WO2018233624A1 - 抗Her2纳米抗体及其编码序列和用途 - Google Patents

抗Her2纳米抗体及其编码序列和用途 Download PDF

Info

Publication number
WO2018233624A1
WO2018233624A1 PCT/CN2018/091953 CN2018091953W WO2018233624A1 WO 2018233624 A1 WO2018233624 A1 WO 2018233624A1 CN 2018091953 W CN2018091953 W CN 2018091953W WO 2018233624 A1 WO2018233624 A1 WO 2018233624A1
Authority
WO
WIPO (PCT)
Prior art keywords
her2
nanobody
antibody
immunoconjugate
protein
Prior art date
Application number
PCT/CN2018/091953
Other languages
English (en)
French (fr)
Inventor
丁航海
黄仲廉
黄文华
Original Assignee
和迈生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 和迈生物科技有限公司 filed Critical 和迈生物科技有限公司
Priority to US16/624,403 priority Critical patent/US11517632B2/en
Priority to JP2020520703A priority patent/JP7097636B2/ja
Priority to EP18819877.4A priority patent/EP3643726A4/en
Publication of WO2018233624A1 publication Critical patent/WO2018233624A1/zh
Priority to US18/047,327 priority patent/US20230263916A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1051Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from breast, e.g. the antibody being herceptin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6845Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a cytokine, e.g. growth factors, VEGF, TNF, a lymphokine or an interferon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to the field of biomedical or biopharmaceutical technology, and more particularly to Nanobodies against Her2 and their coding sequences and uses.
  • Her2/ErbB2 Human epidermal growth factor receptor-2
  • Her2/Neu Her2/Neu, ErbB-2, CD340 or p185
  • Her2 is a tyrosine kinase receptor (RTK) belonging to the epidermal growth factor receptor (EGFR/ErbB) family, which consists of 1255 amino acids, including four extracellular domains (I, II, III and IV), one The transmembrane region, a domain having tyrosine kinase activity and a carboxy terminal tail comprising a tyrosine residue and an anchor point of an intracellular signaling molecule, having a molecular weight of about 185 kD.
  • RTK tyrosine kinase receptor
  • EGFR family Members of the EGFR family have a similar structure in which extracellular domains I and III are involved in receptor-ligand binding, causing receptor conformational changes to cause receptor activation, and extracellular domains II and IV involved in receptor dimerization.
  • Her2 has a special open structure, which can activate itself without the participation of specific ligands, form homodimers or form heterodimers with other receptors of the EGFR family, and is a heterologous member of the family. The preferred molecule for polymerization.
  • Her2 is overexpressed in a variety of epithelial-derived malignancies such as breast cancer, ovarian cancer, prostate cancer, gastric cancer, and lung cancer, but the expression level is low or not expressed in normal tissues.
  • the receptor protein When the expression level of Her2 is low, the receptor protein is generally present in the form of a monomer, and its tyrosine kinase activity is low; when Her2 is overexpressed, it can lead to a long-lasting and enhanced activation of the receptor tyrosine kinase, resulting in A series of downstream cascades that activate mitogen-activated protein kinase (MAPK), phosphatidylinositol-3-kinase-protein kinase B/Akt (PI3K-PKB/Akt), phospholipase C-protein kinase C (PLC) -PKC) and multiple major signaling pathways such as transcriptional signal transduction and activated protein (STAT).
  • MAPK mitogen-activated protein
  • Her2-mediated signaling pathway regulates the expression of tumor-associated genes, such as vascular endothelial growth factor, urokinase-type plasminogen activator, cyclooxygenase-2, chemotactic cytokine receptor CXCR-4, etc.
  • tumor-associated genes such as vascular endothelial growth factor, urokinase-type plasminogen activator, cyclooxygenase-2, chemotactic cytokine receptor CXCR-4, etc.
  • Expression, down-regulation of MMP inhibitory factor RECK expression can promote tumor invasion and metastasis.
  • Amplification or overexpression of Her2 plays an important role in the pathogenesis and development of certain invasive breast cancers, and it has become an important marker and therapeutic target for breast cancer.
  • Trastuzumab and Pertuzumab are traditional targeted therapies for breast cancer patients with Her2/neu gene amplification and overexpression.
  • Trastuzumab binds to the membrane proximal region of Her2 extracellular structure, undergoes endocytosis into the nucleus of tumor cells, blocks the process of Her2 recycling to the membrane, accelerates the degradation of Her2 protein, and inhibits Her2 to tumor cells. Transduction of a malignant phenotype. It is very effective for patients with primary invasive breast cancer whose Her2 is overexpressed.
  • Pertuzumab The antigen binding site of Pertuzumab is located in the II subregion of the extracellular domain of Her2, which blocks the cell signaling by interfering with Her2 to form a dimer with other ErbB members, and it can exert anti-tumor effects regardless of the presence or absence of Her2 overexpression.
  • Nanobodies are currently the smallest antibody molecules with a molecular weight of 1/10 of that of a normal antibody.
  • nano-antibodies possess some unique functional properties, such as small molecular mass, strong stability, good solubility, easy expression, weak immunogenicity, strong penetrability and strong targeting. It is simple in humanization, low in preparation cost, etc. It almost completely overcomes the defects of long development cycle, low stability and harsh storage conditions of traditional antibodies.
  • a radioisotope targeting carrier it can quickly and specifically penetrate the tumor tissue to bind the target, and the non-binding antibody can be quickly cleared from the blood, reducing the body radiation dose, compared with traditional antibodies as a tracer and Targeted internal radiotherapy drugs have many obvious advantages.
  • VHH chain of an anti-Her2 Nanobody the amino acid sequence of which is set forth in any one of SEQ ID NO.: 1-40.
  • amino acid sequence of the VHH chain is SEQ ID NO.: 8, 7, 15, 12, 27, 11, 32, 13, 14, 9, 21, 30, 17, 24, 16, 6, 28, 25, 10, 1 is shown.
  • amino acid sequence of the VHH chain is as set forth in any one of SEQ ID NO.: 8, 7, 15, 12, 27, 11, 32, 13.
  • amino acid sequence of the VHH chain is as shown in any one of SEQ ID NO.: 9, 10, 13, 17, 22, 23, 26.
  • the Her2 is human Her2.
  • VHH chain of an anti-Her2 Nanobody comprising a framework region FR and a complementarity determining region CDR, wherein the CDR comprises a corresponding one of the sequences of any one of SEQ ID NO.: 1-40 CDR1, CDR2 and CDR3, and FR1, FR2, FR3 and FR4 separated by said CDR1-3.
  • a heavy chain variable region of an anti-human Her2 antibody comprising three complementarity determining regions CDR1, CDR2, and CDR3, and three CDRs comprising SEQ ID NO.: Corresponding CDR1, CDR2 and CDR3 in any of the 40 sequences.
  • a CDR region of a complementarity determining region of an anti-human Her2 antibody comprising CDR1, CDR2 and CDR3 underlined in the amino acid sequence set forth in SEQ ID NO.: 1-40 is also provided. (The three underlined portions of each VHH amino acid sequence represent CDR1, CDR2 and CDR3 in turn).
  • an anti-Her2 Nanobody which is a Nanobody directed against a Her2 epitope and which has a VHH chain of the amino acid sequence set forth in any one of SEQ ID NO.: 1-40.
  • a preferred anti-Her2 Nanobody has the amino acid sequence of the VHH chain preferred in the first aspect.
  • a polynucleotide is provided, the polynucleotide encoding a protein selected from the group consisting of the VHH chain of the anti-Her2 Nanobody of the first aspect, or the second aspect Anti-Her2 Nanobody.
  • the polynucleotide comprises DNA or RNA.
  • the polynucleotide has a nucleotide sequence as set forth in any one of SEQ ID NO.: 41-80.
  • an expression vector comprising the polynucleotide of the third aspect is provided.
  • the expression vector is selected from the group consisting of DNA, RNA, viral vectors, plasmids, transposons, other gene transfer systems, or a combination thereof.
  • the expression vector comprises a viral vector, such as a lentivirus, an adenovirus, an AAV virus, a retrovirus, or a combination thereof.
  • a host cell comprising the expression vector of the fourth aspect, or a polynucleotide thereof according to the third aspect, is integrated in the genome.
  • the host cell comprises a prokaryotic cell or a eukaryotic cell.
  • the host cell is selected from the group consisting of E. coli, yeast cells.
  • a method for producing an anti-Her2 Nanobody comprising the steps of:
  • the anti-Her2 Nanobody has the amino acid sequence set forth in SEQ ID NO.: 1-40.
  • an immunoconjugate comprising:
  • a coupling moiety selected from the group consisting of a detectable label, a drug, a toxin, a cytokine, a radionuclide, or an enzyme, a gold nanoparticle/nanorod, a nanomagnetic particle, a viral coat protein or a VLP, or combination.
  • the radionuclide comprises:
  • diagnostic isotope the diagnostic isotope being selected from the group consisting of Tc-99m, Ga-68, F-18, I-123, I-125, I-131, In-111, Ga-67, Cu-64, Zr-89, C-11, Lu-177, Re-188, or a combination thereof; and/or
  • therapeutic isotope the therapeutic isotope being selected from the group consisting of Lu-177, Y-90, Ac-225, As-211, Bi-212, Bi-213, Cs-137, Cr-51, Co-60, Dy-165, Er-169, Fm-255, Au-198, Ho-166, I-125, I-131, Ir-192, Fe-59, Pb-212, Mo-99, Pd- 103, P-32, K-42, Re-186, Re-188, Sm-153, Ra223, Ru-106, Na24, Sr89, Tb-149, Th-227, Xe-133Yb-169, Yb-177, Or a combination thereof.
  • the coupling moiety is a drug or a toxin.
  • the drug is a cytotoxic drug.
  • the cytotoxic drug is selected from the group consisting of anti-tubulin drugs, DNA minor groove binding reagents, DNA replication inhibitors, alkylating agents, antibiotics, folic acid antagonists, antimetabolites, chemotherapy A sensitizer, a topoisomerase inhibitor, a vinca alkaloid, or a combination thereof.
  • cytotoxic drugs include, for example, DNA minor groove binding reagents, DNA alkylating agents, and tubulin inhibitors, typical cytotoxic drugs including, for example, auristatin, camptothecin (camptothecins), docamycin/duocarmycins, etoposides, maytansines and maytansinoids (eg DM1 and DM4), taxanes ( Taxanes), benzodiazepines or benzodiazepine containing drugs (eg pyrrolo[1,4]benzodiazepines (PBDs), porphyrin benzodiazepines Classes (indolinobenzodiazepines) and oxazolidinobenzodiazepines, vinca alkaloids, or combinations thereof.
  • typical cytotoxic drugs including, for example, auristatin, camptothecin (camptothecins), docamycin/duocarmycins, etoposides, maytansines and maytansinoids (eg DM1 and
  • the toxin is selected from the group consisting of:
  • Auristatins eg, auristatin E, auristatin F, MMAE, and MMAF
  • chlortetracycline metanicol
  • ricin e.g., auristatin E, auristatin F, MMAE, and MMAF
  • doxorubicin daunorubicin
  • paclitaxel cisplatin
  • cc1065 ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, autumn Narcissus, dihydroxy anthrax dione, actinomycin, diphtheria toxin, pseudomonas exotoxin (PE) A, PE40, abrin toxin, abrin toxin A chain, alfalfa toxin A chain, ⁇ - sarcococci, leucotoxin, mitogellin, retstrictocin, phenolic acid, enoxamycin, cu
  • the coupled moiety is a detectable label.
  • the conjugate is selected from the group consisting of: a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computer tomography) contrast agent, or is capable of producing a detectable agent
  • Product enzymes radionuclides, biotoxins, cytokines (such as IL-2, etc.), antibodies, antibody Fc fragments, antibody scFv fragments, gold nanoparticles/nanorods, viral particles, liposomes, nanomagnetic particles, pre- A drug activating enzyme (eg, DT-diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)), a chemotherapeutic agent (eg, cisplatin) or any form of nanoparticles, and the like.
  • DTD DT-diaphorase
  • BPHL biphenyl hydrolase-like protein
  • the immunoconjugate comprises: a multivalent (e.g., bivalent) VHH chain of an anti-Her2 Nanobody of the first aspect of the invention, an anti-antibody according to the second aspect of the invention Her2 Nanobody.
  • a multivalent (e.g., bivalent) VHH chain of an anti-Her2 Nanobody of the first aspect of the invention an anti-antibody according to the second aspect of the invention Her2 Nanobody.
  • the multivalent protein refers to a VHH chain comprising a plurality of repeating anti-Her2 Nanobodies according to the first aspect of the present invention in the amino acid sequence of the immunoconjugate, and the present invention The anti-Her2 Nanobody described in the two aspects.
  • the immunoconjugate is used for the diagnosis or prognosis of cancer, especially for the expression of Her2 (i.e., Her2-positive tumor).
  • the detection is in vivo or in vitro.
  • the immunoconjugate is used to diagnose and/or treat a tumor expressing a Her2 protein.
  • the use of the anti-Her2 Nanobody of the second aspect of the invention, or the immunoconjugate of the seventh aspect of the invention for preparing (a) a reagent for detecting Her2 molecule (b) a drug for treating a tumor.
  • the coupled portion of the immunoconjugate is a diagnostic isotope.
  • the reagent is one or more reagents selected from the group consisting of an isotope tracer, a contrast agent, a flow detection reagent, a cellular immunofluorescence detection reagent, a nanomagnetic particle, and an imaging agent. .
  • the reagent for detecting the Her2 molecule is a (in vivo) contrast agent for detecting the Her2 molecule.
  • the detection is in vivo or in vitro.
  • the detection comprises flow detection, cellular immunofluorescence detection.
  • a pharmaceutical composition comprising:
  • the coupled portion of the immunoconjugate is a drug, a toxin, and/or a therapeutic isotope.
  • the pharmaceutical composition further comprises other drugs for treating tumors, such as cytotoxic drugs.
  • the pharmaceutical composition is for treating a tumor that expresses a Her2 protein (i.e., Her2 positive).
  • the pharmaceutical composition is in the form of an injection.
  • the pharmaceutical composition is used for preparing a medicament for treating a tumor, the tumor being selected from the group consisting of gastric cancer, liver cancer, leukemia, kidney tumor, lung cancer, small bowel cancer, bone cancer, prostate cancer, Colorectal cancer, breast cancer, colorectal cancer, prostate cancer, cervical cancer, lymphoma, adrenal tumor, or bladder tumor.
  • the tumor is a tumor that expresses a Her2 protein (ie, Her2 positive).
  • the use is non-diagnostic and non-therapeutic.
  • an antibody comprising: the heavy chain variable region VHH according to the first aspect of the invention.
  • the antibody is an antibody specific for the Her2 protein.
  • the antibody is a Nanobody.
  • a recombinant protein comprising:
  • the tag sequence comprises a 6His tag and an HA tag.
  • the recombinant protein specifically binds to the Her2 protein.
  • a thirteenth aspect of the invention provides the use of the VHH chain according to the first aspect of the invention, the Nanobody of the second aspect of the invention, or the immunoconjugate of the seventh aspect of the invention, Used to prepare medicaments, reagents, test plates or kits;
  • the reagent, the detection plate or the kit is used for: detecting the Her2 protein in the sample;
  • the agent is for treating or preventing a tumor expressing a Her2 protein (ie, Her2 positive).
  • the tumor comprises: melanoma, gastric cancer, lymphoma, liver cancer, leukemia, kidney tumor, lung cancer, small bowel cancer, bone cancer, prostate cancer, colorectal cancer, breast cancer, colon cancer, prostate cancer , or adrenal tumors.
  • a fourteenth aspect of the invention there is provided a method of detecting a Her2 protein in a sample, the method comprising the steps of:
  • a method of treating a disease comprising administering a Nanobody of the second aspect of the invention or the immunoconjugate of the seventh aspect of the invention to a subject in need thereof.
  • the subject comprises a mammal, such as a human.
  • a Her2 protein detecting reagent comprising the immunoconjugate of the seventh aspect of the invention and a testably acceptable carrier.
  • the coupled portion of the immunoconjugate is a diagnostic isotope.
  • the detectably acceptable carrier is a non-toxic, inert, aqueous carrier medium.
  • the detection reagent is one or more reagents selected from the group consisting of an isotope tracer, a contrast agent, a flow detection reagent, a cellular immunofluorescence detection reagent, nanomagnetic particles, and imaging. Agent.
  • the detection reagent is a contrast agent
  • the contrast agent further comprises other preparations for contrast.
  • the contrast agent is a contrast agent for MRI (magnetic resonance imaging) or CT (computer tomography).
  • the imaging agent simultaneously sequesters two or more signals, such as Ga-68 and Gd, for both PET/CT and MRI; or Tc-99m and a fluorescent agent, simultaneously For SPECT/CT and fluorescence detection.
  • the detection reagent is for in vivo detection.
  • the test agent is in the form of a liquid or a powder (e.g., a liquid, an injection, a lyophilized powder, a tablet, a dose, a aerosol).
  • a liquid or a powder e.g., a liquid, an injection, a lyophilized powder, a tablet, a dose, a aerosol.
  • kits for detecting a Her2 molecule comprising the immunoconjugate of the seventh aspect of the invention and instructions.
  • the instructions recite the kit for non-invasive detection of Her2 expression in a subject.
  • the kit is for detection of a tumor expressing a Her2 protein (i.e., Her2 positive).
  • an eighteenth aspect of the invention there is provided the use of the immunoconjugate of the seventh aspect of the invention for the preparation of a contrast agent for detecting Her2 molecules in vivo.
  • the detection is for diagnosis or prognosis of cancer.
  • a CAR-T cell in a nineteenth aspect of the invention, is provided, the CAR-T cell expressing a chimeric antigen receptor CAR, the antigen binding domain of the CAR having the first aspect of the invention A VHH chain, or a Nanobody of the second aspect of the invention.
  • a formulation comprising the CAR-T cell of the nineteenth aspect of the invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the formulation is a liquid formulation.
  • the dosage form of the formulation includes an injection.
  • the concentration of the CAR-T cells in the preparation is 1 ⁇ 10 3 - 1 ⁇ 10 8 cells / ml, preferably 1 ⁇ 10 4 - 1 ⁇ 10 7 cells / ml. .
  • Figure 1 is a SDS-PAGE diagram of antigen protein purification.
  • the three electrophoresis lanes in the figure are nucleic acid molecular standards from left to right, purified human Her2 (ECD)-Fc protein, human Her2 (ECD) protein after TEV digestion, and the above proteins are expressed by HEK293F cells.
  • Figure 2 is a diagram of library construction and its quality detection.
  • Figure A shows the first PCR amplification product, and the taper recovers the target band of about 700 bp in size;
  • Figure B shows the second PCR amplification product, and obtains the VHH gene fragment of about 400 bp in size;
  • Figure C shows the storage capacity of the library.
  • FIG constructed by dilutions of the library were plated, taken 1/5 10 3-fold serial dilution, 104 times, 105 times the number of clones, 106 times, calculating a library size determines the number of monoclonal 2x10 9 CFU;
  • Figure D shows the library insertion rate detection map. 24 clones of the library were randomly picked for PCR identification.
  • the DNA bands from the left to the right gel were: the first is the DNA molecular marker, and the other is the detection insert.
  • the PCR product, the PCR product band was about 500 bp, and the VHH insertion rate of the library was 100%.
  • Figure 3 shows the Her2 Nanobody Screening Enrichment Process. The library did not show double enrichment in the first round of panning, 3.75 times enrichment in the second round of panning, and 110 times enrichment in the third round of panning.
  • Figure 4 shows the results of purification of the Her2 Nanobody.
  • the nano-antibody is purified by nickel column ion affinity chromatography in one step, and the purity can reach more than 95%.
  • Figure 5 shows the affinity of the Her2 Nanobody to different species Her2.
  • the Nanobody reacted with the human and murine antigenic protein Her2 at different gradient concentrations, and the results showed that the Nanobody only binds to human Her2.
  • Figure 6 shows the results of SPECT-CT imaging of the HER-2 high expression tumor-bearing mice with the I-125-labeled Her2 Nanobody. Multiple Nanobodies are effective in the efficient accumulation of Her2 high expression tumors, while non-binding antibodies can be rapidly cleared from the blood through the kidneys and bladder.
  • Figure 7 shows the results of SPECT-CT imaging of 99m-Tc-labeled Her2 Nanobody in HER-2 high expression tumor-bearing mice. Nanobodies are capable of specifically accumulating tumors with high expression of Her2, and non-binding antibodies can be rapidly cleared from the blood through the kidneys and bladder without competing with trastuzumab or pertuzumab.
  • the inventors have successfully obtained a class of anti-Her2 Nanobodies through extensive screening through extensive and in-depth research.
  • the experimental results show that the Her2 Nanobody obtained by the present invention can effectively bind to Her2.
  • the present invention utilizes a human Her2 antigen protein to immunize a camel to obtain a high quality immuno Nanobody gene library. Then, the Her2 protein molecule was coupled to the ELISA plate to display the correct spatial structure of the Her2 protein. The antigen of this form was screened by the phage display technology to screen the immuno Nanobody gene library (the camelid heavy chain antibody phage display gene library), thereby obtaining Her2 specific Nanobody gene. This gene was further transferred to Escherichia coli to obtain a highly specific Nanobody strain which was highly expressed in Escherichia coli.
  • the present invention also surprisingly finds an immunoconjugate which is particularly suitable for detecting Her2 molecules, which comprises a specific anti-Her2 Nanobody VHH chain and a radionuclide, which can be used for Her2 expression in a subject to be tested. Non-invasive testing of the situation.
  • the immunoconjugate of the invention has small size and high specificity, and is suitable for simultaneous systemic detection of primary and metastatic tumors with high accuracy and small radiation dose.
  • the present invention also provides immunoconjugates that are effective for treating Her2-positive tumors.
  • Nanobody of the invention As used herein, the terms " Nanobody of the invention”, “anti-Her2 Nanobody of the invention”, “Her2 Nanobody of the invention” are used interchangeably and refer to a nanoparticle that specifically recognizes and binds to Her2 (including human Her2). antibody. Particularly preferred is the amino acid sequence of the VHH chain as set forth in SEQ ID NO.: 1-40.
  • antibody or "immunoglobulin” is an isotetrameric glycoprotein of about 150,000 daltons having the same structural features, consisting of two identical light chains (L) and two identical heavy chains. (H) Composition. Each light chain is linked to the heavy chain by a covalent disulfide bond, and the number of disulfide bonds between the heavy chains of different immunoglobulin isotypes is different. Each heavy and light chain also has regularly spaced intrachain disulfide bonds. Each heavy chain has a variable region (VH) at one end followed by a plurality of constant regions.
  • VH variable region
  • Each light chain has a variable region (VL) at one end and a constant region at the other end; the constant region of the light chain is opposite the first constant region of the heavy chain, and the variable region of the light chain is opposite to the variable region of the heavy chain .
  • Particular amino acid residues form an interface between the variable regions of the light and heavy chains.
  • single domain antibody VHH
  • nanobody a single domain antibody consisting of only one heavy chain variable region.
  • VHH single domain antibody
  • CH1 light chain and heavy chain constant region 1
  • variable means that certain portions of the variable regions of an antibody differ in sequence, which form the binding and specificity of various specific antibodies for their particular antigen. However, the variability is not evenly distributed throughout the variable region of the antibody. It is concentrated in three segments in the variable region of the light and heavy chains called the complementarity determining region (CDR) or hypervariable region. The more conserved portion of the variable region is referred to as the framework region (FR).
  • the variable regions of the native heavy and light chains each comprise four FR regions which are substantially in a beta-sheet configuration and are joined by three CDRs forming a linker, in some cases forming a partial beta sheet structure.
  • the CDRs in each chain are closely joined together by the FR region and together with the CDRs of the other chain form the antigen binding site of the antibody (see Kabat et al, NIH Publ. No. 91-3242, Vol. I, pp. 647-669). (1991)).
  • the constant regions are not directly involved in the binding of the antibody to the antigen, but they exhibit different effector functions, such as antibody-dependent cytotoxicity of the participating antibodies.
  • immunoconjugates and fusion expression products include: drugs, toxins, cytokines, radionuclides, enzymes, and other diagnostic or therapeutic molecules that are combined with the antibodies or fragments thereof of the invention to form Conjugate.
  • the invention also encompasses cell surface markers or antigens that bind to the anti-Her2 protein antibody or fragment thereof.
  • variable region are used interchangeably with “complementarity determining region (CDR).
  • the heavy chain variable region of the antibody comprises three complementarity determining regions, CDR1, CDR2, and CDR3.
  • the heavy chain of the antibody comprises the heavy chain variable region and the heavy chain constant region described above.
  • the terms "antibody of the invention”, “protein of the invention”, or “polypeptide of the invention” are used interchangeably and refer to a polypeptide which specifically binds to a Her2 protein, such as a protein or polypeptide having a heavy chain variable region. . They may or may not contain an initial methionine.
  • the invention also provides other proteins or fusion expression products having the antibodies of the invention.
  • the invention encompasses any protein or protein conjugate having a heavy chain comprising a variable region and a fusion expression product (ie, an immunoconjugate and a fusion expression product), so long as the variable region is linked to the heavy chain of an antibody of the invention
  • the variable regions are identical or at least 90% homologous, preferably at least 95% homologous.
  • variable regions which are divided into four framework regions (FR), four FR amino acid sequences. Relatively conservative, not directly involved in the binding reaction.
  • CDRs form a cyclic structure in which the ⁇ -sheets formed by the FRs are spatially close to each other, and the CDRs on the heavy chain and the CDRs on the corresponding light chain constitute the antigen-binding site of the antibody.
  • the amino acid sequence of the same type of antibody can be compared to determine which amino acids constitute the FR or CDR regions.
  • variable regions of the heavy chains of the antibodies of the invention are of particular interest because at least some of them are involved in binding antigen.
  • the invention includes those molecules having an antibody heavy chain variable region with a CDR, as long as the CDRs thereof have 90% or more (preferably 95% or more, optimally 98% or more) homology to the CDRs identified herein. Sex.
  • the present invention encompasses not only intact antibodies, but also fragments of immunologically active antibodies or fusion proteins formed by antibodies with other sequences. Accordingly, the invention also includes fragments, derivatives and analogs of the antibodies.
  • fragment refers to a polypeptide that substantially retains the same biological function or activity of an antibody of the invention.
  • the polypeptide fragment, derivative or analog of the present invention may be (i) a polypeptide having one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) substituted, and such substituted amino acid residues It may or may not be encoded by the genetic code, or (ii) a polypeptide having a substituent group in one or more amino acid residues, or (iii) a mature polypeptide and another compound (such as a compound that extends the half-life of the polypeptide, for example Polyethylene glycol) a polypeptide formed by fusion, or (iv) a polypeptide formed by fused an additional amino acid sequence to the polypeptide sequence (such as a leader or secretion sequence or a sequence or proprotein sequence used to purify the polypeptide, or a fusion protein formed by the 6
  • the antibody of the present invention refers to a polypeptide comprising the above CDR region having Her2 protein binding activity.
  • the term also encompasses variant forms of a polypeptide comprising the above-described CDR regions that have the same function as the antibodies of the invention. These variants include, but are not limited to, one or more (typically 1-50, preferably 1-30, more preferably 1-20, optimally 1-10) amino acid deletions , Insertion and/or Substitution, and the addition of one or several (usually within 20, preferably within 10, more preferably within 5) amino acids at the C-terminus and/or N-terminus.
  • the function of the protein is generally not altered.
  • the addition of one or several amino acids at the C-terminus and/or N-terminus will generally not alter the function of the protein.
  • the term also encompasses active fragments and active derivatives of the antibodies of the invention.
  • Variant forms of the polypeptide include: homologous sequences, conservative variants, allelic variants, natural mutants, induced mutants, DNA capable of hybridizing to the DNA encoding the antibody of the present invention under high or low stringency conditions.
  • the encoded protein, and the polypeptide or protein obtained using an antiserum against the antibody of the present invention.
  • the invention also provides other polypeptides, such as fusion proteins comprising Nanobodies or fragments thereof.
  • the invention also includes fragments of the Nanobodies of the invention.
  • the fragment will have at least about 50 contiguous amino acids, preferably at least about 50 contiguous amino acids, more preferably at least about 80 contiguous amino acids, and most preferably at least about 100 contiguous amino acids of the antibody of the invention.
  • “conservative variant of the antibody of the present invention” means having up to 10, preferably up to 8, more preferably up to 5, and most preferably up to 3, compared to the amino acid sequence of the antibody of the present invention. Amino acids are replaced by amino acids of similar or similar nature to form a polypeptide. These conservative variant polypeptides are preferably produced by amino acid substitution according to Table 1.
  • the present invention also provides a polynucleotide molecule encoding the above antibody or a fragment thereof or a fusion protein thereof.
  • the polynucleotide of the present invention may be in the form of DNA or RNA.
  • DNA forms include cDNA, genomic DNA or synthetic DNA.
  • DNA can be single-stranded or double-stranded.
  • the DNA can be a coding strand or a non-coding strand.
  • Polynucleotides encoding mature polypeptides of the invention include: coding sequences encoding only mature polypeptides; coding sequences for mature polypeptides and various additional coding sequences; coding sequences for mature polypeptides (and optionally additional coding sequences) and non-coding sequences .
  • polynucleotide encoding a polypeptide can be a polynucleotide comprising the polypeptide, or a polynucleotide further comprising additional coding and/or non-coding sequences.
  • the invention also relates to polynucleotides which hybridize to the sequences described above and which have at least 50%, preferably at least 70%, more preferably at least 80% identity between the two sequences.
  • the invention particularly relates to polynucleotides that hybridize to the polynucleotides of the invention under stringent conditions.
  • stringent conditions means: (1) hybridization and elution at a lower ionic strength and higher temperature, such as 0.2 x SSC, 0.1% SDS, 60 ° C; or (2) hybridization a denaturing agent such as 50% (v/v) formamide, 0.1% calf serum/0.1% Ficoll, 42 ° C, etc.; or (3) at least 90% identity between the two sequences, more It is good that hybridization occurs more than 95%.
  • the polypeptide encoded by the hybridizable polynucleotide has the same biological function and activity as the mature polypeptide.
  • the full-length nucleotide sequence of the antibody of the present invention or a fragment thereof can be usually obtained by a PCR amplification method, a recombinant method or a synthetic method.
  • One possible method is to synthesize related sequences by artificial synthesis, especially when the fragment length is short.
  • a long sequence of fragments can be obtained by first synthesizing a plurality of small fragments and then performing the ligation.
  • the coding sequence of the heavy chain and the expression tag (such as 6His) can be fused together to form a fusion protein.
  • the recombinant sequence can be used to obtain the relevant sequences in large quantities. This is usually done by cloning it into a vector, transferring it to a cell, and then isolating the relevant sequence from the proliferated host cell by conventional methods.
  • the biomolecule (nucleic acid, protein, etc.) to which the present invention relates includes biomolecules existing in an isolated form.
  • DNA sequence encoding the protein of the present invention (or a fragment thereof, or a derivative thereof) completely by chemical synthesis.
  • the DNA sequence can then be introduced into various existing DNA molecules (or vectors) and cells known in the art.
  • mutations can also be introduced into the protein sequences of the invention by chemical synthesis.
  • the invention also relates to vectors comprising the appropriate DNA sequences described above, as well as appropriate promoters or control sequences. These vectors can be used to transform appropriate host cells to enable them to express proteins.
  • the host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher eukaryotic cell, such as a mammalian cell.
  • a prokaryotic cell such as a bacterial cell
  • a lower eukaryotic cell such as a yeast cell
  • a higher eukaryotic cell such as a mammalian cell.
  • Representative examples are: Escherichia coli, Streptomyces; bacterial cells of Salmonella typhimurium; fungal cells such as yeast; insect cells of Drosophila S2 or Sf9; animal cells of CHO, COS7, 293 cells, and the like.
  • Transformation of host cells with recombinant DNA can be carried out using conventional techniques well known to those skilled in the art.
  • the host is a prokaryote such as E. coli
  • competent cells capable of absorbing DNA can be harvested after the exponential growth phase and treated by the CaCl 2 method, and the procedures used are well known in the art.
  • Another method is to use MgCl 2 .
  • Conversion can also be carried out by electroporation if desired.
  • the host is a eukaryote, the following DNA transfection methods can be used: calcium phosphate coprecipitation, conventional mechanical methods such as microinjection, electroporation, liposome packaging, and the like.
  • the obtained transformant can be cultured by a conventional method to express the polypeptide encoded by the gene of the present invention.
  • the medium used in the culture may be selected from various conventional media depending on the host cell used.
  • the cultivation is carried out under conditions suitable for the growth of the host cell.
  • the selected promoter is induced by a suitable method (such as temperature conversion or chemical induction) and the cells are cultured for a further period of time.
  • the recombinant polypeptide in the above method can be expressed intracellularly, or on the cell membrane, or secreted outside the cell.
  • the recombinant protein can be isolated and purified by various separation methods using its physical, chemical, and other properties. These methods are well known to those skilled in the art. Examples of such methods include, but are not limited to, conventional renaturation treatment, treatment with a protein precipitant (salting method), centrifugation, osmotic sterilizing, super treatment, ultracentrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography, high performance liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
  • the antibodies of the invention may be used alone or in combination or in combination with a detectable label (for diagnostic purposes), a therapeutic agent, a PK (protein kinase) modifying moiety, or a combination of any of these.
  • Detectable labels for diagnostic purposes include, but are not limited to, fluorescent or luminescent labels, radioactive labels, MRI (magnetic resonance imaging) or CT (computer tomography) contrast agents, or capable of producing detectable products. Enzyme.
  • Therapeutic agents that can be bound or conjugated to the antibodies of the invention include, but are not limited to: 1. radionuclides; 2. biotoxic; 3. cytokines such as IL-2, etc.; 4. gold nanoparticles/nanorods; Particles; 6. liposomes; 7. nanomagnetic particles; 8. drug activating enzymes (eg, DT-diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)); 9. therapeutic agents (eg , cisplatin) or any form of nanoparticles, etc.
  • DTD DT-diaphorase
  • BPHL biphenyl hydrolase-like protein
  • the invention also provides an immunoconjugate comprising:
  • a coupling moiety selected from the group consisting of a radionuclide, an enzyme antibody, a cell, or a combination thereof.
  • the immunoconjugate is as described in the seventh aspect of the invention.
  • the immunoconjugate of the invention can be used for non-invasive detection of Her2 expression of a test subject, has small size and high specificity, is suitable for simultaneous detection of primary and metastatic tumors, and has high accuracy and small radiation dose. .
  • Coupling moieties constituting the antibody conjugates of the invention include toxins, such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.
  • cytotoxic agents include, but are not limited to, auristatins (eg, auristatin E, auristatin F, MMAE, and MMAF), chlortetracycline, etometanol, ricin, ricin A-chain, Butatin, doxymethine, dolastatin, doxorubicin, daunorubicin, paclitaxel, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide , vincristine, vinblastine, colchicine, dihydroxy anthrax dione, actinomycin, diphtheria toxin, pseudomonas exotoxin (PE) A, PE40,
  • Preferred small molecule drugs are compounds having high cytotoxicity, preferably monomethylaustatatin, calicheamicin, maytansinoids, or a combination thereof; more preferably selected from: monomethyl au Statin-E (MMAE), monomethyl auristatin-D (MMAD), monomethyl auristatin-F (MMAF), or a combination thereof.
  • MMAE monomethyl au Statin-E
  • MMAD monomethyl auristatin-D
  • MMAF monomethyl auristatin-F
  • the invention also provides a composition.
  • the composition is a pharmaceutical composition comprising the above antibody or active fragment thereof or a fusion protein or immunoconjugate thereof, and a pharmaceutically acceptable carrier.
  • these materials can be formulated in a non-toxic, inert, and pharmaceutically acceptable aqueous carrier medium wherein the pH is usually from about 5 to about 8, preferably from about 6 to about 8, although the pH may be The nature of the formulation and the condition to be treated vary.
  • the formulated pharmaceutical compositions can be administered by conventional routes including, but not limited to, intratumoral, intraperitoneal, intravenous, or topical administration.
  • the pharmaceutical composition of the present invention can be directly used for binding to a Her2 protein molecule, and thus can be used for treating tumors.
  • other therapeutic agents can be used simultaneously.
  • the pharmaceutical composition of the present invention contains a safe and effective amount (e.g., 0.001 to 99% by weight, preferably 0.01 to 90% by weight, more preferably 0.1 to 80% by weight) of the above-described Nanobody of the present invention (or a conjugate thereof) and pharmaceutically An acceptable carrier or excipient.
  • a safe and effective amount e.g., 0.001 to 99% by weight, preferably 0.01 to 90% by weight, more preferably 0.1 to 80% by weight
  • Such carriers include, but are not limited to, saline, buffer, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the pharmaceutical preparation should be matched to the mode of administration.
  • the pharmaceutical composition of the present invention can be prepared in the form of an injection, for example, by a conventional method using physiological saline or an aqueous solution containing glucose and other adjuvants.
  • compositions such as injections and solutions are preferably prepared under sterile conditions.
  • the active ingredient is administered in a therapeutically effective amount, for example, about 10 ng/kg body weight to about 50 mg/kg body weight per day, more preferably 50 ng/kg body weight to about 1 mg/kg body weight or 10 ⁇ g/kg body weight. - about 10 mg / kg body weight.
  • polypeptides of the invention or conjugates thereof can also be used with other therapeutic agents, such as anti-tumor agents or immunomodulators.
  • a safe and effective amount of the immunoconjugate is administered to the mammal, wherein the safe and effective amount is usually at least about 10 nanograms per kilogram of body weight, and in most cases no more than about 50 milligrams per kilogram of body weight.
  • the dosage is from about 50 nanograms per kilogram of body weight to about 1 milligram per kilogram of body weight.
  • specific doses should also consider factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • the Nanobody is provided with a detectable label. More preferably, the label is selected from the group consisting of an isotope, a colloidal gold label, a colored label or a fluorescent label.
  • the colloidal gold label can be carried out by methods known to those skilled in the art.
  • the anti-Her2 Nanobody is labeled with colloidal gold to obtain a colloidal gold-labeled Nanobody.
  • the anti-Her2 Nanobody of the present invention has good specificity and high potency.
  • CAR-T cell As used herein, the terms "CAR-T cell”, “CAR-T”, “CAR-T cell of the invention” all refer to the CAR-T cell of the nineteenth aspect of the invention.
  • a chimeric antigen receptor includes an extracellular domain, an optional hinge region, a transmembrane domain, and an intracellular domain.
  • the extracellular domain includes an optional signal peptide and a target-specific binding element (also referred to as an antigen binding domain).
  • the intracellular domain includes a costimulatory molecule and a purine chain portion.
  • a costimulatory signaling region refers to a portion of an intracellular domain that includes a costimulatory molecule. Costimulatory molecules are cell surface molecules required for efficient response of lymphocytes to antigens, rather than antigen receptors or their ligands.
  • antigen binding domain refers to a Fab fragment, Fab' fragment, F(ab') 2 fragment, or a single Fv fragment having antigen binding activity.
  • the Fv antibody contains the antibody heavy chain variable region, the light chain variable region, but no constant region, and has the smallest antibody fragment of the entire antigen binding site.
  • Fv antibodies also comprise a polypeptide linker between the VH and VL domains and are capable of forming the desired structure for antigen binding.
  • the antigen binding domain is typically a scFv (single-chain variable fragment).
  • the single chain antibody is preferably a sequence of one amino acid strand encoded by one nucleotide chain.
  • the scFv comprises the VHH chain of the first aspect of the invention, or the Nanobody of the second aspect of the invention.
  • the CAR can be designed to include a transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain that is naturally associated with one of the domains in the CAR is used.
  • transmembrane domains may be selected or modified by amino acid substitutions to avoid binding such domains to the transmembrane domain of the same or different surface membrane proteins, thereby minimizing complexes with receptors. The interaction of other members.
  • a linker can be incorporated between the extracellular domain and the transmembrane domain of the CAR, or between the cytoplasmic domain and the transmembrane domain of the CAR.
  • the term "linker” generally refers to any oligopeptide or polypeptide that functions to link a transmembrane domain to the extracellular domain or cytoplasmic domain of a polypeptide chain.
  • the linker may comprise from 0 to 300 amino acids, preferably from 2 to 100 amino acids and most preferably from 3 to 50 amino acids.
  • the extracellular domain When CAR is expressed in T cells, the extracellular domain recognizes a specific antigen and then transduces the signal through the intracellular domain, causing activation and proliferation of cells, cytolysis toxicity, and secretion of cytokines such as IL-2 and IFN- ⁇ . Etc., affecting tumor cells, causing tumor cells to not grow, cause death or otherwise be affected, and cause the patient's tumor burden to shrink or eliminate.
  • the antigen binding domain is preferably fused to an intracellular domain from one or more of a costimulatory molecule and a sputum chain.
  • the invention also relates to methods of detecting Her2 protein.
  • the method steps are substantially as follows: obtaining a cell and/or tissue sample; dissolving the sample in a medium; detecting the level of Her2 protein in the dissolved sample.
  • the sample to be used is not particularly limited, and a representative example is a cell-containing sample present in the cell preservation solution.
  • the present invention also provides a kit comprising the antibody (or a fragment thereof) or a test plate of the present invention.
  • the kit further comprises a container, instructions for use, a buffer, and the like.
  • the invention also provides a detection kit for detecting Her2 levels, comprising an antibody for recognizing Her2 protein, a lysis medium for dissolving a sample, detecting a common reagent and a buffer required, such as various buffers, detection Mark, detect substrates, etc.
  • the test kit can be an in vitro diagnostic device.
  • the invention also provides a kit comprising the immunoconjugate of the invention, in a preferred embodiment of the invention, the kit further comprises a container, instructions for use, an isotope tracer and a group selected from the group consisting of One or more reagents: contrast agent, flow detection reagent, cellular immunofluorescence detection reagent, nanomagnetic particle and imaging agent.
  • the kit of the present invention is an in vivo diagnostic kit for non-invasively detecting Her2 expression of a subject.
  • the nano-antibody of the present invention has a wide range of biological application value and clinical application value, and its application involves various fields such as diagnosis and treatment of Her2-related diseases, basic medical research, and biological research.
  • a preferred application is for clinical diagnosis and targeted therapy for Her2.
  • Nanobody of the present invention is highly specific for a human Her2 protein having the correct spatial structure.
  • the nanobody of the present invention has a strong affinity.
  • the Nanobody of the present invention is capable of specifically binding to human Her2, and is efficiently accumulated in the Her2 high expression tumor model, and does not compete with trastuzumab or pertuzumab.
  • the Her2 Nanobody of the present invention is very suitable for Her2 targeted cancer diagnosis and therapeutic evaluation, and a new generation of Her2 targeted in vivo radiotherapy.
  • the plasmid was mixed with the transfection reagent PEI 1:5 and allowed to stand for 10 min, then added to HEK293F cells, and cultured for 5-6 days at 37 ° C in a 6% CO 2 shaker incubator;
  • the eluted protein was ultrafiltered into PBS, and the yield was measured and sampled for SDS-PAGE (detection results are shown in Fig. 1).
  • the purity of the antigen was 95% or more, and it can be used for subsequent immune reaction.
  • the protein was digested with a TEV enzyme to obtain an unlabeled antigenic protein Her2 (ECD) for subsequent antibody screening.
  • ECD unlabeled antigenic protein Her2
  • FIG. 2A is the first PCR amplification product, and the 700-bp VHH-hinge-CH2-CH3 fragment is recovered by tapping, and Figure 2B is the second PCR amplification product. a 400 bp VHH fragment);
  • Phage-enzyme-linked immunosorbent assay (ELISA) for screening specific single positive clones Phage-enzyme-linked immunosorbent assay (ELISA) for screening specific single positive clones:
  • a crude antibody was obtained by an infiltration method, and the antibody was transferred to an antigen-coated ELISA plate and allowed to stand at room temperature for 1 hour.
  • Example 4 Nanobody expression and purification in host strain Escherichia coli:
  • Example 3 (1) Different clones obtained by sequencing analysis of Example 3 (7 Nanobodies were randomly selected), each corresponding plasmid was electrotransformed into Escherichia coli WK6, and coated with LA+glucose containing ampicillin and The culture plate of glucose was cultured overnight at 37 ° C;
  • the purification results are shown in Figure 4.
  • the anti-Her2 Nanobody has a purity of more than 95% after the purification process.
  • Example 5 Enzyme-linked immunosorbent assay (ELISA) to identify the affinity of Nanobodies to different species Her2
  • Antigen protein HER2 coated with human and mouse species The antigenic protein Her2 of human and murine species was added at 4 ° C overnight.
  • the Nanobody of the present invention binds only to human Her2.
  • Example 6 Flow cytometry to detect the binding of Nanobodies to cells
  • Cell type BT474 and MDA-MB-231. The cells were washed twice with PBS.
  • Nanobody was diluted with PBS and diluted to a concentration of 0.1 ug/uL.
  • the cells were washed twice with PBS, resuspended in 100 uL PBS, and then added with mouse anti-HA Alexa Fluor 488 labeled antibody, 1 uL of each sample, mixed, and placed at 4 ° C for 20 min.
  • Antibody number BT474 MDA-MB-231 9 99.8% 9.5% 10 99.7% 10.6% 13 99.9% 14.2% 17 99.9% 12.3% twenty two 99.7% 5.8% twenty three 99.9% 11.5% 26 99.9% 16.7%
  • Chip regeneration was performed by rinsing with 10 mM Glycine for the next antibody assay
  • nanobodies of the present invention all have an affinity for Her2 above the nanomolar level and do not compete with trastuzumab or pertuzumab for binding to Her2.
  • the data for some of the Nanobodies are shown in Table 5.
  • NOD/SCID mice were implanted with estrogen tablets subcutaneously on the right side of the right side of the cells one day before cell inoculation, and 1 ⁇ 10 7 Her2 high expression (BT474) cells were inoculated on the right breast fat pad until the tumor grew to 150- 200mm 3 is used for formal experimental research.
  • Nanobodies of the present invention are effective for the effective accumulation of the Her2 high expression tumor model, and are applied to cancer diagnosis and treatment.
  • non-binding antibodies can be quickly cleared from the blood through the kidneys and bladder, reducing the body's radiation dose.
  • NOD/SCID mice were implanted with estrogen tablets subcutaneously on the right side of the right side of the cells one day before cell inoculation, and 1 ⁇ 10 7 Her2 high expression (BT474) cells were inoculated on the right breast fat pad until the tumor grew to 150- 200mm 3 is used for formal experimental research.
  • mice Anesthetized tumor-bearing mice were injected with isoflurane, Tc-99m-labeled Nanobody ( ⁇ 10ug, 5MBq) was injected into the tail vein or 20x unlabeled Nanobody was mixed at the time of injection, or 20x trastuzumab or 72 hours before. 20x pertuzumab was pretreated with tail vein injection.
  • Nanobodies of the present invention are capable of specifically accumulating specifically in the Her2 high expression tumor model and do not compete with trastuzumab or pertuzumab. It can be applied to Her2 targeted cancer diagnosis and efficacy evaluation, while developing a new generation of Her2 targeted therapy.

Abstract

本发明提供了抗 Her2 纳米抗体及其编码序列和用途。具体地,本发明提供了一类针对人表皮生长因子受体- 2(Her2/ERBB2) 的纳米抗体。本发明公开了这种纳米抗体及编码该纳米抗体的基因序列,相应的表达载体和能够表达该纳米抗体的宿主细胞,以及本发明纳米抗体的生产方法及其相关用途。本发明还可提供了所述纳米抗体的免疫偶联物及其应用,尤其是在诊断和治疗Her2 阳性肿瘤方面的应用。

Description

抗Her2纳米抗体及其编码序列和用途 技术领域
本发明涉及生物医学或生物制药技术领域,更具体地涉及针对Her2的纳米抗体及其编码序列和用途。
背景技术
人表皮生长因子受体-2(human epidermal growth factor receptor 2,Her2/ErbB2)又称Her2/Neu,ErbB-2,CD340或p185,是人Her2基因编码的蛋白质。Her2是一种酪氨酸激酶受体(RTK),属于表皮生长因子受体(EGFR/ErbB)家族,其由1255个氨基酸组成,包括四个胞外域(I,II,III和IV),一个跨膜区,具有酪氨酸激酶活性的结构域和一个包含酪氨酸残基和胞内信号分子锚定位点的羧基末端尾巴,分子量约为185kD。EGFR家族成员具有相似的结构,其中胞外域I和III参与受体与配体的结合,引起受体构像变化从而引起受体激活,胞外域II和IV则参与受体二聚化。Her2具有特殊的开放式结构,不需要特异性配体的参与便可发生自身激活,形成同源二聚体或与EGFR家族的其他受体形成异二聚体,是家族成员间发生异源二聚化的首选分子。
Her2在乳腺癌、卵巢癌、***癌、胃癌、肺癌等多种上皮细胞来源的恶性肿瘤中过表达,而在正常组织中表达水平很低或不表达。当Her2表达水平较低时,受体蛋白一般以单体的形式存在,其酪氨酸激酶活性较低;而当Her2过表达时,可导致持久而增强的受体酪氨酸激酶活化,引起下游一系列的级联反应,激活促***原活化蛋白激酶(MAPK)、磷脂酰肌醇-3-激酶-蛋白激酶B/Akt(PI3K-PKB/Akt)、磷脂酶C-蛋白激酶C(PLC-PKC)及转录信号转导和活化蛋白(STAT)等多条主要的信号通路。Her2介导的信号通路可调控肿瘤相关基因的表达,如上调血管内皮(细胞)生长因子、尿激酶型纤溶酶激活物、环氧合酶-2、趋化细胞因子受体CXCR-4等表达,下调MMP抑制因子RECK表达,可促进肿瘤的侵袭和转移。Her2的扩增或过度表达在某些浸润性乳腺癌的发病和发展过程中具有重要作用,其已成为乳腺癌的重要的标志物和治疗靶标。
人源化重组单克隆抗体曲妥珠单抗(Trastuzumab)和帕妥珠单抗(Pertuzumab)是传统的应用于Her2/neu基因扩增和过表达的乳腺癌患者的靶向治疗药物。Trastuzumab通过与Her2胞外结构的近膜区域结合,发生内吞作用进入肿瘤细胞核内,阻断了Her2再循环到胞膜的过程,加速了Her2蛋白的旁路降解,从而抑制Her2对肿瘤细胞向恶性表型的转导。对于Her2过表达的原发性浸润性乳腺癌患者非常有效。Pertuzumab的抗原结合点位于Her2胞外域的II亚区,通过干扰Her2与其它ErbB成员形成二聚体从而阻断细胞信号传递,无论是否存在Her2过表达,它均能发挥抗瘤作用。
传统单克隆抗体生产流程复杂,成本过高;其分子质量过大,难穿透组织,造成肿 瘤区域的有效浓度较低,治疗效果不充分;传统的抗体具有很高的免疫原性,而改造的抗体很难达到原来的亲和力。此外,完全人源化的传统抗体开发周期长,生产成本高,稳定性不够等诸多因素限制其在临床中的应用及普及。然而,纳米抗体是目前最小的抗体分子,其分子量是普通抗体的1/10。纳米抗体除具备单克隆抗体的抗原反应性外,还拥有一些独特的功能特性,如分子质量小,稳定性强、可溶性好、易表达、免疫原性弱、穿透性强、靶向性强、人源化简单,制备成本低廉等,几乎完美克服了传统抗体开发周期长,稳定性较低,保存条件苛刻等缺陷。另外因其结构特殊,作为放射性同位素靶向载体,能够快速、特异性地穿透肿瘤组织结合靶标,而无结合抗体能很快从血液清除,減低身体放射性剂量,对比传统抗体作为示踪剂及靶向内放疗药物具有众多明显优势。
然而,目前本领域尚缺乏令人满意的针对Her2的纳米抗体。因此,本领域迫切需要开发新的有效针对Her2的特异性纳米抗体。
发明内容
本发明的目的就是提供一类有效针对Her2的特异性纳米抗体。
在本发明的第一方面,提供了一种抗Her2纳米抗体的VHH链,所述VHH链的氨基酸序列如SEQ ID NO.:1-40中任一所示。
在另一优选例中,所述VHH链的氨基酸序列如SEQ ID NO.:8、7、15、12、27、11、32、13、14、9、21、30、17、24、16、6、28、25、10、1中任一所示。
在另一优选例中,所述VHH链的氨基酸序列如SEQ ID NO.:8、7、15、12、27、11、32、13中任一所示。
在另一优选例中,所述VHH链的氨基酸序列如SEQ ID NO.:9、10、13、17、22、23、26中任一所示。
在另一优选例中,所述的Her2为人Her2。
此外,还提供一种抗Her2纳米抗体的VHH链,所述的VHH包括框架区FR和互补决定区CDR,其中,所述的CDR包括SEQ ID NO.:1-40中任一序列中相应的CDR1、CDR2和CDR3,以及被所述CDR1-3所隔开的FR1、FR2、FR3和FR4。
此外,还提供一种抗人Her2抗体的重链可变区,所述的重链可变区包括三个互补决定区CDR1、CDR2、和CDR3,并且3个CDR包括SEQ ID NO.:1-40中任一序列中相应的CDR1、CDR2和CDR3。
此外,还提供一种抗人Her2抗体的互补决定区CDR区,所述的互补决定区CDR区包括如SEQ ID NO.:1-40所示的氨基酸序列中下划线所示的CDR1、CDR2和CDR3(每个VHH氨基酸序列中的三个下划线部分依次代表CDR1、CDR2和CDR3)。
在本发明的第二方面,提供了一种抗Her2纳米抗体,它是针对Her2表位的纳米抗体,并且具有如SEQ ID NO.:1-40中任一所示的氨基酸序列的VHH链。
在另一优选例中,优选的抗Her2纳米抗体具有第一方面中优选的VHH链的氨基酸 序列。
在本发明的第三方面,提供了一种多核苷酸,所述多核苷酸编码选自下组的蛋白质:第一方面所述的抗Her2纳米抗体的VHH链,或第二方面所述的抗Her2纳米抗体。
在另一优选例中,所述的多核苷酸包括DNA或RNA。
在另一优选例中,所述的多核苷酸具有如SEQ ID NO.:41-80中任一所示的核苷酸序列。
在本发明的第四方面,提供了一种表达载体,所述表达载体含有第三方面所述的多核苷酸。
在另一优选例中,所述的表达载体选自下组:DNA、RNA、病毒载体、质粒、转座子、其他基因转移***、或其组合。优选地,所述表达载体包括病毒载体,如慢病毒、腺病毒、AAV病毒、逆转录病毒、或其组合。
在本发明的第五方面,提供了一种宿主细胞,所述宿主细胞含有第四方面所述的表达载体,或其基因组中整合有第三方面所述的多核苷酸。
在另一优选例中,所述的宿主细胞包括原核细胞或真核细胞。
在另一优选例中,所述的宿主细胞选自下组:大肠杆菌、酵母细胞。
本发明第六方面,提供了一种产生抗Her2纳米抗体的方法,包括步骤:
(a)在适合产生纳米抗体的条件下,培养本发明第五方面所述的宿主细胞,从而获得含所述抗Her2纳米抗体的培养物;以及
(b)从所述培养物中分离或回收所述的抗Her2纳米抗体。
在另一优选例中,所述的抗Her2纳米抗体具有如SEQ ID NO.:1-40所示的氨基酸序列。
本发明第七方面,提供了一种免疫偶联物,该免疫偶联物含有:
(a)如本发明第一方面所述的抗Her2纳米抗体的VHH链、或如本发明第二方面所述的抗Her2纳米抗体;和
(b)选自下组的偶联部分:可检测标记物、药物、毒素、细胞因子、放射性核素、或酶、金纳米颗粒/纳米棒、纳米磁粒、病毒外壳蛋白或VLP、或其组合。
在另一优选例中,所述的放射性核素包括:
(i)诊断用同位素,所述的诊断用同位素选自下组:Tc-99m、Ga-68、F-18、I-123、I-125、I-131、In-111、Ga-67、Cu-64、Zr-89、C-11、Lu-177、Re-188、或其组合;和/或
(ii)治疗用同位素,所述的治疗用同位素选自下组:Lu-177、Y-90、Ac-225、As-211、Bi-212、Bi-213、Cs-137、Cr-51、Co-60、Dy-165、Er-169、Fm-255、Au-198、Ho-166、I-125、I-131、Ir-192、Fe-59、Pb-212、Mo-99、Pd-103、P-32、K-42、Re-186、Re-188、Sm-153、Ra223、Ru-106、Na24、Sr89、Tb-149、Th-227、Xe-133Yb-169、Yb-177、或其组合。
在另一优选例中,所述偶联部分为药物或毒素。
在另一优选例中,所述的药物为细胞毒性药物。
在另一优选例中,所述的细胞毒性药物选自下组:抗微管蛋白药物、DNA小沟结合试剂、DNA复制抑制剂、烷化试剂、抗生素、叶酸拮抗物、抗代谢药物、化疗增敏剂、拓扑异构酶抑制剂、长春花生物碱、或其组合。
特别有用的细胞毒性药物类的例子包括,例如,DNA小沟结合试剂、DNA烷基化试剂、和微管蛋白抑制剂、典型的细胞毒性药物包括、例如奥瑞他汀(auristatins)、喜树碱(camptothecins)、多卡霉素/倍癌霉素(duocarmycins)、依托泊甙(etoposides)、美登木素(maytansines)和美登素类化合物(maytansinoids)(例如DM1和DM4)、紫杉烷(taxanes)、苯二氮卓类(benzodiazepines)或者含有苯二氮卓的药物(benzodiazepine containing drugs)(例如吡咯并[1,4]苯二氮卓类(PBDs),吲哚啉苯并二氮卓类(indolinobenzodiazepines)和噁唑烷并苯并二氮卓类(oxazolidinobenzodiazepines))、长春花生物碱(vinca alkaloids)、或其组合。
在另一优选例中,所述的毒素选自下组:
耳他汀类(例如,耳他汀E、耳他汀F、MMAE和MMAF)、金霉素、类美坦西醇、篦麻毒素、篦麻毒素A-链、考布他汀、多卡米星、多拉司他汀、阿霉素、柔红霉素、紫杉醇、顺铂、cc1065、溴化乙锭、丝裂霉素、依托泊甙、替诺泊甙(tenoposide)、长春新碱、长春碱、秋水仙素、二羟基炭疽菌素二酮、放线菌素、白喉毒素、假单胞菌外毒素(PE)A、PE40、相思豆毒素、相思豆毒素A链、蒴莲根毒素A链、α-八叠球菌、白树毒素、迈托毒素(mitogellin)、局限曲菌素(retstrictocin)、酚霉素、依诺霉素、麻疯树毒蛋白(curicin)、巴豆毒素、卡奇霉素、肥皂草(Sapaonaria officinalis)抑制剂、糖皮质激素、或其组合。
在另一优选例中,所述偶联部分为可检测标记物。
在另一优选例中,所述偶联物选自:荧光或发光标记物、放射性标记物、MRI(磁共振成像)或CT(电子计算机X射线断层扫描技术)造影剂、或能够产生可检测产物的酶、放射性核素、生物毒素、细胞因子(如IL-2等)、抗体、抗体Fc片段、抗体scFv片段、金纳米颗粒/纳米棒、病毒颗粒、脂质体、纳米磁粒、前药激活酶(例如,DT-心肌黄酶(DTD)或联苯基水解酶-样蛋白质(BPHL))、化疗剂(例如,顺铂)或任何形式的纳米颗粒等。
在另一优选例中,所述免疫偶联物含有:多价(如二价)的如本发明第一方面所述的抗Her2纳米抗体的VHH链、如本发明第二方面所述的抗Her2纳米抗体。
在另一优选例中,所述多价是指,在所述免疫偶联物的氨基酸序列中包含多个重复的如本发明第一方面所述的抗Her2纳米抗体的VHH链、本发明第二方面所述的抗Her2纳米抗体。
在另一优选例中,所述的免疫偶联物用于癌症的诊断或预后,尤其是用于表达Her2的肿瘤(即Her2阳性的肿瘤)。
在另一优选例中,所述的检测为体内检测或体外检测。
在另一优选例中,所述免疫偶联物用于诊断和/或治疗表达Her2蛋白的肿瘤。
本发明第八方面,提供了本发明第二方面所述的抗Her2纳米抗体、或本发明第七方 面所述的免疫偶联物的用途,用于制备(a)用于检测Her2分子的试剂;(b)用于***的药物。
在另一优选例中,所述的免疫偶联物的偶联部分为诊断用同位素。
在另一优选例中,所述的试剂为选自下组的一种或多种试剂:同位素示踪剂、造影剂、流式检测试剂、细胞免疫荧光检测试剂、纳米磁粒和显像剂。
在另一优选例中,所述检测Her2分子的试剂为(体内)检测Her2分子的造影剂。
在另一优选例中,所述的检测为体内检测或体外检测。
在另一优选例中,所述的检测包括流式检测、细胞免疫荧光检测。
本发明第九方面,提供了一种药物组合物,含有:
(i)本发明第一方面所述的抗Her2纳米抗体的VHH链、或如本发明第二方面所述的抗Her2纳米抗体、或本发明第七方面所述的免疫偶联物;以及
(ii)药学上可接受的载体。
在另一优选例中,所述的免疫偶联物的偶联部分为药物、毒素、和/或治疗用同位素。
在另一优选例中,所述的药物组合物中还含有***的其他药物,如细胞毒性药物。
在另一优选例中,所述的药物组合物用于治疗表达Her2蛋白(即Her2阳性)的肿瘤。
在另一优选例中,所述的药物组合物为注射剂型。
在另一优选例中,所述的药物组合物用于制备***的药物,所述的肿瘤选自下组:胃癌、肝癌、白血病、肾脏肿瘤、肺癌、小肠癌、骨癌、***癌、结直肠癌、乳腺癌、大肠癌、***癌、***、淋巴癌、肾上腺肿瘤、或***。
本发明第十方面,提供了本发明第二方面所述抗Her2纳米抗体、或本发明第七方面所述的免疫偶联物的一种或多种的用途:
(i)用于检测人Her2分子;
(ii)用于流式检测;
(iii)用于细胞免疫荧光检测;
(iv)用于***;
(v)用于肿瘤诊断。
在另一优选例中,所述肿瘤为表达Her2蛋白(即Her2阳性)的肿瘤。
在另一优选例中,所述用途为非诊断的和非治疗的。
在本发明的第十一方面,还提供了一种抗体,所述抗体具有:如本发明第一方面所述的重链可变区VHH。
在另一优选例中,所述的抗体为特异性抗Her2蛋白的抗体。
在另一优选例中,所述抗体为纳米抗体。
本发明第十二方面,提供了一种重组蛋白,所述的重组蛋白具有:
(i)如本发明第一方面所述的重链可变区VHH的序列或如本发明第二方面所述的纳米抗体的序列;以及
(ii)任选的协助表达和/或纯化的标签序列。
在另一优选例中,所述的标签序列包括6His标签和HA标签在另一优选例中,所述的重组蛋白特异性结合于Her2蛋白。
本发明第十三方面,提供了如本发明第一方面所述的VHH链、如本发明第二方面所述的纳米抗体、或本发明第七方面所述的免疫偶联物的用途,它们被用于制备药剂、试剂、检测板或试剂盒;
其中,所述试剂、检测板或试剂盒用于:检测样品中Her2蛋白;
其中,所述药剂用于治疗或预防表达Her2蛋白(即Her2阳性)的肿瘤。
在另一优选例中,所述肿瘤包括:黑色素瘤、胃癌、淋巴瘤、肝癌、白血病、肾脏肿瘤、肺癌、小肠癌、骨癌、***癌、结直肠癌、乳腺癌、大肠癌、***癌、或肾上腺肿瘤。
本发明第十四方面,提供了一种检测样品中Her2蛋白的方法,所述方法包括步骤:
(1)将样品与本发明第二方面所述的纳米抗体接触;
(2)检测是否形成抗原-抗体复合物,其中形成复合物就表示样品中存在Her2蛋白。
本发明第十五方面,提供了一种治疗疾病的方法,所述方法包括,给需要的对象施用本发明第二方面所述的纳米抗体或本发明第七方面所述的免疫偶联物。
在另一优选例中,所述的对象包括哺乳动物,如人。
本发明第十六方面,提供了一种Her2蛋白检测试剂,所述的检测试剂包含本发明第七方面所述的免疫偶联物和检测学上可接受的载体。
在另一优选例中,所述的免疫偶联物的偶联部分为诊断用同位素。
在另一优选例中,所述的检测学上可接受的载体为无毒的、惰性的水性载体介质。
在另一优选例中,所述的检测试剂为选自下组的一种或多种试剂:同位素示踪剂、造影剂、流式检测试剂、细胞免疫荧光检测试剂、纳米磁粒和显像剂。
在另一优选例中,所述的检测试剂为造影剂,并且,所述的造影剂还包含用于造影的其他制剂。
在另一优选例中,所述的造影剂是用于MRI(磁共振成像)或CT(电子计算机X射线断层扫描技术)的造影剂。
在另一优选例中,所述的显像剂同时螯合了两种或多种信号,如Ga-68和Gd,同时用于PET/CT和MRI;或Tc-99m和荧光剂,同时用于SPECT/CT和荧光检测。
在另一优选例中,所述的检测试剂用于体内检测。
在另一优选例中,所述的检测试剂的剂型为液态或粉状(如水剂,针剂,冻干粉,片剂,含服剂,吸雾剂)。
本发明的第十七方面,提供了一种检测Her2分子的试剂盒,所述试剂盒含有本发明第七方面所述的免疫偶联物和说明书。
在另一优选例中,所述的说明书记载,所述的试剂盒用于非侵入性地检测待测对象的 Her2表达。
在另一优选例中,所述的试剂盒用于表达Her2蛋白(即Her2阳性)的肿瘤的检测。
在本发明的十八方面,提供了一种本发明第七方面所述的免疫偶联物的用途,用于制备体内检测Her2分子的造影剂。
在另一优选例中,所述检测用于癌症的诊断或预后。
在本发明的第十九方面,提供了一种CAR-T细胞,所述CAR-T细胞表达嵌合抗原受体CAR,所述CAR的抗原结合结构域具有如本发明第一方面所述的VHH链、或本发明第二方面所述的纳米抗体。
在本发明的第二十方面,提供了一种制剂,所述制剂含有本发明第十九方面所述的CAR-T细胞,以及药学上可接受的载体、稀释剂或赋形剂。
在另一优选例中,所述制剂为液态制剂。
在另一优选例中,所述制剂的剂型包括注射剂。
在另一优选例中,所述制剂中所述CAR-T细胞的浓度为1×10 3-1×10 8个细胞/ml,较佳地1×10 4-1×10 7个细胞/ml。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1是抗原蛋白纯化SDS-PAGE图。图中三条电泳泳道从左至右依次为核酸分子标准,纯化的人Her2(ECD)-Fc蛋白,经TEV酶切后的人Her2(ECD)蛋白,以上蛋白均由HEK293F细胞表达。
图2是文库构建及其质量检测图。图A为第一次PCR扩增产物,割胶回收大小约为700bp的目的条带;图B为第二次PCR扩增产物,获得大小约为400bp的VHH基因片段;图C为文库的库容检测图,构建好的文库被梯度稀释后涂板,取1/5梯度稀释10 3倍、10 4倍、10 5倍、10 6倍的克隆数目,计算单克隆数确定文库大小为2x10 9CFU;图D为文库***率检测图,随机挑取文库的24个单克隆进行PCR鉴定,从左到右凝胶孔的DNA条带分别是:第一道为DNA分子标记,其余孔道为检测***片段的PCR产物,PCR产物带约为500bp,经检测该文库的VHH***率为100%。
图3显示了Her2纳米抗体筛选富集过程。文库经第一轮淘选未出现倍富集,第二轮淘选出现3.75倍富集,第三轮淘选出现110倍富集。
图4显示了Her2纳米抗体的纯化结果。纳米抗体经过镍柱离子亲和层析一步制备纯化,纯度可达到95%以上。
图5显示了Her2纳米抗体与不同种属Her2的亲和力。纳米抗体在不同梯度浓度下分别与人源及鼠源抗原蛋白Her2进行反应,结果显示纳米抗体只与人源Her2结合。
图6显示了I-125标记Her2纳米抗体在HER-2高表达荷瘤鼠SPECT-CT成像结果。多个纳米抗体均能够有效于Her2高表达肿瘤有效积聚,同时非结合抗体能够快速透过肾脏和膀胱从血液清除。
图7显示了99m-Tc标记Her2纳米抗体在HER-2高表达荷瘤鼠SPECT-CT成像结果。纳米抗体均能够特异性地于Her2高表达肿瘤有效积聚,非结合抗体能够快速透过肾脏和膀胱从血液清除,同时并不与曲妥珠单抗或帕妥珠单抗竞争。
具体实施方式
本发明人通过广泛而深入的研究,经过大量的筛选,成功获得一类抗Her2纳米抗体,实验结果表明,本发明获得的Her2纳米抗体能有效与Her2结合。
具体地,本发明利用人源的Her2抗原蛋白免疫骆驼,获得高质量的免疫纳米抗体基因文库。然后将Her2蛋白分子偶联在酶标板上,展示Her2蛋白的正确空间结构,以此形式的抗原利用噬菌体展示技术筛选免疫纳米抗体基因库(骆驼重链抗体噬菌体展示基因库),从而获得了Her2特异性的纳米抗体基因。再将此基因转至大肠杆菌中,从而获得了能在大肠杆菌中高效表达的、且特异性高的纳米抗体株。
本发明还首次意外地发现一种特别适合检测Her2分子的免疫偶联物,所述的免疫偶联物包含特定的抗Her2纳米抗体的VHH链和放射性核素,可以用于待测对象Her2表达情况的非侵入性检测。本发明的免疫偶联物具有小尺寸和高度特异性,适合同时靶向原发及转移肿瘤的全身检测,准确度高,辐射剂量小。
此外,本发明还提供了可有效治疗Her2阳性肿瘤的免疫偶联物。
如本文所用,术语“本发明纳米抗体”、“本发明的抗Her2纳米抗体”、“本发明Her2纳米抗体”可互换使用,均指特异性识别和结合于Her2(包括人Her2)的纳米抗体。特别优选的是VHH链的氨基酸序列如SEQ ID NO.:1-40所示的纳米抗体。
如本文所用,术语“抗体”或“免疫球蛋白”是有相同结构特征的约150000道尔顿的异四聚糖蛋白,其由两个相同的轻链(L)和两个相同的重链(H)组成。每条轻链通过一个共价二硫键与重链相连,而不同免疫球蛋白同种型的重链间的二硫键数目不同。每条重链和轻链也有规则间隔的链内二硫键。每条重链的一端有可变区(VH),其后是多个恒定区。每条轻链的一端有可变区(VL),另一端有恒定区;轻链的恒定区与重链的第一个恒定区相对,轻链的可变区与重链的可变区相对。特殊的氨基酸残基在轻链和重链的可变区之间形成界面。
如本文所用,术语“单域抗体(VHH)”、“纳米抗体”(nanobody)具有相同的含义,指克隆抗体重链的可变区,构建仅由一个重链可变区组成的单域抗体(VHH),它是具有完整功能的最小的抗原结合片段。通常先获得天然缺失轻链和重链恒定区1(CH1)的抗体后,再 克隆抗体重链的可变区,构建仅由一个重链可变区组成的单域抗体(VHH)。
如本文所用,术语“可变”表示抗体中可变区的某些部分在序列上有所不同,它形成了各种特定抗体对其特定抗原的结合和特异性。然而,可变性并不均匀地分布在整个抗体可变区中。它集中于轻链和重链可变区中称为互补决定区(CDR)或超变区中的三个片段中。可变区中较保守的部分称为构架区(FR)。天然重链和轻链的可变区中各自包含四个FR区,它们大致上呈β-折叠构型,由形成连接环的三个CDR相连,在某些情况下可形成部分β折叠结构。每条链中的CDR通过FR区紧密地靠在一起并与另一链的CDR一起形成了抗体的抗原结合部位(参见Kabat等,NIH Publ.No.91-3242,卷I,647-669页(1991))。恒定区不直接参与抗体与抗原的结合,但是它们表现出不同的效应功能,例如参与抗体的依赖于抗体的细胞毒性。
如本领域技术人员所知,免疫偶联物及融合表达产物包括:药物、毒素、细胞因子(cytokine)、放射性核素、酶和其他诊断或治疗分子与本发明的抗体或其片段结合而形成的偶联物。本发明还包括与所述的抗Her2蛋白抗体或其片段结合的细胞表面标记物或抗原。
如本文所用,术语“重链可变区”与“V H”可互换使用。
如本文所用,术语“可变区”与“互补决定区(complementarity determining region,CDR)”可互换使用。
在本发明的一个优选的实施方式中,所述抗体的重链可变区包括三个互补决定区CDR1、CDR2、和CDR3。
在本发明的一个优选的实施方式中,所述抗体的重链包括上述重链可变区和重链恒定区。
在本发明中,术语“本发明抗体”、“本发明蛋白”、或“本发明多肽”可互换使用,都指特异性结合Her2蛋白的多肽,例如具有重链可变区的蛋白或多肽。它们可含有或不含起始甲硫氨酸。
本发明还提供了具有本发明抗体的其他蛋白质或融合表达产物。具体地,本发明包括具有含可变区的重链的任何蛋白质或蛋白质偶联物及融合表达产物(即免疫偶联物及融合表达产物),只要该可变区与本发明抗体的重链可变区相同或至少90%同源性,较佳地至少95%同源性。
一般,抗体的抗原结合特性可由位于重链可变区的3个特定区域来描述,称为可变区域(CDR),将该段间隔成4个框架区域(FR),4个FR的氨基酸序列相对比较保守,不直接参与结合反应。这些CDR形成环状结构,通过其间的FR形成的β折叠在空间结构上相互靠近,重链上的CDR和相应轻链上的CDR构成了抗体的抗原结合位点。可以通过比较同类型的抗体的氨基酸序列来确定是哪些氨基酸构成了FR或CDR区域。
本发明抗体的重链的可变区特别令人感兴趣,因为它们中至少部分涉及结合抗原。因此,本发明包括那些具有带CDR的抗体重链可变区的分子,只要其CDR与此处鉴定的 CDR具有90%以上(较佳地95%以上,最佳地98%以上)的同源性。
本发明不仅包括完整的抗体,还包括具有免疫活性的抗体的片段或抗体与其他序列形成的融合蛋白。因此,本发明还包括所述抗体的片段、衍生物和类似物。
如本文所用,术语“片段”、“衍生物”和“类似物”是指基本上保持本发明抗体相同的生物学功能或活性的多肽。本发明的多肽片段、衍生物或类似物可以是(i)有一个或多个保守或非保守性氨基酸残基(优选保守性氨基酸残基)被取代的多肽,而这样的取代的氨基酸残基可以是也可以不是由遗传密码编码的,或(ii)在一个或多个氨基酸残基中具有取代基团的多肽,或(iii)成熟多肽与另一个化合物(比如延长多肽半衰期的化合物,例如聚乙二醇)融合所形成的多肽,或(iv)附加的氨基酸序列融合到此多肽序列而形成的多肽(如前导序列或分泌序列或用来纯化此多肽的序列或蛋白原序列,或与6His标签形成的融合蛋白)。根据本文的教导,这些片段、衍生物和类似物属于本领域熟练技术人员公知的范围。
本发明抗体指具有Her2蛋白结合活性的、包括上述CDR区的多肽。该术语还包括具有与本发明抗体相同功能的、包含上述CDR区的多肽的变异形式。这些变异形式包括(但并不限于):一个或多个(通常为1-50个,较佳地1-30个,更佳地1-20个,最佳地1-10个)氨基酸的缺失、***和/或取代,以及在C末端和/或N末端添加一个或数个(通常为20个以内,较佳地为10个以内,更佳地为5个以内)氨基酸。例如,在本领域中,用性能相近或相似的氨基酸进行取代时,通常不会改变蛋白质的功能。又比如,在C末端和/或N末端添加一个或数个氨基酸通常也不会改变蛋白质的功能。该术语还包括本发明抗体的活性片段和活性衍生物。
该多肽的变异形式包括:同源序列、保守性变异体、等位变异体、天然突变体、诱导突变体、在高或低的严紧度条件下能与本发明抗体的编码DNA杂交的DNA所编码的蛋白、以及利用抗本发明抗体的抗血清获得的多肽或蛋白。
本发明还提供了其他多肽,如包含纳米抗体或其片段的融合蛋白。除了几乎全长的多肽外,本发明还包括了本发明纳米抗体的片段。通常,该片段具有本发明抗体的至少约50个连续氨基酸,较佳地至少约50个连续氨基酸,更佳地至少约80个连续氨基酸,最佳地至少约100个连续氨基酸。
在本发明中,“本发明抗体的保守性变异体”指与本发明抗体的氨基酸序列相比,有至多10个,较佳地至多8个,更佳地至多5个,最佳地至多3个氨基酸被性质相似或相近的氨基酸所替换而形成多肽。这些保守性变异多肽最好根据表1进行氨基酸替换而产生。
表1
最初的残基 代表性的取代 优选的取代
Ala(A) Val;Leu;Ile Val
Arg(R) Lys;Gln;Asn Lys
Asn(N) Gln;His;Lys;Arg Gln
Asp(D) Glu Glu
Cys(C) Ser Ser
Gln(Q) Asn Asn
Glu(E) Asp Asp
Gly(G) Pro;Ala Ala
His(H) Asn;Gln;Lys;Arg Arg
Ile(I) Leu;Val;Met;Ala;Phe Leu
Leu(L) Ile;Val;Met;Ala;Phe Ile
Lys(K) Arg;Gln;Asn Arg
Met(M) Leu;Phe;Ile Leu
Phe(F) Leu;Val;Ile;Ala;Tyr Leu
Pro(P) Ala Ala
Ser(S) Thr Thr
Thr(T) Ser Ser
Trp(W) Tyr;Phe Tyr
Tyr(Y) Trp;Phe;Thr;Ser Phe
Val(V) Ile;Leu;Met;Phe;Ala Leu
本发明还提供了编码上述抗体或其片段或其融合蛋白的多核苷酸分子。本发明的多核苷酸可以是DNA形式或RNA形式。DNA形式包括cDNA、基因组DNA或人工合成的DNA。DNA可以是单链的或是双链的。DNA可以是编码链或非编码链。
编码本发明的成熟多肽的多核苷酸包括:只编码成熟多肽的编码序列;成熟多肽的编码序列和各种附加编码序列;成熟多肽的编码序列(和任选的附加编码序列)以及非编码序列。
术语“编码多肽的多核苷酸”可以是包括编码此多肽的多核苷酸,也可以是还包括附加编码和/或非编码序列的多核苷酸。
本发明还涉及与上述的序列杂交且两个序列之间具有至少50%,较佳地至少70%,更佳地至少80%相同性的多核苷酸。本发明特别涉及在严格条件下与本发明所述多核苷酸可杂交的多核苷酸。在本发明中,“严格条件”是指:(1)在较低离子强度和较高温度下的杂交和洗脱,如0.2×SSC,0.1%SDS,60℃;或(2)杂交时加有变性剂,如50%(v/v)甲酰胺,0.1%小牛血清/0.1%Ficoll,42℃等;或(3)仅在两条序列之间的相同性至少在90%以上,更好是95%以上时才发生杂交。并且,可杂交的多核苷酸编码的多肽与成熟多肽有相同的生物学功能和活性。
本发明的抗体的核苷酸全长序列或其片段通常可以用PCR扩增法、重组法或人工合成的方法获得。一种可行的方法是用人工合成的方法来合成有关序列,尤其是片段长度较短时。通常,通过先合成多个小片段,然后再进行连接可获得序列很长的片段。此外,还可将重链的编码序列和表达标签(如6His)融合在一起,形成融合蛋白。
一旦获得了有关的序列,就可以用重组法来大批量地获得有关序列。这通常是将其克隆入载体,再转入细胞,然后通过常规方法从增殖后的宿主细胞中分离得到有关序列。本发明所涉及的生物分子(核酸、蛋白等)包括以分离的形式存在的生物分子。
目前,已经可以完全通过化学合成来得到编码本发明蛋白(或其片段,或其衍生物)的DNA序列。然后可将该DNA序列引入本领域中已知的各种现有的DNA分子(或如载体)和细胞中。此外,还可通过化学合成将突变引入本发明蛋白序列中。
本发明还涉及包含上述的适当DNA序列以及适当启动子或者控制序列的载体。这些载体可以用于转化适当的宿主细胞,以使其能够表达蛋白质。
宿主细胞可以是原核细胞,如细菌细胞;或是低等真核细胞,如酵母细胞;或是高等真核细胞,如哺乳动物细胞。代表性例子有:大肠杆菌,链霉菌属;鼠伤寒沙门氏菌的细菌细胞;真菌细胞如酵母;果蝇S2或Sf9的昆虫细胞;CHO、COS7、293细胞的动物细胞等。
用重组DNA转化宿主细胞可用本领域技术人员熟知的常规技术进行。当宿主为原核生物如大肠杆菌时,能吸收DNA的感受态细胞可在指数生长期后收获,用CaCl 2法处理,所用的步骤在本领域众所周知。另一种方法是使用MgCl 2。如果需要,转化也可用电穿孔的方法进行。当宿主是真核生物,可选用如下的DNA转染方法:磷酸钙共沉淀法,常规机械方法如显微注射、电穿孔,脂质体包装等。
获得的转化子可以用常规方法培养,表达本发明的基因所编码的多肽。根据所用的宿主细胞,培养中所用的培养基可选自各种常规培养基。在适于宿主细胞生长的条件下进行培养。当宿主细胞生长到适当的细胞密度后,用合适的方法(如温度转换或化学诱导)诱导选择的启动子,将细胞再培养一段时间。
在上面的方法中的重组多肽可在细胞内、或在细胞膜上表达、或分泌到细胞外。如果需要,可利用其物理的、化学的和其它特性通过各种分离方法分离和纯化重组的蛋白。这些方法是本领域技术人员所熟知的。这些方法的例子包括但并不限于:常规的复性处理、用蛋白沉淀剂处理(盐析方法)、离心、渗透破菌、超处理、超离心、分子筛层析(凝胶过滤)、吸附层析、离子交换层析、高效液相层析(HPLC)和其它各种液相层析技术及这些方法的结合。
本发明的抗体可以单独使用,也可与可检测标记物(为诊断目的)、治疗剂、PK(蛋白激酶)修饰部分或任何以上这些物质的组合结合或偶联。
用于诊断目的可检测标记物包括但不限于:荧光或发光标记物、放射性标记物、MRI(磁共振成像)或CT(电子计算机X射线断层扫描技术)造影剂、或能够产生可检测产物的酶。
可与本发明抗体结合或偶联的治疗剂包括但不限于:1.放射性核素;2.生物毒;3.细胞因子如IL-2等;4.金纳米颗粒/纳米棒;5.病毒颗粒;6.脂质体;7.纳米磁粒;8.药激活酶(例如,DT-心肌黄酶(DTD)或联苯基水解酶-样蛋白质(BPHL));9.疗剂(例如,顺 铂)或任何形式的纳米颗粒等。
免疫偶联物
本发明还提供了一种免疫偶联物,所述的免疫偶联物含有:
(a)如本发明第一方面所述的抗Her2纳米抗体的VHH链、或如本发明第二方面所述的抗Her2纳米抗体;和
(b)选自下组的偶联部分:放射性核素、酶抗体、细胞、或其组合。
在另一优选例中,所述免疫偶联物如本发明第七方面所述。
本发明的免疫偶联物可以用于非侵入性地检测待测对象的Her2表达,具有小尺寸和高度特异性,适合同时靶向原发及转移肿瘤的全身检测,准确度高,辐射剂量小。
细胞毒剂
构成本发明抗体偶联物的偶联部分包括:毒素,如细菌、真菌、植物或动物来源的小分子毒素或酶活性毒素,包括其片段和/或变体。细胞毒剂的例子包括但不限于:耳他汀类(例如,耳他汀E、耳他汀F、MMAE和MMAF)、金霉素、类美坦西醇、篦麻毒素、篦麻毒素A-链、考布他汀、多卡米星、多拉司他汀、阿霉素、柔红霉素、紫杉醇、顺铂、cc1065、溴化乙锭、丝裂霉素、依托泊甙、替诺泊甙(tenoposide)、长春新碱、长春碱、秋水仙素、二羟基炭疽菌素二酮、放线菌素、白喉毒素、假单胞菌外毒素(PE)A、PE40、相思豆毒素、相思豆毒素A链、蒴莲根毒素A链、α-八叠球菌、白树毒素、迈托毒素(mitogellin)、局限曲菌素(retstrictocin)、酚霉素、依诺霉素、麻疯树毒蛋白(curicin)、巴豆毒素、卡奇霉素、肥皂草(Sapaonaria officinalis)抑制剂以及糖皮质激素和其它化学治疗剂,以及放射性同位素,如At211、I131、I125、Y90、Re186、Re188、Sm153、Bi212或213、P32和包括Lu177在内的Lu的放射性同位素。抗体也可与能够将前药转化成其活性形式的抗癌前药活化酶偶联。
优选的小分子药物为具有高细胞毒性的化合物,优选单甲基澳瑞他汀(monomethylauristatin)、加利车霉素、美登素类、或其组合;更佳地选自:单甲基澳瑞他汀-E(MMAE)、单甲基澳瑞他汀-D(MMAD)、单甲基澳瑞他汀-F(MMAF)、或其组合。
药物组合物
本发明还提供了一种组合物。优选地,所述的组合物是药物组合物,它含有上述的抗体或其活性片段或其融合蛋白或免疫偶联物,以及药学上可接受的载体。通常,可将这些物质配制于无毒的、惰性的和药学上可接受的水性载体介质中,其中pH通常约为5-8,较佳地pH约为6-8,尽管pH值可随被配制物质的性质以及待治疗的病症而有所变化。配制好的药物组合物可以通过常规途径进行给药,其中包括(但并不限于):瘤内、腹膜内、静脉内、或局部给药。
本发明的药物组合物可直接用于结合Her2蛋白分子,因而可用于***。此外,还可同时使用其他治疗剂。
本发明的药物组合物含有安全有效量(如0.001-99wt%,较佳地0.01-90wt%,更佳地0.1-80wt%)的本发明上述的纳米抗体(或其偶联物)以及药学上可接受的载体或赋形剂。这类载体包括(但并不限于):盐水、缓冲液、葡萄糖、水、甘油、乙醇、及其组合。药物制剂应与给药方式相匹配。本发明的药物组合物可以被制成针剂形式,例如用生理盐水或含有葡萄糖和其他辅剂的水溶液通过常规方法进行制备。药物组合物如针剂、溶液宜在无菌条件下制造。活性成分的给药量是治疗有效量,例如每天约10纳克/千克体重-约50毫克/千克体重,更佳地50纳克/千克体重-约1毫克/千克体重或10微克/千克体重-约10毫克/千克体重。
此外,本发明的多肽或其偶联物还可与其他治疗剂(如抗肿瘤剂或免疫调节剂)一起使用。
使用药物组合物时,是将安全有效量的免疫偶联物施用于哺乳动物,其中该安全有效量通常至少约10纳克/千克体重,而且在大多数情况下不超过约50毫克/千克体重,较佳地该剂量是约50纳克/千克体重-约1毫克/千克体重。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。
标记的纳米抗体
在本发明的一个优选例中,所述纳米抗体带有可检测标记物。更佳地,所述的标记物选自下组:同位素、胶体金标记物、有色标记物或荧光标记物。
胶体金标记可采用本领域技术人员已知的方法进行。在本发明的一个优选的方案中,抗Her2的纳米抗体用胶体金标记,得到胶体金标记的纳米抗体。
本发明的抗Her2纳米抗体具有很好的特异性,很高的效价。
CAR-T细胞
如本文所用,术语“CAR-T细胞”、“CAR-T”、“本发明CAR-T细胞”均指本发明第十九方面所述的CAR-T细胞。
如本文所用,嵌合免疫抗原受体(Chimeric antigen receptor,CAR)包括细胞外结构域、任选的铰链区、跨膜结构域、和细胞内结构域。胞外结构域包括任选的信号肽和靶-特异性结合元件(也称为抗原结合结构域)。细胞内结构域包括共刺激分子和ζ链部分。共刺激信号传导区指包括共刺激分子的细胞内结构域的一部分。共刺激分子为淋巴细胞对抗原的有效应答所需要的细胞表面分子,而不是抗原受体或它们的配体。
如本文所用,“抗原结合结构域”“单链抗体片段”均指具有抗原结合活性的Fab片段,Fab’片段,F(ab’) 2片段,或单一Fv片段。Fv抗体含有抗体重链可变区、轻链可变区,但没有恒定区,并具有全部抗原结合位点的最小抗体片段。一般的,Fv抗体还包含 VH和VL结构域之间的多肽接头,且能够形成抗原结合所需的结构。抗原结合结构域通常是scFv(single-chain variable fragment)。单链抗体优选是由一条核苷酸链编码的一条氨基酸链序列。作为本发明的优选方式,所述scFv包含本发明第一方面所述的VHH链、或本发明第二方面所述的纳米抗体。
对于绞链区和跨膜区(跨膜结构域),CAR可被设计以包括融合至CAR的胞外结构域的跨膜结构域。在一个实施方式中,使用天然与CAR中的结构域之一相关联的跨膜结构域。在一些例子中,可选择跨膜结构域,或通过氨基酸置换进行修饰,以避免将这样的结构域结合至相同或不同的表面膜蛋白的跨膜结构域,从而最小化与受体复合物的其他成员的相互作用。
在CAR的胞外结构域和跨膜结构域之间,或在CAR的胞浆结构域和跨膜结构域之间,可并入接头。如本文所用的,术语“接头”通常指起到将跨膜结构域连接至多肽链的胞外结构域或胞浆结构域作用的任何寡肽或多肽。接头可包括0-300个氨基酸,优选地2至100个氨基酸和最优选地3至50个氨基酸。
CAR在T细胞中表达时,胞外段可识别一个特异的抗原,随后通过胞内结构域转导该信号,引起细胞的活化增殖、细胞溶解毒性和分泌细胞因子如IL-2和IFN-γ等,影响肿瘤细胞,导致肿瘤细胞不生长、被促使死亡或以其他方式被影响,并导致患者的肿瘤负荷缩小或消除。抗原结合结构域优选与来自共刺激分子和ζ链中的一个或多个的细胞内结构域融合。
检测方法
本发明还涉及检测Her2蛋白的方法。该方法步骤大致如下:获得细胞和/或组织样本;将样本溶解在介质中;检测在所述溶解的样本中Her2蛋白的水平。
在本发明的检测方法中,所使用的样本没有特别限制,代表性的例子是存在于细胞保存液中的含细胞的样本。
试剂盒
本发明还提供了一种含有本发明的抗体(或其片段)或检测板的试剂盒,在本发明的一个优选例中,所述的试剂盒还包括容器、使用说明书、缓冲剂等。
本发明还提供了用于检测Her2水平的检测试剂盒,该试剂盒包括识别Her2蛋白的抗体,用于溶解样本的裂解介质,检测所需的通用试剂和缓冲液,如各种缓冲液、检测标记、检测底物等。该检测试剂盒可以是体外诊断装置。
本发明还提供了一种含有本发明的免疫偶联物的试剂盒,在本发明的一个优选例中,所述的试剂盒还包括容器、使用说明书、同位素示踪剂以及选自下组的一种或多种试剂:造影剂、流式检测试剂、细胞免疫荧光检测试剂、纳米磁粒和显像剂。
优选的,本发明的试剂盒是体内诊断试剂盒,用于非侵入性地检测待测对象的Her2 表达。
应用
如上所述,本发明的纳米抗体有广泛生物应用价值和临床应用价值,其应用涉及到与Her2相关的疾病的诊断和治疗、基础医学研究、生物学研究等多个领域。一个优选的应用是用于针对Her2的临床诊断和靶向治疗。
本发明的主要优点包括:
(a)本发明纳米抗体高特异性针对人的具有正确空间结构的Her2蛋白。
(b)本发明纳米抗体的亲和力强。
(c)本发明纳米抗体的生产简便。
(d)本发明纳米抗体能夠特异性结合人源Her2,在Her2高表达肿瘤模型有效积聚,并不与曲妥珠单抗或帕妥珠单抗进行竞争。本发明Her2纳米抗体非常适合应用于Her2靶向癌症诊断及疗效评估,以及新一代Her2的靶向体内放疗。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数是重量百分比和重量份数。
实施例1:人Her2蛋白的表达纯化
(1)将人Her2的核苷酸序列合成在pCDNA3.1(-)载体上,然后将其胞外段序列亚克隆至pFUSE-IgG1载体上,其中在hHer2(ECD)的C端引入一个TEV酶切位点,便于制备无Fc标签的hHer2(ECD)蛋白;
(2)用Omega质粒大提试剂盒提取构建的pFUSE-IgG1-hHer2(ECD)质粒;
(3)培养HEK293F细胞至OD为2.0×10 6个/mL;
(4)将质粒与转染试剂PEI 1:5混合均匀后静置10min,然后加入到HEK293F细胞中,37℃,6%CO 2摇床培养箱中培养5-6天;
(5)收集细胞上清,与Protein A珠子在室温下结合1h;
(6)用磷酸盐缓冲液pH 7.0洗涤珠子后,再用0.1M pH 3.0Glycine洗脱蛋白;
(7)将洗脱的蛋白超滤至PBS中,测定产量后取样进行SDS-PAGE检测(检测结果如图1所示),该抗原纯度达到95%以上,能够用于后续的免疫反应。
(8)随后,利用TEV酶将该蛋白进行酶切,获得没有标签的抗原蛋白Her2(ECD)用于后续抗体筛选。
实施例2:Her2纳米抗体文库的构建
(1)将1mg hHer2(ECD)-Fc抗原与弗氏佐剂等体积混合,免疫一只新疆双峰驼,每周一次,共免疫3次,刺激B细胞表达抗原特异性的纳米抗体;
(2)3次免疫结束后,提取100mL骆驼外周血淋巴细胞并提取总RNA;
(3)合成cDNA并利用套式PCR扩增VHH(图2A为第一次PCR扩增产物,割胶回收700bp大小的VHH-hinge-CH2-CH3片段,图2B为第二次PCR扩增产物,为400bp的VHH片段);
(4)利用限制性内切酶Pst I及Not I酶切20μg pMECS噬菌体展示载体(Biovector供应)及10μg VHH并连接两个片段;
(5)将连接产物转化至电转感受态细胞TG1中,构建Her2纳米抗体文库并测定库容。结果如图2C所示,构建好的文库被梯度稀释后涂板,取1/5梯度稀释10 3倍、10 4倍、10 5倍、10 6倍的克隆数目,计算单克隆数,确定库容大小为2×10 9CFU。
(6)与此同时,随机挑取24颗克隆进行菌落PCR检测,图2D显示菌落PCR结果,结果表明所建文库的***率为100%。
实施例3:Her2纳米抗体的筛选及鉴定
抗体筛选:
(1)将溶解在100mM NaHCO 3、pH 8.2中的10μg hHer2(ECD)-Fc抗原(10μg Fc in NaHCO 3作为对照)偶联在NUNC酶标板上,4℃放置过夜;
(2)第二天加入100μL 0.1%BSA,室温封闭2h;
(3)2h后,加入100μL噬菌体(2×10 11CFU免疫骆驼纳米抗体噬菌展示基因库),室温作用1h;
(4)用0.05%PBS+Tween-20洗5遍,以洗掉非特异的噬菌体;
(5)用100mM三乙醇胺将与Her2特异性结合的噬菌体解离下,并感染处于对数期生长的大肠杆菌TG1细胞,37℃培养1h,产生并纯化噬菌体用于下一轮的筛选,相同筛选过程重复3轮,其富集结果如图3所示:经三轮bio-panning过程后最终出现110倍富集。
噬菌体的酶联免疫方法(ELISA)筛选特异性单个阳性克隆:
(1)从上述2-3轮筛选后含有噬菌体的细胞培养皿中,挑选600个单个菌落并接种于含有100μg/mL的氨苄青霉素的TB培养基(1L TB培养基中含有2.3g KH 2PO 4,12.52g K 2HPO 4,12g蛋白胨,24g酵母提取物,4mL甘油)中,生长至对数期后,加终浓度1mM的IPTG,28℃培养过夜。
(2)利用渗透法获得粗提抗体,并将抗体转移到经抗原包被的ELISA板中,在室温下放置1h。
(3)用PBST洗去未结合的抗体,加入鼠抗HA抗体,(购自北京康为世纪生物科技有限公司),在室温下放置1h。
(4)用PBST洗去未结合的抗体,加入山羊抗小鼠碱性磷酸酶标记抗体,在室温下放置1h。
(5)用PBST洗去未结合的抗体,加入碱性磷酸酶显色液,于ELISA仪上,在405nm波长,读取吸收值。
(6)当样品孔OD值大于对照孔OD值3倍以上时(Ratio+/->3),判为阳性克隆孔。PE-ELISA结果共出现486颗阳性克隆,其比值(Ratio:+/-)在3-30之间,随后,将所有阳性克隆转至LA培养基中培,提取质粒并进行测序。由于测序结果较多,且测序结果多为重复序列,因此只展示出最终获得的40株纳米抗体相应的ELISA结果,如表2。
表2
抗体编号 1 2 3 4 5 6 7 8
A405+ 1.7152 1.5341 1.3097 1.9694 1.1044 2.143 2.2361 2.5615
A405- 0.0769 0.0794 0.1009 0.0929 0.0871 0.0914 0.0816 0.0915
Ratio(+/-) 22.30 19.32 12.98 21.20 12.68 23.45 27.40 27.99
抗体编号 9 10 11 12 13 14 15 16
A405+ 2.1524 1.6302 1.8819 2.1761 2.2359 1.9576 2.2728 1.7853
A405- 0.0886 0.0728 0.0739 0.0851 0.0891 0.0804 0.087 0.0761
Ratio(+/-) 24.29 22.39 25.47 25.57 25.09 24.35 26.12 23.46
抗体编号 17 18 19 20 21 22 23 24
A405+ 1.8811 1.5904 1.7713 1.4735 2.0862 1.7769 1.7703 2.6757
A405- 0.0791 0.0829 0.0906 0.1439 0.1029 0.0833 0.1034 0.1127
Ratio(+/-) 23.78 19.18 19.55 10.24 20.27 21.33 17.12 23.74
抗体编号 25 26 27 28 29 30 31 32
A405+ 1.9731 1.8971 1.6555 1.857 1.4811 2.3129 1.6545 2.3609
A405- 0.0879 0.09 0.0808 0.0798 0.079 0.0966 0.0774 0.0934
Ratio(+/-) 22.45 21.08 20.49 23.27 18.75 23.94 21.38 25.28
抗体编号 33 34 35 36 37 38 39 40
A405+ 2.0527 2.2628 1.99 2.2236 2.095 2.1419 1.8579 1.5695
A405- 0.0965 0.1129 0.0947 0.1029 0.1067 0.0981 0.094 0.0824
Ratio(+/-) 21.27 20.04 21.01 21.61 19.63 21.83 17.76 19.05
上述40株抗体VHH链的核苷酸序列分别如SEQ ID NO.:1-40所示。表3中,编号为n的VHH(n=1-40的正整数),其氨基酸序列为SEQ ID NO.:n,相应的编码序列为SEQ ID NO.:40+n。
表3
Figure PCTCN2018091953-appb-000001
40株纳米抗体的序列如下,40株纳米抗体的三个CDR区分别用下划线标出。
Figure PCTCN2018091953-appb-000002
Figure PCTCN2018091953-appb-000003
Figure PCTCN2018091953-appb-000004
Figure PCTCN2018091953-appb-000005
Figure PCTCN2018091953-appb-000006
Figure PCTCN2018091953-appb-000007
Figure PCTCN2018091953-appb-000008
实施例4:纳米抗体在宿主菌大肠杆菌中表达、纯化:
(1)对于实施例3测序分析所获得不同克隆株(随机筛选出7个纳米抗体),将各相应质粒电转化到大肠杆菌WK6中,并将其涂布在LA+glucose即含有氨苄青霉素和葡萄糖的培养平板上,37℃培养过夜;
(2)挑选单个菌落接种在5mL含有氨苄青霉素的LB培养液中,37℃摇床培养过夜;
(3)接种1mL的过夜菌种至330mL TB培养液中,37℃摇床培养,培养到OD值达到0.6-1时,加入IPTG,28℃摇床培养过夜;
(4)离心,收菌;
(5)利用渗透法,获得抗体粗提液;
(6)经镍柱离子亲和层析制备纯化的纳米抗体。
纯化结果见图4,抗Her2纳米抗体经过该纯化过程,其纯度可达到95%以上。
实施例5:酶联免疫法(ELISA)鉴定纳米抗体与不同种属Her2的亲和力
(1)包被人和鼠种属的抗原蛋白HER2:加入人和鼠种属的抗原蛋白Her2,4℃过夜。
(2)第二天用PBST洗涤3次,加入1%BSA室温下封闭2小时。
(3)将纯化的纳米抗体进行梯度稀释,与包被的Her2抗原,在室温下放置1小时。
(4)用PBST洗去未结合的抗体,加入鼠抗HA抗体,在室温下放置1小时。
(5)用PBST洗去未结合的抗体,加入山羊抗小鼠碱性磷酸酶标记抗体,在室温下放置1小时。
(6)用PBST洗去未结合的抗体,加入碱性磷酸酶显色液,于ELISA仪上,在405nm波长,读取吸收值,根据吸收值判断纳米抗体的特异性。
检测结果如图5所示,本发明的纳米抗体只与人源Her2结合。
实施例6:流式细胞术检测纳米抗体与细胞的结合
(1)细胞类型:BT474及MDA-MB-231。用PBS将细胞洗涤2次。
(2)用PBS稀释纳米抗体,浓度稀释为0.1ug/uL。
(3)将洗涤好的细胞分装至96孔板中,每个样品的细胞数为3x10 5个,然后每孔加入稀释的纳米抗体,混匀,置于4℃,20min。
(4)用PBS洗涤细胞2次,100uL PBS重悬细胞,然后加入鼠抗HA Alexa Fluor488标记抗体,每个样品1uL,混匀,置于4℃,20min。
(5)用PBS洗涤细胞2次,再用300uL PBS重悬细胞至流式管中,避光至于冰上,上机检测。
结果显示,在Her2高表达(BT474)的细胞株,本发明纳米抗体的阳性率均为>99%,在Her2低表达(MDA-MB-231)的细胞株,纳米抗体的阳性率为6-14%,两者相差至少约6倍,这进一步提示本发明纳米抗体具有非常优异针对Her2的特异性。部分纳米抗体的结果见表4。
表4
抗体编号 BT474 MDA-MB-231
9 99.8% 9.5%
10 99.7% 10.6%
13 99.9% 14.2%
17 99.9% 12.3%
22 99.7% 5.8%
23 99.9% 11.5%
26 99.9% 16.7%
实施例7:Biacore 3K亲和力及曲妥珠单抗及帕妥珠单抗竞争测定
(1)固定:利用羧基氨基反应将固定相抗原蛋白Her2固定在CM-5传感芯片表面;
(2)结合:将纳米抗体用HBS缓冲液稀释成不同浓度,观察纳米抗体在有曲妥珠单抗或帕妥珠单抗或单独与抗原结合过程;
(3)芯片再生:进行下一个抗体测定时用10mM Glycine冲洗进行再生;
结果表明,本发明纳米抗体均与Her2的亲和力达到纳摩尔浓度级别以上,并且不与曲妥珠单抗或帕妥珠单抗竞争结合Her2。部分纳米抗体的数据示于表5。
表5
抗体编号 K on(1/Ms) K off(1/s) KD(M) 曲妥珠单抗竞争 帕妥珠单抗竞争
9 1.11E+06 3.97E-03 3.60E-09 - -
10 7.06E+05 4.57E-03 6.47E-09 - -
13 1.27E+06 1.50E-03 1.18E-09 - -
17 6.31E+05 4.21E-03 6.68E-09 - -
22 9.57E+05 1.90E-03 1.98E-09 - -
23 6.82E+05 2.55E-03 3.73E-09 - -
26 8.57E+05 1.12E-03 1.31E-09 - -
实施例8纳米抗体I-125标记,分离纯化及SPECT显像扫描
(1)取抗体原液150μL,加入100μL 0.02mol/L pH7.4磷酸盐缓冲溶液和50μL Na125I溶液,混匀,加入20μL 5mg/mL氯胺T溶液,混匀器上室温下反应70s,加入200μL偏重亚硫酸钠溶液(5mg/mL)作用5分钟。
(2)用PD10柱分离,洗脱液为0.02mol/L pH7.4磷酸缓冲溶液,每管收集10滴,用纸层析对I-125标记纳米抗体进行放射纯度鉴定。
(3)NOD/SCID小鼠在细胞接种前一天于右侧背部皮下植入***药片,在右侧乳腺脂肪垫接种1×10 7个Her2高表达(BT474)细胞,待肿瘤长至150-200mm 3用于正式实验研究。
(4)通过异氟烷麻醉荷瘤鼠,尾静脉注射I-125标记纳米抗体(~50ug,5MBq)
(5)给药后30min进行扫描,采集方式为静态15min SPECT、中分辨率全身CT。
结果显示,本发明的多个纳米抗体均能够有效于Her2高表达肿瘤模型有效积聚,应用于癌症诊断及治疗。同时非结合抗体能够快速透过肾脏和膀胱从血液清除,减低身体放射性剂量。
部分纳米抗体在荷瘤鼠30min SPECT扫描图片及身体内分布数据示于图6和表6。
表6
Figure PCTCN2018091953-appb-000009
实施例9纳米抗体Tc-99m标记,分离纯化及SPECT显像扫描
(1)分别加入Na2CO3 5.5mg、酒石酸钾钠15.2mg和NaBH4 20.5mg至10mL无菌瓶,通入20min CO,再加入1mL(35mCi)Na[99mTcO4],密封反应瓶,于80℃下加热30min。
(2)取抗体原液溶液50μl,加入500μl 99mTc(CO)3(H2O)3反应液(6.56mCi),于45-50℃下反应90min。
(3)用PD10柱分离,洗脱液为0.02mol/L pH7.4磷酸缓冲溶液,用薄层色谱法对99mTc标记纳米抗体进行放射纯度鉴定。
(4)NOD/SCID小鼠在细胞接种前一天于右侧背部皮下植入***药片,在右侧乳腺脂肪垫接种1×10 7个Her2高表达(BT474)细胞,待肿瘤长至150-200mm 3用于正式实验研究。
(5)通过异氟烷麻醉荷瘤鼠,尾静脉注射Tc-99m标记纳米抗体(~10ug,5MBq)或注射时同时混合20x未标记纳米抗体,或72小时前进行20x曲妥珠单抗或20x帕妥珠单抗尾静脉注射预处理。
(6)给药后30min进行扫描,采集方式为静态15min SPECT、中分辨率全身CT。
结果显示,本发明的纳米抗体均能够特异性地于Her2高表达肿瘤模型有效积聚,并不与曲妥珠单抗或帕妥珠单抗竞争。可应用于Her2靶向的癌症诊断及疗效评估,同时开发新一代Her2靶向治疗。
部分纳米抗体在荷瘤鼠30min SPECT扫描图片及身体内分布数据示于图7和表7。
表7
Figure PCTCN2018091953-appb-000010
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (17)

  1. 一种抗Her2纳米抗体的VHH链,其特征在于,所述VHH链的氨基酸序列如SEQ ID NO.:1-40中任一所示。
  2. 一种抗Her2纳米抗体,其特征在于,它是针对Her2表位的纳米抗体,并且具有如SEQ ID NO.:1-40中任一所示的氨基酸序列的VHH链。
  3. 一种多核苷酸,其特征在于,所述多核苷酸编码选自下组的蛋白质:权利要求1所述的抗Her2纳米抗体的VHH链,或权利要求2所述的抗Her2纳米抗体。
  4. 如权利要求3所述的多核苷酸,其特征在于,具有如SEQ ID NO.:41-80中任一所示的核苷酸序列。
  5. 一种表达载体,其特征在于,所述表达载体含有权利要求3所述的多核苷酸。
  6. 一种宿主细胞,其特征在于,所述宿主细胞含有权利要求5所述的表达载体,或其基因组中整合有权利要求3所述的多核苷酸。
  7. 一种产生抗Her2纳米抗体的方法,其特征在于,包括步骤:
    (a)在适合产生纳米抗体的条件下,培养权利要求6所述的宿主细胞,从而获得含所述抗Her2纳米抗体的培养物;
    (b)从所述培养物中分离或回收所述的抗Her2纳米抗体。
  8. 一种免疫偶联物,其特征在于,该免疫偶联物含有:
    (a)如权利要求1所述的抗Her2纳米抗体的VHH链、或如权利要求2所述的抗Her2纳米抗体;
    (b)选自下组的偶联部分:可检测标记物、药物、毒素、细胞因子、放射性核素、酶、金纳米颗粒/纳米棒、纳米磁粒、病毒外壳蛋白或VLP、或其组合。
  9. 如权利要求8所述的免疫偶联物,其特征在于,所述的放射性核素包括:
    (i)诊断用同位素,所述的诊断用同位素选自下组:Tc-99m、Ga-68、F-18、I-123、I-125、I-131、In-111、Ga-67、Cu-64、Zr-89、C-11、Lu-177、Re-188、或其组合;和/或
    (ii)治疗用同位素,所述的治疗用同位素选自下组:Lu-177、Y-90、Ac-225、As-211、Bi-212、Bi-213、Cs-137、Cr-51、Co-60、Dy-165、Er-169、Fm-255、Au-198、Ho-166、I-125、I-131、Ir-192、Fe-59、Pb-212、Mo-99、Pd-103、P-32、K-42、Re-186、Re-188、Sm-153、Ra223、Ru-106、Na24、Sr89、Tb-149、Th-227、Xe-133 Yb-169、Yb-177、或其组合。
  10. 如权利要求8所述的免疫偶联物,其特征在于,所述的药物为细胞毒性药物,较佳地,所述的细胞毒性药物选自下组:
    抗微管蛋白药物、DNA小沟结合试剂、DNA复制抑制剂、烷化试剂、抗生素、叶酸拮抗物、抗代谢药物、化疗增敏剂、拓扑异构酶抑制剂、长春花生物碱、或其组合。
  11. 如权利要求8所述的免疫偶联物,其特征在于,所述的毒素选自下组:
    耳他汀类(例如,耳他汀E、耳他汀F、MMAE和MMAF)、金霉素、类美坦西醇、篦麻毒素、篦麻毒素A-链、考布他汀、多卡米星、多拉司他汀、阿霉素、柔红霉素、紫杉醇、顺铂、cc1065、溴化乙锭、丝裂霉素、依托泊甙、替诺泊甙(tenoposide)、长春新碱、长春碱、秋水仙素、二羟基炭疽菌素二酮、放线菌素、白喉毒素、假单胞菌外毒素(PE)A、PE40、相思豆毒素、相思豆毒素A链、蒴莲根毒素A链、α-八叠球菌、白树毒素、迈托毒素(mitogellin)、局限曲菌素(retstrictocin)、酚霉素、依诺霉素、麻疯树毒蛋白(curicin)、巴豆毒素、卡奇霉素、肥皂草(Sapaonaria officinalis)抑制剂、糖皮质激素、或其组合。
  12. 一种Her2蛋白或Her2癌症的检测试剂,其特征在于,所述的检测试剂包含权利要求8所述的免疫偶联物和检测学上可接受的载体。
  13. 如权利要求12所述的检测试剂,其特征在于,所述的检测试剂为选自下组的一种或多种试剂:同位素示踪剂、造影剂、流式检测试剂、细胞免疫荧光检测试剂、纳米磁粒和显像剂。
  14. 权利要求2所述的抗Her2纳米抗体的用途,其特征在于,用于制备(a)用于检测Her2分子的试剂;(b)用于***的药物。
  15. 一种药物组合物,其特征在于,含有:
    (i)权利要求1所述的抗Her2纳米抗体的VHH链、或如权利要求2所述的抗Her2纳米抗体、或如权利要求8所述的免疫偶联物;以及
    (ii)药学上可接受的载体。
  16. 一种CAR-T细胞,其特征在于,所述CAR-T细胞表达嵌合抗原受体CAR,所述CAR的抗原结合结构域具有如权利要求1所述的抗Her2纳米抗体的VHH链、或如权利要求2所述的抗Her2纳米抗体。
  17. 一种制剂,其特征在于,所述制剂含有权利要求16所述的CAR-T细胞,以及药学上可接受的载体、稀释剂或赋形剂。
PCT/CN2018/091953 2017-06-20 2018-06-20 抗Her2纳米抗体及其编码序列和用途 WO2018233624A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US16/624,403 US11517632B2 (en) 2017-06-20 2018-06-20 Anti-Her2 single chain antibody and coding sequence and use thereof
JP2020520703A JP7097636B2 (ja) 2017-06-20 2018-06-20 抗Her2ナノボディ及びそのコード序列と用途
EP18819877.4A EP3643726A4 (en) 2017-06-20 2018-06-20 ANTI-HER2 NANOCBODIES, CODING SEQUENCE AND ASSOCIATED USE
US18/047,327 US20230263916A1 (en) 2017-06-20 2022-10-18 Anti-her2 nanobody and coding sequence and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710471319 2017-06-20
CN201710471319.6 2017-06-20

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/624,403 A-371-Of-International US11517632B2 (en) 2017-06-20 2018-06-20 Anti-Her2 single chain antibody and coding sequence and use thereof
US18/047,327 Continuation US20230263916A1 (en) 2017-06-20 2022-10-18 Anti-her2 nanobody and coding sequence and use thereof

Publications (1)

Publication Number Publication Date
WO2018233624A1 true WO2018233624A1 (zh) 2018-12-27

Family

ID=64735901

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/091953 WO2018233624A1 (zh) 2017-06-20 2018-06-20 抗Her2纳米抗体及其编码序列和用途

Country Status (5)

Country Link
US (2) US11517632B2 (zh)
EP (1) EP3643726A4 (zh)
JP (1) JP7097636B2 (zh)
CN (1) CN109096401B (zh)
WO (1) WO2018233624A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021163490A1 (en) * 2020-02-14 2021-08-19 University Of Washington Targeted depletion of bacteria from mixed populations through programmable cell-cell adhesion
WO2022031884A3 (en) * 2020-08-05 2022-03-24 Synthekine, Inc. Il2rg binding molecules and methods of use

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109627319B (zh) * 2019-01-11 2022-06-21 北京协同创新研究院 一种抗her-2的重链抗体及其应用
CN109793750B (zh) * 2019-03-27 2021-07-02 四川大学 一种抗肿瘤纳米药物
US20240076403A1 (en) * 2020-12-18 2024-03-07 Simcere Zaiming Pharmaceutical Co., Ltd. Her2 antibody and application thereof
CN114649094B (zh) * 2022-03-30 2022-11-15 广东省人民医院 一种基于核磁共振的乳腺癌多参数临床决策辅助装置
WO2023194988A1 (en) * 2022-04-04 2023-10-12 The National Institute for Biotechnology in the Negev Ltd. Binding molecules specific to human epidermal growth factor receptor 2
WO2023230488A1 (en) * 2022-05-23 2023-11-30 Cereius, Inc. Her2-binding agents and uses thereof
CN117362440A (zh) * 2022-06-30 2024-01-09 复旦大学 Her2纳米抗体及偶联物的制备方法和用途

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009068625A2 (en) * 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against her2 and polypeptides comprising the same for the treatment of cancers and/or tumors
CN106866823A (zh) * 2017-03-08 2017-06-20 康众(北京)生物科技有限公司 一种抗Her2的纳米抗体
CN107236046A (zh) * 2017-05-15 2017-10-10 江苏吴中医药集团有限公司苏州中凯生物制药厂 一种重组人内皮抑素融合蛋白及其制备方法和应用

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101143902B (zh) * 2007-05-18 2010-06-02 中国医学科学院医药生物技术研究所 抗HER2单链抗体-力达霉素强化融合蛋白HER2(Fv-LDM)
US20090258005A1 (en) * 2007-05-29 2009-10-15 Trubion Pharmaceuticals Inc. Therapeutic compositions and methods
WO2009055074A2 (en) * 2007-10-25 2009-04-30 Wyeth Erbb2 binding proteins and use thereof
CA2720368C (en) * 2008-04-02 2017-08-22 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
EP2575880B1 (en) 2010-05-27 2019-01-16 Genmab A/S Monoclonal antibodies against her2 epitope
AU2012245116A1 (en) * 2011-04-20 2013-11-07 Genmab A/S Bispecific antibodies against HER2 and CD3
EP3014277A4 (en) * 2013-06-27 2017-04-19 GE Healthcare Limited Use of her2 binders
US20180036442A1 (en) * 2014-07-29 2018-02-08 Vrije Universiteit Brussel Radio-labelled antibody fragments for use in the prognosis, diagnosis of cancer as well as for the prediction of cancer therapy response
US11298433B2 (en) * 2015-07-17 2022-04-12 Vrije Universiteit Brussel Radiolabelled antibody fragments for use in treating cancer
WO2017075537A1 (en) 2015-10-30 2017-05-04 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
CN106883297B (zh) * 2015-12-16 2019-12-13 苏州康宁杰瑞生物科技有限公司 基于ch3结构域的异二聚体分子、其制备方法及用途
CN112500480B (zh) * 2020-07-17 2022-04-01 上海洛启生物医药技术有限公司 针对新型冠状病毒的纳米抗体及其应用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009068625A2 (en) * 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against her2 and polypeptides comprising the same for the treatment of cancers and/or tumors
CN106866823A (zh) * 2017-03-08 2017-06-20 康众(北京)生物科技有限公司 一种抗Her2的纳米抗体
CN107236046A (zh) * 2017-05-15 2017-10-10 江苏吴中医药集团有限公司苏州中凯生物制药厂 一种重组人内皮抑素融合蛋白及其制备方法和应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ARVIND CHOPRA: "(99mTc) Epidermal growth factor receptor-specific nanobody", MOLECULAR IMAGING AND CONTRAST AGENT DATABASE (MICAD), 13 May 2008 (2008-05-13), XP055651466, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/books/NBK23690/pdf/Bookshelf_NBK23690.pdf> *
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021163490A1 (en) * 2020-02-14 2021-08-19 University Of Washington Targeted depletion of bacteria from mixed populations through programmable cell-cell adhesion
WO2022031884A3 (en) * 2020-08-05 2022-03-24 Synthekine, Inc. Il2rg binding molecules and methods of use

Also Published As

Publication number Publication date
CN109096401A (zh) 2018-12-28
JP2020525048A (ja) 2020-08-27
CN109096401B (zh) 2021-07-27
EP3643726A1 (en) 2020-04-29
US20230263916A1 (en) 2023-08-24
US20200306392A1 (en) 2020-10-01
EP3643726A4 (en) 2021-09-01
JP7097636B2 (ja) 2022-07-08
US11517632B2 (en) 2022-12-06

Similar Documents

Publication Publication Date Title
CN109096401B (zh) 抗Her2纳米抗体及其编码序列和用途
CN110452297B (zh) 抗vegf单域抗体及其应用
JP7007758B2 (ja) 癌の予後と診断における放射性標識抗pd-l1ナノボディの応用
US20230399395A1 (en) Anti-il5 nanoantibody and use thereof
CN114106190A (zh) 一种抗vegf/pd-l1双特异性抗体及其用途
EP4257605A1 (en) Anti-tslp nanobody and use thereof
US20230002503A1 (en) Nano-antibody targeting caix antigen and application thereof
WO2023020551A1 (zh) 抗ptk7单域抗体及其应用
CN114539415B (zh) 一种抗PD-L1/VEGF/TGF-β多特异性抗体及其用途
WO2024099310A1 (zh) 抗il-13长效纳米抗体序列及其应用
CN116120450A (zh) 靶向人cd38分子的驼科纳米抗体制备和应用
CN117186226A (zh) 抗cd38纳米抗体及其应用
CN117659194A (zh) 靶向egfr的纳米抗体、药物偶联物及其用途
CN118005784A (zh) 抗il-13长效纳米抗体序列及其应用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18819877

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020520703

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018819877

Country of ref document: EP

Effective date: 20200120