9基因编辑技术对不同CD7 gRNA的敲除效率:CD7 gRNA-2、gRNA-5、gRNA-6、gRNA-8、gRNA-9、gRNA-10、gRNA-11和gRNA-12的敲除效率为54.5%、87.9%、42.7%、42.7、80.1%、60.5%、92%和88.6%, 而CD7 gRNA-1、gRNA-3、gRNA-4和gRNA-7几乎没有敲除作用。
6.第12天,电穿孔抗CD7 CAR mRNA,产生CD7 KO抗CD7 CAR-T细胞;或
电穿孔抗CD7 CAR mRNA与LACO mRNA(本试验中采用的LACO分子是A40C.CD28(A40C28)),产生CD7 KO抗CD7 CAR+A40C28 T细胞。
检测CAR-T细胞中表达的抗CD7 scFv与CD7-Fc蛋白的结合能力,其中Mock是没有CAR分子的对照T细胞。FACS染色结果(图4)显示,抗CD7 scFv-H1、-H4、-H5、-H6、-H7、-H8、-H9、-H10、-H12、-H13、-H14、-H15、-H17和-H18对CD7-Fc重组蛋白有结合能力,而抗CD7 scFv-H2、-H3、-H11和-H16对CD7-Fc重组蛋白没有结合能力或结合能力较弱。
用同型对照和抗CD7 mAb对17个不同种类的肿瘤细胞系进行FACS染色,图12显示,MOLM14、SupT1和CEM肿瘤细胞高水平表达CD7,而U87、A549、ASPC1、H226、SKOV3、786O、OVCAR3、U251、Caski、Raji、Nalm6、DOHH2、RPMI8226和Jurkat不表达CD7或低水平表达CD7而无法检测。
实施例8抗CD7 CAR-T细胞的体外细胞毒性试验
1.将表达EGFP的肿瘤细胞系或电穿孔的EGFP-A549细胞接种到平底96孔板,3000个细胞/100ul/孔。
2.将CAR-T细胞稀释到适当的浓度,用100ul/孔的细胞接种到不同E/T比例的肿瘤细胞上,如10:1,3:1,1:1。
3.将共培养板放入IncuCyte S3机器,并设置扫描参数。
4.扫描3天后,分析绿色物体总集成强度(GCU xμm2/Well),计算杀伤效率。
结果如图6-8所示,除抗CD7 scFv-H2 CAR-T、抗CD7 scFv-H3 CAR-T、抗CD7 scFv-H11 CAR-T、抗CD7 scFv-H14 CAR-T和抗CD7 scFv-H16CAR-T细胞之外,其余抗CD7 scFv CAR-T对表达CD7的A549肿瘤细胞均有杀伤力。
其中,图9显示了抗CD7 scFv-H1、-H7、-H9、-H10和-H17 CAR-T细胞对CD7过度表达的A549肿瘤细胞都有很强的杀伤力。
图10显示了抗CD7 CAR(H1,H7,H9,H10,H17)+A40C28 T细胞对CD7过度表达的A549肿瘤细胞都有很强的杀伤力。
实施例9 CAR-T细胞的CD107a染色
1.在96孔板的每个孔中加入20ul PE-CD107a mAb。
2.稀释肿瘤细胞至2×10e6/ml,用100ul/孔/96孔圆板播种细胞。
3. 3.稀释CAR-T细胞至1×10e6/ml,用100ul/孔/96孔圆板播种细胞。
4.在500rpm下离心5分钟,使细胞附着在上面。
5.在37℃下培养细胞1小时。
6.用培养基稀释高尔基止血剂1500倍,在每个孔中加入20ul稀释的高尔基止血剂。
7.在37℃下再培养细胞2.5小时。
8.用抗CD3-APC和抗CD8-FITC抗体在37℃下染色30分钟。
9.清洗并通过流式细胞仪进行分析。
CD107a是一个早期激活的标记物。图11显示,抗CD7 CAR(H1、-H7、-H9、-H10和-H17)T细胞可以被CD7抗原特异性激活。
在与H226、OVCAR3、SKOV3、Caski、U87、U251、ASPC1、A549和A549+10ug CD7 mRNA的共培养和杀伤试验中,不同mRNA的抗CD7 CAR-T细胞的CD107a染色,包括模拟T细胞(NO EP)、用抗CD7 CAR-H1、-H7、-H9、-H10和-H17电穿孔的T细胞。图13-14显示,所有五个抗CD7的CAR(H1、H7、H9、H10、H17)T细胞都被CD7抗原表达的细胞特异性激活,但CD7阴性的肿瘤细胞系却没有。
在gRNA筛选部分,在12个候选gRNA中,CD7 gRNA-2、gRNA-5、gRNA-6、gRNA-8、gRNA-9、gRNA-10、gRNA-11和gRNA-12的CD7敲除效率分别为54.5%、87.9%、42.7%、42.7、80.1%、60.5%、92%和88.6%,而CD7 gRNA-1、gRNA-3、gRNA-4和gRNA-7几乎没有敲除效果。
在18个抗CD7 scFv序列中,抗CD7 scFv-H1、-H4、-H5、-H6、-H7、-H8、-H9、-H10、-H12、-H13、-H14、-H15、-H17和-H18具有与CD7-Fc重组蛋白的结合能力,而抗CD7 scFv-H2、-H3、-H11和-H16对CD7-Fc重组蛋白没有结合能力或结合能力较弱。进一步的结果显示,在肿瘤杀伤和CD107a染色实验中,五个抗CD7的CAR(H1、H7、H9、H10、H17)T细胞都被CD7抗原表达的细胞特异性激活,而不是CD7阴性肿瘤细胞系。
实施例10淋巴细胞-APCs共刺激分子构建
设计并构建了四种不同形式的同时针对CD28和CD40的融合蛋白(淋巴细胞-APC共刺激分子(LACO-Stim))(图15)。
1)由细胞外CD40配体(CD40L)和细胞内CD28组成的融合蛋白。
2)由抗CD28 scFv和CD40L胞外区组成的融合蛋白(基于Ab的可溶性蛋白)。
3)由抗CD28和CD40的scFv组成的双特异性抗体。
4)由抗CD40 scFv作为细胞外区与CD28(或4-1BB)细胞内区组成的嵌合蛋白(基于Ab的膜融合蛋白)。
实施例11共表达LACO-stim与CAR—T细胞或TCR-T细胞的构建
通过RNA电穿孔将表3中的Her2 CAR(4D5.BBZ)或针对NY-ESO-1 TCR和LACO-stim共同转移至T细胞,通过流式细胞仪检查CAR/LACO-stim的表达。所有与CAR和LACO-stim共转的T细胞都检测到CAR染色和CD40-Fc蛋白的结合(图16)
在Incucyte活细胞分析系统中测试了T细胞的抗肿瘤功能,该系统是一个实时定量的活细胞成像和分析平台,能够对细胞行为进行可视化和量化。使用转导了HLA-A2和NY-ESO-1(A549-ESO)的Her2阳性癌系A549作为靶向肿瘤细胞系,发现与单独的CAR或与转换分子PD1-CD28共表达的CAR相比,与CAR共表达的T细胞和1412.T4.CD40L,或1412-F2.103,或1412-F5.157或1412-F5.77显示控制肿瘤生长的能力有所提高,表明可溶性形式的LACO-stim为T细胞提供了额外的CD28信号,以进一步激活T细胞、增殖和生存,导致控制肿瘤生长的能力提高(图17)。当LACO-stim与NY-ESO-1 TCR共同引入时,发现与单独的TCR或与PD1-CD28共同转移的TCR相比,所有LACO-stim分子都能帮助TCR T细胞进一步控制肿瘤生长(图18)。
表3

实施例12 CirRNA电转细胞制备与表征
1.CD7 H1.BBZ CAR线性mRNA的体外转录(IVT)
1.1通过用BspQ1酶消化使pDA-CD7 H1.BBZ CAR(即CAR-H1)质粒线性化。其中,CAR-H1的氨基酸序列如SEQ ID NO.180所示。
1.2用PCR Cleanup kit(Qiagen)纯化线性化的载体,用RNase-free水洗脱。
1.3 DNA浓度用nanodrop测定,并通过琼脂糖DNA凝胶电泳检测。
1.4 IVT按照说明书操作(Thermofisher,目录号:AMB13455)。简单来说,将1μg模板DNA、NTP/ARCA缓冲液、T7缓冲液、GTP、T7酶和RNase free H2O以20μl体积加入0.2ml PCR管中,并在37℃下孵育4小时。
1.5 4小时后,每次反应加入2μl DNase I,37℃孵育15分钟。
1.6然后进行加尾程序(tailing procedure)。
1.7使用RNasy试剂盒(Qiagen)纯化IVT mRNA。
1.8 RNA浓度采用nanodrop法测定,PAGE凝胶电泳检测。
2.CD7 H1.BBZ CAR circRNA的体外转录(IVT)
2.1通过用BspQ1酶消化使pCA-CD7 H1.BBZ、pCA45-CD7 H1.BBZ、pCA70-CD7 H1.BBZ CAR质粒线性化。
2.2用PCR Cleanup kit(Qiagen)纯化线性化载体,用RNase-free水洗脱。
2.3 DNA浓度用nanodrop测定,并通过琼脂糖DNA凝胶电泳检测。
2.4 IVT按照说明书操作(Thermofisher,目录号:AMB13345)。简而言之,将1μg模板DNA、NTP混合物、T7缓冲液、GTP、T7酶和RNase free H2O以20μl体积加 入0.2ml PCR管中,并在37℃下孵育4小时。
2.5 4小时后,每次反应加入2μl DNase I,37℃孵育15分钟。
2.6在DNase处理后,加入额外的GTP至终浓度为2mM,然后将反应在55℃下加热15分钟。
2.7 circRNA通过柱或AKTA系统进行纯化。
2.8 RNA浓度采用nanodrop法测定,并且通过PAGE凝胶电泳检测。
其中,CD7 H1.BBZ CAR线性mRNA和CD7 H1.BBZ CAR circRNA的结构均参见图19。其中,T7启动子的核苷酸序列如SEQ ID NO.258所示;同源臂1的核苷酸序列如SEQ ID NO.259所示;内含子1的核苷酸序列如SEQ ID NO.260所示;间隔子1的核苷酸序列如SEQ ID NO.261所示;CVB3IRES的核苷酸序列如SEQ ID NO.262所示;;间隔子2的核苷酸序列如SEQ ID NO.263所示;内含子2的核苷酸序列如SEQ ID NO.264所示;同源臂2的核苷酸序列如SEQ ID NO.265所示;70A的核苷酸序列如SEQ ID NO.266所示。
CD7 H1.BBZ CAR线性mRNA(CD7 CAR-H1 linear mRNA)的核苷酸序列如SEQ ID NO.255所示;CD7 H1.BBZ CAR circRNA(CD7 CAR-H1 circRNA)的核苷酸序列如SEQ ID NO.256所示;CD7 H1.BBZ CAR circRNA(CD7 CAR-H1 circRNA-70A)的核苷酸序列如SEQ ID NO.257所示。
3.通过CIMmultus Oligo dT柱纯化circRNA
3.1缓冲液制备:缓冲液A,平衡/洗涤缓冲液:50mM磷酸钠、250mM氯化钠、5mM EDTA,pH 7.0;缓冲液B,第二洗涤缓冲液:50mM磷酸钠,5mM EDTA,pH 7.0;缓冲液C,洗脱缓冲液:10mM Tris,pH 8.0;缓冲液D,清洁缓冲液:3M盐酸胍,5mM EDTA,pH 7.0。
3.2用缓冲液A平衡柱:泵平衡缓冲液(A)通过CIMmultus Oligo dT柱,直到输出pH和电导率与输入缓冲液相同。流速:每分钟5个柱体积(CV/min)。
3.3注入样品。根据说明书进样。
3.4使用缓冲液A进行第一轮洗脱(Wash 1):10CV的平衡缓冲液。
3.5使用缓冲液B进行第二轮洗脱(Wash 2):10CV的第二洗涤缓冲液。
3.6使用缓冲液C稀释:洗脱并收集circRNA样品,直至UV吸光度回到基线。
3.7使用缓冲液D清洗。用至少10CV的清洗缓冲液清洗柱系统。
4.将RNA电穿孔转染至A549-GFP和T细胞
4.1收集A549-GFP肿瘤细胞和T细胞,用Opti-MEM培养基洗涤3次。
4.2用Opti-MEM培养基重悬细胞沉淀,调整细胞浓度至1×10e7/ml。
4.3分装5μg MSLN mRNA至1.5ml EP管中,加入100μl A549细胞,混匀。对于T细胞电穿孔,将10μg CD7 H1.BBZ mRNA或circRNA等分到1.5ml EP管中,然后加入100μl A549细胞,混匀。
4.4设置BTX机器参数:
a)T细胞:500伏,0.7毫秒
b)A549肿瘤细胞:300伏,0.5毫秒;
4.5在BTX电穿孔杯中加入100μl混有RNA的细胞,轻敲避免气泡。
4.6进行电穿孔,然后将细胞转移到预热的培养基中,37℃培养。
5.CD7 H1.BBZ CAR-T细胞的体外细胞毒性测定
5.1共培养前12小时,将用5μg CD7 mRNA电穿孔的A549-EGFP细胞以3000个细胞/100μl/孔接种到平底96孔板中。
5.2电穿孔后约12-24小时,计数并稀释CAR-T细胞至合适浓度,种子细胞100μl/孔加入不同E/T比例如3:1和1:1的肿瘤细胞。
5.3将共培养板放入IncuCyte S3机器中,设置扫描参数。
5.4扫描4天后,分析总绿色物体综合强度(GCU x μm2/Well),计算杀灭效率。
结果如图20A-20B所示,其分别显示CD7 H1.BBZ CAR线性mRNA和CD7 H1.BBZ CAR circRNA对肿瘤细胞有特异性的杀伤效果。其中,图20B中的A40C28是指同时电转CD7 H1.BBZ CAR线性mRNA和A40C28线性mRNA的T细胞;或者,同时电转CD7 H1.BBZ CAR circRNA和A40C28 cirRNA的T细胞。20A显示,cirRNA组效果优于线性mRNA组,cirRNA ployA组效果优于cirRNA组;20B显示,共表达A40C28的细胞中,cirRNA ployA组效果优于线性mRNA组。
参考文献
1.Morgan RA,Dudley ME,Wunderlich JR,Hughes MS,Yang JC,Sherry RM,Royal RE, Topalian SL,Kammula US,Restifo NP,Zheng Z et al:Cancer regression in patients after transfer of genetically engineered lymphocytes.Science(2006)314(5796):126-129.
2.Robbins PF,Morgan RA,Feldman SA,Yang JC,Sherry RM,Dudley ME,Wunderlich JR,Nahvi AV,Helman LJ,Mackall CL,Kammula US et al:Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with ny-eso-1.J Clin Oncol(2011)29(7):917-924.
3.Rapoport AP,Stadtmauer EA,Binder-Scholl GK,Goloubeva O,Vogl DT,Lacey SF,Badros AZ,Garfall A,Weiss B,Finklestein J,Kulikovskaya I et al:Ny-eso-1-specific tcr-engineered t cells mediate sustained antigen-specific antitumor effects in myeloma.Nature medicine(2015)21(8):914-921.
4.Kochenderfer JN,Wilson WH,Janik JE,Dudley ME,Stetler-Stevenson M,Feldman SA,Maric I,Raffeld M,Nathan DA,Lanier BJ,Morgan RA et al:Eradication of b-lineage cells and regression of lymphoma in a patient treated with autologous t cells genetically engineered to recognize cd19.Blood(2010)116(20):4099-4102.
5.Kalos M,Levine BL,Porter DL,Katz S,Grupp SA,Bagg A,June CH:T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia.Science translational medicine(2011)3(95):95ra73.
6.Anderson KG,Stromnes IM,Greenberg PD:Obstacles posed by the tumor microenvironment to t cell activity:A case for synergistic therapies.Cancer Cell(2017)31(3):311-325.
7.Boutros C,Tarhini A,Routier E,Lambotte O,Ladurie FL,Carbonnel F,Izzeddine H,Marabelle A,Champiat S,Berdelou A,Lanoy E et al:Safety profiles of anti-ctla-4 and anti-pd-1 antibodies alone and in combination.Nature reviews Clinical oncology(2016)13(8):473-486.
8.Stephan MT,Ponomarev V,Brentjens RJ,Chang AH,Dobrenkov KV,Heller G,Sadelain M:T cell-encoded cd80 and 4-1bbl induce auto-and transcostimulation,resulting in potent tumor rejection.Nat Med(2007)13(12):1440-1449.
9.Topp MS,Riddell SR,Akatsuka Y,Jensen MC,Blattman JN,Greenberg PD:Restoration of cd28 expression in cd28-cd8+memory effector t cells reconstitutes antigen-induced il-2 production.J Exp Med(2003)198(6):947-955.
10.Daniel-Meshulam I,Horovitz-Fried M,Cohen CJ:Enhanced antitumor activity mediated by human 4-1bb-engineered t cells.International journal of cancer(2013)133(12):2903-2913.
11.Ankri C,Cohen CJ:Out of the bitter came forth sweet:Activating cd28-dependent co-stimulation via pd-1 ligands.Oncoimmunology(2014)3(1):e27399.
12.Prosser ME,Brown CE,Shami AF,Forman SJ,Jensen MC:Tumor pd-l1 co-stimulates primary human cd8(+)cytotoxic t cells modified to express a pd1:Cd28 chimeric receptor.Molecular immunology(2012)51(3-4):263-272.
13.Ankri C,Shamalov K,Horovitz-Fried M,Mauer S,Cohen CJ:Human t cells engineered to express a programmed death 1/28 costimulatory retargeting molecule display enhanced antitumor activity.J Immunol(2013)191(8):4121-4129.
14.Kobold S,Grassmann S,Chaloupka M,Lampert C,Wenk S,Kraus F,Rapp M,Duwell P,Zeng Y,Schmollinger JC,Schnurr M et al:Impact of a new fusion receptor on pd-1-mediated immunosuppression in adoptive t cell therapy.J Natl Cancer Inst(2015)107(8).
15.Liu X,Ranganathan R,Jiang S,Fang C,Sun J,Kim S,Newick K,Lo A,June CH,Zhao Y,Moon EK:A chimeric switch-receptor targeting pd1 augments the efficacy of second-generation car t cells in advanced solid tumors.Cancer research(2016)76(6):1578-1590.
16.Schlenker R,Olguin-Contreras LF,Leisegang M,Schnappinger J,Disovic A,Ruhland S,Nelson PJ,Leonhardt H,Harz H,Wilde S,Schendel DJ et al:Chimeric pd-1:28 receptor upgrades low-avidity t cells and restores effector function of tumor-infiltrating lymphocytes for adoptive cell therapy.Cancer research(2017)77(13):3577-3590.
17.Li S,Siriwon N,Zhang X,Yang S,Jin T,He F,Kim YJ,Mac J,Lu Z,Wang S,Han X et al:Enhanced cancer immunotherapy by chimeric antigen receptor-modified t cells engineered to secrete checkpoint inhibitors.Clinical cancer research:an official journal of the American Association for Cancer Research(2017)23(22):6982-6992.
18.Rafiq S,Yeku OO,Jackson HJ,Purdon TJ,van Leeuwen DG,Drakes DJ,Song M,Miele MM,Li Z,Wang P,Yan S et al:Targeted delivery of a pd-1-blocking scfv by car-t cells enhances anti-tumor efficacy in vivo.Nature biotechnology(2018)36(9):847-856.
19.Liu X,Jiang S,Fang C,Yang S,Olalere D,Pequignot EC,Cogdill AP,Li N,Ramones  M,Granda B,Zhou L et al:Affinity-tuned erbb2 or egfr chimeric antigen receptor t cells exhibit an increased therapeutic index against tumors in mice.Cancer Res(2015)75(17):3596-3607.
20.Zhao Y,Zheng Z,Robbins PF,Khong HT,Rosenberg SA,Morgan RA:Primary human lymphocytes transduced with ny-eso-1 antigen-specific tcr genes recognize and kill diverse human tumor cell lines.J Immunol(2005)174(7):4415-4423.
21.Zhao Y,Moon E,Carpenito C,Paulos CM,Liu X,Brennan AL,Chew A,Carroll RG,Scholler J,Levine BL,Albelda SM et al:Multiple injections of electroporated autologous t cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor.Cancer research(2010)70(22):9053-9061.

Claims (151)

  1. 分离的抗原结合蛋白,其特异性结合CD7蛋白,其中所述分离的抗原结合蛋白包含重链可变区(VH),所述VH包含HCDR1、HCDR2和HCDR3,其中HCDR1包含SEQ ID NO:1所示的氨基酸序列。
  2. 根据权利要求1所述的分离的抗原结合蛋白,其中所述HCDR1包含SEQ ID NO:2至SEQ ID NO:9中任一项所示的氨基酸序列。
  3. 根据权利要求1-2中任一项所述的分离的抗原结合蛋白,其中所述HCDR2包含SEQ ID NO:10至SEQ ID NO:17中任一项所示的氨基酸序列。
  4. 根据权利要求1-3中任一项所述的分离的抗原结合蛋白,其中所述HCDR3包含SEQ ID NO:18或SEQ ID NO:27所示的氨基酸序列。
  5. 根据权利要求1-4中任一项所述的分离的抗原结合蛋白,其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列。
  6. 根据权利要求1-5中任一项所述的分离的抗原结合蛋白,其中所述VH包括框架区HFR1,HFR2、HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,且所述HFR1包含SEQ ID NO:28至SEQ ID NO:38中任一项所示的氨基酸序列。
  7. 根据权利要求1-6中任一项所述的分离的抗原结合蛋白,其中所述HFR2位于所述HCDR1与所述HCDR2之间,且所述HFR2包含SEQ ID NO:39至SEQ ID NO:47中任一项所示的氨基酸序列。
  8. 根据权利要求1-7中任一项所述的分离的抗原结合蛋白,其中所述HFR3位于所述HCDR2与所述HCDR3之间,且所述HFR3包含SEQ ID NO:48至SEQ ID NO:56中任一项所示的氨基酸序列。
  9. 根据权利要求1-8中任一项所述的分离的抗原结合蛋白,其中所述HFR4的N末端与所述HCDR3的C末端直接或间接相连,且所述HFR4包含SEQ ID NO:57至SEQ ID NO:60中任一项所示的氨基酸序列。
  10. 根据权利要求1-9所述的分离的抗原结合蛋白,其包含VH,其中所述VH包括框架区HFR1,HFR2,HFR3和HFR4,所述HFR1的C末端与所述HCDR1的N末端直接或间接相连,所述HFR2位于所述HCDR1与所述HCDR2之间,所述HFR3位于所述HCDR2与所述HCDR3之间,所述HFR4的N末端与所述HCDR3的C末端直接或间接相连;其中,所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:57所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:29所示的氨基酸序列,所述HFR2包含SEQ ID NO:40所示的氨基酸序列,所述HFR3包含SEQ ID NO:49所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58示的氨基酸序列;或
    所述SEQ ID NO:31所示的氨基酸序列,所述HFR2包含SEQ ID NO:41所示的氨 基酸序列,所述HFR3包含SEQ ID NO:50所示的氨基酸序列,HFR4包含SEQ ID NO:59所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:32所示的氨基酸序列,所述HFR2包含SEQ ID NO:42所示的氨基酸序列,所述HFR3包含SEQ ID NO:51所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:33所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:34所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:35所示的氨基酸序列,所述HFR2包含SEQ ID NO:43所示的氨基酸序列,所述HFR3包含SEQ ID NO:52所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:28所示的氨基酸序列,所述HFR2包含SEQ ID NO:39所示的氨基酸序列,所述HFR3包含SEQ ID NO:48所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:30所示的氨基酸序列,所述HFR2包含SEQ ID NO:44所示的氨基酸序列,所述HFR3包含SEQ ID NO:53所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:36所示的氨基酸序列,所述HFR2包含SEQ ID NO:45所示的氨基酸序列,所述HFR3包含SEQ ID NO:54所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:37所示的氨基酸序列,所述HFR2包含SEQ ID NO:46所示的氨基酸序列,所述HFR3包含SEQ ID NO:55所示的氨基酸序列,HFR4包含SEQ ID NO:60所示的氨基酸序列;或
    所述HFR1包含SEQ ID NO:38所示的氨基酸序列,所述HFR2包含SEQ ID NO:47所示的氨基酸序列,所述HFR3包含SEQ ID NO:56所示的氨基酸序列,HFR4包含SEQ ID NO:58所示的氨基酸序列。
  11. 根据权利要求1-10中任一项所述的分离的抗原结合蛋白,其包含VH,其中所述VH包含SEQ ID NO:61至SEQ ID NO:73所示的氨基酸序列。
  12. 根据权利要求1-11中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR1包含SEQ ID NO:74至SEQ ID NO:86中任一项所示的氨基酸序列。
  13. 根据权利要求1-12中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR2包含SEQ ID NO:87至SEQ ID NO:98中任一项所示的氨基酸序列。
  14. 根据权利要求1-13中任一项所述的分离的抗原结合蛋白,其包括VL,其中所述VL包含LCDR1、LCDR2、LCDR3,所述LCDR3包含SEQ ID NO:99至SEQ ID NO:111中任一项所示的氨基酸序列。
  15. 根据权利要求1-14中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含LCDR1,LCDR2和LCDR3,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:78所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
    所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
  16. 根据权利要求1-15中任一项所述的分离的抗原结合蛋白,其包含VH和VL,其中所述VH包含HCDR1,HCDR2和HCDR3,所述VL包含LCDR1,LCDR2和LCDR3;其中所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:74所示的氨基酸序列,所述LCDR2包含SEQ ID NO:87所示的氨基酸序列,所述LCDR3包含SEQ ID NO:99所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:3所示的氨基酸序列,所述HCDR2包含SEQ ID NO:11所示的氨基酸序列,所述HCDR3包含SEQ ID NO:19所示的氨基酸序列,所述LCDR1包含SEQ ID NO:75所示的氨基酸序列,所述LCDR2包含SEQ ID NO:88所示的氨基酸序列,所述LCDR3包含SEQ ID NO:100所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:20所示的氨基酸序列,所述LCDR1包含SEQ ID NO:76所示的氨基酸序列,所述LCDR2包含SEQ ID NO:89所示的氨基酸序列,所述LCDR3包含SEQ ID NO:101所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:4所示的氨基酸序列,所述HCDR2包含SEQ ID NO:12所示的氨基酸序列,所述HCDR3包含SEQ ID NO:21所示的氨基酸序列,所述LCDR1包含SEQ ID NO:77所示的氨基酸序列,所述LCDR2包含SEQ ID NO:90所示的氨基酸序列,所述LCDR3包含SEQ ID NO:102所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:5所示的氨基酸序列,所述HCDR2包含SEQ ID NO:13所示的氨基酸序列,所述HCDR3包含SEQ ID NO:22所示的氨基酸序列,所述LCDR1包含SEQ ID NO:91所示的氨基酸序列,所述LCDR2包含SEQ ID NO:91所示的氨基酸 序列,所述LCDR3包含SEQ ID NO:103所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:79所示的氨基酸序列,所述LCDR2包含SEQ ID NO:92所示的氨基酸序列,所述LCDR3包含SEQ ID NO:104所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:80所示的氨基酸序列,所述LCDR2包含SEQ ID NO:93所示的氨基酸序列,所述LCDR3包含SEQ ID NO:105所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:6所示的氨基酸序列,所述HCDR2包含SEQ ID NO:14所示的氨基酸序列,所述HCDR3包含SEQ ID NO:23所示的氨基酸序列,所述LCDR1包含SEQ ID NO:81所示的氨基酸序列,所述LCDR2包含SEQ ID NO:94所示的氨基酸序列,所述LCDR3包含SEQ ID NO:106所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:18所示的氨基酸序列,所述LCDR1包含SEQ ID NO:82所示的氨基酸序列,所述LCDR2包含SEQ ID NO:95所示的氨基酸序列,所述LCDR3包含SEQ ID NO:107所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:2所示的氨基酸序列,所述HCDR2包含SEQ ID NO:10所示的氨基酸序列,所述HCDR3包含SEQ ID NO:24所示的氨基酸序列,所述LCDR1包含SEQ ID NO:83所示的氨基酸序列,所述LCDR2包含SEQ ID NO:96所示的氨基酸序列,所述LCDR3包含SEQ ID NO:108所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:7所示的氨基酸序列,所述HCDR2包含SEQ ID NO:15所示的氨基酸序列,所述HCDR3包含SEQ ID NO:25所示的氨基酸序列,所述LCDR1包含SEQ ID NO:84所示的氨基酸序列,所述LCDR2包含SEQ ID NO:97所示的氨基酸序列,所述LCDR3包含SEQ ID NO:109所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:8所示的氨基酸序列,所述HCDR2包含SEQ ID NO:16所示的氨基酸序列,所述HCDR3包含SEQ ID NO:26所示的氨基酸序列,所述LCDR1包含SEQ ID NO:85所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:110所示的氨基酸序列;或
    所述HCDR1包含SEQ ID NO:9所示的氨基酸序列,所述HCDR2包含SEQ ID NO:17所示的氨基酸序列,所述HCDR3包含SEQ ID NO:27所示的氨基酸序列,所述LCDR1包含SEQ ID NO:86所示的氨基酸序列,所述LCDR2包含SEQ ID NO:98所示的氨基酸序列,所述LCDR3包含SEQ ID NO:111所示的氨基酸序列。
  17. 根据权利要求1-16中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,且所述LFR1包含SEQ ID NO:112至SEQ ID NO:123中任一项所示的氨基酸序列。
  18. 根据权利要求1-17中任一项所述的分离的抗原结合蛋白,其中所述LFR2位于所述LCDR1与所述LCDR2之间,且所述LFR2包含SEQ ID NO:124至SEQ ID NO:130中任一项所示的氨基酸序列。
  19. 根据权利要求1-18中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR3,所述LFR3位于所述LCDR2与所述LCDR3之间,且所述LFR3包含SEQ ID NO:131至SEQ ID NO:141中任一项所示的氨基酸序列。
  20. 根据权利要求1-19中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR4,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连,且所述LFR4包含SEQ ID NO:142至SEQ ID NO:147中任一项所示的氨基酸序列。
  21. 根据权利要求1-20中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包括框架区LFR1,LFR2,LFR3和LFR4,其中所述LFR1的C末端与所述LCDR1的N末端直接或间接相连,所述LFR2位于所述LCDR1与所述LCDR2之间,所述LFR3位于所述LCDR2与所述LCDR3之间,所述LFR4的N末端与所述LCDR3的C末端直接或间接相连;其中所述LFR1包含SEQ ID NO:112所示的氨基酸序列,所述LFR2包含SEQ ID NO:124所示的氨基酸序列,所述LFR3包含SEQ ID NO:131所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:113所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:132所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:133所示的氨基酸序列,所述LFR4 包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:115所示的氨基酸序列,所述LFR2包含SEQ ID NO:127所示的氨基酸序列,所述LFR3包含SEQ ID NO:134所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:116所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:135所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:117所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:136所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:118所示的氨基酸序列,所述LFR2包含SEQ ID NO:128所示的氨基酸序列,所述LFR3包含SEQ ID NO:137所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:119所示的氨基酸序列,所述LFR2包含SEQ ID NO:125所示的氨基酸序列,所述LFR3包含SEQ ID NO:138所示的氨基酸序列,所述LFR4包含SEQ ID NO:143所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:120所示的氨基酸序列,所述LFR2包含SEQ ID NO:129所示的氨基酸序列,所述LFR3包含SEQ ID NO:139所示的氨基酸序列,所述LFR4包含SEQ ID NO:142所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:114所示的氨基酸序列,所述LFR2包含SEQ ID NO:126所示的氨基酸序列,所述LFR3包含SEQ ID NO:140所示的氨基酸序列,所述LFR4包含SEQ ID NO:144所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:121所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:145所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:122所示的氨基酸序列,所述LFR2包含SEQ ID NO:130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:146所示的氨基酸序列;或
    所述LFR1包含SEQ ID NO:123所示的氨基酸序列,所述LFR2包含SEQ ID NO: 130所示的氨基酸序列,所述LFR3包含SEQ ID NO:141所示的氨基酸序列,所述LFR4包含SEQ ID NO:147所示的氨基酸序列。
  22. 根据权利要求1-21中任一项所述的分离的抗原结合蛋白,其包含VL,其中所述VL包含SEQ ID NO:148至SEQ ID NO:160中任一项所示的氨基酸序列。
  23. 根据权利要求1-22中任一项所述的分离的抗原结合蛋白,其包含VH和VL,其中所述VH包含SEQ ID NO:61所示的氨基酸序列,所述VL包含SEQ ID NO:148所示的氨基酸序列;或
    所述VH包含SEQ ID NO:62所示的氨基酸序列,所述VL包含SEQ ID NO:149所示的氨基酸序列;或
    所述VH包含SEQ ID NO:63所示的氨基酸序列,所述VL包含SEQ ID NO:150所示的氨基酸序列;或
    所述VH包含SEQ ID NO:64所示的氨基酸序列,所述VL包含SEQ ID NO:151所示的氨基酸序列;或
    所述VH包含SEQ ID NO:65所示的氨基酸序列,所述VL包含SEQ ID NO:152所示的氨基酸序列;或
    所述VH包含SEQ ID NO:66所示的氨基酸序列,所述VL包含SEQ ID NO:153所示的氨基酸序列;或
    所述VH包含SEQ ID NO:67所示的氨基酸序列,所述VL包含SEQ ID NO:154所示的氨基酸序列;或
    所述VH包含SEQ ID NO:68所示的氨基酸序列,所述VL包含SEQ ID NO:155所示的氨基酸序列;或
    所述VH包含SEQ ID NO:69所示的氨基酸序列,所述VL包含SEQ ID NO:156所示的氨基酸序列;或
    所述VH包含SEQ ID NO:70所示的氨基酸序列,所述VL包含SEQ ID NO:157所示的氨基酸序列;或
    所述VH包含SEQ ID NO:71所示的氨基酸序列,所述VL包含SEQ ID NO:158所示的氨基酸序列;或
    所述VH包含SEQ ID NO:72所示的氨基酸序列,所述VL包含SEQ ID NO:159所示的氨基酸序列;或
    所述VH包含SEQ ID NO:73所示的氨基酸序列,所述VL包含SEQ ID NO:160所示的氨基酸序列。
  24. 根据权利要求1-23中任一项所述的分离的抗原结合蛋白,其包括抗体或其抗原结合片段。
  25. 根据权利要求1-24中任一项所述的分离的抗原结合蛋白,所述抗体包括单克隆抗体、多克隆抗体、二聚体、多聚体、多特异性抗体、完整抗体、抗体片段、人抗体、人源化抗体或嵌合抗体。
  26. 根据权利要求1-25中任一项所述的分离的抗原结合蛋白,其中所述抗原结合片段包括Fab,Fab’,Fv片段,F(ab’)2,scFv,di-scFv和/或dAb。
  27. 根据权利要求1-26所述的抗原结合蛋白,其包括scFv。
  28. 根据权利要求1-27所述的抗原结合蛋白,其中所述VL和VH通过接头连接。
  29. 根据权利要求28所述的抗原结合蛋白,其中所述接头包括多肽接头。
  30. 根据权利要求29所述的抗原结合蛋白,其中所述多肽接头包含(GGGGS)n所示的氨基酸序列,其中n为1至5的任意整数。
  31. 根据权利要求1-30中任一项所述的分离的抗原结合蛋白,其包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
  32. 分离的多肽,其包含权利要求1-31中任一项所述的分离的抗原结合蛋白。
  33. 免疫缀合物,其包含权利要求1-31中任一项所述的抗原结合蛋白。
  34. 嵌合抗原受体,其包含至少一个包含权利要求1-31中任一项的抗原结合蛋白的细胞外抗原结合结构域。
  35. 根据权利要求34所述的嵌合抗原受体,其中所述细胞外抗原结合结构域包括scFv。
  36. 根据权利要求1-35中任一项所述的嵌合抗原受体,其包括跨膜域,所述跨膜域包含源自选自下组中的一种或多种蛋白的跨膜域:CD8、CD28、CD3ε(CD3e)、4-1BB、CD4、CD27、CD7、PD-1、TRAC、TRBC、CD3ζ、CTLA-4、LAG-3、CD5、ICOS、OX40、NKG2D、2B4(CD244)、FcεRIγ、BTLA、CD30、GITR、HVEM、DAP10、CD2、NKG2C、LIGHT、DAP12,CD40L(CD154)、TIM1、CD226、DR3、CD45、CD80、CD86、CD9、CD16、CD22、CD33、CD37、CD64和SLAM。
  37. 根据权利要求36所述的嵌合抗原受体,其中所述跨膜域包含源自CD8的跨膜域。
  38. 根据权利要求34-37中任一项所述的嵌合抗原受体,其中所述跨膜域包含SEQ ID  NO:177所示的氨基酸序列。
  39. 根据权利要求34-38中任一项所述的嵌合抗原受体,其包括胞内共刺激信号传导结构域,所述胞内共刺激信号传导结构域包含源自选自下组中的一种或多种蛋白的胞内共刺激信号传导结构域:CD28、CD137、CD27、CD2、CD7、CD8、OX40、CD226、DR3、SLAM、CDS、ICAM-1、NKG2D、NKG2C、B7-H3、2B4、FcεRIγ、BTLA、GITR、HVEM、DAP10、DAP12、CD30、CD40、CD40L、TIM1、PD-1、LFA-1、LIGHT、JAML、CD244、CD100、ICOS、CD40和MyD88。
  40. 根据权利要求39所述的嵌合抗原受体,其中所述胞内共刺激信号传导结构域源自4-1BB的共刺激信号传导结构域。
  41. 根据权利要求39-40中任一项所述的嵌合抗原受体,其中所述胞内共刺激信号传导结构域包含SEQ ID NO:178中任一项所示的氨基酸序列。
  42. 根据权利要求34-41中任一项所述的嵌合抗原受体,其包括胞内信号转导结构域,所述胞内信号转导结构域包含源自选自下组中的一种或多种蛋白的胞内信号转导结构域:CD3ζ、CD3δ、CD3γ、CD3ε、CD79a、CD79b、FceRIγ、FceRIβ、FcγRIIa、牛白血病病毒gp30、Epstein-Barr病毒(EBV)LMP2A、猿免疫缺陷病毒PBj14 Nef、DAP10、DAP-12和至少包含一个ITAM的结构域。
  43. 根据权利要求42所述的嵌合抗原受体,其中所述胞内信号转导结构域包含源自CD3ζ的信号传导结构域。
  44. 根据权利要求42-43中任一项所述的嵌合抗原受体,其中所述胞内信号转导结构域包含SEQ ID NO:179所示的氨基酸序列。
  45. 根据权利要求34-44中任一项所述的嵌合抗原受体,其在细胞外抗原结合结构域和跨膜域之间包括铰链区,所述铰链区包含源自选自下组中的一种或多种蛋白的铰链区:CD28、IgG1、IgG4、IgD、4-1BB、CD4、CD27、CD7、CD8、PD-1、ICOS、OX40、NKG2D、NKG2C、FcεRIγ、BTLA、GITR、DAP10、TIM1、SLAM、CD30和LIGHT。
  46. 根据权利要求45所述的嵌合抗原受体,所述铰链区包含源自CD8的铰链区。
  47. 根据权利要求45-46中任一项所述的嵌合抗原受体,所述铰链区包含SEQ ID NO:176所示的氨基酸序列。
  48. 根据权利要求34-47中任一项所述的嵌合抗原受体,所述嵌合抗原受体的非靶向部分包含跨膜域、铰链区、胞内共刺激信号传导结构域和胞内信号传导结构域。
  49. 根据权利要求34-48中任一项所述的嵌合抗原受体,所述嵌合抗原受体的非靶向部分包含CD8分子跨膜域、CD8的铰链区、4-1BB的胞内共刺激信号传导结构域和CD3ζ胞内信号传导结构域。
  50. 根据权利要求34-49中任一项所述的嵌合抗原受体,其还包含信号肽片段,所述信号肽片段的C端与所述细胞外抗原结合结构域的N端连接。
  51. 根据权利要求50所述的嵌合抗原受体,所述信号肽片段包括CD8信号肽片段。
  52. 根据权利要求中50-51中任一项所述的嵌合抗原受体,所述信号肽片段包含如SEQ ID NO:175所示的氨基酸序列。
  53. 根据权利要求34-52中任一项所述的嵌合抗原受体,其包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
  54. 分离的核酸分子,其编码权利要求1-31中任一项所述的分离的抗原结合蛋白、权利要求32所述的多肽或权利要求34-53中任一项所述的嵌合抗原受体。
  55. 表达载体,其包含根据权利要求54所述的核酸分子。
  56. 细胞,所述细胞包含根据权利要求54所述的核酸分子或根据权利要求55所述的表达载体,和/或ii)所述细胞表达根据权利要求1-31中任一项所述的抗原结合蛋白、权利要求32所述的多肽或权利要求34-53中任一项所述的嵌合抗原受体。
  57. 制备权利要求1-31中任一项所述的分离的抗原结合蛋白的方法,所述方法包括在使得权利要求1-31中任一项所述的分离的抗原结合蛋白表达的条件下,培养根据权利要求56所述的细胞。
  58. 工程化的细胞,其包含权利要求1-31中任一项所述的抗原结合蛋白,权利要求32所述的多肽,权利要求34-53中任一项所述的嵌合抗原受体,权利要求54所述的核酸分子,和/或表达权利要求55中任一项所述的载体。
  59. 根据权利要求58所述的工程化的细胞,所述细胞包括免疫效应细胞。
  60. 根据权利要求59所述的工程化的细胞,所述的免疫效应细胞包括人细胞。
  61. 根据权利要求59-60中任一项所述的工程化的细胞,所述免疫效应细胞包括T细胞、B细胞、天然杀伤细胞(NK细胞)、巨噬细胞、NKT细胞、单核细胞、树突状细胞、粒细胞、淋巴细胞、白细胞和/或外周血单个核细胞。
  62. 根据权利要求59-61中任一项所述的工程化的细胞,所述免疫效应细胞包括自体或同种异体的免疫效应细胞。
  63. 根据权利要求59-62中任一项所述的工程化的细胞,其中所述免疫效应细胞包括同种异体T细胞或自体T细胞。
  64. 根据权利要求59-63中任一项所述的工程化的细胞,所述的免疫效应细胞包括经修饰的免疫效应细胞。
  65. 根据权利要求64所述的工程化的细胞,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
  66. 根据权利要求64-65中任一项所述的工程化的细胞,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
  67. 根据权利要求59-66中任一项所述的工程化的细胞,所述免疫效应细胞包括CAR-T细胞。
  68. 根据权利要求67所述的工程化的细胞,所述CAR-T细胞不诱导自相残杀。
  69. 一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
    a.对T细胞群中的CD7基因进行修饰;
    b.激活所述T细胞群;
    c.用权利要求34-53中任一项所述的嵌合抗原受体转导所述T细胞;以及
    d.扩增所述嵌合抗原受体T细胞。
  70. 根据权利要求69所述的方法,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
  71. 根据权利要求69-70中任一项所述的方法,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
  72. 根据权利要求69-71中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
  73. 根据权利要求69-72中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
  74. 根据权利要求69-73中任一项所述的方法,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
  75. 根据权利要求74所述的方法,其中所述靶向CD7基因的gRNA包含SEQ ID NO: 211至SEQ ID NO:218中任一项所示的核苷酸序列。
  76. 根据权利要求74所述的方法,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
  77. 根据权利要求74所述的方法,其中所述gRNA与所述Cas9同时递送。
  78. 根据权利要求77所述的方法,其中所述递送是通过电穿孔进行的。
  79. 根据权利要求69-78所述的方法,其中所述转导嵌合抗原受体包括向转导circRNA嵌合抗原受体。
  80. 根据权利要求79所述的方法,其中所述cirRNA嵌合抗原受体按以下顺序包含内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA)。
  81. 药物组合物,其包含权利要求1-31中任一项所述的分离的抗原结合蛋白、权利要求32所述的多肽、权利要求33所述的免疫缀合物、权利要求54所述的核酸分子、权利要求55所述的表达载体、权利要求56所述的细胞、权利要求34-53中任一项所述的嵌合抗原受体和/或权利要求58-68中任一项所述的工程化细胞,以及任选地药学上可接受的载体。
  82. 试剂盒,其包含权利要求1-31中任一项所述的分离的抗原结合蛋白、权利要求32所述的多肽、权利要求33所述的免疫缀合物、权利要求54所述的核酸分子、权利要求55所述的表达载体、权利要求56所述的细胞、权利要求34-53中任一项所述的嵌合抗原受体、权利要求58-68中任一项所述的工程化细胞、或权利要求79所述的药物组合物。
  83. 权利要求1-31中任一项所述的分离的抗原结合蛋白、权利要求32所述的多肽、权利要求33所述的免疫缀合物、权利要求54所述的核酸分子、权利要求55所述的表达载体、权利要求56所述的细胞、权利要求34-53中任一项所述的嵌合抗原受体、权利要求58-68中任一项所述的工程化细胞、或权利要求81所述的药物组合物在制备药物中的用途,所述药物用于预防和/或治疗CD7相关的疾病或病症。
  84. 根据权利要求83所述的用途,其中所述CD7相关的疾病或病症包括肿瘤。
  85. 根据权利要求84所述的用途,其中所述肿瘤包括表达CD7的肿瘤。
  86. 根据权利要求84-85中任一项所述的用途,其中所述肿瘤包括血液瘤。
  87. 根据权利要求84-86中任一项所述的用途,其中所述肿瘤包括CD7阳性的恶性血液瘤。
  88. 根据权利要求84-87中任一项所述的用途,其中所述肿瘤包括T细胞恶性肿瘤。
  89. 一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的权利要求1-31中任一项所述的分离的抗原结合蛋白、权利要求32所述的多肽、权利要求33所述的免疫缀合物、权利要求34-53中任一项所述的嵌合抗原受体、权利要求54所述的核酸分子、权利要求55所述的表达载体、权利要求56所述的细胞、权利要求58-68中任一项所述的工程化细胞、或权利要求81所述的药物组合物。
  90. 根据权利要求89所述的方法,其中所述肿瘤包括表达CD7的肿瘤。
  91. 根据权利要求89所述的方法,其中所述肿瘤包括血液瘤。
  92. 根据权利要求89所述的方法,其中所述肿瘤包括CD7阳性的恶性血液瘤。
  93. 根据权利要求89所述的方法,其中所述肿瘤包括T细胞恶性肿瘤。
  94. 根据权利要求93所述的方法,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
  95. 一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与权利要求58-68中任一项所述的工程化的细胞接触。
  96. 经修饰的免疫效应细胞,其包含编码融合蛋白的核酸分子,所述融合蛋白包含激活抗原呈递细胞(APC)的第一结构域和激活免疫效应细胞的第二结构域,其中(i)第一结构域包括(a)结合APC的激活受体配体或其受体结合片段,或(b)结合APC的激活受体抗体或其抗原结合片段;和(ii)第二结构域包括(a)免疫效应细胞的共刺激配体或其受体结合片段,(b)结合免疫效应细胞的共刺激受体的抗体或其抗原结合片段,或(c)免疫效应细胞的共刺激受体或其功能片段,所述免疫效应细胞包括权利要求59-67中任一项所述的免疫效应细胞。
  97. 根据权利要求96所述的经修饰的免疫效应细胞,其中所述第一结构域的N端与第二结构域的C端连接。
  98. 根据权利要求96所述的经修饰的免疫效应细胞,其中所述第一结构域的C端与第二结构域的N端连接。
  99. 根据权利要求96-98中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域与第二结构域直接连接或间接连接。
  100. 根据权利要求96-99中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域与第二结构域通过连接子连接。
  101. 根据权利要求100所述的经修饰的免疫效应细胞,其中所述连接子包括肽连接子。
  102. 根据权利要求96-101中任一项所述的经修饰的免疫效应细胞,其中所述抗体或其抗原结合片段为scFv。
  103. 根据权利要求96-102中任一项所述的经修饰的免疫效应细胞,其中所述APC选自由树突细胞、巨噬细胞、髓源性抑制细胞、单核细胞、B细胞、T细胞和朗格汉斯细胞组成的组。
  104. 根据权利要求96-103中任一项所述的经修饰的免疫效应细胞,其中所述APC的激活受体选自CD40、CD80、CD86、CD91、DEC-205和DC-SIGN。
  105. 根据权利要求96-142中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含结合CD40、CD80、CD86、CD91、DEC-205、DC-SIGN的配体或它们的受体结合片段。
  106. 根据权利要求96-105中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含CD40配体(CD40L)的受体结合片段。
  107. 根据权利要求96-106中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域包含结合所述APC的激活受体的抗体或其抗原结合片段。
  108. 根据权利要求96-107中任一项所述的经修饰的免疫效应细胞,其中所述第一结结构域为抗CD40抗体或其抗原结合片段。
  109. 根据权利要求96-108中任一项所述的经修饰的免疫效应细胞,其中所述免疫效应细胞选自由T细胞、NK细胞、NKT细胞、巨噬细胞、嗜中性粒细胞和粒细胞组成的组。
  110. 根据权利要求96-109中任一项所述的经修饰的免疫效应细胞,其中第二结构域包含共刺激受体的胞内结构域。
  111. 根据权利要求96-110中任一项所述的经修饰的免疫效应细胞,其中所述共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
  112. 根据权利要求96-111中任一项所述的经修饰的免疫效应细胞,其中所述共刺激受体为CD28。
  113. 根据权利要求96-112中任一项所述的经修饰的免疫效应细胞,其中所述第二 结构域是免疫效应细胞的共刺激配体或其受体结合片段。
  114. 根据权利要求96-113中任一项所述的经修饰的免疫效应细胞,其中共刺激配体选自CD58、CD70、CD83、CD80、CD86、CD137L、CD252、CD275、CD54、CD49a、CD112、CD150、CD155、CD265、CD270、TL1A、CD127、IL-4R、GITR-L、TIM-4、CD153、CD48、CD160、CD200R和CD44。
  115. 根据权利要求96-114中任一项所述的经修饰的免疫效应细胞,其中第二结构域是结合共刺激受体的抗体或其抗原结合片段。
  116. 根据权利要求96-115中任一项所述的经修饰的免疫效应细胞,其中共刺激受体选自CD28、4-1BB、ICOS、CD27、OX40、DAP10、2B4、CD30、CD2、LIGHT、GITR、TLR、DR3和CD43。
  117. 根据权利要求116所述的经修饰的免疫效应细胞,其中共刺激受体为CD28或4-1BB。
  118. 根据权利要求96-117中任一项所述的经修饰的免疫效应细胞,其中所述第一结构域与第二结构域选自以下组合:
    i)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28或4-1BB胞内结构域;
    ii)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
    iii)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28胞内结构域;
    iv)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含抗CD28抗体或其抗原结合片段;
    v)所述第一结构域包含CD40L或其受体结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段;和
    vi)所述第一结构域包含抗CD40抗体或其抗原结合片段,且第二结构域包含CD28共刺激配体或其受体结合片段。
  119. 根据权利要求96-118中任一项所述的经修饰的免疫效应细胞,其中所述融合蛋白包含SEQ ID NO:234至SEQ ID NO:250中任一项所示的氨基酸序列。
  120. 根据权利要求96-119中任一项所述的经修饰的免疫效应细胞,其中所述免疫 效应细胞包括工程化的免疫效应细胞。
  121. 根据权利要求120所述的经修饰的免疫效应细胞,其中所述工程化的免疫效应细胞包括CAR-T细胞、CAR-NK或TCR-T细胞。
  122. 根据权利要求96-121中任一项所述的经修饰的免疫效应细胞,与相应的免疫细胞相比,其中所述经修饰的免疫效应细胞CD7表面表达降低,并表达抗CD7 CAR。
  123. 根据权利要求122所述的经修饰的免疫效应细胞,其中所述工程化的免疫效应细胞共表达CD7 CAR。
  124. 根据权利要求123所述的经修饰的免疫效应细胞,其中所述CD7 CAR包含细胞外抗原结合结构域,所述细胞外抗原结合结构域包含SEQ ID NO:162至SEQ ID NO:174中任一项所示的氨基酸序列。
  125. 根据权利要求124所述的经修饰的免疫效应细胞,其中所述CAR包含SEQ ID NO:180至SEQ ID NO:192中任一项所示的氨基酸序列。
  126. 根据权利要求96-125中任一项所述的经修饰的免疫效应细胞,其中所述经修饰的免疫效应细胞的CD7的表达缺失或受抑制。
  127. 根据权利要求96-126中任一项所述的经修饰的免疫效应细胞,所述经修饰的免疫效应细胞不诱导自相残杀。
  128. 药物组合物,其包含权利要求96-127中任一项所述的经修饰的免疫效应细胞,以及药学上可接受的载体。
  129. 权利要求96-127中任一项所述的经修饰的免疫效应细胞,权利要求128所述的药物组合物在制备药物中的用途,所述药物用于治疗肿瘤。
  130. 根据权利要求129所述的用途,其中所述肿瘤包括血液瘤和实体瘤。
  131. 根据权利要求129-130中任一项所述的用途,其中所述肿瘤包括表达CD7的肿瘤。
  132. 根据权利要求129-131中任一项所述的用途,其中所述肿瘤包括CD7阳性的恶性血液瘤。
  133. 根据权利要求129-132中任一项所述的用途,其中所述肿瘤包括T细胞恶性肿瘤。
  134. 根据权利要求129-133中任一项所述的用途,其中所述T细胞恶性肿瘤包括急性T淋巴细胞白血病(T-ALL)、急性髓细胞白血病或NK/T细胞淋巴瘤。
  135. 一种治疗肿瘤的方法,所述方法包括向有此需要的受试者施用有效量的权利要求96-127中任一项所述的经修饰的免疫效应细胞、权利要求128所述的药物组合物。
  136. 一种杀伤恶性T细胞的方法,所述方法包括将所述恶性T细胞与权利要求96-127中任一项所述的经修饰的免疫效应细胞、权利要求128所述的药物组合物接触。
  137. 一种制备嵌合抗原受体T(CAR-T)细胞群的方法,其中所述CAR靶向CD7,其包括以下步骤:
    a.对T细胞群中的CD7基因进行修饰;
    b.激活所述T细胞群;
    c.将权利要求96-127中任一项所述的嵌合抗原受体和融合蛋白转导入所述T细胞群;以及
    d.扩增所述嵌合抗原受体T细胞群。
  138. 根据权利要求137所述的方法,与未经修饰的相应细胞相比,经修饰的所述T细胞群中CD7缺失或受抑制。
  139. 根据权利要求137-138中任一项所述的方法,其中所述修饰包括向所述T细胞群施用一种或多种选自下组的物质:反义RNA、siRNA、shRNA、转录激活因子样效应物核酸酶(TALEN)、锌指核酸酶(ZFN)和CRISPR/Cas系统。
  140. 根据权利要求137-139中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas系统。
  141. 根据权利要求137-140中任一项所述的方法,其中所述修饰包括向所述T细胞群施用CRISPR/Cas9系统。
  142. 根据权利要求141所述的方法,其中所述修饰包括向所述T细胞群施用Cas9和靶向CD7基因的gRNA。
  143. 根据权利要求142所述的经修饰的方法,其中所述靶向所述CD7基因的gRNA包含SEQ ID NO:211至SEQ ID NO:218中任一项所示的核苷酸序列。
  144. 根据权利要求143所述的方法,其中所述Cas9作为mRNA或蛋白质递送到所述细胞中。
  145. 根据权利要求144所述的方法,其中所述gRNA与所述Cas9同时递送。
  146. 根据权利要求145所述的方法,其中所述递送是通过电穿孔进行的。
  147. 根据权利要求137-146中任一项所述的方法,其中所述将抗CD7 CAR和融 合蛋白转导入所述T细胞包括将编码所述抗CD7 CAR的核酸分子和编码所述融合蛋白的核酸分子引入T细胞。
  148. 根据权利要求147所述的方法,其中所述CD7 CAR核酸分子和所述融合蛋白的核酸分子为mRNA。
  149. 根据权利要求147-148任一项所述的方法,其中所述CD7 CAR核酸分子和所述融合蛋白的核酸分子为circRNA。
  150. 根据权利要求147-149任一项所述的方法,其中所述cirRNA嵌合抗原受体包含按以下顺序排列的元件:内部核糖体进入位点(IRES)元件、靶向CD7的蛋白质编码序列和聚腺苷酸(polyA),circRNA融合蛋白包含按以下顺序排列的元件:内部核糖体进入位点(IRES)元件、融合蛋白编码序列和聚腺苷酸(polyA)。
  151. 包含权利要求96-127中任一项所述的经修饰的免疫效应细胞,其中所述细胞群来源于外周血单核细胞(PBMC)、外周血血液白细胞(PBL)、肿瘤浸润淋巴细胞(TIL)、细胞因子诱导的杀伤细胞(CIK)、淋巴因子激活的杀伤细胞(LAK)或骨髓浸润淋巴细胞(MIL)。
PCT/CN2023/076567 2022-02-17 2023-02-16 经修饰的免疫效应细胞及其用途 WO2023155852A1 (zh)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN2022076628 2022-02-17
CNPCT/CN2022/076628 2022-02-17
CNPCT/CN2022/076635 2022-02-17
CN2022076635 2022-02-17
CN2022076634 2022-02-17
CNPCT/CN2022/076634 2022-02-17

Publications (1)

Publication Number Publication Date
WO2023155852A1 true WO2023155852A1 (zh) 2023-08-24

Family

ID=87577589

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/076567 WO2023155852A1 (zh) 2022-02-17 2023-02-16 经修饰的免疫效应细胞及其用途

Country Status (1)

Country Link
WO (1) WO2023155852A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051926A2 (en) * 2001-12-14 2003-06-26 Friedrich-Alexander-Universitaet Erlangen-Nuernberg Anti-cd7 immunotoxin as fusion protein
US20180148506A1 (en) * 2016-11-22 2018-05-31 National University Of Singapore Blockade of cd7 expression and chimeric antigen receptors for immunotherapy of t-cell malignancies
CN109652379A (zh) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 Cd7嵌合抗原受体修饰的nk-92mi细胞及其应用
US20200055948A1 (en) * 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
CN112300282A (zh) * 2020-11-03 2021-02-02 南京北恒生物科技有限公司 靶向cd7的人源化抗体及其用途

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051926A2 (en) * 2001-12-14 2003-06-26 Friedrich-Alexander-Universitaet Erlangen-Nuernberg Anti-cd7 immunotoxin as fusion protein
US20180148506A1 (en) * 2016-11-22 2018-05-31 National University Of Singapore Blockade of cd7 expression and chimeric antigen receptors for immunotherapy of t-cell malignancies
US20200055948A1 (en) * 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
CN109652379A (zh) * 2018-12-29 2019-04-19 博生吉医药科技(苏州)有限公司 Cd7嵌合抗原受体修饰的nk-92mi细胞及其应用
CN112300282A (zh) * 2020-11-03 2021-02-02 南京北恒生物科技有限公司 靶向cd7的人源化抗体及其用途

Similar Documents

Publication Publication Date Title
JP2022116230A (ja) 免疫療法用改変細胞
JP2021040652A (ja) Gタンパク質共役受容体を標的化するキメラ抗原受容体およびその使用
JP2022105192A (ja) B細胞成熟抗原を標的化するキメラ抗原受容体およびその使用
KR20200130709A (ko) 전립선-특이 막 항원 car 및 이의 사용 방법
KR20170057298A (ko) Cd19에 특이적인 항체 및 키메라 항원 수용체
WO2017165683A1 (en) Cell secreted minibodies and uses thereof
CN113412117A (zh) 嵌合抗原和t细胞受体及使用的方法
CN111511383A (zh) 多功能免疫细胞疗法
JP2021524268A (ja) Pd−l1及びcd137に結合する抗体分子
KR102412805B1 (ko) 세포 면역 요법을 위한 조성물 및 방법
US20230149460A1 (en) Methods for generating engineered memory-like nk cells and compositions thereof
US20230183342A1 (en) Antibodies to nkp46 and constructs thereof for treatment of cancers and infections
BR112021000397A2 (pt) Fragmentos de ligação fc que compreendem um sítio de ligação a antígeno cd137
JP2024510739A (ja) 新規な細胞治療システム
CA3177488A1 (en) Compositions and methods for tcr reprogramming using cd70 specific fusion proteins
WO2018132508A1 (en) Methods and compositions relating to ex vivo culture and modulation of t cells
WO2023155852A1 (zh) 经修饰的免疫效应细胞及其用途
CN115322257A (zh) Bcma靶向抗体、嵌合抗原受体及其应用
CA3228546A1 (en) Antigen recognizing receptors targeting cd33 and uses thereof
KR20220068232A (ko) Cd371을 표적화하는 항원 인식 수용체 및 그의 용도
WO2023180511A1 (en) Improved chimeric receptors
KR20210148823A (ko) Cd22에 특이적인 항체 및 이의 용도
CN115335123A (zh) 人4-1bb激动剂抗体及其使用方法
EP3820901A1 (en) Two chimeric antigen receptors specifically binding cd19 and igkappa

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: 23755853

Country of ref document: EP

Kind code of ref document: A1